You are on page 1of 14

ll

Review
Self-Eating for Muscle Fitness:
Autophagy in the Control of Energy Metabolism
David Sebastián1,2,3,* and Antonio Zorzano1,2,3,*
1Institutefor Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
2Departament de Bioquı́mica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
3Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III,

Madrid, Spain
*Correspondence: david.sebastian@irbbarcelona.org (D.S.), antonio.zorzano@irbbarcelona.org (A.Z.)
https://doi.org/10.1016/j.devcel.2020.06.030

Cellular processes that sense and transmit metabolic changes are crucial for adaptation to external signals.
In this regard, autophagy provides energy upon nutrient deprivation and represents a quality control mech-
anism that eliminates damaged organelles or proteins. Here, we review recent findings on the metabolic path-
ways controlling autophagy in skeletal muscle, a plastic tissue that undergoes major changes in energy de-
mands. We also analyze the implications of autophagy in the regulation of energy metabolism in muscle and
how alterations in this process affect energy homeostasis at the whole-body level and the development of
metabolic diseases and aging.

Introduction in muscle adaptation to exercise. Interestingly, alterations in


The maintenance of cellular homeostasis is essential for tissue muscle autophagy have also been shown to be associated
function and cell survival. Comprising about 40% of whole- with changes in whole-body energy metabolism. Therefore, the
body lean mass, skeletal muscle plays a key role in the regulation aim of this review is to summarize research findings on the role
of energy metabolism. Muscle is a highly plastic tissue, respond- of autophagy in the regulation of energy metabolism in skeletal
ing and adapting to changes in energy demands, such as those muscle and how this process impacts on muscle plasticity and
occurring during nutrient deprivation or physical activity and ex- systemic energy balance.
ercise. Therefore, cellular processes aimed to sense and trans-
mit metabolic changes within skeletal muscle are crucial for Autophagy: Molecular Regulation
cellular homeostasis in this tissue and at the whole-body level. Autophagy is an evolutionarily conserved process of recycling
In this context, autophagy has emerged as a crucial process. and degradation of cellular components, such as cytoplasmic
Autophagy operates under basal conditions and can be induced components, protein aggregates, and damaged organelles.
under appropriate stress conditions. Basal autophagy is respon- These components, also named the cargo, are engulfed within
sible for the removal of damaged organelles and maintenance of a double-membrane vesicle, the so-called autophagosome.
basal energy homeostasis, which is especially important in long- These vesicles are then fused with the lysosome, forming the au-
lived post-mitotic cells, such as skeletal muscle fibers. In tolysosome, where the cargo is finally degraded by the action of
contrast, induced autophagy is characterized by the degradation lysosomal hydrolases. Autophagy can be either non-selective
of intracellular macromolecules and organelles to meet energy (also named bulk) or selective. In the former, an entire region of
requirements in situations of nutrient deficiency or high-energy the cytoplasm is engulfed by the autophagosome and then
demands, such as fasting and exercise or physical activity. Three degraded in the lysosome. In contrast, in selective autophagy,
main forms of autophagy have been described, namely macro, a specific substrate or organelle is preferentially targeted for
micro, and chaperone mediated, which differ in the mechanism degradation based on its interaction with autophagy receptors.
of substrate selection and delivery into the lysosome for degra- Autophagy is a tightly regulated process that involves more
dation. In this regard, macroautophagy (hereafter referred to as than 40 autophagy-related (ATG) proteins in yeast, of which 19
autophagy) involves the formation of a double-membrane proteins are required for autophagosome formation (Kim and
vesicle named autophagosome, which engulfs the cargo to be Lee, 2014). Most of them have mammalian homologs, suggest-
degraded and transports it to the lysosome for its clearance. Mi- ing that they are evolutionary conserved in mammals (Kim and
croautophagy and chaperone-mediated autophagy participate Lee, 2014). Key steps in autophagy include the following: initia-
in the degradation of small portions of cytosol and selected pro- tion, vesicle nucleation or formation of the isolation membrane,
teins by directly delivering them to lysosomes in a chaperone-in- vesicle elongation, fusion with the lysosome, and degradation
dependent or -dependent manner. (Figure 1). Details of the autophagic machinery and its regulation
The importance of autophagy in skeletal muscle function and have been reviewed elsewhere (Galluzzi et al., 2017; Yin et al.,
metabolic homeostasis has been revealed by studies on various 2016), and therefore, only the most relevant aspects are high-
mouse models deficient in this process. These studies have lighted here. Autophagy is triggered by activation of the ULK1
demonstrated a tight interconnection between muscle meta- complex, formed by ULK1, ATG13, FIP200, and ATG101.
bolism and autophagy, as well as an essential role of autophagy Upon induction of autophagy, the serine threonine kinase

268 Developmental Cell 54, July 20, 2020 ª 2020 Elsevier Inc.
ll
Review

1. AUTOPHAGY INITIATION 2. MEMBRANE NUCLEATION AND 3. PHAGOPHORE EXPANSION 4. FUSION WITH THE LYSOSOME 5. DEGRADATION
PHAGOPHORE FORMATIOM

BECLIN1 Late endosome Lysosome


VPS34
ATG14
VPS34
VPS15

Class III PI3K complex ESCRT


complex

AMPK

ULK1 ATG13 Amphisome

FIP2OO ATG101

WIPI 2
ULK1 complex Isolation
mTORC1 membrane Phagophore Autophagosome Autolysosome
ATG9

Delivery of
lipids in ATG2 ATG12 LC3
ATG9- ATG5 conjugation
containing ATG18 ATG16L system Lysosome
vesicles conjugation
system

Figure 1. Molecular Regulation of Autophagy


Upon activation of the ULK1 complex, different complexes are recruited to the isolation membrane. Class III PI3K complex catalyzes the formation of PI3P, which
recruits WIPI2 and ATG2/ATG18 complex. ATG9-containing vesicles are thought to be a source of lipids for isolation membrane formation. Once formed, the
isolation membraneor phagophore maturates by action of ATG12-ATG5-ATG16L and LC3-PE. Autophagosome closure is mediated by ESCRT members. Finally,
autophagosome could either fuse directly with the lysosome to form the autolysosome or fuse first with late endosomes forming the amphisome. Once the
autolysosome is formed, degradation of the cargo occurs.

ULK1 is activated, thereby phosphorylating ATG13 and FIP200 mature autophagosome fuses with the lysosome to form the au-
(Hosokawa et al., 2009; Jung et al., 2009). Once activated, the tolysosome, where the cargo is degraded by the action of
ULK1 complex recruits the Beclin 1-Vps34 complex (class III several lysosomal acid hydrolases. In addition, autophago-
PI3K complex) to the site of autophagosome formation. In addi- somes can merge with late endosomes prior to fusion with
tion, ULK1 directly phosphorylates Beclin 1, which can interact lysosomes to form amphisomes, which deliver cargo in the
with Ambra 1 or ATG14L (Nazio et al., 2013; Russell et al., autophagic vesicle (Nakamura and Yoshimori, 2017). Finally,
2013). Vps34 is a lipid kinase responsible for the formation of cargo-degradation components are recycled for biosynthetic
phosphatidylinositol-3 phosphate (PI3P), which recruits proteins purposes or permanently removed (Figure 1).
such as DFCP1 and WIPI to promote the initiation of double-
membrane vesicle nucleation (Axe et al., 2008; Lu et al., 2011). Metabolic Control of Autophagy in Muscle
The WIPI family of proteins is comprised of four members, with Skeletal muscle is a key tissue in the integration of energy meta-
WIPI1 and WIPI2 being the most studied. WIPI2 appears to be bolism. It shows remarkable plasticity, which allows for rapid
involved in the recruitment of the autophagosome maturation adaptation to changes in energetic demands, in which auto-
complex ATG12-ATG5-ATG16L to the isolation membrane by phagy has been described to play a major role. In this regard,
direct binding to ATG16L, whereas WIPI1 may operate as a skeletal muscle is one of the tissues with either the highest basal
negative regulator of autophagosomes (Polson et al., 2010; Proi- or the highest stimulated autophagic flux, as determined using
kas-Cezanne et al., 2015). Other steps that are important in the GFP-LC3 transgenic mice (Mizushima et al., 2004). Muscle is
formation of isolation membrane involve the Atg2/Atg18 com- composed by different muscle fibers subtypes, which show
plex (Suzuki et al., 2007), which binds to PIP3, and Atg9-contain- distinct metabolic properties and differences in autophagy. Skel-
ing membranes, which are suggested to provide the starting ma- etal muscle fibers are characterized as one type of slow-twitch
terial for the isolation membrane formation (Orsi et al., 2012). fibers (type I) and three types of fast-twitch fibers (type IIa, IIx/
Vesicle elongation and completion of the autophagosome is d, and IIb), of which types I and IIa are oxidative, whereas types
mediated by two ubiquitin-like conjugation systems, namely IIx and IIb are primary glycolytic. Importantly, autophagy has
ATG12-ATG5-ATG16L and ATG8 (LC3)-phosphatidylethanol- been shown to be higher in fast-twitch fibers than in slow-twitch
amine (Mizushima et al., 1998), leading to the formation of the fibers (Mizushima et al., 2004). The main conditions associated
lipid-conjugated form of LC3 (LC3-phosphatidylethanolamine, with metabolic adaptations in muscle that are shown to regulate
or LC3-II). LC3-II then localizes to the autophagosomal mem- autophagy are detailed below.
branes and participates in the formation and elongation of the Regulation of Autophagy by Nutrient Availability
autophagosome. LC3 also plays a role in selective autophagy At a cellular level, autophagy is a key process in energy produc-
by interacting with various autophagy receptors, such as p62, tion and is tightly regulated by nutrient availability via the modu-
NBR1, optineurin, NDP52, BNIP3, and FUNDC1. Finally, closure lation of a number of signaling pathways. Skeletal muscle is a
of the autophagosome has been recently shown to be regulated high-energy-demanding tissue that primarily relies on glucose
by members of the endosomal sorting complex required for and fatty acids for ATP generation. Glucose and fatty acid oxida-
transport (ESCRT) (Takahashi et al., 2018). Once formed, the tion in skeletal muscle are subjected to negative regulation by

Developmental Cell 54, July 20, 2020 269


ll
Review
AA
Glucose (Amino acids)

Leucine Arginine Methionine


GCN2
Sestrins
CASTOR 1 SAMTOR
Glucose Sestrins eIF2α
CASTOR 1
GATOR 2
AMP/ATP ATF4
GATOR 1
SAMTOR
AMPK
ATF4 Autophagy genes
FOXO3
mTORC1

MUSCLE CELL TFEB FOXO3 Autophagy genes

ULK1 ATG13 Lysosomal and


TFEB
autophagy genes
FIP2OO ATG101

ULK1 complex
Nucleus
BECLIN1 AMBRA1
VPS34
ATG14
VPS15

Class III PI3K complex


Activation of
Autophagy

Figure 2. Metabolic Regulation of Autophagy by Amino Acids and Glucose


Amino acids are sensed by mTORC1 through a mechanism depending on sestrins, CASTOR, and SAMTOR, although this has not been validated specifically in
skeletal muscle. In conditions of amino acid availability, activation of mTORC1 inhibits autophagy via phosphorylation and inactivation of ULK1 complex and then
inhibition of ATG14-Beclin 1-Vps34. mTORC1 also inhibits the ULK1 activator Ambra1 and TFEB. In conditions of amino acid deficiency, mTORC1 is inactivated
and then autophagy induced. Amino acid deficiency also activates GCN2, which phosphorylates and activates eIF2a-inducing ATF4 and the transcription of
autophagy genes. Glucose starvation induces autophagic flux through activation of AMPK, which phosphorylates and inactivates mTORC1, activating the ULK1
complex. In addition, AMPK can activate ULK1 and Beclin 1 independently of mTORC1 and phosphorylate and activate FoxO3, leading to the induction of
autophagy gene expression.

