You are on page 1of 15

REviEws

Autophagy in metabolic disease


and ageing
Munehiro Kitada   1,2 and Daisuke Koya   1,2,3 ✉
Abstract | Autophagy is an evolutionarily conserved, lysosome-​dependent catabolic process
whereby cytoplasmic components, including damaged organelles, protein aggregates and
lipid droplets, are degraded and their components recycled. Autophagy has an essential role
in maintaining cellular homeostasis in response to intracellular stress; however, the efficiency
of autophagy declines with age and overnutrition can interfere with the autophagic process.
Therefore, conditions such as sarcopenic obesity, insulin resistance and type 2 diabetes mellitus
(T2DM) that are characterized by metabolic derangement and intracellular stresses (including
oxidative stress, inflammation and endoplasmic reticulum stress) also involve the accumulation
of damaged cellular components. These conditions are prevalent in ageing populations. For
example, sarcopenia is an age-​related loss of skeletal muscle mass and strength that is involved
in the pathogenesis of both insulin resistance and T2DM, particularly in elderly people. Impairment
of autophagy results in further aggravation of diabetes-​related metabolic derangements in insulin
target tissues, including the liver, skeletal muscle and adipose tissue, as well as in pancreatic
β-​cells. This Review summarizes the role of autophagy in the pathogenesis of metabolic diseases
associated with or occurring in the context of ageing, including insulin resistance, T2DM and
sarcopenic obesity, and describes its potential as a therapeutic target.

The increasing incidence of metabolic derangements, and dysfunction of cellular metabolic homeostasis, which
including sarcopenic obesity, insulin resistance and are closely associated with ageing5. Under starvation
type 2 diabetes mellitus (T2DM), among elderly people conditions, autophagy can provide nutrients to maintain
in the past decade is a subject of serious concern world- cellular functions through the degradation of large mole­
wide1. Ageing causes many physiological changes in body cules, organelles, proteins and end products. In addition,
composition, resulting in an increase in body fat mass, autophagy removes damaged organelles, misfolded pro-
including increased visceral fat, and a decrease in muscle teins and lipid droplets, and thus plays a crucial role in
mass and strength; these changes are closely related to maintaining cellular homeostasis. Autophagic activity
metabolic disease2. The accumulation of excessive fat decreases with age in many species, and adequate auto-
1
Department of Diabetology
and Endocrinology,
in visceral adipose depots and in non-​adipose tissues, phagy is recognized as a central biological pathway that
Kanazawa Medical including the liver and skeletal muscle, is an important promotes health and longevity6.
University, Uchinada, factor in the pathogenesis of obesity-​related metabolic Under conditions of metabolic derangement, prom-
Ishikawa, Japan. disease2. In addition, obesity and sarcopenia often coexist, inent accumulation of lipid droplets, protein aggregates
2
Division of Anticipatory and it is recognized that sarcopenic obesity worsens and damaged organelles (all major substrates of auto-
Molecular Food Science meta­bolic disease in older people3. Moreover, impaired phagy) is observed in β-​cells and insulin target tissues,
and Technology, Medical
Research Institute,
pancreatic β-​cell function is crucial for the pathogenesis including skeletal muscle, liver and adipose tissue.
Kanazawa Medical of T2DM, which is also associated with ageing4. Impaired autophagy is closely related to the patho-
University, Uchinada, Other hallmarks of ageing include an imbalance in genesis of several metabolic diseases, including sarco-
Ishikawa, Japan. intercellular communication, stem cell exhaustion, dys- penic obesity, insulin resistance and T2DM. Therefore,
3
Department of General regulated nutrient sensing, genomic instability, telomere induction of adequate autophagy is a potential therapeu-
Internal Medicine, Kusatsu attrition, epigenetic alterations, loss of proteostasis, tic target in the amelioration of age-​related metabolic
General Hospital, Kusatsu,
Shiga, Japan.
cellular senescence and mitochondrial dysfunction5. derangements.
✉e-​mail: koya0516@ Progressive accumulation of cellular damage owing to In this Review, we outline the mechanism of auto-
kanazawa-​med.ac.jp intracellular stressors, such as oxidative stress, inflam- phagy and its regulation; discuss the relationship bet­
https://doi.org/10.1038/ mation, endoplasmic reticulum (ER) stress and an inad- ween impaired autophagy and age-​related metabolic
s41574-021-00551-9 equate response to hypoxia, ultimately lead to disruption derangements, including insulin resistance, T2DM and

Nature Reviews | Endocrinology

0123456789();:
Reviews

Key points Phagophore elongation


At the omegasome, increased local production of PI3P
• Autophagic activity decreases with age in many species, and adequate autophagy is results in recruitment of the effector proteins WD
recognized as an important biological pathway that promotes health and longevity. repeat domain phosphoinositide-​interacting protein 2
• Basal autophagy and appropriate adaptive autophagy responses induced by (WIPI2) and zinc-​f inger FYVE domain-​containing
intracellular stress and changes in nutrient status enable elimination of damaged protein 1 (DFCP1) to the omegasome via the interac-
cellular components and contribute to cellular homeostasis. tion of PI3P with their PI3P-​binding domains. In the
• Nutrient-​sensing pathways, including those involving mTORC1, AMPK and SIRT1, ATG12 conjugation system, ATG7 and ATG10 conju-
are involved in the regulation of autophagy at multiple steps during autophagic flux. gate ATG12 to ATG5. The ATG12–ATG5 complex then
• Impairment of autophagy results in further aggravation of diabetes-​related metabolic binds to ATG16L1 and mobilizes to the phagophore as
derangements in insulin target tissues, including the liver, skeletal muscle and adipose the ATG12–ATG5–ATG16L1 complex, which binds to
tissue, as well as in pancreatic β-​cells.
WIPI2 via ATG16L1. ATG2 connects the ER membrane
• Calorie restriction, exercise and pharmacological interventions, including several to the isolation membrane and delivers phospholipids
antidiabetic medicines, induce autophagy and are, therefore, recognized as candidate
contained in the ER membrane to the cytoplasmic layer
therapies for age-​related metabolic disease.
of the isolation membrane12. Thereafter, phospholipids
delivered by ATG2 are flipped and redistributed by
sarcopenic obesity; and discuss candidate therapies for ATG9, which is a lipid scramblase, thereby driving
regulation of autophagy. phagophore membrane elongation13,14.
In addition to the ER, several membranous orga-
Types of autophagy nelles, including the plasma membrane, mitochondria,
Three types of autophagy have been distinguished on recycling endosomes and Golgi complex, contribute
the basis of their differing mechanisms of cargo seques- to elongation of the phagophore by donating mem-
tration. Microautophagy involves sequestration of cyto- brane materials, and parts of these lipid bilayers are
plasmic components directly into lysosomes, whereas delivered by ATG9-​containing vesicles. Microtubule-
chaperone-​mediated autophagy involves selective deg- associated proteins 1A/1B light chain 3 (LC3) is cleaved
radation of cargo proteins, which are recognized and by ATG4 to form LC3I, and LC3I then conjugates with
delivered to lysosomes by chaperone complexes. Finally, phosphatidylethanolamine via ATG7 and ATG3 to
macroautophagy is mediated by many proteins encoded form LC3II. The ATG12 complex promotes LC3II for-
by autophagy-​related genes (ATGs). As macroautophagy mation and its conjugation to the phagosome. LC3II
is the most extensively studied of these types in the field interacts with cargo via the LC3-​interacting regions
of ageing and metabolic disease, the four sequential (LIRs) of autophagy receptors and is also required for
stages of macroautophagy are discussed in detail in the elongation and closure of the phagophore membrane
next sections (Fig. 1)7–11. as autophagosomes mature. In addition, LC3II is
removed by ATG4 from the surface of autophagosomes
Initiation and is recycled for the next round of the conjugation
Intracellular signalling pathways that activate initia- reaction.
tion of the autophagic process are triggered by vari-
ous intracellular stresses and alterations in nutrient Autophagosome–lysosome fusion
status. The target of these pathways is the UNC-​like PI3KC3-​CII, which is required for autophagosome
autophagy-​activating kinase 1 (ULK1) initiation com- maturation, contains ultraviolet radiation resistance-
plex, which consists of ULK1, ATG13, focal adhesion associated gene protein (UVRAG). Interaction of
kinase family interacting protein of 200 kDa (FIP200) UVRAG with the homotypic fusion and vacuole pro-
and ATG101. Activation of this complex triggers tein sorting (HOPS) tethering complex stimulates the
membrane nucleation. fusion of autophagosomes and lysosomes. Rubicon
(encoded by RUBCN) suppresses the formation of
Phagophore formation autolysosomes by interacting with UVRAG. Fusion
Nucleation of the membrane requires the lipid kinase of the autophagosome membrane with the lysosome
activity of class III phosphatidylinositol-3 kinase com- membrane requires both the HOPS complex, which
plex I (PI3KC3-​CI), which generates phosphatidylino­ tethers autophagosomes to lysosomes, and syntaxin-17
sitol 3-​phosphate (PI3P) at the phagophore. PI3KC3-​CI (STX17), which facilitates membrane fusion. Pacer
consists of catalytic subunit vacuolar protein sorting 34 interacts with STX17 and recruits the HOPS com-
(VPS34), beclin-1, p115, autophagy-​related protein plex to autophagosomes. Autophagosome–lysosome
14-​like protein (ATG14L), activating molecule in fusion is mediated by trafficking of several proteins,
beclin-1-​regulated autophagy protein 1 (AMBRA1) and including small GTPase family proteins such as
nuclear receptor-​binding factor 2 (NRBF2). Once the Ras-​related protein Rab-7a. Effector proteins, such
ULK1 initiation complex is activated, ULK1 binds as soluble N-​ethylmaleimide-​sensitive fusion attach-
to and phosphorylates ATG14 at Ser19, VPS34 at Ser249 ment protein receptors (SNAREs), within the auto-
and beclin-1 at Ser15 (as well as at other sites), which phagic cargo are degraded and released into the
causes activation of PI3KC3-​CI; these proteins then cytoplasm.
translocate to the phagophore assembly site, which Lysosomes are regenerated from the autolysosomal
is located at a characteristic ER structure called the membrane in a process known as autophagic lysosome
omegasome. re-​formation (ALR), which sets the stage for a new cycle

www.nature.com/nrendo

0123456789();:
Reviews

of autophagosome initiation. The increase in intracellu- are recycled to maintain lysosome homeostasis during
lar amino acid levels resulting from autophagic activity autophagy, a process that marks the last stage of auto-
causes mTORC1 to reactivate, thereby not only shut- phagy. This step is needed to provide a functional pool
ting down the generation of autophagosomes but also of lysosomes for the generation of autolysosomes during
promoting ALR. During ALR, lysosomal membranes prolonged starvation.

Plasma membrane Cytoplasm Degradation


• Intracellular stress
• Alteration of Autophagic lysosome Recycling of nutrients
nutrient status reformation
Membrane Mitochondrion
nucleation and
Initiation phagophore
formation Acidic
Protein hydrolases
Membrane sources aggregate
PI3KC3 complex I
• Plasma membrane Lysosome
ULK1 complex VPS34 Beclin-1 • Golgi Autolysosome
ULK1 ATG14L AMBRA1 p115 • Mitochondria
• Recycling endosomes
ATG13 FIP200 • ATG9-containing vesicles
ATG101 SNAREs
Lipid
droplet Rab-7a
Cargo
PI3KC3 sequestration
complex I Phagophore
elongation
ATG12
ULK1 complex
PI3P

2
IPI
G3

ATG10
G1

conjugation Autophagosome–
W I3P
AT

ATG12
1 AT

P lysosome fusion
DFCP1 W 16L G5

Isolation
P1

LC3II
P
PI3P PI3

G AT

membrane
DFC

PE ATG7 PI3KC3-CII
ATG5 HOPS
2

Beclin-1
IPI
AT

ATG16L AT12
ATG2 VPS34
ATG9 ATG5 STX17
Omegasome UVRAG
Nucleus Expansion Pacer VPS15
ATG9
Rough
ER
Sealing Rubicon
Autophagosome ATG4
ATG3

