You are on page 1of 18

REVIEWS

Autophagy in acute brain injury


Lorenzo Galluzzi1–5*, José Manuel Bravo-San Pedro1–5*, Klas Blomgren6 and
Guido Kroemer1–4,6–8
Abstract | Autophagy is an evolutionarily ancient mechanism that ensures the lysosomal
degradation of old, supernumerary or ectopic cytoplasmic entities. Most eukaryotic cells,
including neurons, rely on proficient autophagic responses for the maintenance of homeostasis in
response to stress. Accordingly, autophagy mediates neuroprotective effects following some
forms of acute brain damage, including methamphetamine intoxication, spinal cord injury and
subarachnoid haemorrhage. In some other circumstances, however, the autophagic machinery
precipitates a peculiar form of cell death (known as autosis) that contributes to the aetiology of
other types of acute brain damage, such as neonatal asphyxia. Here, we dissect the
context-specific impact of autophagy on non-infectious acute brain injury, emphasizing the
possible therapeutic application of pharmacological activators and inhibitors of this catabolic
process for neuroprotection.

The term autophagy (from ancient Greek autos, ‘self ’ degradation — to cope with perturbations in both the
1
Equipe 11 Labellisée Ligue
and phago, ‘to eat’ — literally meaning ‘self-eating’) refers intracellular and the extracellular microenvironments7,8.
Contre le Cancer, Centre de
Recherche des Cordeliers, to a group of evolutionarily old mechanisms whereby Importantly, such adaptive macroautophagic responses
75006 Paris, France. eukary­otic cells eliminate cytoplasmic material through can be driven by an excess of autophagic substrates or
2
INSERM, U1138, lysosomal degradation1. Three main types of autophagy by an accrued demand for autophagic products or func-
75006 Paris, France. have been described so far, and these differ from each tions, and this functional dichotomy determines cargo
3
Université Paris Descartes/
Paris V, Sorbonne Paris Cité,
other in the modalities through which autophagic sub- specificity 9. Thus, cells exposed to a chemical agent that
75006 Paris, France. strates are delivered to lysosomes. Specifically, cargo depolarizes mitochondria mount a macroautophagic
4
Université Pierre et Marie delivery during microautophagy involves the direct response specifically targeting damaged mitochondria
Curie/Paris VI, 75006 Paris, invagination of the lysosomal membrane2. By contrast, (which is known as mitophagy) and aimed at removing
France.
chaperone-mediated autophagy (CMA) depends on the an excess of potentially dangerous autophagic substrates.
5
Gustave Roussy
Comprehensive Cancer recognition of autophagic substrates bearing a KFERQ By contrast, cells subjected to nutrient deprivation activate
Institute, 94805 Villejuif, motif by cytosolic chaperones of the heat-shock protein a relatively nonspecific variant of macroautophagy to sat-
France. family, followed by the lysosomal-associated membrane isfy an increased need for autophagic products9. From here
6
Karolinska Institute, protein 2 (LAMP2)-dependent translocation of these sub- onwards, the term autophagy refers to macroautophagy
Department of Women’s and
Children’s Health, Karolinska
strates across the lysosomal membrane3. Finally, macro­ unless otherwise specified.
University Hospital Q2:07, autophagy relies on the progressive sequestration of Adaptive autophagy often mediates prominent
17176 Stockholm, Sweden. autophagic cargoes by double-membraned vesicles known cytoprotective and anti-inflammatory effects10 (BOX 1).
7
Metabolomics and Cell as autophagosomes, which, upon their closure, fuse with Accordingly, defects in autophagic responses contrib-
Biology Platforms, Gustave
lysosomes to form so‑called autolysosomes4 (FIG. 1). ute to various human pathologies that involve the loss
Roussy Comprehensive
Cancer Institute, Macroautophagy (the best characterized form of of cellular homeostasis and consequent demise of post­
94805 Villejuif, France. autophagy in mammalian cells) operates at baseline mitotic cells; such pathologies include cardiovascular and
8
Pôle de Biologie, Hopitâl levels to preserve the structure and activity of several renal diseases, as well as acute and chronic neurological
Européen George Pompidou, subcellular compartments, including the endoplasmic disorders11,12. Moreover, autophagic defects compromise
AP‑HP, 75015 Paris, France.
reticulum (ER) and mitochondria5. Accordingly, nor- inflammatory homeostasis and generally increase the
*These authors contributed
mal autophagic functions in the CNS are required for susceptibility of the host to infectious diseases13. In addi-
equally to this work.
neuronal survival in the absence of external perturba- tion, accumulating evidence indicates that functional
Correspondence to L.G.
and G.K.
tions6. Moreover, the molecular machinery for macro­ autophagic responses can, at least in some situations, pre-
deadoc@vodafone.it; autophagy responds to a wide panel of sensors that cipitate a peculiar form of regulated cell death (RCD) that
kroemer@orange.fr continuously monitor indicators of homeostasis7,8. Thus, has been called autosis14 (BOX 2). This evidence implies
doi:10.1038/nrn.2016.51 cells can adjust autophagic flux — that is, an ongoing that autophagy de facto mediates cytotoxic rather
Published online 3 Jun 2016 autophagic response functionally coupled to lysosomal than cytoprotective effects in specific settings15. The