each other, and glucose utilization inhibits lipid oxidation and gard, during a meal, increased levels of amino acids activate pro-
vice versa by the so-called Randle or glucose-fatty acid cycle tein synthesis and the inhibition of autophagy, ultimately leading
(Hue and Taegtmeyer, 2009; Randle et al., 1963). In contrast to to an increase in muscle mass (Zhang et al., 2014). The mecha-
glucose and fatty acids, amino acids are not a significant energy nism by which amino acids modulate autophagy involve the acti-
source for skeletal muscle, where their oxidation accounts for vation of mTORC1 (Figure 2). Essential amino acids, such as
3%–6% of ATP generation during prolonged exercise in humans leucine, are sensed by mTORC1 by a mechanism that is not fully
and is negligible during resting conditions (Gibala, 2001). How- understood. For a deep understanding of the biology of mTOR
ever, variations in the relative amount of these substrates have and how it senses amino acid availability, we refer the readers
an impact on the regulation of autophagy within the muscle by to excellent reviews (Kim and Guan, 2019; Wolfson and Sabatini,
acting on several energy-sensing pathways (Figures 2 and 3). 2017). Briefly, leucine is sensed by sestrins and arginine by
Amino Acids. The amino acid pool in skeletal muscle is main- CASTOR1, which, upon amino acid binding, dissociate from
tained by the coordination of protein synthesis and protein and release their suppressive effects on GATOR2 (a positive
degradation. Only six amino acids are metabolized in resting regulator of mTORC1), thereby activating mTORC1. Methionine
skeletal muscle: aspartate, asparagine, glutamate, and the is sensed in the form of SAM (S-adenosylmethionine) by SAM-
branched-chain amino acids (BCAAs) valine, leucine, and isoleu- TOR, which, upon SAM binding, dissociates from and inhibits
cine. Of these, leucine and part of the isoleucine molecule can be GATOR1 (a negative regulator of mTORC1), thus leading to
oxidized in muscle, via their conversion to acetyl-CoA. The car- mTORC1 activation. Whether those mechanisms of amino acid
bon skeleton of BCAAs and aspartate are used for the novo syn- sensing operate in muscle cells remains to be determined.
thesis of tricarboxylic acid (TCA) cycle intermediates and gluta- Canonically, mTORC1 inhibits autophagy through interaction
mine (Wagenmakers, 1998). However, in addition to their role with the ULK1/FIP200/Atg13 complex by phosphorylating and
as energy substrates, amino acids obtained from the circulation inhibiting ULK1 (Kim et al., 2011). In addition, it has been shown
or from protein degradation in muscle play a crucial role in the that mTORC1 also inhibits AMBRA1 and ATG14 (Nazio et al.,
regulation of signaling pathways that modulate muscle growth, 2013; Yuan et al., 2013), two autophagy-stimulatory interactors
metabolism, and function (Torre-Villalvazo et al., 2019). In this re- of BECN1, and the transcription factor EB (TFEB), a crucial

270 Developmental Cell 54, July 20, 2020


ll
Review
FFA
(Free fatty acids)

PKR eIF2α
FFA
JNK

DAG PKC
mTORC1

ULK1 complex

MUSCLE CELL
Autophagosome

Class III PI3K complex

Lysosome

Activation of
Autophagy

Autolysosome

Figure 3. Metabolic Regulation of Autophagy by Lipids


Contradictory results have been found for the effect of lipids on autophagy depending on the duration of fatty acid exposure or cell context. High levels of fatty
acids could activate autophagy by inhibiting mTORC1 or by activating different pathways depending on PKR-eIF2a, JNK, and PKC, leading to an increase in
autophagosome formation, although the molecular mechanisms and whether they are preserved in skeletal muscle are not known. In addition, high levels of lipids
have been shown to impair autophagosome-lysosome fusion.

transcription factor involved in the expression of genes involved Glucose. Glucose availability regulates autophagy by control-
in the autophagy-lysosome pathway (Settembre et al., 2013). ling the energetic charge of the cell. Thus, in conditions of
However, studies using either pharmacological or genetic ap- glucose deprivation, cellular levels of ATP decrease, leading to
proaches have shown that mTORC1 is responsible only for the an increase in the AMP/ATP ratio and activation of AMPK (Hardie
regulation of 10% of autophagic flux in muscle (Sandri, 2010b). et al., 2012), which in turn promotes autophagy by various mech-
In this respect, accurate biochemical analyses have determined anisms (Figure 2). AMPK inhibits mTORC1 by directly phosphor-
that rapamycin-mediated mTOR inhibition increases protein ylating Raptor (a component of mTORC1 complex) (Gwinn et al.,
breakdown by only 10% in myotubes—a much smaller response 2008) or by phosphorylating and activating TSC2 (an upstream
than the 50% increase in protein breakdown after Akt inhibition inhibitor of mTORC1) (Inoki et al., 2003), finally leading to the acti-
(Zhao et al., 2007). In addition, in vivo inhibition of mTOR by ra- vation of autophagy. Independently of mTORC1 inhibition,
pamycin or by RNAi approaches is not sufficient to induce mus- AMPK also triggers autophagy by phosphorylation and activa-
cle atrophy or autophagosome formation (Mammucari et al., tion of ULK1 and BECN1 (Egan et al., 2011a; Kim et al., 2013a;
2007; Zhao et al., 2007). Moreover, knockout (KO) mice for Kim et al., 2011). Although the exact serine residues in ULK1
mTOR or Raptor (an mTORC1 component) show muscular dys- phosphorylated by AMPK remain unclear, Ser317, -467, -555,
trophy rather than muscle atrophy (Risson et al., 2009). There- -574, -637, and -777 may have a role (Bach et al., 2011). Howev-
fore, these data suggest that mTORC1 is not the major energy er, it has been reported that phosphorylation of Ser555 is not suf-
sensor involved in the regulation of autophagy in skeletal muscle. ficient to increase autophagosome content in rodent muscle or in
Importantly, amino acids can regulate autophagy indepen- human myotubes (Fritzen et al., 2016). Activation of AMPK by
dently of mTORC1 signaling (Figure 2). In this context, the glucose deprivation is also responsible for the phosphorylation
eIF2a kinase GCN2, a sensor of amino acid availability, induces and activation of the transcription factor FoxO3 (Greer et al.,
the expression of a set of genes involved in the formation, elon- 2007; Sanchez et al., 2012), which is responsible for inducing
gation, and function of the autophagosome by activating the eI- key autophagic genes such as LC3, Bnip3, Vps34, Atg12, and
F2a-ATF4 pathway (B’chir et al., 2013). In addition, the lack of GABARAP (Mammucari et al., 2007) (Figure 2). Contrary to
some amino acids, such as leucine, glutamate, and glutamine, glucose starvation, high glucose availability inhibits autophagy.
leads to a reduction of acetyl-CoA pools, which has been shown In this regard, glucose has been described to inhibit autophagy
to induce autophagy (Mariño et al., 2014). However, whether via activation of mTORC1 independently of AMPK, via the forma-
these mechanisms are preserved in skeletal muscle is still tion of a lysosomal v-ATPase-Ragulator-Rag GTPase complex
not known. (Efeyan et al., 2013). In all, glucose regulates autophagy by

Developmental Cell 54, July 20, 2020 271


ll
Review

distinct signaling pathways, which might depend on the cellular myotubes of severely obese subjects, which is a model of
context or the type or duration of changes in glucose concen- chronic lipid overload (Bollinger et al., 2015). Therefore, lipid
tration. oversupply might have a biphasic role on autophagy in muscle,
During fasting, the combined effect of low amino acid and first increasing in order to protect from insulin resistance. How-
glucose availability leads to the activation of autophagy. In this ever, if lipid excess persists, this adaptive role is lost and auto-
regard, the effects of fasting-mimetic dietary interventions phagy decreases, thereby exacerbating insulin resistance.
known to expand health span, such as caloric restriction, inter- Regulation of Autophagy by Insulin
mittent fasting, and time-restricted feeding, have been Under conditions of high nutrient availability, increased insulin
described to mediate their benefits, in part, through regulation levels are a major regulator of autophagy in muscle (Figure 4).
of autophagy. Long-term caloric restriction (3–15 years) in hu- A recent multi-dimensional transcriptomic analysis has shown
mans is associated with an increase in autophagy mediators that, under euglycemic clamp conditions, physiological levels
such as Beclin 1 and LC3 in skeletal muscle (Yang et al., of insulin are associated with the downregulation of an auto-
2016). In addition, a time-restricted feeding model (isocaloric phagy transcriptional network in muscle (Batista et al., 2019).
twice a day feeding) increases autophagic flux in skeletal muscle In addition, several KO mouse models have shed light on the
and other tissues, leading to an increase in muscle mass, energy involvement of insulin in the regulation of autophagy and the
expenditure, and protection from obesity and diabetes (Marti- possible molecular players involved. Hence, muscle-specific
nez-Lopez et al., 2017). Low-carbohydrate diets, such ketogenic ablation of insulin receptor (IR) in mice leads to a moderate
diets, are known to mimic calorie restriction and extend longevity reduction in muscle mass, and this is further impaired in double
and health span (Roberts et al., 2017) and have been shown to KO mice for IR and IGF-1 receptor (IGF1R), thereby reflecting the
reverse high-fat-diet-induced alterations in muscle by activating importance of insulin signaling in muscle mass maintenance
autophagy through inhibition of Akt and mTOR signaling (Li (O’Neill et al., 2016). Importantly, these effects are associated
et al., 2018). with an increase in autophagy, which is dependent on FoxO1,
Lipids. The role of lipids in the regulation of autophagy is less FoxO3, and FoxO4, as illustrated by a reversal in increased auto-
characterized than that of amino acids or glucose. Several phagy upon combined deletion of these transcription factors in
studies have shown that an increase in lipids is associated with double KO mice (O’Neill et al., 2016). In addition to IR and
a reduction in autophagy (Figure 3). In this regard, an in vitro IGF1R, a main actor of the insulin signaling pathway is the protein
fusion assay of autophagosomes and lysosomes obtained kinase Akt, which controls approximately 50% of autophagy in
from mouse liver has shown that changes in membrane lipid skeletal muscle (Zhao et al., 2007). Akt phosphorylates FoxO3,
composition reduces autophagosome-lysosome fusion by up preventing its nuclear translocation and thus the transcription
to 70% (Koga et al., 2010). Moreover, the long-term incubation of FoxO3-dependent autophagic genes (Mammucari et al.,
of b cells with a high concentration of palmitate impairs auto- 2007). In addition, Akt phosphorylates and inhibits TSC2, a nega-
phagy and lysosomal acidification and activity (Las et al., tive regulator of mTORC1 (Manning and Toker, 2017), thus acti-
2011). In contrast, other studies in b cells have shown that 12 h vating mTORC1 and inhibiting autophagy. Therefore, upon acti-
of treatment with palmitic acid triggers autophagy by blocking vation of the insulin signaling pathway, insulin represses
mTORC1 or activating the PKR-JNK pathway (Choi et al., autophagic gene expression, which is mediated mainly by inhibi-
2009; Komiya et al., 2010). Moreover, palmitate has also been tion of FoxO transcription factors.
shown to induce autophagy in human osteosarcoma U2OS cells Regulation of Autophagy by Exercise and Physical
by disrupting the interaction of STAT3 and PKR and leading to Activity
PKR-dependent phosphorylation of eIF-2a (Shen et al., 2012). Exercise training induces multiple adaptations in skeletal mus-
In skeletal muscle cells, long exposure to palmitate may inhibit cle, and these depend on the type of exercise. Resistance exer-
autophagy, as indicated by decreased levels of LC3II and cise is associated with increased muscle mass, while endurance
increased levels of p62 (Choi et al., 2018). However, other exercise is characterized by metabolic adaptations oriented to
studies have shown that increased levels of fatty acids activate increase muscle oxidative capacity. Importantly, in both types
autophagy (Niso-Santano et al., 2015; Tan et al., 2012). In this of adaptation, autophagy seems to play an important role by
respect, in human cancer cells, short-term incubation with maintaining cellular energy homeostasis and regulating organ-
palmitate induces canonical autophagy, whereas oleate triggers elle and protein turnover after exercise (Halling and Pile-
non-canonical autophagy dependent on an intact Golgi appa- gaard, 2017).
ratus (Niso-Santano et al., 2015). In contrast, in MEF (mouse em- The first observation of a role of autophagy in exercise was
bryonic fibroblasts) and HepG2 cells, palmitate, but not oleate, observed in mouse muscle fibers 2 to 7 days after a single stren-
increases autophagic flux by a mechanism involving PKC activa- uous treadmill running bout (Salminen and Vihko, 1984). This was
tion (Shen et al., 2012). A possible explanation for this discrep- followed by a number of studies showing that acute endurance
ancy could be the duration of fatty acid exposure (acute versus exercise activates autophagy in mouse skeletal muscle (Grumati
chronic) and/or the cellular context. However, these mecha- et al., 2011; He et al., 2012; Jamart et al., 2012b; Pagano et al.,
nisms have not been described yet in muscle. In skeletal muscle 2014). In contrast, the role of resistance exercise on autophagy
in vivo, contradictory results have been obtained. For instance, in rodents is less well defined to date. Resistance exercise has
upon high-fat feeding, autophagy has been found to be unal- been associated either with an activation of autophagy with a
tered in rats (Campbell et al., 2015) and either decreased (Choi reduced number of autophagosomes or with no changes in
et al., 2018) or increased in mice (Liu et al., 2015b; Moresi LC3II protein levels during the recovery period after exercise
et al., 2012). In humans, autophagic flux is repressed in primary (Luo et al., 2013; Ogasawara et al., 2016). Studies in humans