ATG7
LC3I ATG4

LC3 conjugation LC3


system

Fig. 1 | Autophagy. On initiation of autophagy, intracellular stress and LC3-​interacting regions and is required for elongation and closure of the
alterations in nutrient status activate UNC-​like autophagy activating phagophore membrane as autophagosomes mature. LC3II is removed by
kinase 1 (ULK1). ULK1 then binds to class III phosphatidylinositol-3 ATG4 from the surface of autophagosomes. PI3KC3-​CII contains UV
kinase (PI3KC3) complex 1 (PI3KC3-​C I), causing generation of radiation resistance-​associated gene protein (UVRAG), which interacts
phosphatidylinositol-3-​phosphate (PI3P) at the phagophore assembly site with homotypic fusion and vacuole protein sorting (HOPS) and stimulate
(located at the omegasome) after membrane isolation from the autophagosome–lysosome fusion. Rubicon suppresses the formation of
endoplasmic reticulum (ER). Local PI3P production drives PI3P-​mediated autolysosomes by interacting with UVRAG. Fusion of the autophagosome
recruitment of the effector proteins WD repeat domain phosphoinositide-​ membrane with the lysosome membrane requires both the HOPS complex,
interacting protein 2 (WIPI2) and zinc-​finger FYVE domain-​containing which tethers autophagosomes to lysosomes, and syntaxin-17 (STX17).
protein 1 (DFCP1). In the autophagy-​related gene (ATG) 12 conjugation Pacer interacts with STX17 and recruits HOPS to autophagosomes.
system, ATG12–ATG5–ATG16L1 complex binds to WIPI2 via ATG16LI. ATG2 Autophagosome–lysosome fusion is mediated by trafficking proteins,
delivers phospholipids contained in the ER membrane to the cytoplasmic including small GTPase-​family proteins, such as Rab-7a. Within the
layer of the isolation membrane. Then, ATG9 drives membrane expansion. autophagic cargo, effector proteins (such as soluble N-​ethylmaleimide-​
Membrane materials from other organelles, including the plasma sensitive fusion attachment protein receptors (SNAREs)) are degraded and
membrane, mitochondria, recycling endosomes and Golgi complex, released into the cytoplasm for nutrient recycling. Lysosomes are reformed
also contribute to phagophore elongation and are delivered by from the autolysosomal membrane, setting the stage for a new cycle of
ATG9-​containing vesicles. The ATG12 complex promotes microtubule-​ autolysosome formation. AMBRA1, activating molecule in beclin-
associated protein light chain (LC)-3II formation via LC3I from LC3, and its 1-​regulated autophagy protein 1; FIP200, focal adhesion kinase family
conjugation to the phagosome via phosphatidylethanolamine (PE). LC3II interacting protein of 200 kDa; VPS, vacuolar protein sorting. Adapted
interacts with cargo through selective autophagy receptors via from ref.8, Springer Nature Limited.

Nature Reviews | Endocrinology

0123456789();:
Reviews

For  instance, an accumulation of autophagosomes


might indicate either generally increased autophagic
activity or, conversely, decreased activity of one or more
Ubiquitin-independent down­stream steps, such as inefficient autophagosome–
• Mitophagy lysosome fusion or decreased lysosomal degradation.
• ERphagy Therefore, accurate estimation of autophagic activity
requires a determination of the overall rate of autophagic
degradation, which is termed autophagic flux. This
dynamic parameter represents the amount of autophagic
degradation per unit time.
Ubiquitin-dependent The most commonly used methods for assessing auto-
• Mitophagy
• Aggrephagy phagic flux involve measurement of the turnover rate of
ATG proteins such as LC3 (Supplementary Table 1).
For example, the amount of LC3II roughly correlates
Phagosome with the number of autophagosomes. However, merely
showing increased levels of LC3II is insufficient to
Selective autophagy Protein demon­strate increased autophagic flux, because an
LIR UBD
receptor aggregate
increase in LC3II levels could also occur in the context
Protein with Damaged
LIR
integral LIR motif mitochondrion of decreased autophagic throughput downstream of
autophagosome–lysosome fusion. Determination
LC3II Damaged ER
of autophagic flux mandates that the amount of degraded
Ubiquitin
LC3II in lysosomes is also measured, which can be done
by comparing the amount of LC3II present in cells in
Fig. 2 | Selective autophagy. Selective autophagy is the presence and absence of lysosomal inhibitors such
the process by which specific types of cell components as bafilomycin A1, chloroquine, pepstatin A or E64d.
(including protein aggregates, lipid droplets, mitochon-
Under conditions of increased autophagic flux, the
dria, peroxisomes, lysosomes and endoplasmic reticulum)
are removed and recycled. Efficient recognition and level of LC3II in autophagosomes will be increased in
sequestration of these components within autophago- the presence of lysosomal inhibitors owing to blockade
somes involves a diverse array of ubiquitin-​dependent and of autophagosome–lysosome fusion, which causes
ubiquitin-​independent cargo signals that physically link LC3II to accumulate. Autophagic flux can also be eva­
their targets to selective autophagy receptors on the luated by monitoring the levels of selective autophagy
autophagosome via microtubule-​associated proteins substrates such as sequestosome-1. Similarly to the
1A/1B light chain 3 (LC3)-​interacting regions (LIRs, which LC3 flux assay, the amount of sequestosome-1 degra­
bind to lipidated and/or autophagosome membrane-​ dation can be measured using lysosomal inhibitors
associated LC3II), thereby targeting them for degradation. (Supplementary Table 1).
ER, endoplasmic reticulum; UBD, ubiquitin D.
Other methods for measuring autophagic flux involve
transfection with fluorescent reporters that are capable
Selective autophagy of indicating the cumulative amount of degradation of
Selective autophagy removes and recycles specific types cytoplasm-​derived material within lysosomes. Such
of cell components. For example, protein aggregates are reporters have been used successfully in cultured cells
removed by aggrephagy, lipid droplets by lipophagy and and model organisms19–21 (Box 1 and Supplementary
damaged or superfluous mitochondria by mitophagy. Table 1). However, monitoring autophagic flux in vivo
Other forms of selective autophagy remove and recycle in humans is currently not feasible.
peroxisomes, lysosomes and ER.
Selective autophagy involves efficient recognition and Regulation of autophagy
sequestration of the cargo within autophagosomes15,16. All organisms respond to intracellular stresses (includ-
The cargo signals recognized by selective autophagy ing oxidative stress, inflammation, hypoxia and ER
receptors are diverse, but can be broadly grouped stress) and to nutrients that are available in the environ­
into ubiquitin-​dependent and ubiquitin-​independent ment by implementing adaptive changes to cellular
types (Fig. 2; Table 1). These proteins physically link activities including metabolism, growth and autophagy.
their targets to the autophagosome via LIR motifs Basal autophagy as well as adaptive autophagy responses
that bind to lipidated LC3II and/or autophagosome induced by intracellular stress and changes in nutrient
membrane-​associated LC3II, thereby targeting them status eliminate damaged cellular components and have
for degradation. In addition to autophagy receptors, an important role in cellular homeostasis (Fig. 3). By con-
lipids (including phospholipids, such as cardiolipin and trast, impaired or excessive autophagy leads to cell senes-
ceramide) have been shown to mediate mitophagy17,18. cence or death owing to the accumulation of damaged
cellular components (Fig. 3).
Autophagic flux Nutrient-sensing pathways, including those involving
Autophagy is a dynamic, multistage process. Therefore, mechanistic target of rapamycin complex 1 (mTORC1)22–39,
estimating the rate at which autophagy occurs by AMP-​a ctivated kinase (AMPK) 23,40–53 and SIRT1
static enumeration of autophagic structures at a sin- (NAD+-​dependent protein deacetylase sirtuin-1)54–57,
gle time point can lead to inaccurate interpretations. are involved in the regulation of autophagic flux at

www.nature.com/nrendo

0123456789();:
Reviews

multiple steps (Table 2) and show substantial crosstalk58–62 expression of rubicon (a negative regulator of autophagy)
(Fig.  3) . During starvation, autophagy is intensively is increased in C. elegans, Drosophila and mouse tissues
induced by activation of AMPK and SIRT1 and sup- from aged individuals at the transcript and/or protein
pression of mTORC1 (refs58–62) (Fig. 3). By contrast, auto- level, whereas expression of this protein is suppressed
phagy is suppressed under conditions of excess nutrition in several long-​lived C. elegans models and in mice sub-
through activation of mTORC1 and reductions in jected to calorie restriction75. Knockdown of the gene
AMPK and SIRT1 activity (Fig. 3). Most of the processes encoding rubicon in C. elegans and Drosophila extends
through which intracellular stress induces autophagy are lifespan and (in mice) ameliorates several age-​associated
mediated by nutrient-​sensing pathways57,63,64. In addition, phenotypes in the heart, kidneys and brain75.
lysosome-​mediated degradative pathways are regulated By contrast, several reports published in the past
by genes in the coordinated lysosomal expression and 4 years suggest that autophagy can be detrimental in some
regulation (CLEAR) network65,66, which includes both different model systems under certain circumstances. For
genes related to the lysosomal complement (that is, instance, elevated autophagy accompanied by increased
encoding hydrolases, transporters and accessory pro- mitochondrial permeability shortens lifespan in C. elegans
teins) and genes involved in the regulation of lysosomal and in mice76. Moreover, although autophagy is indis­
biogenesis and function. pensable during development, its attenuation late in
life can extend healthy lifespan in C. elegans77. Finally,
Autophagy, ageing and lifespan whereas moderate overexpression of Atg1 substantially
Autophagy is part of a convergent mechanism shared by extends lifespan in Drosophila, strong induction of its
various pathways involved in longevity and the regula- expression is toxic78.
tion of lifespan. The expression of ATG-​related proteins
and other proteins required for the induction of auto- Pathophysiological roles of autophagy
phagy, such as SIRT1, is decreased in tissues from older Overnutrition and lack of exercise cause sarcopenic
individuals. The capacity of autophagy also declines obesity, insulin resistance and T2DM, which are asso-
with ageing, which contributes to the accumulation of ciated with increased mortality. Ageing can accelerate
damaged macromolecules and organelles and results in these meta­bolic derangements and is also a recognized
adverse effects on lifespan. risk factor for T2DM. Chronic calorie surplus results in
Inactivation of autophagy owing to knockdown of storage of fat in adipose tissue; moreover, the accumula-
various ATGs leads to reduced lifespan as a result of the tion of fat — in non-​adipose tissues, including the liver
premature onset of age-​related symptoms in several and skeletal muscle, as well as excess fat in visceral adi-
model organisms, including Saccharomyces cerevisiae, pose tissue — also provokes insulin resistance. In addi-
Caenorhabditis elegans and Drosophila melanogaster 67–69. tion, excessive ectopic fat accumulation can elevate serum
However, knockout of Atg3, Atg5 and Atg7 in mice is levels of free fatty acids, resulting in systemic lipotoxicity
embryonically or neonatally lethal, which makes it impos- and β-​cell dysfunction. T2DM develops as a consequence
sible to examine the role of autophagy in mammalian of the combination of insulin resistance and relative insu-
longevity70–72. Conversely, mice that overexpress Atg5 lin deficiency. Glucolipotoxicity-​related mitochondrial
exhibit ubiquitously enhanced autophagy and are normal dysfunction, oxidative stress, inflammation and ER stress
at birth but develop leanness and loss of weight with in pancrea­tic β-​cells, the liver, skeletal muscle and adipose
ageing despite the same food intake as their wild-​type tissue are closely related to the pathogenesis of T2DM.
littermates73. However, the lean phenotype of mice Thus, autophagy has an important homeostatic role in
that overexpress Atg5 is accompanied by an extended mitigating the adverse cellular effects of chronic calorie
lifespan. Mice with a knock-​in gain-​of-​function point excess through organelle quality control and removal of
mutation in Becn1 that disrupts the interaction between protein aggregates and lipid droplets.
beclin-1 and BCL2, thereby constitutively activating The importance of autophagy in the pathogenesis
autophagy, also have an extended lifespan74. By contrast, of metabolic derangements has been shown in mouse

Table 1 | Selective autophagy


Selective Mammalian cargo proteins
autophagy type
Ubiquitin-​dependent Ubiquitin-​independent
Mitophagy Optineurin, calcium-​binding and coiled-​coil BCL2/adenovirus E1B 19 kDa protein-​interacting
domain-​containing protein 2 (also known as protein 3 (BNIP3), BCL2/adenovirus E1B 19 kDa
NDP52), TAX1-​binding protein 1 (TAX1BP1), protein-​interacting protein 3-​like (BNIP3L, also
sequestosome-1 known as NIX) and FUN14 domain-​containing
protein 1 (FUNDC1)
Aggrephagy Neighbour of BRCA1 gene 1 protein (NBR1), Optineurin
optineurin, Toll-nteracting protein (TOLLIP)
ERphagy NR FAM134B (also known as reticulophagy regulator
1 (RETREG1))
Lipophagy NR Patatin-​like phospholipase domain-​containing
enzyme 8 (PNPLA8)
ER, endoplasmic reticulum; NR, not reported