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Chaperone-mediated autophagy (CMA) b Microautophagy


HSP70
KFERQ Bulk delivery
Acid hydrolase

Unfolded Cargo
protein
Chaperone

Selective
delivery
HSP90

LAMP2
Fragment of
unfolded protein

c Macroautophagy
Lysosomal degradation Autolysosome

Isolation
membrane

Mitochondrion

Phagophore

LC3

Autophagosome

Autophagic cargo

Figure 1 | General organization of autophagic responses. All types of autophagy culminate in theReviews
Nature delivery| Neuroscience
of
autophagic substrates to the lysosome and the degradation of these substrates by lysosomal hydrolases.
a | Chaperone-mediated autophagy involves the recognition of autophagic substrates bearing a KFERQ motif by
members of the heat shock protein (HSP) family. This step is followed by the lysosomal-associated membrane protein 2
(LAMP2)-dependent translocation of chaperoned autophagic cargoes across the lysosomal membrane. b | By contrast,
cargo delivery during microautophagy occurs upon the direct invagination of the lysosomal membrane. c | During
macroautophagy, cytoplasmic entities destined for disposal are progressively enwrapped by a double-membrane
organelle known as an autophagosome. Following closure, mature autophagosomes can fuse with lysosomes to form
so‑called autolysosomes. LC3 (official name: MAP1LC3B), microtubule-associated protein 1 light chain 3β.

precise mechanisms through which proficient autophagic nucleation, elongation and closure of autophagosomes, as
responses accelerate rather than retard RCD remain to well as the fusion of autophagosomes with lysosomes16.
be elucidated. ATG9 mediates an essential, yet poorly characterized,
In this Review, we discuss recent findings suggesting function in early autophagosome formation. ATG3, ATG4
that autophagy has a context-specific impact on differ- and ATG7 cooperate to catalyse the lipidation of micro-
ent types of non-infectious acute brain injury — includ- tubule-associated protein 1 light chain 3β (MAP1LC3B;
ing excitotoxic challenges and epileptogenesis, exposure also known as LC3), which subsequently accumulates
to neurotoxins, neonatal asphyxia, adult stroke and in autophagosomal membranes and binds to substrates
neuro­trauma — with particular emphasis on the possi- tagged for autophagic degradation. Moreover, ATG7
Regulated cell death
(RCD). A form of cell death bility of using pharmacological modulators (activators cooperates with ATG10 to promote the formation of an
that depends on genetically or inhibitors) of autophagy for neuroprotection. ATG5–ATG12–ATG16L1 complex, which supports the
encoded machinery. In elongation of autophagosomes16.
contrast to its accidental General regulation of autophagy Most of the signal transduction cascades that control
counterpart, RCD can be
retarded or accelerated via
Autophagy mechanistically depends on the coordinated autophagy operate by influencing the activation status
specific pharmacological or activity of various members of the autophagy-­related of either of two multiprotein complexes: mechanistic
genetic interventions. (ATG) protein family, which together underlie the target of rapamycin (MTOR) complex 1 (MTORC1) or