272 Developmental Cell 54, July 20, 2020


ll
Review
Insulin
IGF1 Insulin
Receptor (IR)

Akt

TSC2 FOXO3

mTORC1

MUSCLE CELL ULK1 complex FOXO3 Autophagy genes

Class III PI3K complex


Nucleus

Activation of
Autophagy

Figure 4. Regulation of Muscle Autophagy by Insulin


Under high-nutrient conditions, insulin activates Akt, which, by activating mTORC1 and inhibiting FoxO3, inhibits autophagy and autophagic gene expression,
respectively.

have also given contradictory results, suggesting either activa- is less clear because distinct forms of exercise have different ef-
tion or inhibition of autophagy during endurance or resistance fects on Akt activation. Thus, following a single bout of exercise,
exercise. This discrepancy could be due to the lack of robust Akt is dephosphorylated and then inactivated (He et al., 2012).
measurements of autophagic flux, differences in exercise proto- However, during a sprint, endurance and resistance exercise,
cols, and variations in times of muscle sampling (Fritzen et al., Akt is activated, preferentially when exercise is performed in a
2016; Fry et al., 2013; Jamart et al., 2012b; Møller et al., 2015; fed state (Camera et al., 2010; Guerra et al., 2010), which would
Ogborn et al., 2015; Schwalm et al., 2015; Smiles et al., 2015). negatively regulate FoxO3 and inhibit autophagic gene expres-
Nonetheless, it is becoming apparent that endurance exercise sion. Interestingly, increased expression and activity of FoxO3
induces autophagy, probably in an exercise duration-dependent has been observed in human and mouse skeletal muscle
manner, whereas autophagy does not seem to be essential for following acute endurance exercise (Jamart et al., 2012a; Louis
muscle adaptations to resistance exercise. et al., 2007). These data suggest that Akt and AMPK-mediated
The mechanisms by which exercise regulates autophagy in FoxO3 regulation contributes to the transcriptional regulation
muscle involve mTOR, AMPK, Akt, and FoxO3 signaling path- of autophagy in skeletal muscle in response to exercise. In addi-
ways (Figure 5). Hence, inactivation of mTORC1 leads to a tion to FoxO3, other transcription factors participate in the regu-
decrease in ULK1 Ser757 phosphorylation and activation of lation of autophagy during exercise. In this regard, lysosomal
autophagy in mouse skeletal muscle during exercise (Pagano Ca2+ release through mucolipin 1 (MCOLN1) activates the phos-
et al., 2014) and 10 h post exercise (Halling et al., 2016). In addi- phatase calcineurin, which dephosphorylates TFEB, leading to
tion, AMPK has been shown to be necessary for exercise- its nuclear translocation and activation of lysosomal genes in
induced autophagy (Egan et al., 2011b) by phosphorylation of response to exercise (Medina et al., 2015). It has also been pro-
ULK1 in Ser317 and Ser555, which are increased in response posed that PGC1a plays a role in basal and exercise-induced
to a single exercise bout. This association of exercise-induced autophagy. In this regard, muscle PGC1a overexpression is
AMPK and autophagy is also supported by results from some associated with an increase in BNIP3 protein expression and
human studies (Møller et al., 2015; Schwalm et al., 2015), an increased basal autophagic flux (Lira et al., 2013), whereas
although this correlation has not been detected in other work both total and muscle-specific PGC1a KO mice show a reduc-
(Fritzen et al., 2016). AMPK can also activate autophagy by tion in exercise-induced autophagy (Halling et al., 2016; Vain-
phosphorylation and activation of FoxO3, which is induced in ul- shtein et al., 2015). The precise mechanism by which PGC1a
tra-endurance exercise (Jamart et al., 2012b; Zhao et al., 2007) regulates autophagy is unknown, but a direct regulation of auto-
and which activates the expression of autophagic genes in mus- phagy gene expression or an indirect effect by controlling mus-
cle (Mammucari et al., 2007). The role of Akt in muscle autophagy cle oxidative capacity has been proposed.

Developmental Cell 54, July 20, 2020 273


ll
Review

Exercise/Muscle contraction

Endurance/
Acute Resistance
exercise exercise
AMPK

mTORC1 Ca2+ Akt PGC1α

FOXO3

ULK1 complex
Calcineurin PGC1α
Autophagy genes
MUSCLE CELL

FOXO3 Autophagy genes


Class III PI3K complex

TFEB TFEB Lysosomal and


autophagy genes

Activation of
Autophagy Nucleus

Figure 5. Metabolic Regulation of Autophagy by Exercise


Muscle contraction and exercise lead to activation of autophagy by inhibiting mTORC1 and activating AMPK, which can also inhibit mTORC1 and activate UKL1.
Moreover, AMPK activates FoxO3, inducing the expression of autophagy genes. Different forms of exercise have been found to modulate Akt. Hence, acute
exercise blocks Akt, relieving the inactivation of FoxO3 and therefore increasing FoxO3-dependent gene transcription. In contrast, endurance and resistance
exercise activate Akt, inhibiting FoxO3. Increases in calcium following exercise activate the phosphatase calcineurin, which activates TFEB and the expression of
lysosomal and autophagy genes. Finally, PGC1a is activated during exercise and induces the expression of autophagy genes.

Together, these data reflect the complexity of exercise- lysosomal storage diseases (Margeta, 2020). In this context,
induced autophagy in muscle, which appears to involve the inter- Pompe disease, caused by loss-of-function mutations in the
connection of various signaling pathways. In addition, the lysosomal hydrolase acid a-glucosidase (GAA) responsible for
different types and duration of exercise, as well as the distinct glycogen catabolism in the lysosome, and Danon disease,
molecular markers and techniques used for monitoring auto- caused by mutations in LAMP2 gene, are associated with the
phagy, increase the difficulty of data interpretation. accumulation of glycogen granules and with a deficient delivery
of glycogen accumulated inside autophagosomes to lysosomes
Autophagy and Regulation of Skeletal Muscle Energy for its degradation (Nishino et al., 2000). Interestingly, muscle-
Metabolism specific ablation of Vps15 in mouse leads to a severe myopathy
As described in previous sections, autophagy is regulated in associated with defects in autophagosome and lysosome fusion
different physiological and pathological conditions, such as fast- and the accumulation of glycogen. In contrast, the overexpres-
ing, atrophy, and exercise (Figure 6). Importantly, many of the sion of the Vps37-Vps15 complex in muscle cells derived from
pathways leading to autophagy regulation are performed via Danon disease patients results in a partial amelioration of
the control of skeletal muscle metabolism. glycogen overload (Nemazanyy et al., 2013).
In addition to its role in quality control, autophagy serves to Loss of muscle homeostasis due to alterations in autophagy
mobilize cellular energy stores to provide nutrients in times of has been suggested to underlie the development of several my-
need. In this regard, autophagy is involved in glycogen degrada- opathies and muscular dystrophies. Decreased autophagy in
tion via a process called glycophagy, which is crucial for the skeletal muscle-specific ATG7 KO mice was associated with
maintenance of glucose homeostasis in skeletal muscle (Kotou- an accumulation of protein aggregates, abnormal mitochon-
las et al., 2006). In this process, it has been suggested that the dria, and oxidative stress, leading to muscle atrophy, weak-
interaction of STBD1 (starch-binding domain-containing protein ness, and several features of myopathy (Masiero et al., 2009).
1) with GABARAPL1 plays an important role by tethering In addition, autophagy has been shown to be decreased in
glycogen to autophagic membrane structures for lysosomal several muscular dystrophies, such as collagen VI muscular
degradation (Jiang et al., 2010). The importance of autophagy dystrophy (Grumati et al., 2010) and Duchenne muscular dys-
in glycogen breakdown has been demonstrated by a number trophy (De Palma et al., 2012), as well as in a variety of lyso-
of studies showing that alterations in this process underlie the somal storage disorders such as the aforementioned Pompe
basis of several muscular disorders, previously described as and Danon diseases (Margeta, 2020). Patients affected by