Nature Reviews | Endocrinology

0123456789();:
Reviews

Box 1 | Autophagic flux measurement using fluorescent reporters accumulation of damaged organelles or lipid droplets
is crucial for the pathogenesis of metabolic disease. The
Autophagic flux determination using tandem fluorescent reporters (in which red contribution of impaired autophagy to the development
fluorescent protein (RFP) and green fluorescent protein (GFP) are sequentially bound of metabolic disease is illustrated in Fig. 4.
to the cargo protein of interest) does not require lysosome inhibition, and the tandem
fluorescent module can be adapted to determine whether individual autophagic
Pancreatic β-​cell function
structures are degraded after fusion with lysosomes. Examples of such reporters
include RFP–GFP–LC3 and RFP–GFP–sequestosome-1, in which a monomeric RFP The contribution of autophagy to β-​cell physiology and
(such as mCherry) and GFP are bound to the N terminus of LC3. When autophagosomes metabolic adaptation has been demonstrated in mice with
develop, cells transfected with these reporters exhibit yellow fluorescence due to the autophagy-​deficient β-​cells. Mice with β-​cell-​specific
overlap of red and green fluorescence. Within autolysosomes, GFP fluorescence is ablation of Atg7 develop glucose intolerance and
immediately quenched by the acidic environment, leaving only the red fluorescent decreased insulin secretion81,82, which indicates that basal
signal, which is unaffected by acidic conditions. Confocal microscopy can be used autophagy is crucial for insulin secretion. Under physio­
to observe dynamic changes in the fluorescent signal indicative of autophagic flux. logical fasting conditions, insulin secretion decreases
Selective autophagy substrates such as sequestosome-1 can be used instead of LC3. because the early secretory granules of insulin are
In transfected cells, the GFP–LC3–-​RFP–LC3ΔG reporter (in which GFP–LC3 is fused degraded by lysosomes through inactivation of protein
to the N terminus of an RFP–LC3 molecule whose C-​terminal glycine is deleted) is
kinase D. This process, which is called starvation-​induced
co-​translationally processed by ATG4 to produce GFP–LC3 and free RFP–LC3ΔG.
The GFP–LC3 moiety acts as an autophagic substrate and serves to measure autophagic nascent granule degradation83, results in mTORC1
activity. By contrast, free RFP–LC3ΔG lacks the C-​terminal glycine needed for conjuga- activation and increased levels of intracellular amino
tion to the autophagosome membrane and remains in the cytoplasm where it acts as acids despite fasting, which lead to suppression of auto-
an internal control for reporter expression and cellular health. The ratio of GFP–LC3 phagy. Moreover, excess autophagy in β-​cells degrades
to RFP–LC3ΔG is inversely correlated with cumulative autophagic degradation activity insulin-​c ontaining vesicles through vesicophagy,
and can be evaluated using flow cytometry or a fluorescence microplate reader. The resulting in a decrease in insulin content and glucose
consumption of autophagosomes can be assessed by simply measuring the amount intolerance84. Although basal autophagy and appropri-
of reduction in GFP–LC3 via flow cytometry. However, because this system lacks an ate adaptive autophagy responses are both required for
internal control, another reporter must be used to simultaneously evaluate changes the maintenance of β-​cell mass and insulin secretion,
in cellular health.
insulin secretion is inhibited by starvation-induced
An LC3–HiBiT reporter can be used to measure autophagic flux by monitoring LC3
reporter levels in cell lysates. This reporter employs an 11 amino acid HiBiT peptide nascent granule degradation-​mediated suppression
linked to human LC3B. The HiBiT peptide binds with high affinity to the luciferase of autophagy, which prevents hypoglycaemia under
subunit LgBiT, producing active luciferase, which generates luminescence proportional physiological fasting conditions.
to the amount of autophagy in the presence of furimazine. The amount of LC3 reporter Pancreatic β-​cells from patients with T2DM exhibit
within the cell can be measured using a microplate reader. more abundant autophagic vacuoles and autophago-
Keima is a coral-​derived acid-​stable fluorescent protein. Fluorescence microscopy somes and have lower expression of lysosomal genes
detects ionized Keima within lysosomes as a red fluorescent signal at pH <7.0 and than do β-​cells from people without diabetes, which
neutral Keima in the cytoplasm as a green fluorescent signal at pH ≥7.0. Delivery of suggests that T2DM causes alterations at the level of
cytosolic Keima to the lysosome reflects non-​selective macroautophagy, whereas autophagic structure removal85. Indeed, in cultured
delivery of Keima fused to a specific (for example, mitochondrial) protein reflects
β cells, simulation of glucolipotoxicity via treatment
selective autophagy (mitophagy).
Several transgenic mouse models have been engineered to express tandem fluorescent with palmitate and high glucose levels suppresses auto-
reporters, such as GFP–LC3 (systemic and cardiac-​specific models), mCherry–LC3 phagy via decreased lysosome-​related gene expression,
(cardiac-​specific model), mCherry–GFP–LC3 (cardiac-​specific model), monomeric impaired autophagosome–lysosome fusion or decreased
RFP–GFP–LC3 (systemic and cardiac-​specific models), GFP–LC3–RFP–LC3∆G lysosomal acidification86–89. In these β-​cell models,
(systemic model), and mitochondria-​targeted Keima (systemic model). pharmacological lysosome reacidification or autophagy
induction restores autophagic flux even under lipotoxic
models of genetically altered autophagic function. conditions90, whereas blocking autophagy exacerbates
Although Atg7+/− mice show no metabolic abnorma­ lipotoxicity91. Moreover, simultaneous activation of
lities, Atg7+/−ob/ob mice and Atg7+/− mice fed a high-​fat autophagy (by intracellular stresses, such as ER stress)
diet exhibit insulin resistance, increased lipid storage and blockade of autophagic flux causes the accumula-
and elevated inflammation in the liver, skeletal tion of defective lysosomes, which leads to the induction
muscle and adipose tissue79. In addition, Atg4b–/– mice of β-​cell death89. Thus, glucolipotoxicity is involved in
(which have limited autophagic competence) exhibit the β-​c ell dysfunction mediated by dysregulated
a considerable increase in body weight, expanded autophagic flux; however, the detailed mechanism
adipocytes in their visceral fat, increased liver steatosis underlying this process remains unknown.
and insulin intolerance upon metabolic challenge with In addition, ER stress contributes to the production
either a high-fat diet or sugar-​enriched drinking water80. of human islet amyloid polypeptide (hIAPP), which
Moreover, Atg4b–/– mice have increased vulnerability forms amyloid aggregates associated with β-​cell death
to (and increased severity of) streptozotocin-​induced and with the development of T2DM92. Several studies
T1DM owing to their compromised pancreatic β-​cell have demonstrated that autophagy is essential to protect
homeostasis80. Autophagy-​inducing treatment rescues β-​cells from toxic hIAPP oligomer accumulation93,94.
the diabetic pheno­types of these mice79. By contrast,
Atg5-​overexpressing lean mice exhibit increased insulin Liver function
sensitivity, resistance to oxidative stress and enhanced The liver is one of the most metabolically active organs
motor function alongside their extended lifespan73. and controls both glucose and lipid metabolism.
Thus, autophagy at levels insufficient to prevent the Non-​a lcoholic fatty liver disease (NAFLD) caused

www.nature.com/nrendo

0123456789();:
Reviews

a by hepatic lipid accumulation due to overnutrition is


• Intracellular stress • Hypoxia
• Mitochondrial dysfunction • ER stress responsible for hepatic insulin resistance and contri­
• Oxidative stress • Change in nutrient status butes to T2DM95. Hepatic autophagy is now known to
• Inflammation play a crucial role in the regulation of glucose and/or
lipid metabolism, insulin resistance and energy balance
in the whole body.
In mouse models of obesity or diabetes (such as db/db
Level of autophagy or ob/ob mice, and mice fed a high-​fat diet96) liver cells
exhibit decreased autophagic flux similar to that in
Impaired (insufficient) Basal autophagy or adequate adaptive responses Excessive humans with NAFLD97. Mice with conditional knockout
of Atg7 in hepatocytes exhibit accumulations of swollen
and deformed mitochondria and an increased number
of ubiquitylated protein aggregates and lipid droplets72.
Experiments in mice with hepatocyte-​specific knockout
of Atg7 or cultured mouse hepatocytes with knock­
down of Atg7 or Atg5 expression have revealed that
inhibition of autophagy increases triglyceride storage
↑ Damaged organelles ↓ Clearance of damaged or ↑ Cell death in lipid droplets98. In addition, mice with hepatocyte-
↑ Protein aggregates excess cellular components specific knockout of Vps34 develop hepatomegaly and
↑ Lipid droplets
hepatosteatosis along with impaired protein turnover99.
Moreover, another study demonstrated that small hair-
• Maintenance of pin (sh)RNA-​mediated knockdown of liver Atg7 in mice
• Cellular dysfunction cellular homeostasis
↑ Senescence ↓ Senescence results in insulin resistance, glucose intolerance and ER
stress96. Conversely, restoration of Atg7 expression via
adeno­viral gene delivery in the livers of ob/ob mice or
b mice fed a high-​fat diet improves whole-​body insulin
Overnutrition
↑ Insulin–IGF-I signalling Starvation
sensitivity through the suppression of hepatic glucose
↑ Intracellular amino acids Energy depletion production and enhancement of insulin-​stimulated
glucose disposal in the periphery96.
Transcription factor EB (TFEB) is a member
↑ mTORC1 ↓ mTORC1 of the basic helix–loop–helix leucine-​zipper family of

Fig. 3 | Effects of autophagy on cellular senescence and


regulation of autophagy by nutrient-sensing pathways.
↓ Autophagy ↑ Autophagy
a | Intracellular stresses, including mitochondrial dys­
function, oxidative stress, inflammation, hypoxia, endo-
plasmic reticulum (ER) stress and changes in nutrient
↓ LKB1 ↑ LKB1 status regulate autophagy. Basal autophagy and adequate
↓ AMPK ↓ SIRT1 ↑ AMPK ↑ SIRT1 adaptive autophagy responses promote homeostasis at
↓ NAD+ ↑ NAD+ the cell and organelle levels, resulting in protection from
senescence and prolonged cell survival. Inadequate or
impaired autophagy leads to cellular dysfunction and
c Overnutrition Starvation senescence owing to the accumulation of damaged
TFEB organelles, protein aggregates or lipid droplets, whereas
mTORC1 TFE3 TFEB excessive autophagy leads to reduced cell survival or
TFE3 P mTORC1 P P cell death. b | Excess nutrition decreases autophagy via
mechanistic target of rapamycin complex 1 (mTORC1)
activation and reduces the function of AMP-​activated
Lysosome Cytosol
kinase (AMPK) and sirtuin-1 (SIRT1). By contrast, starvation
or energy depletion induces autophagy via reduced
mTORC1 activity and reduced activation of AMPK and
SIRT1. Crosstalk between nutrient-​sensing pathways that
↑ Lysosomal ↓ Lysosomal converge on mTORC1, AMPK and SIRT1 is mediated by
component genes component genes changes in NAD+ levels or in the activity of liver kinase B1
Nucleus ↑ Autophagy- ↓ Autophagy-
related genes related genes (LKB1). c | The transcription factors TFEB and TFE3 control
TFEB lysosomal numbers and function by positively regulating
TFE3 genes in the coordinated lysosomal expression and regula-
Promoters of Promoters of
tion (CLEAR) network. In overnutrition conditions, TFEB and
CLEAR-related genes CLEAR-related genes TFE3 translocate to the nucleus and drive the expression of
CLEAR-​related genes (which encode autophagy-​related
and lysosomal proteins), resulting in increased lysosomal
biogenesis and increased autophagic flux. By contrast,
under starvation conditions these transcription factors
↑ Autophagic flux ↓ Autophagic flux
remain phosphorylated and inactive in the cytosol.
IGF-​I, insulin-​like growth factor-1.