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a supramolecular complex containing beclin 1 (BECN1) stimulates autophagy by releasing BECN1 from inhibi-
and phosphatidylinositol 3‑kinase catalytic subunit tory interactions with anti-apoptotic members of the B ell
type 3 (PIK3C3; also known as VPS34). MTORC1 lymphoma2 (BCL‑2) protein family 22.
contains MTOR and regulatory-associated protein of Mitophagic responses to depolarized mitochondria
MTOR (RAPTOR) and operates as a central suppres- are generally associated with the PTEN-induced puta-
sor of autophagy. Conversely, the BECN1–VPS34 com- tive kinase 1 (PINK1)-dependent and parkin RBR E3
plex promotes autophagy by catalysing the formation ubiquitin protein ligase (PARK2)-dependent ubiqui-
of phosphatidylinositol 3‑phosphate16. For instance, tylation of outer mitochondrial membrane proteins,
AMP-activated protein kinase (AMPK), which responds including mitofusin 1 (MFN1) and MFN2 (REF. 23).
to decreasing ATP levels upon phosphorylation by Ubiquitylated MFN1 and MFN2 are degraded by the
serine/threonine kinase 11 (STK11; also known as LKB1) proteasome, resulting in mitochondrial fragmentation23.
as well as to phosphorylation by calcium/calmodulin- Moreover, ubiquitylated structures, including mito-
dependent protein kinase kinase 2 (CAMKK2; also chondria, are bound by the autophagic adaptor seques-
known as CAMKKβ)17, triggers autophagy by inacti- tosome 1 (SQSTM1; also known as p62), which enables
vating MTORC1 (REF. 18) and by activating VPS34 or their uptake by autophagosomes through lipidated
unc‑51‑like kinase 1 (ULK1), which is a key kinase for LC3 (REF. 24). Other signal transduction cascades reg-
the initiation of autophagic responses19–21. Conversely, the ulate autophagy in many cell types, including neurons,
stress sensor death-associated protein kinase 1 (DAPK1) astrocytes and glial cells1,7. However, these pathways are

Box 1 | Cytoprotection, hormesis and autophagy


One of the primary functions of autophagy in mammalian organisms is to support cellular viability in the course of
adaptive responses to stress10. In line with this notion, the pharmacological or genetic inhibition of autophagy usually
sensitizes mammalian cells (including neurons) to a wide panel of stressful conditions, including nutritional, physical and
chemical cues10.
The precise mechanisms through which autophagy exerts cytoprotective functions exhibit a high degree of context-
dependency. These mechanisms include, but are not limited to, the following: the provision of catabolic substrates for
bioenergetic reactions (for example, in response to nutrient deprivation); the supply of substrates for anabolic
reactions (such as DNA repair, upon exposure to DNA-damaging agents); the preservation of optimal energy
metabolism (associated with the co‑activation of mitophagy and mitochondrial biogenesis); the elimination of
potential sources of oxidative damage (such as permeabilized mitochondria and redox-active protein aggregates); and
the direct inhibition of lethal signal transducers or processes (such as mitochondrial outer membrane
permeabilization)1,192,193.
Notably, adaptive autophagic responses can lead to robust hormetic effects; that is, they do not simply contribute to the
restoration of baseline physiological conditions, but also establish a novel state of homeostasis characterized by an
accrued fitness and resistance to stress194 (see part a of the figure). The hormetic effects of autophagy are relatively broad,
meaning that cells mounting an adaptive autophagic response upon exposure to a specific insult normally exhibit
increased resistance to various stressful conditions194. However, although autophagy underlies the hormetic effects of
various sublethal insults, it rarely mediates the cytotoxicity of the same challenges imposed in a lethal manner. Thus, the
hormetic and lethal mechanisms triggered by specific insults often differ from each other, and the former (frequently
autophagy) generally inhibit the latter (frequently apoptosis or some forms of regulated necrosis)192.
Autophagy has also been associated with non-adaptive hormesis195 (see part b of the figure). Thus, cells that are
pharmacologically driven into an autophagic response in the absence of potentially dangerous insults can also develop an
accrued fitness and broad resistance to future stress195. The hormetic effects of autophagy explain (at least in part) why
several nutritional or pharmacological manipulations that induce autophagic responses — for example, caloric restriction
or the administration of the sirtuin 1 activator resveratrol or the mechanistic target of rapamycin complex 1 (MTORC1)
inhibitor rapamycin — extends lifespan in several model organisms, including mice196. Moreover, non-adaptive autophagy­-
dependent hormesis may provide the basis for using pharmacological activators of autophagy as neuroprotective agents in
individuals at increased risk of acute brain injury.

a b
Hormetic Hormetic Hormetic
Stress capacity stimulus capacity
Fitness

Fitness

Autophagic adaptor
Adaptive Adaptive
capacity capacity
A protein that physically
interacts with lipidated LC3 as
well as with autophagic
receptors, hence facilitating the
sequestration of autophagic Physiological Adaptive response Hormetic response Physiological Hormetic response
cargoes within forming conditions conditions
autophagosomes. Time Time