274 Developmental Cell 54, July 20, 2020


ll
Review

LOCAL EFFECTS SYSTEMIC EFFECTS

Impaired lysosomal
degradation of glycogen
Myopathies Protection against
(Pompe and Danon disease) diet-induced obesity
Decreased insulin and and glucose intolerance
Activation of FGF21
exercise-stimulated Exercise intolerance
Autophagy levels
glucose uptake
Muscle atrophy
Browning of white
Insulin resistance adipose tissue
Mitochondrial dysfunction and
reduced energy metabolism

Figure 6. Muscle Autophagy and Regulation of Energy Metabolism


Alterations in muscle autophagy are associated with abnormalities in muscle metabolism (local effects) and to changes in whole-body energy metabolism
(systemic effects). Deficient autophagy in muscle has been shown to be associated with the development of myopathies such as Pompe and Danon diseases.
Furthermore, impaired autophagy could lead to mitochondrial dysfunction, decreased insulin-stimulated and exercise-induced glucose uptake, and reduced
energy metabolism in muscle, leading to muscle atrophy, exercise intolerance, and insulin resistance. Importantly, mitochondrial dysfunction induced by reduced
autophagy in muscle leads to increased expression and secretion of FGF21, which has been shown to have beneficial effects at the whole-body level, such as
protection from diet-induced obesity and glucose intolerance.

multisystem disorder Vici syndrome show a pattern of vacuolar muscle atrophy under conditions that favor protein loss in skel-
myopathy with glycogen storage and immature, hypoplastic, etal muscle (Sala et al., 2014). Ablation of the transcription factor
and atrophic muscle fibers (Hedberg-Oldfors et al., 2017). Simi- Runx1 also leads to excessive autophagy and potentiates mus-
larly, Epg5-deficient mice show muscle denervation and myo- cle atrophy in denervated muscles (Wang et al., 2005), suggest-
fiber degeneration (Zhao et al., 2013). Interestingly, EPG5 is ing that Runx1 is required to preserve muscle mass upon dener-
the human homolog of the metazoan-specific autophagy vation. Interestingly, the response to these different stimuli that
gene epg-5, encoding a key autophagy regulator implicated induce muscle atrophy and weakness differs between fiber
in the formation of autolysosomes (Tian et al., 2010) by interact- types. For example, oxidative type I fibers are more resistant
ing with Rab7 and VAMP7/8 proteins (Wang et al., 2016). How- to fasting but more sensitive to inactivity and denervation-
ever, increased autophagy is also associated with some induced atrophy, whereas type II fibers are more vulnerable to
muscular dystrophies, such as MDC1A, a severe form of cancer cachexia, diabetes, and aging (Wang and Pessin,
congenital muscular dystrophy characterized by mutations in 2013). However, the molecular basis for this selectivity and
the gene encoding laminin a2 (Gawlik and Durbeej, 2011). In whether differences in autophagy flux could contribute to that
this regard, an increase in autophagy has been found in the remain unresolved. Regarding autophagy, a skeletal muscle-
dy3k/dy3k mutant mouse model for laminin a2 and muscle bi- specific transgenic mouse model of the tyrosine kinase Fyn,
opsies from two MDC1A patients (Carmignac et al., 2011). whose activity is higher in fast-twitch muscles, shows a marked
Therefore, both reduced and increased autophagy can induce atrophy of glycolytic fibers due to STAT3-dependent reduction in
myopathies, indicating that a fine balance of autophagic flux the Vps34/p150/Beclin 1/ATG14 complex and consequently in
is necessary to preserve muscle homeostasis (Sandri, 2010a). autophagy (Yamada et al., 2012). These data could potentially
Muscle atrophy also occurs in response to denervation or explain the fiber-type-specific muscle wasting observed in
inactivity and in response to fasting and various diseases, this model.
including sepsis, AIDS, renal and cardiac failure, cancer (cancer Autophagy has been shown to regulate glucose utilization in
cachexia), and diabetes. Several studies indicate that activation muscle. Pharmacological inhibition of autophagy in C2C12 mus-
of autophagy is one of the molecular mechanisms underlying cle cells reduces insulin-stimulated glucose uptake (Liu et al.,
muscle atrophy in these conditions (Mammucari et al., 2007; 2015a). In vivo, the role of skeletal muscle autophagy in glucose
Zhao et al., 2007). The decrease in PI3K-Akt signaling associated metabolism has been studied in several mouse models deficient
to these catabolic conditions induces the nuclear translocation for proteins involved in the regulation of autophagy. For instance,
and activity of FoxO transcription factors, such as FoxO3, which BCL2 AAA knockin mutant mice, which have reduced autophagy
activate autophagic gene expression. In this regard, specific due to an inhibition of BCL2-BECN1 dissociation, and ATG6+/
ablation of FoxO transcription factors in skeletal muscle protects mice show reduced GLUT4 translocation to plasma membrane
from fasting- and denervation-induced muscle atrophy (Milan and glucose uptake in response to exercise (He et al., 2012;
et al., 2015). Therefore, an excessive autophagy aggravates Lira et al., 2013). In addition, prolonged iron overload, which re-
muscle loss in muscle-wasting conditions. Muscle-specific over- sults in an autophagy defect in skeletal muscle by altering
expression of DOR/TP53inp2, a positive autophagy regulator mTORC1, has been shown to contribute to impaired insulin-
(Mauvezin et al., 2010), exacerbates muscle wasting in strepto- stimulated glucose uptake and insulin signaling (Jahng et al.,
zotocin-induced diabetes and cancer cachexia in mice (Penna 2019). In contrast, another study has demonstrated that deficient
et al., 2019; Sala et al., 2014). Interestingly, TP53inp2 is naturally autophagy in skeletal muscle by knocking out ATG7 does not
repressed in human insulin resistance and in murine models of have an impact on AMPK activation or glucose homeostasis
diabetes, suggesting that TP53INP2 repression would reduce (Lo Verso et al., 2014). However, this study revealed that

Developmental Cell 54, July 20, 2020 275


ll
Review

autophagy is critical for the preservation of mitochondrial func- by TFEB enhances physical performance and coordinates meta-
tion during damaging muscle contraction. bolic flexibility during exercise (Mansueto et al., 2017). In addi-
The role of autophagy in lipid metabolism has gained consid- tion, decreased autophagy and mitophagy in muscle of Mfn2-
erable interest due to its potential implication in obesity and deficient mice is associated with muscle atrophy and a reduction
metabolic syndrome. In liver, it has been shown that lipid drop- in muscle force and exercise capacity during aging (Sebastián
lets (LDs) can be degraded by autophagy by a process called lip- et al., 2016). Reduced mitophagy in either Parkin or FUNDC1
ophagy, which is activated in response to fasting or sustained KO mice is also linked to reduced exercise performance (Fu
lipid overload (Singh et al., 2009). Several mechanisms have et al., 2018; Peker et al., 2018). Together, these data reveal a
been described to regulate lipophagy, such as Rab7-mediated tight connection between autophagy, the control of muscle
translocation of autophagosomes and lysosomes to the LDs metabolism, exercise-induced muscle adaptations, and condi-
(Schroeder et al., 2015), TFEB-mediated activation of PGC1a tions associated to muscle wasting.
target genes involved in fatty acid catabolism, including lipoph-
agy (Settembre et al., 2013), and FOXO1 activation (Lettieri Bar- Skeletal Muscle Autophagy and the Regulation of
bato et al., 2013). However, in skeletal muscle, lipophagy has Whole-Body Energy Metabolism
been much less studied (Lam et al., 2016). The role of autophagy on muscle metabolism also has marked
Defects in autophagy are also associated with alterations in implications for energy metabolism at the whole-body level, as
mitochondrial function, which could contribute to impaired en- demonstrated by several studies performed in KO mouse
ergy homeostasis in muscle. In this regard, specific ablation of models for various autophagic regulators. In this regard, BCL2
Atg7 in muscle exacerbates mitochondrial dysfunction and AAA knockin mutant mice are more susceptible to high-fat-
oxidative stress during aging (Carnio et al., 2014). Interestingly, diet-induced glucose intolerance, mainly due to an impaired
TFEB, which is involved in autophagy and lysosomal biogenesis, response to the beneficial metabolic effects of exercise (He
has also been shown to regulate mitochondrial mass and func- et al., 2012). Mice lacking AMPK specifically in muscle show
tion in muscle by inducing the expression of genes involved in an impairment in fasting-induced autophagy, which leads to a
mitochondrial biogenesis, fatty acid oxidation, and oxidative downregulation of muscle proteolysis and reduced circulating
phosphorylation (Mansueto et al., 2017). Moreover, modulation levels of alanine, resulting in an impairment in liver gluconeogen-
of autophagy by other proteins that are not constituents of the esis (Bujak et al., 2015). These findings establish an essential
canonical autophagy machinery has also been linked to alter- requirement for skeletal muscle AMPK-mediated autophagy in
ations in mitochondria and energy metabolism in muscle. In preserving blood glucose levels during prolonged fasting. How-
this regard, mice lacking collagen-type VI a (COLA6 KO mice) ever, muscle-specific Atg7 KO mice, which show impaired auto-
show impaired autophagy, which leads to the accumulation of phagy and mitochondrial dysfunction, have a reduced fat mass,
dysfunctional mitochondria and an altered mitochondrial together with enhanced glucose clearance and energy expendi-
network (Grumati et al., 2010, 2011). In addition, ablation of the ture. In addition, these animals are protected from diet-induced
mitochondrial fusion protein Mfn2 inhibits autophagic degrada- obesity and insulin resistance (Kim et al., 2013b). Interestingly,
tion of mitochondria (mitophagy), thereby leading to the accumu- these effects are caused by an ATF4-mediated increase in
lation of damaged mitochondria and to an impairment of mito- FGF21, a cytokine released from Atg7-deficient muscles, which
chondrial function and glucose metabolism in C2C12 increases fatty acid oxidation and the browning of white adipose
myotubes and skeletal muscle in vivo (Sebastián et al., 2012, tissue. Although basal levels of FGF21 expression in muscle are
2016). These results suggest that the regulation of mitochondrial low compared with other tissues, this stress-induced FGF21
turnover by autophagy is crucial for the maintenance of energy secretion constitutes an adaptive endocrine crosstalk between
metabolism in muscle. In this regard, ablation of the mitophagy alterations in muscle autophagy and changes in metabolism in
protein Parkin in muscle leads to an impairment in mitochondrial other tissues (Tezze et al., 2019). This adaptive mechanism has
function (Peker et al., 2018). Moreover, skeletal muscle-specific also been described upon muscle-specific ablation of FUNDC1,
ablation of the mitophagy protein FUNDC1 in mice results in an which impairs mitophagy and mitochondrial function but confers
LC3-mediated mitophagy defect, leading to impaired mitochon- protection from diet-induced obesity, glucose intolerance, and
drial energetics and decreased muscle fat utilization (Fu et al., insulin resistance (Fu et al., 2018). Similar to the ablation of
2018). Interestingly, Urolithin A, a natural compound found in ATG7, FUNDC1 deficiency induces a retrograde response in
pomegranate fruit, has been found to activate mitophagy in mus- muscle involving increased expression of FGF21, thereby pro-
cle and to increase complex-II-dependent mitochondrial respira- moting thermogenic remodeling of adipose tissue (Fu et al.,
tion (Ryu et al., 2016). 2018). These studies reveal a tight connection between the pres-
Importantly, the role of autophagy in regulating metabolism ence of mitochondrial dysfunction in muscle, probably as a
described above has important physiological implications in consequence of impaired autophagy and/or mitophagy, and
skeletal muscle, such as muscle adaptation to exercise, as the triggering of an integrated stress response to promote meta-
well as some of the metabolic benefits associated with it. Hence, bolism in other tissues. In this regard, skeletal muscle-specific
deficient autophagy and glucose uptake in BCL2 AAA knockin ablation of the mitochondrial fusion protein OPA1 leads to severe
mutant mice or ATG6+/ mice is associated with a decrease in mitochondrial dysfunction in skeletal muscle but also to
exercise capacity (He et al., 2012; Lira et al., 2013). Specific abla- improved glucose tolerance and protection from diet-induced
tion of ATG7 in muscle has also been shown to induce muscle obesity, mediated by increased levels of FGF21 (Pereira et al.,
atrophy and reduce muscle power (Kim et al., 2013b; Masiero 2017; Rodrı́guez-Nuevo et al., 2018; Tezze et al., 2017). Some
et al., 2009). Increased mitochondrial mass and function driven of the alterations detected in Opa1-deficient muscles are also