Nature Reviews | Endocrinology

0123456789();:
Reviews

Table 2 | regulation of autophagy via nutrient-​sensing pathways


Target molecules and phosphorylation site(s) Function Effect on autophagy refs
mTORC1
ATG13 phosphorylation at Ser258 Decreased ULK1 complex activity Decreased initiation 22

ULK1 phosphorylation at Ser637 and Ser757 Decreased ULK1 complex activity Decreased initiation 23,24

ATG14 phosphorylation at Ser3, Ser223, Thr303 Decreased PI3KC3-​CI activity Decreased nucleation 25

and Ser440
AMBRA1 phosphorylation at Ser52 Decreased PI3KC3-​CI activity Decreased nucleation 26

NRBF2 phosphorylation at Ser113 and Ser120 Decreased PI3KC3-​CI activity Decreased nucleation 15

Decreased ULK1-​mediated ATG14 phosphorylation Decreased PI3KC3-​CI activity Decreased nucleation 28

at Ser29
Phosphorylation of p300 at Ser2271, Ser2279, Decreased LC3 acetylation leading to Decreased phagophore elongation; 29

Ser2291 and Ser3375 decreased LC3–ATG7 interaction; decreased decreased nucleation


VPS34 acetylation leading to decreased
PI3KC3-​CI activity
WIPI2 phosphorylation at Ser395 Degraded by HUWE1 Decreased phagophore elongation 30

UVRAG phosphorylation at Ser498 Blocks UVRAG–rubicon interaction leading Decreased autolysosome formation 31

to decreased PI3KC3-​CII activity


UVRAG phosphorylation at Ser550 and Ser571 Increased VPS34 and increased PI3P at the Increased lysosome re-​formation 32

lysosome
Pacer phosphorylation at Ser157 Decreased HOPS–STX17 interaction Decreased autolysosome formation 33

TFEB phosphorylation at Ser211, Ser122 and Decreased translocation into the nucleus Downregulation of genes involved in 34–36

Ser142 lysosomal biogenesis and autophagy


TFE3 phosphorylation at Ser321 Decreased translocation into the nucleus Downregulation of genes involved in 37–39

lysosomal biogenesis and autophagy


AMPK
TSC2 phosphorylation at Thr1227 and Ser1345 Decreased mTORC1 activation Increased flux 40

Raptor phosphorylation at Ser722 and Ser792 Decreased mTORC1 activation Increased flux 41,42

ULK1 phosphorylation at Ser467 , Ser555, Ser574, Increased ULK1 complex activity Increased initiation 43–46

Ser637 and Thr574


ULK1 phosphorylation at Ser317 and Ser777 Increased ULK1 complex activity Increased initiation 23

ULK1 phosphorylation at Ser224 Decreased ULK1 complex activity Decreased initiation 22

ATG9 phosphorylation at Ser761 Increased ATG9-​containing vesicles Increased phagophore elongation 53

VPS34 phosphorylation at Thr163 and Ser165 Non-​autophagic (in the absence of ATG14); Increased nucleation 52

decreased PI3KC3-​CI activity


Beclin-1 phosphorylation at Ser91 and Ser94 Increased PI3KC3-​CI activity Increased nucleation 52

DAPK2 phosphorylation at Ser289 Decreased DAPK2-​mediated Increased nucleation 48

phosphorylation of beclin-1 at Thr119


PAQR3 phosphorylation at Thr32 Increased PI3KC3-​CI activity Increased nucleation 49

RACK1 phosphorylation at Thr50 Increased PI3KC3-​CI activity Increased nucleation 50

SIRT1
ATG5, ATG7 and decreased LC3 acetylation Increased ATG12 conjugation Increased nucleation and phagophore 54,55

elongation
Decreased FOXO1 acetylation Increased Rab-7a levels Increased autolysosome formation 56

Decreased FOXO3 acetylation Increased BNIP3 and NIX levels Increased mitophagy 57

AMBRA1, activating molecule in beclin 1-​regulated autophagy protein 1; AMPK, AMP-​activated kinase; ATG, autophagy related gene; BNIP3, BCL2 and adenovirus
E1B 19kDa-​interacting protein 3; DAPK2, death-​associated protein kinase 2; FOXO, forkhead box O; HOPS, homotypic fusion and protein sorting; HUWE1, E3
ubiquitin ligase; LC3, microtubule-​associated protein 1A/1B-​light chain 3; NRBF2, nuclear receptor binding factor 2; PAQR3, progestin and adipoQ receptor family
member 3; PI3KC3-​C, class III phosphatidylinositol 3-​kinase-​complex; PI3P, phosphatidylinositol-3-​phosphate; Rab-7a, Ras-​related protein; RACK1, receptor of
activated protein C kinase 1; SIRT1, sirtuin-1; STX17 , syntaxin-17; TFE3, transcription factor E3; TFEB, transcription factor EB; TSC2, tuberous sclerosis complex 2;
ULK1, UNC-​like autophagy activating kinase 1; UVRAG, ultraviolet (UV) irradiation resistance-​associated gene; VPS34, vacuolar protein sorting 34; WIPI2, WD-​repeat
domain effector protein 2.

transcription factors. TFEB controls lysosomal bio- starvation conditions, TFEB translocates to the nucleus
genesis and autophagy by regulating the expression where it induces the expression of lysosomal component
of TFEB target genes in the coordinated lysosomal genes and ATGs, resulting in new lysosome generation
expression and regulation (CLEAR) network66. Under and increased autophagic flux. Livers from mice with

www.nature.com/nrendo

0123456789();:
Reviews

liver-​specific knockout of Tfeb are large and pale, and attenuates obesity in mice fed a high-​fat diet and the
their hepatocytes are filled with lipid vacuoles, consis­ metabolic syndrome phenotype in ob/ob mice 100.
tent with impairment of lipid degradation pathways100. Similarly to TFEB, TFE3 is another transcription factor
By contrast, livers from mice fed a high-​fat diet that were that activates lysosomal biogenesis and autophagy.
injected with an adenoviral expression vector including The presence of rubicon protein, which inhibits
human TFEB have a normal dark-​red colour and autophagosome and lysosome fusion, is also thought
markedly less lipid accumulation than do livers from to be involved in the pathogenesis of NAFLD. In mice
wild-​t ype controls 100. Liver overexpression of Tfeb with hepatocyte-​specific knockout of Rubcn, autophagy
suppression is abolished. Moreover, in Rubcn-​knockout
mice fed a high-​fat diet, hepatotoxicity, lipid droplet
↑ Nutrients Obesity ↓ Exercise
accumulation and hepatic steatosis are all attenuated101.
In addition, expression of Rubcn is increased in cultured
↑ Ectopic and visceral fat accumulation mouse hepatocytes after treatment with palmitate and in
human hepatocytes from patients with NAFLD101. These
Lipotoxicity
results suggest that the pathophysiology of NAFLD
is related to suppression of autophagic flux, which is
mediated by increased expression of RUBCN.
Oxidative stress, inflammation, ER stress
Adipose tissue function
Basal autophagy plays an important part in the regula-
tion of adipocyte development and differentiation. The
Normal autophagy role of basal autophagy in adult adipocytes has been elu-
response
Impaired autophagy cidated using mice in which autophagy has been geneti­
response cally altered. Mice with adipocyte-​specific knockout of
Atg7 exhibit relatively small lipid deposits throughout
Glucotoxicity ↑ Damaged organelles, lipid droplets, protein aggregates the whole body, multilocular lipid droplets in adipocytes
and an increased number of mitochondria102. Ablation
of autophagy in white adipose tissue induces adipocyte
Impaired cellular homeostasis browning, which has several results: increased energy
expenditure; increased insulin sensitivity; reduced levels
of adipokines, such as leptin; and a lean and obesity-
resistant phenotype. However, the loss of normal lipid
β-cell Liver Adipose tissue Skeletal muscle
↑ Fat Adipocyte ↓ Muscle mass deposits leads to increased perinatal and postnatal
↓ Insulin
secretion deposition dysfunction ↓ Muscle strength mortality, possibly because lipid storage is insufficient
to meet the energy demands associated with acute
stress. When essential autophagy genes (such as Atg7)
NAFLD Sarcopenia, are subjected to conditional knockout, the resulting
sarcopenic obesity
adipocytes are dysfunctional and tend to lose their lipid
storage and endocrine capabilities. Therefore, basal auto-
phagy is essential for maintaining adipocyte function.
By contrast, Atg4b–/– mice (which show partial auto-
phagic insufficiency) fed a calorie-​rich diet exhibit an
obese phenotype accompanied by large adipocytes80, as
↑ Insulin resistance, described above. This finding indicates that an adaptive
T2DM response of autophagy to metabolic stresses is required
for the suppression of obesity and metabolic diseases.
Fig. 4 | relationships between obesity, insulin resistance, T2dM and autophagy.
With ageing, adipose tissue undergoes substantial
Excessive nutrition and insufficient exercise lead to obesity with ectopic and visceral
fat accumulation, which result in lipotoxicity, oxidative stress, inflammation and endo- changes in abundance, distribution, cellular composi-
plasmic reticulum (ER) stress. Under lipotoxic conditions, cellular homeostasis and tion and endocrine signalling that are associated with
autophagy are impaired and lead to organelle damage, lipid droplet accumulation the development of insulin resistance and metabolic
and protein aggregation in pancreatic β-​cells, liver tissue, adipose tissue and skeletal dysfunction103. Autophagy in subcutaneous and visceral
muscle. As a result, β-​cells exhibit reduced insulin secretion, and increased lipid adipose tissue is upregulated in patients with obesity or
deposition is observed in the liver and is associated with non-​alcoholic fatty liver disease T2DM104–107. In addition, the expression of RUBCN in
(NAFLD). An impaired autophagy response also contributes to adipocyte dysfunction, adipose tissue decreases with age, resulting in increased
such as excess or reduced lipid storage or abnormal adipokine secretion. In skeletal autophagic activity. Mice with adipocyte-​specific knock-
muscle, impaired autophagy contributes to reductions in muscle mass and strength and out of Rubcn exhibit fat atrophy, glucose intolerance,
impaired muscle quality, leading to the development of sarcopenia (sarcopenic obesity).
dyslipidaemia and hepatic fat accumulation related to
Consequently, functional impairment of β-​cells, liver tissue, adipose tissue and skeletal
muscle causes insulin resistance and type 2 diabetes mellitus (T2DM). Glucotoxicity due decreased lipid storage capacity and reduced endocrine
to continuous high glucose levels further exacerbates the oxidative stress, inflammation activity of adipokines such as adiponectin108. Thus,
and ER stress resulting from lipotoxicity. In addition, ageing is involved in both rubicon has an essential role in the maintenance of
impairment of autophagy and increased intracellular stress, and impaired autophagy adipo­cyte function and systemic metabolic homoeostasis
promotes the development and exacerbation of insulin resistance and T2DM. by preventing excessive autophagy.