Nature Reviews | Neuroscience


NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 3
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

elicited by insults that have a limited role in the patho- autophagosomal compartment, meaning that they also
genesis of acute brain disorders, and hence will not be increase when autophagosomes cannot fuse with lyso­
discussed in further detail here. somes or when lysosomal functions are impaired25.
Some of the biochemical reactions that accompany Monitoring autophagic flux is therefore crucial to
autophagy are frequently exploited to experimentally understand whether proficient autophagic responses
monitor autophagic responses in vitro and in vivo 25. are ongoing or whether the autophagic machinery is
These reactions include the following: expression of blocked25. This aspect of autophagic responses has pro-
ATG5, BECN1 and LC3; phosphorylation of AMPK, found therapeutic implications. Indeed, when neuro-
ULK1 or substrates of MTORC1; interaction of BECN1 toxicity is accompanied by a flux blockade, favouring
with BCL‑2 family members; lipidation of LC3; lyso­ autophagosomal clearance or inhibiting the formation
somal acidification; and the degradation of p62 or of autophagosomes may mediate neuroprotection,
autophagic substrates25. Importantly, several metabolic, whereas pharmacologically boosting the formation of
physical and chemical cues can promote morphological autophagosomes or inhibiting lysosomal degradation
and biochemical manifestations of autophagy, includ- has detrimental effects. Unfortunately, autophagic flux
ing the cytoplasmic accumulation of autophagosomes was not monitored in several of the reports presented
and autolysosomes (cytoplasmic vacuolization), as below, considerably complicating data interpretation.
well as LC3 lipidation10. However, cytoplasmic vac- Moreover, the pharmacological agents that are cur-
uolization and LC3 lipidation reflect the size of the rently available for modulating autophagy are relatively

Box 2 | Autophagic cell death and autosis


The term autophagic cell death has long been used to refer to a type of accumulation of autophagosomes (APs), autolysosomes (ALs) and empty
regulated cell death (RCD) that is characterized by extensive cytoplasmic vacuoles (EVs) are observed (see the figure). Phase 1b involves the
vacuolization197. However, the fact that cells die while manifesting separation of the inner and outer nuclear membrane, localized swelling
morphological (or biochemical) markers of autophagy does not necessarily of the perinuclear space (PNS) and the occurrence of membrane-bound
mean that autophagy causally promotes RCD, for at least two reasons15. densities in the PNS. Phase 2 is characterized by focal nuclear concavity
First, the accumulation of autophagosomes or LC3 lipidation may reflect and focal ballooning of the PNS, accompanied by general signs of
the blockade of lysosomal degradation rather than the upregulation of necrosis, including swollen mitochondria, rare autophagosomes and
autophagic flux25. Second, even the activation of proficient autophagic autolysosomes, and endoplasmic reticulum degradation.
responses (as monitored with flux assays) may represent an ultimate, but In addition to depending on components of the autophagic molecular
unsuccessful, attempt of cells to restore homeostasis192. In line with this machinery, as demonstrated with pharmacological inhibitors, such as
notion, the pharmacological or genetic inhibition of autophagy most often 3-methyladenine (3-MA), and small interfering RNAs (siRNAs) targeting
accelerates, rather than retards, stress-induced RCD, at least in components of the autophagy machinery (such as BECN1, ATG13 or
mammalian cells192. ATG14), autosis critically relies on the plasma membrane Na+/K+ ATPase,
Instances of bona fide autophagic cell death (a form of RCD that and hence can be pharmacologically inhibited by various cardiac
mechanistically depends on the molecular machinery for autophagy) glycosides, which target the α1 subunit of the Na+/K+ ATPase (ATP1A1).
were initially characterized in developing Drosophila melanogaster It should be noted that the Na+/K+ ATPase expressed by some mouse
larvae198. Accumulating evidence indicates that a specific variant of and rat tissues (including the cardiac muscle) is markedly more
autophagic cell death known as autosis also occurs in mammalian cells, resistant to inhibition by cardiac glycosides than its human
and may have important implications for acute brain injury14. Indeed, counterpart199 — partly because murine ATP1A1 binds to cardiac
various experimental conditions that promote autophagy, as well as glycosides with reduced affinity. However, the Na+/K+ ATPase expressed
pathophysiologically relevant stimuli such as hypoxia–ischaemia, in the mouse and rat brain is normally sensitive to the inhibitory activity
induce autosis14. Cells undergoing autosis exhibit peculiar and of several cardiac glycosides, including ouabain and neriifolin14,200.
temporally consecutive sets of morphological features, which have Thus, cardiac glycosides may represent valuable pharmacological tools
been categorized into three phases. In phase 1a, mild chromatin for the neuroprotective inhibition of autosis in both preclinical and
condensation, nuclear membrane convolution and cytoplasmic clinical settings.