276 Developmental Cell 54, July 20, 2020


ll
Review

driven by activation of a TLR9-driven inflammatory pathway, eases, such as myopathies and muscular dystrophies, cancer
which may be linked to impaired mitophagy (Rodrı́guez-Nuevo cachexia, and diabetes. However, whether those alterations
et al., 2018). Nonetheless, not all muscle alterations in autophagy are directly attributable to changes in autophagy (i.e., those
or mitochondrial function lead to this inflammatory stress observed in Atg KO mouse models) or may be a consequence
response, and it is still not known whether this stress response of more general alterations that also involve autophagy needs
depends on the type of alteration or the intensity of mitochondrial further investigation. Moreover, the precise role of autophagy
dysfunction. In addition, activation of autophagy has been in physiology and in pathology in humans remains inconclusive,
shown to improve muscle metabolism and function (Ryu et al., as most studies have been conducted in mouse models defec-
2016). However, whether this activation leads to amelioration tive for autophagy proteins. Furthermore, research into auto-
of whole-body energy metabolism and whether it is useful for phagy in human tissue samples is technically challenging,
the treatment of metabolic diseases are unresolved questions. owing to the difficulty in measuring autophagic flux. In addition,
Autophagy participates in a stress response by controlling the the study of autophagy in exercise is particularly complex
production of hormones, cytokines, metabolites, and other fac- because different duration and types of exercise may be asso-
tors that cause the crosstalk between tissues or organs in meta- ciated with distinct upstream or downstream autophagic mech-
bolic homeostasis and in adaptive responses. Muscle auto- anisms. Of particular interest is the connection between de-
phagy participates in such a coordination through the fects in autophagy in muscle and the activation of adaptive
operation of non-cell-autonomous and systemic responses. In responses targeting other tissues. Defective muscle meta-
this connection, in Caenorhabditis elegans, excessive auto- bolism due to impaired autophagy could lead to amelioration
phagy in pharyngeal muscle in gbp-2 mutants leads to death of energy metabolism and protection from metabolic disease
when subjected to starvation, by a mechanism that could be re- at the whole-body level, although more studies are needed to
verted by the presence of certain amino acids and their action on understand this connection and the molecular players involved.
specific neurons, suggesting a crosstalk between muscle auto- In this regard, studies oriented to (1) decipher how autophagy is
phagy and neuronal responses (Kang and Avery, 2009). It has regulated specifically in different types of muscles and in
also been reported that muscle overexpression of the transcrip- response to different factors; (2) discover key molecular players
tion factor FOXO and its target 4E-BP activates autophagy, de- involved in muscle autophagy and their regulation; and (3) un-
lays muscle functional decay, and extends lifespan in Drosophila derstand the factors involved in the multisystemic role of mus-
by decreasing feeding behavior and the release of insulin from cle autophagy would be of great importance in coming years. In
producing cells (Demontis and Perrimon, 2010). all, based on recent advances in the study of autophagy in
Muscle autophagy can be also regulated through non-cell- muscle physiology and metabolic regulation, autophagy
autonomous mechanisms that are initiated in other tissues or or- emerges as a promising therapeutic target to combat meta-
gans. In this connection, intestine-specific AMPK activation in- bolic diseases in the near future.
duces autophagy not only in the intestine but also in brain and
muscle, leading to reduced insulin-like peptide signaling and ACKNOWLEDGMENTS
slower systemic aging in Drosophila (Ulgherait et al., 2014). In
addition, during Caenorhabditis elegans development, loss of Due to space constraints, we have not included all relevant references in this
growing field, so we kindly ask readers and authors for their understanding.
cuticle annular furrow collagens elicits autophagy in the hypo- This study was supported by research grants from the MINECO (SAF2016-
dermis, intestine, and muscle by secretion of DAF-7 by specific 75246R), the Generalitat de Catalunya (Grant 2017SGR1015), INFLAMES
ciliated neurons (Zhang et al., 2019). Together, these data sug- (PIE-14/00045) from the Instituto de Salud Carlos III, CIBERDEM (‘‘Instituto
de Salud Carlos III’’), the Fundación Ramon Areces (CIVP18A3942), the Funda-
gest a bidirectional close multisystemic relationship between
ción BBVA, the Fundació Marató de TV3 (20132330), and EFSD. A.Z. is a recip-
muscle autophagy and metabolism crucial for the maintenance ient of an ICREA ‘‘Academia’’ Award (Generalitat de Catalunya). We gratefully
of organismal homeostasis. acknowledge institutional funding from the MINECO through the Centres of
Excellence Severo Ochoa Award and from the CERCA Programme of the Gen-
eralitat de Catalunya.
Concluding Remarks and Future Perspectives
Numerous studies have provided evidence of a close relation- REFERENCES
ship between autophagy and energy metabolism. Autophagy,
as a catabolic process, is tightly regulated by a variety of meta- Axe, E.L., Walker, S.A., Manifava, M., Chandra, P., Roderick, H.L., Haber-
bolic signals and, on the other hand, has a crucial role in meta- mann, A., Griffiths, G., and Ktistakis, N.T. (2008). Autophagosome formation
from membrane compartments enriched in phosphatidylinositol 3-phosphate
bolic adaptation and energy metabolism. Autophagy is very and dynamically connected to the endoplasmic reticulum. J. Cell Biol. 182,
active in skeletal muscle, which, as a central tissue in energy 685–701.
homeostasis, is a strategic target tissue to combat metabolic
B’chir, W., Maurin, A.C., Carraro, V., Averous, J., Jousse, C., Muranishi, Y.,
diseases. Therefore, the study of the molecular components Parry, L., Stepien, G., Fafournoux, P., and Bruhat, A. (2013). The eIF2a/ATF4
of the close interrelationship between muscle autophagy and pathway is essential for stress-induced autophagy gene expression. Nucleic
Acids Res. 41, 7683–7699.
metabolism has gained attention in recent years. In this regard,
metabolic cues originated from different nutrient conditions and Bach, M., Larance, M., James, D.E., and Ramm, G. (2011). The serine/threo-
physiological conditions such as muscle contraction and exer- nine kinase ULK1 is a target of multiple phosphorylation events. Biochem. J.
440, 283–291.
cise are known to modulate autophagy in muscle. Importantly,
functional autophagy is also necessary for correct metabolic Batista, T.M., Garcia-Martin, R., Cai, W., Konishi, M., O’Neill, B.T., Sakaguchi,
M., Kim, J.H., Jung, D.Y., Kim, J.K., and Kahn, C.R. (2019). Multi-dimensional
adaptation, muscle function, and exercise performance. Alter- Transcriptional Remodeling by Physiological Insulin In Vivo. Cell Rep. 26,
ations in muscle autophagy are associated with several dis- 3429–3443.e3.

Developmental Cell 54, July 20, 2020 277


ll
Review
Bollinger, L.M., Powell, J.J., Houmard, J.A., Witczak, C.A., and Brault, J.J. Gibala, M.J. (2001). Regulation of skeletal muscle amino acid metabolism dur-
(2015). Skeletal muscle myotubes in severe obesity exhibit altered ubiquitin- ing exercise. Int. J. Sport Nutr. Exerc. Metab. 11, 87–108.
proteasome and autophagic/lysosomal proteolytic flux. Obesity (Silver Spring)
23, 1185–1193. Greer, E.L., Oskoui, P.R., Banko, M.R., Maniar, J.M., Gygi, M.P., Gygi, S.P.,
and Brunet, A. (2007). The energy sensor AMP-activated protein kinase
Bujak, A.L., Crane, J.D., Lally, J.S., Ford, R.J., Kang, S.J., Rebalka, I.A., Green, directly regulates the mammalian FOXO3 transcription factor. J. Biol. Chem.
A.E., Kemp, B.E., Hawke, T.J., Schertzer, J.D., and Steinberg, G.R. (2015). 282, 30107–30119.
AMPK activation of muscle autophagy prevents fasting-induced hypoglyce-
mia and myopathy during aging. Cell Metab. 21, 883–890. Grumati, P., Coletto, L., Sabatelli, P., Cescon, M., Angelin, A., Bertaggia, E.,
Blaauw, B., Urciuolo, A., Tiepolo, T., Merlini, L., et al. (2010). Autophagy is
Camera, D.M., Edge, J., Short, M.J., Hawley, J.A., and Coffey, V.G. (2010). defective in collagen VI muscular dystrophies, and its reactivation rescues my-
Early time course of Akt phosphorylation after endurance and resistance exer- ofiber degeneration. Nat. Med. 16, 1313–1320.
cise. Med. Sci. Sports Exerc. 42, 1843–1852.
Grumati, P., Coletto, L., Schiavinato, A., Castagnaro, S., Bertaggia, E., Sandri,
Campbell, T.L., Mitchell, A.S., McMillan, E.M., Bloemberg, D., Pavlov, D., M., and Bonaldo, P. (2011). Physical exercise stimulates autophagy in normal
Messa, I., Mielke, J.G., and Quadrilatero, J. (2015). High-fat feeding does skeletal muscles but is detrimental for collagen VI-deficient muscles. Auto-
not induce an autophagic or apoptotic phenotype in female rat skeletal mus- phagy 7, 1415–1423.
cle. Exp. Biol. Med. (Maywood) 240, 657–668.
Guerra, B., Guadalupe-Grau, A., Fuentes, T., Ponce-González, J.G., Morales-
Carmignac, V., Svensson, M., Körner, Z., Elowsson, L., Matsumura, C., Gaw- Alamo, D., Olmedillas, H., Guillén-Salgado, J., Santana, A., and Calbet, J.A.
lik, K.I., Allamand, V., and Durbeej, M. (2011). Autophagy is increased in lam- (2010). SIRT1, AMP-activated protein kinase phosphorylation and down-
inin a2 chain-deficient muscle and its inhibition improves muscle morphology stream kinases in response to a single bout of sprint exercise: influence of
in a mouse model of MDC1A. Hum. Mol. Genet. 20, 4891–4902. glucose ingestion. Eur. J. Appl. Physiol. 109, 731–743.