Nature Reviews | Endocrinology

0123456789();:
Reviews

Skeletal muscle function senescent state. Regarding this point, basal autophagy
Skeletal muscle has an important role in glucose meta­ is essential for maintenance of the quiescent state in
bolism; therefore, the decreased skeletal muscle mass mouse muscle stem cells122. Dysfunctional autophagy
and strength associated with sarcopenia can increase in aged satellite cells or genetic ablation of autophagy in
the risk of insulin resistance and T2DM, particularly in young satellite cells induces senescence owing to the loss
older people109,110. Insulin resistance in skeletal muscle is of proteostasis as well as increased mitochondrial dys-
closely related to reduced muscle mass111 because insulin function and oxidative stress, which result in declines in
and insulin-​like growth factor-1 (IGF-​I) signalling are the function and number of satellite cells. Restoration of
both crucial for protein synthesis in muscle112,113. Thus, autophagy restores the regenerative function of senescent
an appropriate balance between anabolic and catabolic satellite cells from older individuals122.
states, which is driven by nutrient intake and nutrient-
sensing pathways, is important for maintaining muscle Therapeutic interventions
mass. In addition, obesity and sarcopenia often coexist Calorie restriction
and sarcopenic obesity is recognized to worsen several Calorie restriction supports longevity and aids in the
metabolic diseases, including T2DM3. treatment of age-​related diseases123. Autophagy mediates
The role of basal autophagy in skeletal muscle main- the beneficial effect of calorie restriction on lifespan, and
tenance has been shown in mice with skeletal muscle- inhibition of autophagy largely mitigates its longevity‐
specific knockout of Atg7, which exhibit pheno­types enhancing effects124,125. Humans experiencing long-​term
that include accumulation of abnormal mitochondria moderate calorie restriction (a ~30% reduction com-
and protein aggregates, abnormalities in motor neuron pared with the typical Western diet, for an average of
synapses, muscle atrophy and age-​dependent decreases 9.6 years) show increased transcript levels of various auto-
in muscle strength114,115. In addition, autophagy becomes phagy modulators, including AMPK and sirtuin family
progressively more dysfunctional in the muscles of age- members, as well as downregulation of the insulin–IGF-1
ing rodents and in older individuals with sarcopenia or signalling pathway in skeletal muscle compared to age‐
frailty51,115. Moreover, among elderly people, those who matched controls126. Furthermore, the altered skeletal
are overweight exhibit increased dysregulation of seve­ muscle transcriptional profiles induced by calorie restric-
ral autophagic processes, including autophagosome– tion resembled those of younger control individuals126.
lysosome fusion, in skeletal muscle116. The relationship A separate study demonstrated that ATGs, including
between insulin resistance and impaired autophagy in ULK1 and BECN1, are expressed at higher levels in the
skeletal muscle has been demonstrated in humans and skeletal muscle of people eating a calorie-​restricted diet for
in vitro studies. For example, expression of ATGs is 3–15 years than in the skeletal muscle of age-​matched
decreased in skeletal muscle samples from patients with sedentary individuals eating a typical Western diet127.
T2DM who require more than 100 U of insulin daily, and In animal models of obesity and T2DM (namely, db/db
this decrease is associated with abnormal mitochondrial mice and mice fed a high-​fat diet), calorie restriction
morphology and impaired function in skeletal muscle117. alleviates hepatosteatosis and improves β-​cell function,
Moreover, palmitate-​induced cellular senescence and blood glucose levels and lipid profiles; these changes are
insulin resistance in cultured muscle cells, including associated with improvements in autophagy128–130.
myoblasts and myotubes, are mediated by impairment In addition, modified forms of calorie restriction
of autophagic flux118. (such as intermittent fasting, isocaloric twice-​a-​day
Insufficient or impaired mitophagy might result in the (ITAD) feeding or inter-​meal fasting) can activate auto-
accumulation of damaged mitochondria in skeletal mus- phagy, thereby contributing to improvements in blood
cle during ageing119. In addition, increased lipid deposi- glucose levels and lipid metabolic profiles. Intermittent
tion in skeletal muscle cells can causes an age-​associated fasting preserves organelle quality via the autophagy–
decline in mitochondrial function. As lipophagy has lysosome pathway and enhances β-​cell survival in
an important role in degradation of lipid droplets in individuals with obesity-​induced diabetes131. ITAD
skeletal muscle cells, impaired lipophagy might lead feeding also prevents metabolic defects associated
to the accumulation of lipid droplets and ultimately to with both obesity and ageing, including glucose intole­
lipotoxicity in skeletal muscle120. Thus, impairments in rance and hypertriglyceridaemia, and reduces maxi-
selective autophagy, such as mitophagy and lipophagy, mal oxygen consumption in mice fed a high-​fat diet132.
in skeletal muscle could be involved in the pathogenesis Finally, Roux-​en-​Y gastric bypass rapidly reduces hepatic
of age-​associated insulin resistance and T2DM. However, lipid toxicity and attenuates insulin sensitivity in obese
no direct evidence of reduced mitophagy and lipophagy rats with T2DM133,134. In these rats, activation of hepatic
in skeletal muscle in older humans is available so far; autophagy plays an important part in long-​term control
further studies are necessary to confirm this hypothesis. of hepatic lipid metabolism, which might be related
Because of the large energy demand associated to inhibition of the mTORC1 signalling pathway and
with stellate cell activation, autophagic flux is required upregulation of glucagon-​like peptide-1 (GLP-1)133,134.
for quie­scent stellate cells (also termed muscle stem
cells) to be activated, proliferate and differentiate into Exercise
myoblasts121. Thus, suppression of autophagic flux delays The beneficial effects of exercise include extension of
muscle fibre regeneration. During ageing, the regenera­ lifespan, attenuation of metabolic disease and mitiga-
tive capacity of muscle stem cells declines, which is tion of sarcopenia. Acute endurance exercise increases
associated with switching of the quiescent state into a autophagic activity in the heart, liver, β-​c ells and

www.nature.com/nrendo

0123456789();:
Reviews

adipose tissue in mice135. However, an autophagy‐defi- GLP-1 receptor agonists and DPP-4 inhibitors. GLP-1
cient mouse model that contains knock-​in mutations at facilitates autophagy by preventing the impairment of
three BCL2 phosphorylation sites (Thr69Ala, Ser70Ala autophagosome–lysosome fusion that would other­
and Ser84Ala) does not obtain these exercise-​mediated wise occur under conditions of chronic nutrient
benefits and is not protected from glucose intolerance excess. Numerous studies conducted in animal models
induced by a high-​fat diet135. In addition, mice lacking of diabetes and obesity or in INS-1 cells (an insulin-
collagen VI, another model of compromised autophagy, secreting pancreatic β-​cell line) have shown that the
are exercise-​intolerant and develop an exacerbated protective effects of GLP-1 receptor agonists (including
dystrophic phenotype upon acute and chronic exercise136. liraglutide and exendin-4) against glucolipotoxicity in
Thus, autophagic regulation plays a crucial role in the β-​cells are mediated in part by induction of autophagy via
maintenance of skeletal muscle mass and in adaptation either AMPK and forkhead box O1 (FOXO1) activation
to cellular damage in response to exercise. or restoration of lysosome function89,149–155. Liraglutide
Chronic exercise leads to upregulation of auto- attenuates hepatosteatosis by inducing autophagy via the
phagy not only in skeletal muscle137,138 but also in the AMPK and mTORC1 pathways156,157. In addition, GLP-1,
liver, and this mechanism is involved in amelioration of exendin-4 and liraglutide protect hepatocytes from free
hepatosteatosis139–141. Long-​term adaptations to exercise fatty acid-​related death by prohibiting a dysfunctional
that are potentiated by exercise-​induced autophagy are ER stress response and by reducing fat accumulation
mediated by mild oxidative stress, energetic imbalance, and apoptosis via activation of both macroautophagy and
mitochondrial calcium accumulation and misfolded chaperone-​mediated autophagy158. Moreover, liraglutide
proteins. Exercise-​induced autophagy leads to improve- attenuates hepatosteatosis by restoring autophagic
ments in mitochondrial quality control and protein turn- flux, specifically flux through the GLP-1 receptor and
over as well as metabolic adaptations and angiogenesis, TFEB-​mediated lysosomal autophagy pathway159.
which collectively result in enhanced endurance and Dipeptidyl peptidase-4 (DPP-4) inhibitors can induce
improvements in both glucose and lipid homeostasis. endogenous GLP-1 or block its breakdown, thereby
The beneficial effects of calorie restriction and increasing the levels of this incretin hormone. The
exercise are mediated by shared pathways involved in DPP-4 inhibitor MK-626 restores insulin secretion by
the regu­lation of autophagy, including nutrient-​sensing enhancing autophagy in mice fed a high-​fat diet through
pathways. Accordingly, the benefits of exercise might be upregulation of GLP-1 (ref.160). Long‐term treatment
related to autophagy-​activating mechanisms that also with alogliptin, another DPP-4 inhibitor, improves the
underlie the positive effects of calorie restriction on survival and health of such mice through autophagy
lifespan and age‐related diseases142,143. However, auto­ induction, the mechanism of which is dependent on the
phagy induction is differentially affected by exercise SIRT1–AMPK–mTORC1 cascade161. Moreover, aloglip-
training modes, such as resistance exercise and endurance tin has other beneficial effects associated with increased
exercise, and also by different nutrient intake conditions. longevity; for example, this agent increases insulin sensi­
tivity, attenuates the functional decline of pancreatic
Pharmacological interventions β-​cells, decreases organ pathology in the liver, bone and
Antidiabetic medicines, including metformin, GLP-1 vasculature, preserves hepatic mitochondrial function
receptor agonists and sodium–glucose co-​transporter 2 and reduces oxidative stress161. However, whether inhibi-
(SGLT2) inhibitors can show beneficial effects on glucose tion of DPP-4 itself or other factors, including increased
and lipid metabolism through modulation of autophagy. incretin levels, contribute to longevity remains unclear.
In addition, rapamycin, polyphenols and spermidine Interestingly, IL-6 regulates GLP-1 production by
can extend lifespan through induction of autophagy. pancreatic α-​cells162 and is also able to stimulate β-​cell
autophagy, which protects β-​cells against apoptosis
Metformin. Metformin has strong positive effects on fun- under conditions of metabolic stress163. However, the
damental anti-​ageing mechanisms144, and a previous study regulation of autophagy by IL-6 in β-​cells is specific to
showed lifespan-​extending and health-​extending effects this cell type and differs from that in other tissues.
of metformin in middle-​aged male mice145. Several studies
have demonstrated that metformin is involved in the regu­ SGLT2 inhibitors. In addition to its glucose-​lowering
lation of glucose and lipid metabolism by autophagy in role, SGLT2 inhibitor treatment has multiple beneficial
the liver, adipose tissue and pancreatic β-​cells. For exam- consequences, including cardioprotective and renopro-
ple, metformin attenuates hepatosteatosis via SIRT1 acti- tective effects as well as hepatosteatosis-​ameliorating
vation in ob/ob mice146 and via AMPK activation in mice effects. As one of the mechanisms by which SGLT2
fed a high-​fat diet147, although metformin also inhibits inhibitors exert organ-​protective effects, SGLT2 inhibi­
autophagy in epididymal adipose tissue147. Furthermore, tion is expected to induce autophagy through the modu­
metformin attenuates systemic oxidative stress, visceral lation of nutrient-​s ensing pathways, including the
adipose tissue inflammasome expression and auto- activation of AMPK or SIRT1 resulting from continuous
phagy in patients with T2DM with obesity107. Thus, energy loss due to increased excretion of glucose into
metformin affects the regulation of autophagy in a tissue-​ the urine164,165. Indeed, several studies have shown that
dependent fashion, and these tissue-​specific responses to SGLT2 inhibitors exert renoprotective effects by alleviat-
metformin are modulated by obesity. In addition, met- ing autophagic flux impairment in proximal tubular cells
formin stimulates β-​cell autophagy and prevents β-​cell in obese mice fed a high-​fat or high-​sucrose diet and in
apoptosis under conditions of lipotoxicity in vitro148. mice with streptozotocin-​induced diabetes166. Moreover,

Nature Reviews | Endocrinology

0123456789();:
Reviews

the cardioprotective and anti-​inflammatory effects of ACBP functions as a neuroendocrine factor, such that
SGLT2 inhibitors are partially exerted through auto- its increased levels promote feeding behaviour, conse-
phagy induction167,168. However, whether SGLT2 inhibi- quently mitigating starvation and nutrient deprivation,
tion improves longevity through autophagy induction which are the cause of autophagy induction187. At the
remains unclear. same time, the autophagy-​driven release of ACBP from
cells results in depletion of intracellular ACBP. The
Rapamycin. Rapamycin treatment extends lifespan in depletion of intracellular ACBP and increased extracel-
mice169,170 and promotes cardioprotection171 and pre- lular concentration of ACBP both cause feedback inhibi­
vents obesity172 in rodents. However, prolonged rapa- tion of autophagy in a cell-​autonomous fashion and a
mycin treatment exacerbates insulin resistance and paracrine fashion, respectively186.
diabetes173 and actually reduces the lifespan of diabetic Paradoxically, obese mice fed a high-​fat diet and ob/ob
mice174. An intermittent treatment regimen that avoids mice exhibit elevated expression of the gene encoding
these adverse effects of continuous rapamycin has been ACBP in white adipose tissue and the liver as well as
found to extend the lifespan of aged female mice175. increased levels of ACBP in the circulation186. In these
In addition, rapamycin attenuates insulin resistance and mice, starvation causes a decrease from baseline in
hepatosteatosis in rats with T2DM through activation expression of the gene encoding ACBP in white adipose
of autophagy176. However, whether rapamycin treatment tissue and the liver186. In humans, BMI is strongly and
also improves longevity through autophagy induction is positively correlated with circulating levels of ACBP;
remains unclear, and further studies are necessary to individuals with anorexia nervosa have abnormally
address this point. low circulating levels of this protein186. In addition, in
mice, administration of recombinant ACBP protein or
Spermidine. Spermidine, a natural polyamine whose transgenic overexpression of the gene encoding ACBP
intracellular concentration declines during human in the liver induces hyperphagia and lipoanabolic reac-
ageing, has been found to increase the life expectancy tions along with suppression of autophagy, consequently
of yeast, C. elegans, Drosophila and mice. Enhanced leading to obesity186. By contrast, antibody-​mediated
autophagy contributes to lifespan extension by sper­ neutralization of ACBP reduces food intake and pro-
midine 177–180. In addition, autophagy induction by moted lipo-​catabolic reactions, thereby reducing fat
spermidine protects against the detrimental effects of mass186. These data indicate that ACBP is a pro-​obesity
a high-​fat diet, including weight gain, glucose intole­ factor that promotes hyperphagia. Therefore, ACBP
rance, increased adipocyte size and hepatosteatosis, could be a useful therapeutic target in the prevention
in wild-​type mice80. or treatment of obesity and its comorbidities. However,
the relationship between altered autophagic activity
Polyphenols. Several polyphenols derived from natural and the role of ACBP in the pathogenesis of obesity
sources have been shown to act as autophagy inducers. remains unclear.
Resveratrol, which activates SIRT1 (ref.181), has been
shown to increase lifespan in C. elegans via an auto- Conclusions
phagy‐dependent mechanism182 and to exhibit health‐ Impairment of autophagy results in diverse types of
promoting effects, especially in obese overfed mice183. cellular dysfunction that exacerbate the ageing process,
3,4‐Dimethoxychalcone promotes the nuclear translo- whereas enhancement of autophagy generally promotes
cation of autophagy-​promoting transcription factors, cellular homeostasis and functions to prolong lifespan
including TFEB, resulting in autophagy induction and and improve metabolic health. As reduced autophagy
cardioprotective and anticancer effects in mice184. In addi- is implicated in numerous age-​related diseases, such as
tion, 4,4′-​dimethoxychalcone administration extends metabolic disease and sarcopenia, therapeutic upregu­
lifespan in yeast, C. elegans and Drosophila; retards the lation of autophagy to adequate levels is a potential
development of senescence in cultured cells; and pro- treatment strategy for these diseases. However, the role
tects mice from prolonged myocardial ischaemia185. of autophagy in cellular physiology differs according
The activation of autophagy by 4,4′-​dimethoxychalcone to the cell type and pathological condition. In addi-
depends on inhibition of GATA transcription factors but tion, no established non-​invasive methods are capa-
is independent of mTORC1 (ref.185). ble of detecting autophagic activity in human tissues.
To enable evaluation of the effects of drug treatment on
ACBP blockers. Acyl coenzyme A-​binding protein autophagy in humans, further studies are necessary to
(ACBP) has been identified as an appetite stimula- establish non-​invasive methods for autophagy measure-
tor. Physiologically, starvation causes the autophagy- ment, including methods for analysis of biomarkers in
dependent release of ACBP from many mammalian serum and/or urine samples.
cell types, including adipocytes and hepato­cytes, and
an increase in circulating levels of ACBP186. Circulating Published online xx xx xxxx