Autosis
Nutrient deprivation
Na +

↓ATP Na+/K+ ATPase Phase 1a Phase 1b Phase 2


K+ EV
AP
PNS
↓O2
Hypoxia–ischaemia

↑ BECN1
Mitochondrion AL

Autophagy-inducing ?
peptides ? • 3-MA ?
• BECN1-targeting siRNA
Cardiac glycosides
• ATG13-targeting siRNA
• ATG14-targeting siRNA

Nature Reviews | Neuroscience

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

nonspecific (BOX 3; FIG. 2; TABLE 1), and the use of such glutamate 29, quinolinic acid 30, NMDA 31 and kainic
non-selective compounds constitutes a caveat of several acid32–34 reportedly induce markers of autophagic and/or
studies discussed in this Review. mitophagic responses in rodents. However, whether
autophagy protects neurons from excitotoxic insults
Autophagy in excitotoxicity remains unclear.
The term excitotoxicity refers to the neurotoxic effects Evidence — particularly from in vitro studies —
caused by the prolonged depolarization of postsynap- suggests that autophagy may counteract or prevent the
tic terminals, which results in the sustained influx of detrimental effects of excitotoxicity. Pharmacological
Ca2+ ions from the synaptic cleft26. Abrupt or prolonged activators of autophagy, including rapamycin and
increases in cytoplasmic Ca2+ levels cannot be properly trehalose, protected neonatal rat hippocampal neurons
buffered by the ER, and hence may trigger numerous from glutamate-induced excitotoxic death in vitro,
processes with potentially catastrophic consequences, whereas chloroquine (which inhibits autophagy) had
including the activation of various Ca2+-dependent no beneficial effects in this setting 35. Pyruvate (which
hydrolases, such as calpains, and mitochondrial permeabil- stimulates VPS34 activation and preserves bioenergetic
ity transition (MPT)-driven RCD27. Excitotoxicity has fitness) reduced the neurotoxicity of glutamate applied
been implicated in the neurotoxic effects of seizures, to human neuroblastoma SH‑SY5Y cells — an effect that
which are multifactorial (and generally symptomatic) could be blocked by the administration of the relatively
alterations of brain electrical activity that can occur nonspecific VPS34 inhibitor 3‑methyl­adenine (3‑MA)36.
in short episodes or proceed uninterrupted for hours Lithium (which stimulates autophagy) limited the exci-
(status epilepticus)28. Excitotoxic substances such as totoxic damage induced by kainic acid on spinal cord