Carnio, S., LoVerso, F., Baraibar, M.A., Longa, E., Khan, M.M., Maffei, M., Re- Gwinn, D.M., Shackelford, D.B., Egan, D.F., Mihaylova, M.M., Mery, A., Vas-
ischl, M., Canepari, M., Loefler, S., Kern, H., et al. (2014). Autophagy impair- quez, D.S., Turk, B.E., and Shaw, R.J. (2008). AMPK phosphorylation of raptor
ment in muscle induces neuromuscular junction degeneration and precocious mediates a metabolic checkpoint. Mol. Cell 30, 214–226.
aging. Cell Rep. 8, 1509–1521.
Halling, J.F., and Pilegaard, H. (2017). Autophagy-Dependent Beneficial Ef-
Choi, S.E., Lee, S.M., Lee, Y.J., Li, L.J., Lee, S.J., Lee, J.H., Kim, Y., Jun, H.S., fects of Exercise. Cold Spring Harb. Perspect. Med. 7, 7.
Lee, K.W., and Kang, Y. (2009). Protective role of autophagy in palmitate-
induced INS-1 beta-cell death. Endocrinology 150, 126–134. Halling, J.F., Ringholm, S., Nielsen, M.M., Overby, P., and Pilegaard, H. (2016).
PGC-1a promotes exercise-induced autophagy in mouse skeletal muscle.
Choi, J.W., Ohn, J.H., Jung, H.S., Park, Y.J., Jang, H.C., Chung, S.S., and Physiol. Rep. 4, 4.
Park, K.S. (2018). Carnitine induces autophagy and restores high-fat diet-
induced mitochondrial dysfunction. Metabolism 78, 43–51. Hardie, D.G., Ross, F.A., and Hawley, S.A. (2012). AMPK: a nutrient and energy
sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 13,
De Palma, C., Morisi, F., Cheli, S., Pambianco, S., Cappello, V., Vezzoli, M., 251–262.
Rovere-Querini, P., Moggio, M., Ripolone, M., Francolini, M., et al. (2012).
He, C., Bassik, M.C., Moresi, V., Sun, K., Wei, Y., Zou, Z., An, Z., Loh, J., Fisher,
Autophagy as a new therapeutic target in Duchenne muscular dystrophy.
J., Sun, Q., et al. (2012). Exercise-induced BCL2-regulated autophagy is
Cell Death Dis. 3, e418.
required for muscle glucose homeostasis. Nature 481, 511–515.
Demontis, F., and Perrimon, N. (2010). FOXO/4E-BP signaling in Drosophila
Hedberg-Oldfors, C., Darin, N., and Oldfors, A. (2017). Muscle pathology in Vici
muscles regulates organism-wide proteostasis during aging. Cell 143,
syndrome-A case study with a novel mutation in EPG5 and a summary of the
813–825.
literature. Neuromuscul. Disord. 27, 771–776.
Efeyan, A., Zoncu, R., Chang, S., Gumper, I., Snitkin, H., Wolfson, R.L., Kirak, Hosokawa, N., Hara, T., Kaizuka, T., Kishi, C., Takamura, A., Miura, Y., Iemura,
O., Sabatini, D.D., and Sabatini, D.M. (2013). Regulation of mTORC1 by the S., Natsume, T., Takehana, K., Yamada, N., et al. (2009). Nutrient-dependent
Rag GTPases is necessary for neonatal autophagy and survival. Nature 493, mTORC1 association with the ULK1-Atg13-FIP200 complex required for auto-
679–683. phagy. Mol. Biol. Cell 20, 1981–1991.
Egan, D., Kim, J., Shaw, R.J., and Guan, K.L. (2011a). The autophagy initiating Hue, L., and Taegtmeyer, H. (2009). The Randle cycle revisited: a new head for
kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. an old hat. Am. J. Physiol. Endocrinol. Metab. 297, E578–E591.
Autophagy 7, 643–644.
Inoki, K., Li, Y., Xu, T., and Guan, K.L. (2003). Rheb GTPase is a direct target of
Egan, D.F., Shackelford, D.B., Mihaylova, M.M., Gelino, S., Kohnz, R.A., Mair, TSC2 GAP activity and regulates mTOR signaling. Genes Dev. 17, 1829–1834.
W., Vasquez, D.S., Joshi, A., Gwinn, D.M., Taylor, R., et al. (2011b). Phosphor-
ylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy Jahng, J.W.S., Alsaadi, R.M., Palanivel, R., Song, E., Hipolito, V.E.B., Sung,
sensing to mitophagy. Science 331, 456–461. H.K., Botelho, R.J., Russell, R.C., and Sweeney, G. (2019). Iron overload in-
hibits late stage autophagic flux leading to insulin resistance. EMBO Rep.
Fritzen, A.M., Madsen, A.B., Kleinert, M., Treebak, J.T., Lundsgaard, A.M., 20, e47911.
Jensen, T.E., Richter, E.A., Wojtaszewski, J., Kiens, B., and Frøsig, C.
(2016). Regulation of autophagy in human skeletal muscle: effects of exercise, Jamart, C., Benoit, N., Raymackers, J.M., Kim, H.J., Kim, C.K., and Francaux,
exercise training and insulin stimulation. J. Physiol. 594, 745–761. M. (2012a). Autophagy-related and autophagy-regulatory genes are induced
in human muscle after ultraendurance exercise. Eur. J. Appl. Physiol. 112,
Fry, C.S., Drummond, M.J., Glynn, E.L., Dickinson, J.M., Gundermann, D.M., 3173–3177.
Timmerman, K.L., Walker, D.K., Volpi, E., and Rasmussen, B.B. (2013). Skel-
etal muscle autophagy and protein breakdown following resistance exercise Jamart, C., Francaux, M., Millet, G.Y., Deldicque, L., Frère, D., and Féasson, L.
are similar in younger and older adults. J. Gerontol. A Biol. Sci. Med. Sci. 68, (2012b). Modulation of autophagy and ubiquitin-proteasome pathways during
599–607. ultra-endurance running. J. Appl. Physiol. 112, 1529–1537.

Fu, T., Xu, Z., Liu, L., Guo, Q., Wu, H., Liang, X., Zhou, D., Xiao, L., Liu, L., Liu, Jiang, S., Heller, B., Tagliabracci, V.S., Zhai, L., Irimia, J.M., DePaoli-Roach,
Y., et al. (2018). Mitophagy Directs Muscle-Adipose Crosstalk to Alleviate Di- A.A., Wells, C.D., Skurat, A.V., and Roach, P.J. (2010). Starch binding
etary Obesity. Cell Rep. 23, 1357–1372. domain-containing protein 1/genethonin 1 is a novel participant in glycogen
metabolism. J. Biol. Chem. 285, 34960–34971.
Galluzzi, L., Baehrecke, E.H., Ballabio, A., Boya, P., Bravo-San Pedro, J.M.,
Cecconi, F., Choi, A.M., Chu, C.T., Codogno, P., Colombo, M.I., et al. Jung, C.H., Jun, C.B., Ro, S.H., Kim, Y.M., Otto, N.M., Cao, J., Kundu, M., and
(2017). Molecular definitions of autophagy and related processes. EMBO J. Kim, D.H. (2009). ULK-Atg13-FIP200 complexes mediate mTOR signaling to
36, 1811–1836. the autophagy machinery. Mol. Biol. Cell 20, 1992–2003.

Gawlik, K.I., and Durbeej, M. (2011). Skeletal muscle laminin and MDC1A: Kang, C., and Avery, L. (2009). Systemic regulation of autophagy in Caeno-
pathogenesis and treatment strategies. Skelet. Muscle 1, 9. rhabditis elegans. Autophagy 5, 565–566.

278 Developmental Cell 54, July 20, 2020


ll
Review
Kim, J., and Guan, K.L. (2019). mTOR as a central hub of nutrient signalling and Mammucari, C., Milan, G., Romanello, V., Masiero, E., Rudolf, R., Del Piccolo,
cell growth. Nat. Cell Biol. 21, 63–71. P., Burden, S.J., Di Lisi, R., Sandri, C., Zhao, J., et al. (2007). FoxO3 controls
autophagy in skeletal muscle in vivo. Cell Metab. 6, 458–471.
Kim, K.H., and Lee, M.S. (2014). Autophagy–a key player in cellular and body
metabolism. Nat. Rev. Endocrinol. 10, 322–337. Manning, B.D., and Toker, A. (2017). AKT/PKB Signaling: Navigating the
Network. Cell 169, 381–405.
Kim, J., Kundu, M., Viollet, B., and Guan, K.L. (2011). AMPK and mTOR regu-
late autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13, Mansueto, G., Armani, A., Viscomi, C., D’Orsi, L., De Cegli, R., Polishchuk,
132–141. E.V., Lamperti, C., Di Meo, I., Romanello, V., Marchet, S., et al. (2017). Tran-
scription Factor EB Controls Metabolic Flexibility during Exercise. Cell Metab.
Kim, J., Kim, Y.C., Fang, C., Russell, R.C., Kim, J.H., Fan, W., Liu, R., Zhong, 25, 182–196.
Q., and Guan, K.L. (2013a). Differential regulation of distinct Vps34 complexes
by AMPK in nutrient stress and autophagy. Cell 152, 290–303. Margeta, M. (2020). Autophagy Defects in Skeletal Myopathies. Annu. Rev.
Pathol. 15, 261–285.
Kim, K.H., Jeong, Y.T., Oh, H., Kim, S.H., Cho, J.M., Kim, Y.N., Kim, S.S., Kim,
D.H., Hur, K.Y., Kim, H.K., et al. (2013b). Autophagy deficiency leads to protec- Mariño, G., Pietrocola, F., Eisenberg, T., Kong, Y., Malik, S.A., Andryushkova,
tion from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat. A., Schroeder, S., Pendl, T., Harger, A., Niso-Santano, M., et al. (2014). Regu-
Med. 19, 83–92. lation of autophagy by cytosolic acetyl-coenzyme A. Mol. Cell 53, 710–725.