1. Saeedi, P. et al. Global and regional diabetes 2. Kuk, J. L., Saunders, T. J., Davidson, L. E. & Ross, R. 4. Li, N. et al. Aging and stress induced β cell senescence
prevalence estimates for 2019 and projections Age-​related changes in total and regional fat and its implication in diabetes development. Aging 11,
for 2030 and 2045: results from the International distribution. Ageing Res. Rev. 8, 339–348 (2009). 9947–9959 (2019).
Diabetes Federation Diabetes Atlas, 9th edition. 3. Batsis, J. A. & Villareal, D. T. Sarcopenic obesity in 5. Lopez-​Otin, C., Blasco, M. A., Partridge, L., Serrano, M.
Diabetes Res. Clin. Pract. 157, 107843 older adults: aetiology, epidemiology and treatment & Kroemer, G. The hallmarks of aging. Cell 153,
(2019). strategies. Nat. Rev. Endocrinol. 14, 513–537 (2018). 1194–1217 (2013).

www.nature.com/nrendo

0123456789();:
Reviews

6. Choi, A. M., Ryter, S. W. & Levine, B. Autophagy in autophagosome maturation and lipid metabolism. 60. Cantó, C. et al. AMPK regulates energy expenditure
human health and disease. N. Engl. J. Med. 368, Mol. Cell 73, 788–802.e7 (2019). by modulating NAD+ metabolism and SIRT1 activity.
651–662 (2013). 34. Roczniak-​Ferguson, A. et al. The transcription factor Nature 458, 1056–1060 (2009).
7. Hurley, J. H. & Young, L. N. Mechanisms of autophagy TFEB links mTORC1 signaling to transcriptional 61. Takeda-​Watanabe, A., Kitada, M., Kanasaki, K. &
initiation. Annu. Rev. Biochem. 86, 225–244 (2017). control of lysosome homeostasis. Sci. Signal. 5, ra42 Koya, D. SIRT1 inactivation induces inflammation
8. Dikic, I. & Elazar, Z. Mechanism and medical (2012). through the dysregulation of autophagy in human
implications of mammalian autophagy. Nat. Rev. 35. Martina, J. A., Chen, Y., Gucek, M. & Puertollano, R. THP-1 cells. Biochem. Biophys. Res. Commun. 427,
Mol. Cell Biol. 19, 349–364 (2018). mTORC1 functions as a transcriptional regulator of 191–196 (2012).
9. Nakatogawa, H., Ishii, J., Asai, E. & Ohsumi, Y. Atg4 autophagy by preventing nuclear transport of TFEB. 62. Kume, S., Thomas, M. C. & Koya, D. Nutrient sensing,
recycles inappropriately lipidated Atg8 to promote Autophagy 8, 903–914 (2012). autophagy, and diabetic nephropathy. Diabetes 61,
autophagosome biogenesis. Autophagy 8, 177–186 36. Martina, J. A. et al. The nutrient-​responsive 23–29 (2012).
(2012). transcription factor TFE3 promotes autophagy, 63. Filomeni, G., De Zio, D. & Cecconi, F. Oxidative
10. Melia, T. J., Lystad, A. H. & Simonsen, A. lysosomal biogenesis, and clearance of cellular stress and autophagy: the clash between damage
Autophagosome biogenesis: from membrane debris. Sci. Signal. 7, ra9 (2014). and metabolic needs. Cell Death Differ. 22, 377–388
growth to closure. J. Cell Biol. 219, e202002085 37. Vega-​Rubin-de-​Celis, S., Peña-​Llopis, S., Konda, M. (2015).
(2020). & Brugarolas, J. Multistep regulation of TFEB by 64. Rashid, H. O., Yadav, R. K., Kim, H. R. & Chae, H. J.
11. Lawrence, R. E. & Zoncu, R. The lysosome as a cellular mTORC1. Autophagy 13, 464–472 (2017). ER stress: autophagy induction, inhibition and
centre for signalling, metabolism and quality control. 38. Napolitano, G. & Ballabio, A. TFEB at a glance. selection. Autophagy 11, 1956–1977 (2015).
Nat. Cell Biol. 21, 133–142 (2019). J. Cell Sci. 129, 2475–2481 (2016). 65. Sardiello, M. et al. A gene network regulating
12. Osawa, T. et al. Atg2 mediates direct lipid transfer 39. Napolitano, G. et al. mTOR-​dependent phosphorylation lysosomal biogenesis and function. Science 325,
between membranes for autophagosome formation. controls TFEB nuclear export. Nat. Commun. 9, 3312 473–477 (2009).
Nat. Struct. Mol. Biol. 26, 281–288 (2019). (2018). 66. Settembre, C. et al. TFEB links autophagy to lysosomal
13. Matoba, K. et al. Atg9 is a lipid scramblase that 40. Inoki, K., Zhu, T. & Guan, K. L. TSC2 mediates cellular biogenesis. Science 332, 1429–1433 (2011).
mediates autophagosomal membrane expansion. energy response to control cell growth and survival. 67. Hars, E. S. et al. Autophagy regulates ageing in
Nat. Struct. Mol. Biol. 27, 1185–1193 (2020). Cell 115, 577–590 (2003). C. elegans. Autophagy 3, 93–95 (2007).
14. Maeda, S. et al. Structure, lipid scrambling activity 41. Lee, J. W., Park, S., Takahashi, Y. & Wang, H. G. The 68. Meléndez, A. et al. Autophagy genes are essential
and role in autophagosome formation of ATG9A. association of AMPK with ULK1 regulates autophagy. for dauer development and life-​span extension in
Nat. Struct. Mol. Biol. 27, 1194–1201 (2020). PLoS ONE 5, e15394 (2010). C. elegans. Science 301, 1387–1391 (2003).
15. Mancias, J. D. & Kimmelman, A. C. Mechanisms of 42. Gwinn, D. M. et al. AMPK phosphorylation of raptor 69. Dwivedi, M., Song, H. O. & Ahnn, J. Autophagy
selective autophagy in normal physiology and cancer. mediates a metabolic checkpoint. Mol. Cell 30, genes mediate the effect of calcineurin on life span
J. Mol. Biol. 428, 1659–1680 (2016). 214–226 (2008). in C. elegans. Autophagy 5, 604–607 (2009).
16. Gatica, D., Lahiri, V. & Klionsky, D. J. Cargo 43. Bach, M., Larance, M., James, D. E. & Ramm, G. 70. Kuma, A. et al. The role of autophagy during the early
recognition and degradation by selective autophagy. The serine/threonine kinase ULK1 is a target of neonatal starvation period. Nature 432, 1032–1036
Nat. Cell Biol. 20, 233–242 (2018). multiple phosphorylation events. Biochem. J. 440, (2004).
17. Chu, C. T. et al. Cardiolipin externalization to the outer 283–291 (2011). 71. Sou, Y. S. et al. The Atg8 conjugation system is
mitochondrial membrane acts as an elimination signal 44. Tian, W. et al. Phosphorylation of ULK1 by AMPK indispensable for proper development of autophagic
for mitophagy in neuronal cells. Nat. Cell Biol. 15, regulates translocation of ULK1 to mitochondria and isolation membranes in mice. Mol. Biol. Cell 19,
1197–1205 (2013). mitophagy. FEBS Lett. 589, 1847–1854 (2015). 4762–4775 (2008).
18. Dany, M. & Ogretmen, B. Ceramide induced mitophagy 45. Mack, H. I., Zheng, B., Asara, J. M. & Thomas, S. M. 72. Komatsu, M. et al. Impairment of starvation-​induced
and tumor suppression. Biochim. Biophys. Acta 1853, AMPK-​dependent phosphorylation of ULK1 regulates and constitutive autophagy in Atg7-deficient mice.
2834–2845 (2015). ATG9 localization. Autophagy 8, 1197–1214 (2012). J. Cell Biol. 169, 425–434 (2005).
19. Mizushima, N. & Murphy, L. O. Autophagy assays for 46. Egan, D. F. et al. Phosphorylation of ULK1 (hATG1) 73. Pyo, J. O. et al. Overexpression of Atg5 in mice
biological discovery and therapeutic development. by AMP-​activated protein kinase connects energy activates autophagy and extends lifespan. Nat.
Trends Biochem. Sci. 45, 1080–1093 (2020). sensing to mitophagy. Science 331, 456–461 (2011). Commun. 4, 2300 (2013).
20. Klionsky, D. J. et al. Guidelines for the use and 47. Tamargo-​Gómez, I. & Mariño, G. AMPK: regulation 74. Fernández, Á. F. et al. Disruption of the beclin 1–BCL2
interpretation of assays for monitoring autophagy. of metabolic dynamics in the context of autophagy. autophagy regulatory complex promotes longevity in
Autophagy 17, 1–382 (2021). Int. J. Mol. Sci. 19, 3812 (2018). mice. Nature 558, 136–140 (2018).
21. Moulis, M. & Vindis, C. Methods for measuring 48. Shiloh, R. et al. Non-​canonical activation of DAPK2 by 75. Nakamura, S. et al. Suppression of autophagic activity
autophagy in mice. Cells 6, 14 (2017). AMPK constitutes a new pathway linking metabolic by Rubicon is a signature of aging. Nat. Commun. 10,
22. Puente, C., Hendrickson, R. C. & Jiang, X. stress to autophagy. Nat. Commun. 9, 1759 (2018). 847 (2019).
Nutrient-​regulated phosphorylation of ATG13 49. Xu, D. Q. et al. PAQR3 controls autophagy by 76. Zhou, B. et al. Mitochondrial permeability uncouples
inhibits starvation-​induced autophagy. J. Biol. Chem. integrating AMPK signaling to enhance ATG14L-​ elevated autophagy and lifespan extension. Cell 177,
291, 6026–6035 (2016). associated PI3K activity. EMBO J. 35, 496–514 299–314.e16 (2019).
23. Kim, J., Kundu, M., Viollet, B. & Guan, K. L. AMPK (2016). 77. Wilhelm, T. et al. Neuronal inhibition of the
and mTOR regulate autophagy through direct 50. Zhao, Y. et al. RACK1 promotes autophagy by autophagy nucleation complex extends life span
phosphorylation of Ulk1. Nat. Cell Biol. 13, enhancing the Atg14L-​Beclin 1-Vps34-Vps15 complex in post-​reproductive C. elegans. Genes Dev. 31,
132–141 (2011). formation upon phosphorylation by AMPK. Cell Rep. 1561–1572 (2017).
24. Shang, L. et al. Nutrient starvation elicits an 13, 1407–1417 (2015). 78. Bjedov, I. et al. Fine-​tuning autophagy maximises
acute autophagic response mediated by Ulk1 51. Aas, S. N. et al. The impact of age and frailty on lifespan and is associated with changes in
dephosphorylation and its subsequent dissociation skeletal muscle autophagy markers and specific mitochondrial gene expression in Drosophila.
from AMPK. Proc. Natl Acad. Sci. USA 108, strength: a cross-​sectional comparison. Exp. Gerontol. PLoS Genet. 16, e1009083 (2020).
4788–4793 (2011). 125, 110687 (2019). 79. Lim, Y. M. et al. Systemic autophagy insufficiency
25. Yuan, H. X., Russell, R. C. & Guan, K. L. Regulation 52. Kim, J. et al. Differential regulation of distinct Vps34 compromises adaptation to metabolic stress and
of PIK3C3/VPS34 complexes by MTOR in nutrient complexes by AMPK in nutrient stress and autophagy. facilitates progression from obesity to diabetes.
stress-​induced autophagy. Autophagy 9, 1983–1995 Cell 152, 290–303 (2013). Nat. Commun. 5, 4934 (2014).
(2013). 53. Weerasekara, V. K. et al. Metabolic-​stress-induced 80. Fernández, Á. F. et al. Autophagy couteracts weight
26. Nazio, F. et al. mTOR inhibits autophagy by controlling rearrangement of the 14-3-3ζ interactome promotes gain, lipotoxicity and pancreatic β-​cell death upon
ULK1 ubiquitylation, self-​association and function autophagy via a ULK1- and AMPK-​regulated 14-3-3ζ hypercaloric pro-​diabetic regimens. Cell Death Dis. 8,
through AMBRA1 and TRAF6. Nat. Cell Biol. 15, interaction with phosphorylated Atg9. Mol. Cell Biol. e2970 (2017).
406–416 (2013). 34, 4379–4388 (2014). 81. Ebato, C. et al. Autophagy is important in islet
27. Ma, X. et al. MTORC1-mediated NRBF2 54. Huang, R. et al. Deacetylation of nuclear LC3 drives homeostasis and compensatory increase of β cell mass
phosphorylation functions as a switch for the autophagy initiation under starvation. Mol. Cell 57, in response to high-​fat diet. Cell Metab. 8, 325–332
class III PtdIns3K and autophagy. Autophagy 13, 456–466 (2015). (2008).
592–607 (2017). 55. Lee, I. H. et al. A role for the NAD-​dependent 82. Jung, H. S. et al. Loss of autophagy diminishes
28. Park, J. M. et al. The ULK1 complex mediates deacetylase Sirt1 in the regulation of autophagy. pancreatic β cell mass and function with resultant
mTORC1 signaling to the autophagy initiation Proc. Natl Acad. Sci. USA 105, 3374–3379 hyperglycemia. Cell Metab. 8, 318–324 (2008).
machinery via binding and phosphorylating ATG14. (2008). 83. Goginashvili, A. et al. Insulin granules. Insulin
Autophagy 12, 547–564 (2016). 56. Gu, X. et al. SIRT1-mediated FoxOs pathways protect secretory granules control autophagy in pancreatic
29. Wan, W. et al. mTORC1 phosphorylates against apoptosis by promoting autophagy in β cells. Science 347, 878–882 (2015).
acetyltransferase p300 to regulate autophagy and osteoblast-​like MC3T3-E1 cells exposed to sodium 84. Yamamoto, S. et al. Autophagy differentially regulates
lipogenesis. Mol. Cell 68, 323–335.e6 (2017). fluoride. Oncotarget 7, 65218–65230 (2016). insulin production and insulin sensitivity. Cell Rep. 23,
30. Wan, W. et al. mTORC1-regulated and HUWE1- 57. Kume, S. et al. Calorie restriction enhances cell 3286–3299 (2018).
mediated WIPI2 degradation controls autophagy adaptation to hypoxia through Sirt1-dependent 85. Masini, M. et al. Autophagy in human type 2 diabetes
flux. Mol. Cell 72, 303–315.e6 (2018). mitochondrial autophagy in mouse aged kidney. pancreatic β cells. Diabetologia 52, 1083–1086
31. Kim, Y. M. et al. mTORC1 phosphorylates UVRAG to J. Clin. Invest. 120, 1043–1055 (2010). (2009).
negatively regulate autophagosome and endosome 58. Price, N. L. et al. SIRT1 is required for AMPK 86. Cnop, M. et al. RNA sequencing identifies dysregulation
maturation. Mol. Cell 57, 207–218 (2015). activation and the beneficial effects of resveratrol on of the human pancreatic islet transcriptome by
32. Munson, M. J. et al. mTOR activates the VPS34–UVRAG mitochondrial function. Cell Metab. 15, 675–690 the saturated fatty acid palmitate. Diabetes 63,
complex to regulate autolysosomal tubulation and cell (2012). 1978–1993 (2014).
survival. EMBO J. 34, 2272–2290 (2015). 59. Ghosh, H. S., McBurney, M. & Robbins, P. D. 87. Las, G., Serada, S. B., Wikstrom, J. D., Twig, G. &
33. Cheng, X. et al. Pacer is a mediator of mTORC1 SIRT1 negatively regulates the mammalian target Shirihai, O. S. Fatty acids suppress autophagic turnover
and GSK3–TIP60 signaling in regulation of of rapamycin. PLoS ONE 5, e9199 (2010). in β-​cells. J. Biol. Chem. 286, 42534–42544 (2011).