Box 3 | Pharmacological modulation of autophagy


Nutrient deprivation is perhaps the most physiological stimulus that upregulates autophagic flux. It establishes a state of
bioenergetic and catabolic stress characterized by decreased intracellular levels of various metabolites, including ATP,
acetyl-CoA and several amino acids7. Declining ATP levels are paralleled by an increase in AMP, which promotes
autophagy by stimulating the ability of serine/threonine kinase 11 (STK11; also known as LKB1) to activate AMP-activated
protein kinase (AMPK). Low concentrations of acetyl-CoA stimulate autophagic responses as a consequence of reduced
E1A-binding protein p300 (EP300) activity. Finally, a shortage in amino acids triggers autophagy as it results in the
inactivation of mechanistic target of rapamycin complex 1 (MTORC1)7. As the systemic effects of nutrient deprivation are
pleiotropic and may introduce biases that are difficult to control, the effects of autophagy on acute brain injury have
been investigated with a panel of pharmacological agents with autophagy-inducing or -inhibiting properties25. However,
virtually all of these molecules also exhibit limited specificity, and some of them are sometimes administered
intraperitoneally or intravenously even though they might not readily cross the blood–brain barrier (TABLE 1); these two
issues considerably complicate data interpretation.
Commonly used inducers of autophagy include the following agents: rapamycin and curcumin, which potently inhibit
MTORC1; trehalose, which triggers MTORC1‑independent autophagy via hitherto unrecognized mechanisms; pyruvate,
which replenishes the acetyl-CoA pool and promotes the activation of death-associated protein kinase 1 (DAPK1); lithium
(alone or combined with valproate), which inhibits inositol(myo)-1(or 4)-monophosphatase 1 (IMPA1) and glycogen
synthase kinase 3β (GSK3β) and hence indirectly boosts the activity of phosphatidylinositol 3‑kinase catalytic subunit
type 3 (PIK3C3; also known as VPS34); the GSK3β inhibitor SB216763; and kaempferol, a phytoestrogen that reportedly
activates AMPK16. Additional molecules have been shown to promote signs of autophagy in the CNS (TABLE 1), but the
underlying molecular mechanisms remain to be elucidated. Moreover, autophagic flux has not been monitored in all these
settings, implying that some of these agents may actually inhibit (rather than stimulate) proficient autophagic responses.
Two main approaches are used for the experimental inhibition of autophagy: interfering with the formation of
autophagosomes and blocking lysosomal degradation. Molecules that inhibit VPS34 (hence preventing autophagy
initiation) include 3‑methyladenine (3‑MA), wortmannin, KU55933 and LY294002. Compared to 3‑MA and wortmannin
Calpains (which are considerably nonspecific), KU55933 and LY294002 exhibit an improved potency and selectivity for VPS34
A family of Ca2+-dependent, (REF. 25). Taurine limits autophagy initiation by stimulating MTORC1 activity. β‑asarone inhibits mitogen-activated
non-lysosomal cysteine protein kinase 8 (MAPK8; also known as JNK1), hence limiting the ability of JNK1 to release beclin 1 (BECN1) from
proteases involved in inhibitory interactions with B cell lymphoma (BCL‑2) family members16. Compound C (an AMPK inhibitor) also suppresses
processes as diverse as autophagic responses at initiation, as do several proteins that stimulate AKT1 signalling (and hence MTORC1), such as
long-term synaptic insulin-like growth factor 1 (IGF1), glial cell-derived neurotrophic factor (GDNF) and hepatocyte growth factor (HGF)16.
potentiation and regulated cell Conversely, chloroquine, E64d, pepstatin, bafilomycin A1 and l‑asparagine all inhibit autophagy by blocking the fusion
death.
of autophagosomes with lysosomes or by suppressing lysosomal functions16. As lysosomes participate in many biological
Mitochondrial permeability
processes in addition to autophagy (for example, endocytosis), these molecules have multiple off-target effects25. Several
transition other molecules have been shown to decrease the abundance of autophagic markers in the CNS (TABLE 1). Although
(MPT). An abrupt increase in some of these agents may modulate autophagic responses as a consequence of their antioxidant activity, the molecular
the permeability of the inner mechanisms that underlie these findings have not yet been clarified. Importantly, whereas blocking the formation of
mitochondrial membrane to autophagosomes is associated with a global decrease in autophagic markers, interfering with the fusion of
small solutes, resulting in the autophagosomes with lysosomes or inhibiting lysosomal functions generally causes an increase in the size of the
dissipation of the autophagosomal compartment, and hence manifests as a paradoxical increase in cytoplasmic vacuolization and LC3
mitochondrial transmembrane lipidation. Thus, without robust flux studies, it is difficult to conclude whether a specific intervention actually promotes or
potential and structural
inhibits proficient autophagic responses.
breakdown of the organelle.

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

RTK PI3K I Autophagy inducers* Autophagy inhibitors‡

LKB1 CAMKKβ MTORC1 AKT1 Initiation


↓ATP ↑Ca2+ • Kaempferol • Apelin-derived peptide
ULK1 • Rapamycin • GDNF
ATG13 ATG101 • Simvastatin • HGF
AMPK • IGF1
Initiation FIP200 • Pifitrin-α
• Salubrinal
ULK complex • Taurine
DAPK1 ATG9
AMBRA1
BECN1 BECN1 ATG14 Nucleation
VPS34 VPS15 Nucleation • Lithium • 3-methyladenine
JNK1 BCL-2 • Pyruvate • β-asarone
• SB216763 • KU55933
Pre-autophagosome • Valproate • LY294002
PI3K III complex • Wortmannin
ATG10
Elongation Forming
phagosome
ATG12 ATG12 Elongation
ATG5
ATG16L1 Retinoic acid
preLC3 ATG5 ATG12
ATG7 Fusion and degradation
ATG5–ATG12–ATG16L1 complex
ATG4 ATG3
• Bafilomycin A1
LC3 I LC3 II • Chloroquine
• E64d
• L-asparagine
Mitophagy PE PE LC3 II • Pepstatin
PE LC3 II