Koga, H., Kaushik, S., and Cuervo, A.M. (2010). Altered lipid content inhibits Martinez-Lopez, N., Tarabra, E., Toledo, M., Garcia-Macia, M., Sahu, S., Co-
autophagic vesicular fusion. FASEB J. 24, 3052–3065. letto, L., Batista-Gonzalez, A., Barzilai, N., Pessin, J.E., Schwartz, G.J., et al.
(2017). System-wide Benefits of Intermeal Fasting by Autophagy. Cell Metab.
Komiya, K., Uchida, T., Ueno, T., Koike, M., Abe, H., Hirose, T., Kawamori, R., 26, 856–871.e5.
Uchiyama, Y., Kominami, E., Fujitani, Y., and Watada, H. (2010). Free fatty
acids stimulate autophagy in pancreatic b-cells via JNK pathway. Biochem. Masiero, E., Agatea, L., Mammucari, C., Blaauw, B., Loro, E., Komatsu, M.,
Biophys. Res. Commun. 401, 561–567. Metzger, D., Reggiani, C., Schiaffino, S., and Sandri, M. (2009). Autophagy
is required to maintain muscle mass. Cell Metab. 10, 507–515.
Kotoulas, O.B., Kalamidas, S.A., and Kondomerkos, D.J. (2006). Glycogen
autophagy in glucose homeostasis. Pathol. Res. Pract. 202, 631–638. Mauvezin, C., Orpinell, M., Francis, V.A., Mansilla, F., Duran, J., Ribas, V., Pal-
acı́n, M., Boya, P., Teleman, A.A., and Zorzano, A. (2010). The nuclear cofactor
Lam, T., Harmancey, R., Vasquez, H., Gilbert, B., Patel, N., Hariharan, V., Lee, DOR regulates autophagy in mammalian and Drosophila cells. EMBO Rep.
A., Covey, M., and Taegtmeyer, H. (2016). Reversal of intramyocellular lipid 11, 37–44.
accumulation by lipophagy and a p62-mediated pathway. Cell Death Discov.
Medina, D.L., Di Paola, S., Peluso, I., Armani, A., De Stefani, D., Venditti, R.,
2, 16061.
Montefusco, S., Scotto-Rosato, A., Prezioso, C., Forrester, A., et al. (2015).
Lysosomal calcium signalling regulates autophagy through calcineurin and
Las, G., Serada, S.B., Wikstrom, J.D., Twig, G., and Shirihai, O.S. (2011). Fatty
TFEB. Nat. Cell Biol. 17, 288–299.
acids suppress autophagic turnover in b-cells. J. Biol. Chem. 286,
42534–42544.
Milan, G., Romanello, V., Pescatore, F., Armani, A., Paik, J.H., Frasson, L.,
Seydel, A., Zhao, J., Abraham, R., Goldberg, A.L., et al. (2015). Regulation of
Lettieri Barbato, D., Tatulli, G., Aquilano, K., and Ciriolo, M.R. (2013). FoxO1
autophagy and the ubiquitin-proteasome system by the FoxO transcriptional
controls lysosomal acid lipase in adipocytes: implication of lipophagy during
network during muscle atrophy. Nat. Commun. 6, 6670.
nutrient restriction and metformin treatment. Cell Death Dis. 4, e861.
Mizushima, N., Noda, T., Yoshimori, T., Tanaka, Y., Ishii, T., George, M.D.,
Li, J., Kanasaki, M., Xu, L., Kitada, M., Nagao, K., Adachi, Y., Jinzu, H., Nogu-
Klionsky, D.J., Ohsumi, M., and Ohsumi, Y. (1998). A protein conjugation sys-
chi, Y., Kohno, M., Kanasaki, K., and Koya, D. (2018). A ketogenic amino acid
tem essential for autophagy. Nature 395, 395–398.
rich diet benefits mitochondrial homeostasis by altering the AKT/4EBP1 and
autophagy signaling pathways in the gastrocnemius and soleus. Biochim. Bio- Mizushima, N., Yamamoto, A., Matsui, M., Yoshimori, T., and Ohsumi, Y.
phys. Acta Gen. Subj. 1862, 1547–1555. (2004). In vivo analysis of autophagy in response to nutrient starvation using
transgenic mice expressing a fluorescent autophagosome marker. Mol. Biol.
Lira, V.A., Okutsu, M., Zhang, M., Greene, N.P., Laker, R.C., Breen, D.S., Cell 15, 1101–1111.
Hoehn, K.L., and Yan, Z. (2013). Autophagy is required for exercise training-
induced skeletal muscle adaptation and improvement of physical perfor- Møller, A.B., Vendelbo, M.H., Christensen, B., Clasen, B.F., Bak, A.M., Jørgen-
mance. FASEB J. 27, 4184–4193. sen, J.O., Møller, N., and Jessen, N. (2015). Physical exercise increases auto-
phagic signaling through ULK1 in human skeletal muscle. J. Appl. Physiol. 118,
Liu, X., Niu, Y., Yuan, H., Huang, J., and Fu, L. (2015a). AMPK binds to Sestrins 971–979.
and mediates the effect of exercise to increase insulin-sensitivity through auto-
phagy. Metabolism 64, 658–665. Moresi, V., Carrer, M., Grueter, C.E., Rifki, O.F., Shelton, J.M., Richardson,
J.A., Bassel-Duby, R., and Olson, E.N. (2012). Histone deacetylases 1 and 2
Liu, Y., Palanivel, R., Rai, E., Park, M., Gabor, T.V., Scheid, M.P., Xu, A., and regulate autophagy flux and skeletal muscle homeostasis in mice. Proc.
Sweeney, G. (2015b). Adiponectin stimulates autophagy and reduces oxida- Natl. Acad. Sci. USA 109, 1649–1654.
tive stress to enhance insulin sensitivity during high-fat diet feeding in mice.
Diabetes 64, 36–48. Nakamura, S., and Yoshimori, T. (2017). New insights into autophagosome-
lysosome fusion. J. Cell Sci. 130, 1209–1216.
Lo Verso, F., Carnio, S., Vainshtein, A., and Sandri, M. (2014). Autophagy is not
required to sustain exercise and PRKAA1/AMPK activity but is important to Nazio, F., Strappazzon, F., Antonioli, M., Bielli, P., Cianfanelli, V., Bordi, M.,
prevent mitochondrial damage during physical activity. Autophagy 10, Gretzmeier, C., Dengjel, J., Piacentini, M., Fimia, G.M., and Cecconi, F.
1883–1894. (2013). mTOR inhibits autophagy by controlling ULK1 ubiquitylation, self-asso-
ciation and function through AMBRA1 and TRAF6. Nat. Cell Biol. 15, 406–416.
Louis, E., Raue, U., Yang, Y., Jemiolo, B., and Trappe, S. (2007). Time course
of proteolytic, cytokine, and myostatin gene expression after acute exercise in Nemazanyy, I., Blaauw, B., Paolini, C., Caillaud, C., Protasi, F., Mueller, A.,
human skeletal muscle. J. Appl. Physiol. 103, 1744–1751. Proikas-Cezanne, T., Russell, R.C., Guan, K.L., Nishino, I., et al. (2013). De-
fects of Vps15 in skeletal muscles lead to autophagic vacuolar myopathy
Lu, Q., Yang, P., Huang, X., Hu, W., Guo, B., Wu, F., Lin, L., Kovács, A.L., Yu, and lysosomal disease. EMBO Mol. Med. 5, 870–890.
L., and Zhang, H. (2011). The WD40 repeat PtdIns(3)P-binding protein EPG-6
regulates progression of omegasomes to autophagosomes. Dev. Cell 21, Nishino, I., Fu, J., Tanji, K., Yamada, T., Shimojo, S., Koori, T., Mora, M., Riggs,
343–357. J.E., Oh, S.J., Koga, Y., et al. (2000). Primary LAMP-2 deficiency causes X-
linked vacuolar cardiomyopathy and myopathy (Danon disease). Nature 406,
Luo, L., Lu, A.M., Wang, Y., Hong, A., Chen, Y., Hu, J., Li, X., and Qin, Z.H. 906–910.
(2013). Chronic resistance training activates autophagy and reduces
apoptosis of muscle cells by modulating IGF-1 and its receptors, Akt/mTOR Niso-Santano, M., Malik, S.A., Pietrocola, F., Bravo-San Pedro, J.M., Mariño,
and Akt/FOXO3a signaling in aged rats. Exp. Gerontol. 48, 427–436. G., Cianfanelli, V., Ben-Younès, A., Troncoso, R., Markaki, M., Sica, V., et al.