Nature Reviews | Endocrinology

0123456789();:
Reviews

88. Mir, S. U. et al. Inhibition of autophagic turnover in 115. Carnio, S. et al. Autophagy impairment in muscle 140. Tang, H. et al. Swimming prevents nonalcoholic fatty
β-​cells by fatty acids and glucose leads to apoptotic induces neuromuscular junction degeneration and liver disease by reducing migration inhibitory factor
cell death. J. Biol. Chem. 290, 6071–6085 (2015). precocious aging. Cell Rep. 8, 1509–1521 (2014). through Akt suppression and autophagy activation.
89. Zummo, F. P. et al. Glucagon-​like peptide 1 protects 116. Potes, Y. et al. Overweight in elderly people induces Am. J. Transl. Res. 11, 4315–4325 (2019).
pancreatic β-​cells from death by increasing autophagic impaired autophagy in skeletal muscle. Free Radic. 141. Ghareghani, P. et al. Aerobic endurance training
flux and restoring lysosomal function. Diabetes 66, Biol. Med. 110, 31–41 (2017). improves nonalcoholic fatty liver disease (NAFLD)
1272–1285 (2017). 117. Møller, A. B. et al. Altered gene expression and features via miR-33 dependent autophagy induction
90. Trudeau, K. M. et al. Lysosome acidification by repressed markers of autophagy in skeletal muscle in high fat diet fed mice. Obes. Res. Clin. Pract. 12,
photoactivated nanoparticles restores autophagy of insulin resistant patients with type 2 diabetes. 80–89 (2018).
under lipotoxicity. J. Cell Biol. 214, 25–34 (2016). Sci. Rep. 7, 43775 (2017). 142. Vainshtein, A., Tryon, L. D., Pauly, M. & Hood, D. A.
91. Bugliani, M. et al. Modulation of autophagy influences 118. Chang, Y. C. et al. Resveratrol protects muscle cells Role of PGC-1α during acute exercise-​induced
the function and survival of human pancreatic β cells against palmitate-​induced cellular senescence and autophagy and mitophagy in skeletal muscle.
under endoplasmic reticulum stress conditions and in insulin resistance through ameliorating autophagic Am. J. Physiol. Cell Physiol. 308, C710–C719
type 2 diabetes. Front. Endocrinol. 10, 52 (2019). flux. J. Food Drug Anal. 26, 1066–1074 (2018). (2015).
92. Abedini, A. & Schmidt, A. M. Mechanisms of islet 119. Drake, J. C. & Yan, Z. Mitophagy in maintaining 143. Wohlgemuth, S. E., Seo, A. Y., Marzetti, E., Lees, H. A.
amyloidosis toxicity in type 2 diabetes. FEBS Lett. skeletal muscle mitochondrial proteostasis and & Leeuwenburgh, C. Skeletal muscle autophagy and
587, 1119–1127 (2013). metabolic health with ageing. J. Physiol. 595, apoptosis during aging: effects of calorie restriction
93. Kim, J. et al. Amyloidogenic peptide oligomer 6391–6399 (2017). and life-​long exercise. Exp. Gerontol. 45, 138–148
accumulation in autophagy-​deficient β cells induces 120. Lam, T. et al. Reversal of intramyocellular lipid (2010).
diabetes. J. Clin. Invest. 124, 3311–3324 (2014). accumulation by lipophagy and a p62-mediated 144. Kulkarni, A. S., Gubbi, S. & Barzilai, N. Benefits
94. Shigihara, N. et al. Human IAPP-​induced pancreatic pathway. Cell Death Discov. 2, 16061 (2016). of metformin in attenuating the hallmarks of aging.
β cell toxicity and its regulation by autophagy. J. Clin. 121. Tang, A. H. & Rando, T. A. Induction of autophagy Cell Metab. 32, 15–30 (2020).
Invest. 124, 3634–3644 (2014). supports the bioenergetic demands of quiescent 145. Martin-​Montalvo, A. et al. Metformin improves
95. Perry, R. J., Samuel, V. T., Petersen, K. F. & muscle stem cell activation. EMBO J. 33, 2782–2797 healthspan and lifespan in mice. Nat. Commun. 4,
Shulman, G. I. The role of hepatic lipids in hepatic (2014). 2192 (2013).
insulin resistance and type 2 diabetes. Nature 510, 122. García-​Prat, L. et al. Autophagy maintains stemness 146. Song, Y. M. et al. Metformin alleviates hepatosteatosis
84–91 (2014). by preventing senescence. Nature 529, 37–42 by restoring SIRT1-mediated autophagy induction
96. Yang, L., Li, P., Fu, S., Calay, E. S. & Hotamisligil, G. S. (2016). via an AMP-​activated protein kinase-​independent
Defective hepatic autophagy in obesity promotes ER 123. Fontana, L. & Partridge, L. Promoting health and pathway. Autophagy 11, 46–59 (2015).
stress and causes insulin resistance. Cell Metab. 11, longevity through diet: from model organisms to 147. Li, M., Sharma, A., Yin, C., Tan, X. & Xiao, Y.
467–478 (2010). humans. Cell 161, 106–118 (2015). Metformin ameliorates hepatic steatosis and improves
97. González-​Rodríguez, A. et al. Impaired autophagic flux 124. Jia, K. & Levine, B. Autophagy is required for dietary the induction of autophagy in HFD‑induced obese
is associated with increased endoplasmic reticulum restriction-​mediated life span extension in C. elegans. mice. Mol. Med. Rep. 16, 680–686 (2017).
stress during the development of NAFLD. Cell Death Autophagy 3, 597–599 (2007). 148. Jiang, Y. et al. Metformin plays a dual role in MIN6
Dis. 5, e1179 (2014). 125. Hansen, M. et al. A role for autophagy in the extension pancreatic β cell function through AMPK-​dependent
98. Singh, R. et al. Autophagy regulates lipid metabolism. of lifespan by dietary restriction in C. elegans. PLoS autophagy. Int. J. Biol. Sci. 10, 268–277 (2014).
Nature 458, 1131–1135 (2009). Genet. 4, e24 (2008). 149. Jing Yin, J., Bo Li, Y., Ming Cao, M. & Wang, Y.
99. Jaber, N. et al. Class III PI3K Vps34 plays an essential 126. Mercken, E. M. et al. Calorie restriction in humans Liraglutide improves the survival of INS-1 cells by
role in autophagy and in heart and liver function. inhibits the PI3K/AKT pathway and induces a younger promoting macroautophagy. Int. J. Endocrinol. Metab.
Proc. Natl Acad. Sci. USA 109, 2003–2008 (2012). transcription profile. Aging Cell 12, 645–651 (2013). 11, 184–190 (2013).
100. Settembre, C. et al. TFEB controls cellular lipid 127. Yang, L. et al. Long-​term calorie restriction enhances 150. Fan, M. et al. Liraglutide enhances autophagy and
metabolism through a starvation-​induced cellular quality-​control processes in human skeletal promotes pancreatic β cell proliferation to ameliorate
autoregulatory loop. Nat. Cell Biol. 15, 647–658 muscle. Cell Rep. 14, 422–428 (2016). type 2 diabetes in high-​fat-fed and streptozotocin-​
(2013). 128. Kim, K. E. et al. Caloric restriction of db/db mice treated mice. Med. Sci. Monit. 24, 2310–2316
101. Tanaka, S. et al. Rubicon inhibits autophagy reverts hepatic steatosis and body weight with (2018).
and accelerates hepatocyte apoptosis and lipid divergent hepatic metabolism. Sci. Rep. 6, 30111 151. Miao, X. et al. The glucagon-​like peptide-1
accumulation in nonalcoholic fatty liver disease (2016). analogue liraglutide promotes autophagy through
in mice. Hepatology 64, 1994–2014 (2016). 129. Gao, X., Yan, D., Zhao, Y., Tao, H. & Zhou, Y. the modulation of 5′-AMP-​activated protein kinase in
102. Zhang, Y. et al. Adipose-​specific deletion of autophagy-​ Moderate calorie restriction to achieve normal INS-1 β-​cells under high glucose conditions. Peptides
related gene 7 (Atg7) in mice reveals a role in weight reverses β-​cell dysfunction in diet-​induced 100, 127–139 (2018).
adipogenesis. Proc. Natl Acad. Sci. USA 106, obese mice: involvement of autophagy. Nutr. Metab. 152. Lim, S. W., Jin, L., Jin, J. & Yang, C. W. Effect of
19860–19865 (2009). 12, 34 (2015). exendin-4 on autophagy clearance in β cells of rats
103. Tchkonia, T. et al. Fat tissue, aging, and cellular 130. Cui, M., Yu, H., Wang, J., Gao, J. & Li, J. Chronic with tacrolimus-​induced diabetes mellitus. Sci. Rep. 6,
senescence. Aging Cell 9, 667–684 (2010). caloric restriction and exercise improve metabolic 29921 (2016).
104. Kovsan, J. et al. Altered autophagy in human adipose conditions of dietary-​induced obese mice in 153. Fu, J. et al. Liraglutide protects pancreatic β cells from
tissues in obesity. J. Clin. Endocrinol. Metab. 96, autophagy correlated manner without involving endoplasmic reticulum stress by upregulating MANF
E268–E277 (2011). AMPK. J. Diabetes Res. 2013, 852754 (2013). to promote autophagy turnover. Life Sci. 252, 117648
105. Jansen, H. J. et al. Autophagy activity is up-​regulated 131. Liu, H. et al. Intermittent fasting preserves β-​cell mass (2020).
in adipose tissue of obese individuals and modulates in obesity-​induced diabetes via the autophagy-​lysosome 154. Li, X. D., He, S. S., Wan, T. T. & Li, Y. B. Liraglutide
proinflammatory cytokine expression. Endocrinology pathway. Autophagy 13, 1952–1968 (2017). protects palmitate-​induced INS-1 cell injury by
153, 5866–5874 (2012). 132. Martinez-​Lopez, N. et al. System-​wide benefits of enhancing autophagy mediated via FoxO1. Mol. Med.
106. Kosacka, J. et al. Autophagy in adipose tissue of intermeal fasting by autophagy. Cell Metab. 26, Rep. 23, 147 (2021).
patients with obesity and type 2 diabetes. Mol. Cell 856–871.e5 (2017). 155. Wang, J. et al. Liraglutide protects pancreatic β-​cells
Endocrinol. 409, 21–32 (2015). 133. He, B., Liu, L., Yu, C., Wang, Y. & Han, P. Roux-​en-Y against free fatty acids in vitro and affects glucolipid
107. Abad-​Jiménez, Z. et al. Systemic oxidative stress gastric bypass reduces lipid overaccumulation in liver metabolism in apolipoprotein E–/– mice by activating
and visceral adipose tissue mediators of NLRP3 by upregulating hepatic autophagy in obese diabetic autophagy. Mol. Med. Rep. 12, 4210–4218 (2015).
inflammasome and autophagy are reduced in obese rats. Obes. Surg. 25, 109–118 (2015). 156. He, Q., Sha, S., Sun, L., Zhang, J. & Dong, M. GLP-1
type 2 diabetic patients treated with metformin. 134. Ma, N., Ma, R., Tang, K., Li, X. & He, B. Roux-​en-Y analogue improves hepatic lipid accumulation by
Antioxidants 9, 892 (2020). gastric bypass in obese diabetic rats promotes inducing autophagy via AMPK/mTOR pathway.
108. Yamamuro, T. et al. Age-​dependent loss of adipose autophagy to improve lipid metabolism through Biochem. Biophys. Res. Commun. 476, 196–203
Rubicon promotes metabolic disorders via excess mTOR/p70S6K signaling pathway. J. Diabetes Res. (2016).
autophagy. Nat. Commun. 11, 4150 (2020). 2020, 4326549 (2020). 157. He, Y. et al. The preventive effect of liraglutide
109. Anagnostis, P. et al. Type 2 diabetes mellitus is 135. He, C. et al. Exercise-​induced BCL2-regulated on the lipotoxic liver injury via increasing autophagy.
associated with increased risk of sarcopenia: a autophagy is required for muscle glucose homeostasis. Ann. Hepatol. 19, 44–52 (2020).
systematic review and meta-​analysis. Calcif. Tissue Int. Nature 481, 511–515 (2012). 158. Sharma, S., Mells, J. E., Fu, P. P., Saxena, N. K.
107, 453–463 (2020). 136. Grumati, P. et al. Physical exercise stimulates & Anania, F. A. GLP-1 analogs reduce hepatocyte
110. Wang, T. et al. Type 2 diabetes mellitus is associated autophagy in normal skeletal muscles but is steatosis and improve survival by enhancing
with increased risks of sarcopenia and pre-​sarcopenia detrimental for collagen VI-​deficient muscles. the unfolded protein response and promoting
in Chinese elderly. Sci. Rep. 6, 38937 (2016). Autophagy 7, 1415–1423 (2011). macroautophagy. PLoS ONE 6, e25269 (2011).
111. Srikanthan, P. & Karlamangla, A. S. Relative muscle 137. Lira, V. A. et al. Autophagy is required for exercise 159. Fang, Y. et al. Liraglutide alleviates hepatic steatosis
mass is inversely associated with insulin resistance training-​induced skeletal muscle adaptation and by activating the TFEB-​regulated autophagy-​lysosomal
and prediabetes. Findings from the third National improvement of physical performance. FASEB J. 27, pathway. Front. Cell Dev. Biol. 8, 602574 (2020).
Health and Nutrition Examination Survey. J. Clin. 4184–4193 (2013). 160. Liu, L., Liu, J. & Yu, X. Dipeptidyl peptidase-4 inhibitor
Endocrinol. Metab. 96, 2898–2903 (2011). 138. Luo, L. et al. Chronic resistance training activates MK-626 restores insulin secretion through enhancing
112. O’Neill, B. T. et al. Differential role of insulin/IGF-1 autophagy and reduces apoptosis of muscle cells by autophagy in high fat diet-​induced mice. Biochem.
receptor signaling in muscle growth and glucose modulating IGF-1 and its receptors, Akt/mTOR and Biophys. Res. Commun. 470, 516–520 (2016).
homeostasis. Cell Rep. 11, 1220–1235 (2015). Akt/FOXO3a signaling in aged rats. Exp. Gerontol. 48, 161. Zhu, B. et al. Alogliptin improves survival and health of
113. O’Neill, B. T. et al. Insulin and IGF-1 receptors regulate 427–436 (2013). mice on a high-​fat diet. Aging Cell 18, e12883 (2019).
FoxO-​mediated signaling in muscle proteostasis. 139. Pi, H. et al. Long-​term exercise prevents hepatic 162. Ellingsgaard, H. et al. Interleukin-6 enhances insulin
J. Clin. Invest. 126, 3433–3446 (2016). steatosis: a novel role of FABP1 in regulation of secretion by increasing glucagon-​like peptide-1
114. Masiero, E. et al. Autophagy is required to maintain autophagy-​lysosomal machinery. FASEB J. 33, secretion from L cells and alpha cells. Nat. Med. 17,
muscle mass. Cell Metab. 10, 507–515 (2009). 11870–11883 (2019). 1481–1489 (2011).