LC3 II Mitophagy
p62
PE LC3 II

PARK2 Ub Autophagosome Mdivi-1


Ub

Mitochondrion Changes in autophagic markers


Fragmentation LC3 II PE • Curcumin • Apocynin
Ub Ub • Deferoxamine • Caffeine
Fusion • Melatonin • Carnosine
Ub Ub Ub
PARK2 • N-acetyl-cysteine • Ceftriaxone
Ub MFN2 • Trehalose • GCEE
PINK1 Lysosome
MFN1 • Trichostatin A • Hydrogen sulfide
• Melatonin
Autolysosome • N-acetyl-serotonin
• NAD+
Ca 2+
• Necrostatin-1
PINK1 • Neriifolin
• Propofol
MPT Mitochondrion • Rosiglitazone
depolarization Degradation
• SN50

Figure 2 | Autophagy in the CNS. In response to decreasing ATP autophagy/beclin 1 regulator 1; ATG, autophagy-related; BCL-2, B cell
concentrations or increases in cytosolic Ca2+ levels, neurons and other lymphoma 2; BECN1, beclin 1; CAMKKβ Nature(officialReviews | Neuroscience
name: CAMMK2),
cells of the CNS mount a canonical autophagic response driven by calcium/calmodulin-dependent protein kinase kinase 2; DAPK1,
AMP-dependent protein kinase (AMPK) and unc‑51-like kinase 1 (ULK1), death-associated protein kinase 1; FIP200 (official name: RB1CC1), RB1
a process that is otherwise suppressed by mechanistic target of inducible coiled-coil 1; GCEE, γ‑glutamylcysteinyl ethyl ester; GDNF,
rapamycin complex 1 (MTORC1). The functionality of this response glial cell-derived neurotrophic factor; HGF, hepatocyte growth factor;
depends on the coordinated activation of a refined molecular IGF1, insulin-like growth factor 1; JNK1 (official name: MAPK8),
machinery that supports the nucleation, elongation and closure of mitogen-activated protein kinase 8; LC3 (official name: MAP1LC3B),
autophagosomes, as well as the fusion of autophagosomes with microtubule-associated protein 1 light chain 3β; LKB1 (official name:
lysosomes and lysosomal degradation. High cytosolic Ca2+ levels can STK11), serine/threonine kinase 11; MFN, mitofusin; p62 (official name:
also promote mitochondrial permeability transition (MPT). In this SQSTM1), sequestosome 1; PARK2, parkin RBR E3 ubiquitin protein
setting, several proteins of the outer mitochondrial membrane become ligase; PE, phosphatidylethanolamine; PI3K, phosphoinositide 3‑kinase;
ubiquitylated, eventually resulting in mitochondrial fragmentation and PINK1, PTEN-induced putative kinase 1; RTK, receptor tyrosine kinase;
uptake by forming autophagosomes through lipidated LC3 (LC3 II). This Ub, ubiquitin; VPS15, phosphoinositide 3‑kinase regulatory subunit 4;
mitophagic response generally mediates robust neuroprotective effects. VPS34 (official name: PIK3C3), phosphatidylinositol 3‑kinase catalytic
Common pharmacological modulators of autophagy as well as subunit type 3. *Or molecules associated with increased autophagic
molecules that have been associated with an increase or decrease of markers in the CNS. ‡Or molecules associated with decreased
autophagic markers in the CNS are listed (see also TABLE 1). AMBRA1, autophagic markers in the CNS.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