Developmental Cell 54, July 20, 2020 279


ll
Review
(2015). Unsaturated fatty acids induce non-canonical autophagy. EMBO J. 34, Salminen, A., and Vihko, V. (1984). Autophagic response to strenuous exercise
1025–1041. in mouse skeletal muscle fibers. Virchows Arch. B Cell Pathol. Incl. Mol. Pathol.
45, 97–106.
O’Neill, B.T., Lee, K.Y., Klaus, K., Softic, S., Krumpoch, M.T., Fentz, J., Stan-
ford, K.I., Robinson, M.M., Cai, W., Kleinridders, A., et al. (2016). Insulin and Sanchez, A.M., Csibi, A., Raibon, A., Cornille, K., Gay, S., Bernardi, H., and
IGF-1 receptors regulate FoxO-mediated signaling in muscle proteostasis. Candau, R. (2012). AMPK promotes skeletal muscle autophagy through acti-
J. Clin. Invest. 126, 3433–3446. vation of forkhead FoxO3a and interaction with Ulk1. J. Cell. Biochem. 113,
695–710.
Ogasawara, R., Fujita, S., Hornberger, T.A., Kitaoka, Y., Makanae, Y., Naka-
zato, K., and Naokata, I. (2016). The role of mTOR signalling in the regulation Sandri, M. (2010a). Autophagy in health and disease. 3. Involvement of auto-
of skeletal muscle mass in a rodent model of resistance exercise. Sci. Rep. phagy in muscle atrophy. Am. J. Physiol. Cell Physiol. 298, C1291–C1297.
6, 31142.
Sandri, M. (2010b). Autophagy in skeletal muscle. FEBS Lett. 584, 1411–1416.
Ogborn, D.I., McKay, B.R., Crane, J.D., Safdar, A., Akhtar, M., Parise, G., and
Tarnopolsky, M.A. (2015). Effects of age and unaccustomed resistance exer- Schroeder, B., Schulze, R.J., Weller, S.G., Sletten, A.C., Casey, C.A., and
cise on mitochondrial transcript and protein abundance in skeletal muscle of McNiven, M.A. (2015). The small GTPase Rab7 as a central regulator of hepa-
men. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308, R734–R741. tocellular lipophagy. Hepatology 61, 1896–1907.
Orsi, A., Razi, M., Dooley, H.C., Robinson, D., Weston, A.E., Collinson, L.M., Schwalm, C., Jamart, C., Benoit, N., Naslain, D., Prémont, C., Prévet, J., Van
and Tooze, S.A. (2012). Dynamic and transient interactions of Atg9 with auto- Thienen, R., Deldicque, L., and Francaux, M. (2015). Activation of autophagy in
phagosomes, but not membrane integration, are required for autophagy. Mol. human skeletal muscle is dependent on exercise intensity and AMPK activa-
Biol. Cell 23, 1860–1873. tion. FASEB J. 29, 3515–3526.
Pagano, A.F., Py, G., Bernardi, H., Candau, R.B., and Sanchez, A.M. (2014). Sebastián, D., Hernández-Alvarez, M.I., Segalés, J., Sorianello, E., Muñoz,
Autophagy and protein turnover signaling in slow-twitch muscle during exer- J.P., Sala, D., Waget, A., Liesa, M., Paz, J.C., Gopalacharyulu, P., et al.
cise. Med. Sci. Sports Exerc. 46, 1314–1325. (2012). Mitofusin 2 (Mfn2) links mitochondrial and endoplasmic reticulum func-
tion with insulin signaling and is essential for normal glucose homeostasis.
Peker, N., Donipadi, V., Sharma, M., McFarlane, C., and Kambadur, R. (2018). Proc. Natl. Acad. Sci. USA 109, 5523–5528.
Loss of Parkin impairs mitochondrial function and leads to muscle atrophy.
Am. J. Physiol. Cell Physiol. 315, C164–C185. Sebastián, D., Sorianello, E., Segalés, J., Irazoki, A., Ruiz-Bonilla, V., Sala, D.,
Planet, E., Berenguer-Llergo, A., Muñoz, J.P., Sánchez-Feutrie, M., et al.
Penna, F., Ballarò, R., Martinez-Cristobal, P., Sala, D., Sebastian, D., Bus-
(2016). Mfn2 deficiency links age-related sarcopenia and impaired autophagy
quets, S., Muscaritoli, M., Argilés, J.M., Costelli, P., and Zorzano, A. (2019). to activation of an adaptive mitophagy pathway. EMBO J. 35, 1677–1693.
Autophagy Exacerbates Muscle Wasting in Cancer Cachexia and Impairs
Mitochondrial Function. J. Mol. Biol. 431, 2674–2686. Settembre, C., De Cegli, R., Mansueto, G., Saha, P.K., Vetrini, F., Visvikis, O.,
Huynh, T., Carissimo, A., Palmer, D., Klisch, T.J., et al. (2013). TFEB controls
Pereira, R.O., Tadinada, S.M., Zasadny, F.M., Oliveira, K.J., Pires, K.M.P., Ol-
cellular lipid metabolism through a starvation-induced autoregulatory loop.
vera, A., Jeffers, J., Souvenir, R., Mcglauflin, R., Seei, A., et al. (2017). OPA1
Nat. Cell Biol. 15, 647–658.
deficiency promotes secretion of FGF21 from muscle that prevents obesity
and insulin resistance. EMBO J. 36, 2126–2145. Shen, S., Niso-Santano, M., Adjemian, S., Takehara, T., Malik, S.A., Minoux,
Polson, H.E., de Lartigue, J., Rigden, D.J., Reedijk, M., Urbé, S., Clague, M.J., H., Souquere, S., Mariño, G., Lachkar, S., Senovilla, L., et al. (2012). Cyto-
plasmic STAT3 represses autophagy by inhibiting PKR activity. Mol. Cell 48,
and Tooze, S.A. (2010). Mammalian Atg18 (WIPI2) localizes to omegasome-
anchored phagophores and positively regulates LC3 lipidation. Autophagy 6, 667–680.
506–522.
Singh, R., Kaushik, S., Wang, Y., Xiang, Y., Novak, I., Komatsu, M., Tanaka, K.,
Proikas-Cezanne, T., Takacs, Z., Dönnes, P., and Kohlbacher, O. (2015). WIPI Cuervo, A.M., and Czaja, M.J. (2009). Autophagy regulates lipid metabolism.
proteins: essential PtdIns3P effectors at the nascent autophagosome. J. Cell Nature 458, 1131–1135.
Sci. 128, 207–217.
Smiles, W.J., Areta, J.L., Coffey, V.G., Phillips, S.M., Moore, D.R., Stelling-
Randle, P.J., Garland, P.B., Hales, C.N., and Newsholme, E.A. (1963). The werff, T., Burke, L.M., Hawley, J.A., and Camera, D.M. (2015). Modulation of
glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic distur- autophagy signaling with resistance exercise and protein ingestion following
bances of diabetes mellitus. Lancet 1, 785–789. short-term energy deficit. Am. J. Physiol. Regul. Integr. Comp. Physiol. 309,
R603–R612.
Risson, V., Mazelin, L., Roceri, M., Sanchez, H., Moncollin, V., Corneloup, C.,
Richard-Bulteau, H., Vignaud, A., Baas, D., Defour, A., et al. (2009). Muscle Suzuki, K., Kubota, Y., Sekito, T., and Ohsumi, Y. (2007). Hierarchy of Atg pro-
inactivation of mTOR causes metabolic and dystrophin defects leading to se- teins in pre-autophagosomal structure organization. Genes Cells 12, 209–218.
vere myopathy. J. Cell Biol. 187, 859–874.
Takahashi, Y., He, H., Tang, Z., Hattori, T., Liu, Y., Young, M.M., Serfass, J.M.,
Roberts, M.N., Wallace, M.A., Tomilov, A.A., Zhou, Z., Marcotte, G.R., Tran, Chen, L., Gebru, M., Chen, C., et al. (2018). An autophagy assay reveals the
D., Perez, G., Gutierrez-Casado, E., Koike, S., Knotts, T.A., et al. (2017). A ESCRT-III component CHMP2A as a regulator of phagophore closure. Nat.
Ketogenic Diet Extends Longevity and Healthspan in Adult Mice. Cell Metab. Commun. 9, 2855.
26, 539–546.e5.
Tan, S.H., Shui, G., Zhou, J., Li, J.J., Bay, B.H., Wenk, M.R., and Shen, H.M.
Rodrı́guez-Nuevo, A., Dı́az-Ramos, A., Noguera, E., Dı́az-Sáez, F., Duran, X., (2012). Induction of autophagy by palmitic acid via protein kinase C-mediated
Muñoz, J.P., Romero, M., Plana, N., Sebastián, D., Tezze, C., et al. (2018). signaling pathway independent of mTOR (mammalian target of rapamycin).
Mitochondrial DNA and TLR9 drive muscle inflammation upon Opa1 defi- J. Biol. Chem. 287, 14364–14376.
ciency. EMBO J. 37, 37.
Tezze, C., Romanello, V., Desbats, M.A., Fadini, G.P., Albiero, M., Favaro, G.,
Russell, R.C., Tian, Y., Yuan, H., Park, H.W., Chang, Y.Y., Kim, J., Kim, H., Ciciliot, S., Soriano, M.E., Morbidoni, V., Cerqua, C., et al. (2017). Age-Associ-
Neufeld, T.P., Dillin, A., and Guan, K.L. (2013). ULK1 induces autophagy by ated Loss of OPA1 in Muscle Impacts Muscle Mass, Metabolic Homeostasis,
phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat. Cell Biol. Systemic Inflammation, and Epithelial Senescence. Cell Metab. 25, 1374–
15, 741–750. 1389.e6.

Ryu, D., Mouchiroud, L., Andreux, P.A., Katsyuba, E., Moullan, N., Nicolet-Dit- Tezze, C., Romanello, V., and Sandri, M. (2019). FGF21 as Modulator of Meta-
Félix, A.A., Williams, E.G., Jha, P., Lo Sasso, G., Huzard, D., et al. (2016). Ur- bolism in Health and Disease. Front. Physiol. 10, 419.
olithin A induces mitophagy and prolongs lifespan in C. elegans and increases
muscle function in rodents. Nat. Med. 22, 879–888. Tian, Y., Li, Z., Hu, W., Ren, H., Tian, E., Zhao, Y., Lu, Q., Huang, X., Yang, P.,
Li, X., et al. (2010). C. elegans screen identifies autophagy genes specific to
Sala, D., Ivanova, S., Plana, N., Ribas, V., Duran, J., Bach, D., Turkseven, S., multicellular organisms. Cell 141, 1042–1055.
Laville, M., Vidal, H., Karczewska-Kupczewska, M., et al. (2014). Autophagy-
regulating TP53INP2 mediates muscle wasting and is repressed in diabetes. Torre-Villalvazo, I., Alemán-Escondrillas, G., Valle-Rı́os, R., and Noriega, L.G.
J. Clin. Invest. 124, 1914–1927. (2019). Protein intake and amino acid supplementation regulate exercise

280 Developmental Cell 54, July 20, 2020


ll
Review
recovery and performance through the modulation of mTOR, AMPK, FGF21, cle wasting are regulated by a Fyn/STAT3/Vps34 signaling pathway. Cell Rep.
and immunity. Nutr. Res. 72, 1–17. 1, 557–569.

Ulgherait, M., Rana, A., Rera, M., Graniel, J., and Walker, D.W. (2014). AMPK Yang, L., Licastro, D., Cava, E., Veronese, N., Spelta, F., Rizza, W., Bertozzi,
modulates tissue and organismal aging in a non-cell-autonomous manner. Cell B., Villareal, D.T., Hotamisligil, G.S., Holloszy, J.O., and Fontana, L. (2016).
Rep. 8, 1767–1780. Long-Term Calorie Restriction Enhances Cellular Quality-Control Processes
in Human Skeletal Muscle. Cell Rep. 14, 422–428.
Vainshtein, A., Tryon, L.D., Pauly, M., and Hood, D.A. (2015). Role of PGC-1a
during acute exercise-induced autophagy and mitophagy in skeletal muscle.
Yin, Z., Pascual, C., and Klionsky, D.J. (2016). Autophagy: machinery and
Am. J. Physiol. Cell Physiol. 308, C710–C719.
regulation. Microb. Cell 3, 588–596.
Wagenmakers, A.J. (1998). Muscle amino acid metabolism at rest and during
exercise: role in human physiology and metabolism. Exerc. Sport Sci. Rev. 26, Yuan, H.X., Russell, R.C., and Guan, K.L. (2013). Regulation of PIK3C3/VPS34
287–314. complexes by MTOR in nutrient stress-induced autophagy. Autophagy 9,
1983–1995.
Wang, Y., and Pessin, J.E. (2013). Mechanisms for fiber-type specificity of
skeletal muscle atrophy. Curr. Opin. Clin. Nutr. Metab. Care 16, 243–250. Zhang, Y., Nicholatos, J., Dreier, J.R., Ricoult, S.J., Widenmaier, S.B., Hotami-
sligil, G.S., Kwiatkowski, D.J., and Manning, B.D. (2014). Coordinated regula-
Wang, X., Blagden, C., Fan, J., Nowak, S.J., Taniuchi, I., Littman, D.R., and tion of protein synthesis and degradation by mTORC1. Nature 513, 440–443.
Burden, S.J. (2005). Runx1 prevents wasting, myofibrillar disorganization,
and autophagy of skeletal muscle. Genes Dev. 19, 1715–1722. Zhang, Y., Qi, L., and Zhang, H. (2019). TGFb-like DAF-7 acts as a systemic
signal for autophagy regulation in C. elegans. J. Cell Biol. 218, 3998–4006.
Wang, Z., Miao, G., Xue, X., Guo, X., Yuan, C., Wang, Z., Zhang, G., Chen, Y.,
Feng, D., Hu, J., and Zhang, H. (2016). The Vici Syndrome Protein EPG5 Is a
Rab7 Effector that Determines the Fusion Specificity of Autophagosomes Zhao, J., Brault, J.J., Schild, A., Cao, P., Sandri, M., Schiaffino, S., Lecker,
with Late Endosomes/Lysosomes. Mol. Cell 63, 781–795. S.H., and Goldberg, A.L. (2007). FoxO3 coordinately activates protein degra-
dation by the autophagic/lysosomal and proteasomal pathways in atrophying
Wolfson, R.L., and Sabatini, D.M. (2017). The Dawn of the Age of Amino Acid muscle cells. Cell Metab. 6, 472–483.
Sensors for the mTORC1 Pathway. Cell Metab. 26, 301–309.
Zhao, H., Zhao, Y.G., Wang, X., Xu, L., Miao, L., Feng, D., Chen, Q., Kovács,
Yamada, E., Bastie, C.C., Koga, H., Wang, Y., Cuervo, A.M., and Pessin, J.E. A.L., Fan, D., and Zhang, H. (2013). Mice deficient in Epg5 exhibit selective
(2012). Mouse skeletal muscle fiber-type-specific macroautophagy and mus- neuronal vulnerability to degeneration. J. Cell Biol. 200, 731–741.

Developmental Cell 54, July 20, 2020 281

You might also like