www.nature.com/nrendo

0123456789();:
Reviews

163. Linnemann, A. K. et al. Interleukin 6 protects uncoupled from longevity. Science 335, 1638–1643 185. Carmona-​Gutierrez, D. et al. The flavonoid
pancreatic β cells from apoptosis by stimulation (2012). 4,4′-dimethoxychalcone promotes autophagy-​
of autophagy. FASEB J. 31, 4140–4152 (2017). 174. Sataranatarajan, K. et al. Rapamycin increases mortality dependent longevity across species. Nat. Commun.
164. DeFronzo, R. A., Reeves, W. B. & Awad, A. S. in db/db mice, a mouse model of type 2 diabetes. 10, 651 (2019).
Pathophysiology of diabetic kidney disease: impact J. Gerontol. A Biol. Sci. Med. Sci. 71, 850–857 (2016). 186. Bravo-​San Pedro, J. M. et al. Acyl-​CoA-binding
of SGLT2 inhibitors. Nat. Rev. Nephrol. 17, 319–334 175. Arriola Apelo, S. I. et al. Alternative rapamycin protein is a lipogenic factor that triggers food intake
(2021). treatment regimens mitigate the impact of rapamycin and obesity. Cell Metab. 30, 754–767.e9 (2019).
165. Xu, J., Kitada, M., Ogura, Y., Liu, H. & Koya, D. on glucose homeostasis and the immune system. 187. Bravo-​San Pedro, J. M. et al. Cell-​autonomous,
Dapagliflozin restores impaired autophagy and Aging Cell 15, 28–38 (2016). paracrine and neuroendocrine feedback regulation of
suppresses inflammation in high glucose-​treated 176. Zhou, W. & Ye, S. Rapamycin improves insulin autophagy by DBI/ACBP (diazepam binding inhibitor,
HK-2 cells. Cells 10, 1457 (2021). resistance and hepatic steatosis in type 2 diabetes acyl-​CoA binding protein): the obesity factor. Autophagy
166. Fukushima, K., Kitamura, S., Tsuji, K., Sang, Y. & rats through activation of autophagy. Cell Biol. Int. 42, 15, 2036–2038 (2019).
Wada, J. Sodium glucose co-​transporter 2 inhibitor 1282–1291 (2018).
ameliorates autophagic flux impairment on renal 177. Eisenberg, T. et al. Induction of autophagy by Author contributions
proximal tubular cells in obesity mice. Int. J. Mol. Sci. spermidine promotes longevity. Nat. Cell Biol. 11, M.K. researched data for the article and wrote the manu-
21, 4054 (2020). 1305–1314 (2009). script. M.K. and D.K. contributed to discussion of the article
167. Aragón-​Herrera, A. et al. Empagliflozin reduces the 178. Eisenberg, T. et al. Cardioprotection and lifespan content and editing of the manuscript. Both authors critically
levels of CD36 and cardiotoxic lipids while improving extension by the natural polyamine spermidine. appraised the manuscript for important intellectual content
autophagy in the hearts of Zucker diabetic fatty rats. Nat. Med. 22, 1428–1438 (2016). and approved the final version to be published. M.K. and D.K.
Biochem. Pharmacol. 170, 113677 (2019). 179. Morselli, E. et al. Spermidine and resveratrol induce are responsible for the integrity of the content.
168. Xu, C. et al. Canagliflozin exerts anti-​inflammatory autophagy by distinct pathways converging on the
effects by inhibiting intracellular glucose metabolism acetylproteome. J. Cell Biol. 192, 615–629 (2011). Competing interests
and promoting autophagy in immune cells. Biochem. 180. Madeo, F., Eisenberg, T., Pietrocola, F. & Kroemer, G. The authors declare no competing interests.
Pharmacol. 152, 45–59 (2018). Spermidine in health and disease. Science 359,
169. Harrison, D. E. et al. Rapamycin fed late in life extends eaan2788 (2018). Peer review information
lifespan in genetically heterogeneous mice. Nature 181. Howitz, K. T. et al. Small molecule activators of sirtuins Nature Reviews Endocrinology thanks N. Tavernarakis and
460, 392–395 (2009). extend Saccharomyces cerevisiae lifespan. Nature the other, anonymous, reviewer(s) for their contribution to the
170. Zhang, Y. et al. Rapamycin extends life and health in 425, 191–196 (2003). peer review of this work.
C57BL/6 mice. J. Gerontol. A Biol. Sci. Med. Sci. 69, 182. Morselli, E. et al. Caloric restriction and resveratrol
119–130 (2014). promote longevity through the Sirtuin-1-dependent Publisher’s note
171. Chiao, Y. A. et al. Rapamycin transiently induces induction of autophagy. Cell Death Dis. 1, e10 (2010). Springer Nature remains neutral with regard to jurisdictional
mitochondrial remodeling to reprogram energy 183. Baur, J. A. et al. Resveratrol improves health and claims in published maps and institutional affiliations.
metabolism in old hearts. Aging 8, 314–327 (2016). survival of mice on a high-​calorie diet. Nature 444,
172. Chang, G. R. et al. Rapamycin protects against high fat 337–342 (2006). Supplementary information
diet-​induced obesity in C57BL/6J mice. J. Pharmacol. 184. Chen, G. et al. 3,4-Dimethoxychalcone induces The online version contains supplementary material available
Sci. 109, 496–503 (2009). autophagy through activation of the transcription at https://doi.org/10.1038/s41574-021-00551-9.
173. Lamming, D. W. et al. Rapamycin-​induced insulin factors TFE3 and TFEB. EMBO Mol. Med. 11, e10469
resistance is mediated by mTORC2 loss and (2019). © Springer Nature Limited 2021

Nature Reviews | Endocrinology

0123456789();:

You might also like