motor neurons from chick embryos37 or from mice38. Becn1- or Atg7‑targeting short hairpin RNAs (shRNAs)53.
Furthermore, the depletion of BECN1 or high-mobility Consistent with this result, rat cortical neurons engi-
group box 1 (HMGB1), a nuclear protein that stimulates neered to overexpress BECN1 or ATG7 were sensitized
autophagy upon release in the cytoplasm, increased the to the excitotoxic effects of kainic acid53. Furthermore,
sensitivity of cultured rat neurons to NMDA39,40. Small mice receiving an Atg7‑specific siRNA directly in the
interfering RNA (siRNA)-mediated downregulation of hippocampus were less sensitive to the administration
BECN1 also abolished the protective effects of ischaemic of kainic acid than were animals subjected to control,
preconditioning on mouse cortical neurons that were non-targeting RNA interference 34 (Supplementary
subsequently exposed to a neurotoxic glutamate chal- information S2 (table)).
lenge41,42. Similar results were obtained with 3‑MA and Interestingly, the neurotoxic effects of glutamate
the VPS34 inhibitor KU55933 in mice42. Indirect evi- have been partially attributed to the transcriptional
dence for a neuroprotective role of autophagy in mod- downregulation and calpain-dependent degradation
els of excitotoxicity has been collected in vivo, as mice of MFN2 and consequent mitochondrial dysfunc-
lacking both copies of NHL repeat-containing 1 (Nhlrc1) tion55,56. Accordingly, primary Mfn2−/− mouse motor
exhibited autophagic defects and are more sensitive to neurons displayed mitochondrial defects at baseline
kainic acid‑triggered seizures than wild-type mice43. and were more sensitive to the neurotoxic effects of
Similarly, autophagy-stimulating manganese com- glutamate than were control Mfn2 fl/fl neurons. By
plexes of curcumin and diacetylcurcumin limited kainic contrast, MFN2‑overexpressing neurons (which are
acid‑driven seizures in rats44. In these studies, however, expected to exhibit a reduced propensity to mitophagy)
cell death was not directly monitored, implying that were more resistant to glutamate-mediated excito-
autophagy may affect epileptogenesis independently of toxicity than their wild-type counterparts56. Whole-
its ability to limit the demise of stressed neurons. body overexpression of MFN2 also protected mice
Additional data indicate that autophagic defects have from the loss of spinal cord motor neurons induced
an aetiological role in epileptogenesis. Thus, activators by the intraventricular administration of glutamate56.
of autophagy may mediate therapeutic effects in this set- However, neither 3‑MA nor the lysosomal inhibitor
ting (Supplementary information S1 (table)). In humans, bafilomycin A1 blocked the degradation of MFN2 and
autophagy-suppressing mutations in genes encoding consequent mitochondrial dysfunction triggered by
endogenous MTORC1 inhibitors, including tuberous glutamate in this setting 56, casting additional uncer-
sclerosis 1 (TSC1), TSC2 and phosphatase and tensin tainty on the involvement of autophagy in this pro-
homologue (PTEN), are associated with an increased cess. Notably, rat cortical neurons exposed to a brief
predisposition to epilepsy 28. Accordingly, post-mortem glutamate pulse exhibited PARK2 accumulation in the
or surgical samples from individuals with tuberous scle- ER and mitochondria, but did not show signs of mito-
rosis exhibited profound autophagic defects45, and mice phagy unless an antioxidant (N‑acetyl-cysteine) was
lacking copies of Tsc1, Tsc2 or Pten in cortical neurons, co-administered57.
hippocampal neurons or glial cells exhibited frequent These results suggest that, at least in some circum-
episodes of spontaneous seizures that were associated stances, glutamate and other excitotoxic agents might
with premature mortality 45–48. Tsc1−/−, Tsc2−/− or Pten−/− block autophagic responses by causing mitochondrial
mouse neurons or glial cells also exhibited profound dysfunction and hence establishing profound oxidative
autophagic defects, and the systemic administration of stress. Supporting this notion, a cytoplasmic pool of
rapamycin considerably alleviated the severity of sei- ATM serine/threonine kinase (ATM), which activates
zures in, and improved the survival of, these diseased autophagy in response to oxidative (but not genotoxic)
mice45–49. Finally, the conditional deletion of Atg7 from stress58, has been implicated in the cytotoxic response
cortical and hippocampal mouse neurons also resulted of cortical, hippocampal and cerebellar neurons to
in spontaneous recurrent seizures45. intraperitoneal kainic acid administration in mice59.
Nevertheless, apparently at odds with the notion Unfortunately, however, it has not yet been investigated
that autophagy may help to protect neurons from exci- whether the activation of cytoplasmic ATM by the oxi-
totoxicity, 3‑MA and the lysosomal inhibitor E64d25 dative stress caused by kainic acid indeed triggers an
inhibited manifestations of autophagy, including LC3 autophagic response.
lipidation and p62 degradation, and protected striatal Taken together, these observations indicate that the
neurons (ex vivo and in vivo) from the toxic effects of actual impact of autophagy on excitotoxicity remains to
quinolinic acid and kainic acid30,33,50. Moreover, 3‑MA, be clarified (Supplementary information S1,S2 (tables)).
LY294002 (another VPS34 inhibitor), carnosine (an Although differences in the type, intensity and dura-
endogenous pleiotropic dipeptide) and insulin-like tion of the excitotoxic challenge may explain (at least
growth factor 1 (IGF1; which promotes MTORC1 partially) the heterogeneity of the findings discussed
signalling) limited various autophagic markers and above29, we fear that methodological issues have a prom-
Ischaemic preconditioning inhibited the demise of cultured neurons from the rat inent confounding role in this setting. Thus, additional
A hormetic process in which striatum, cerebellum and cortex exposed to NMDA or investigations involving specific strategies of autophagy
sublethal hypoxic challenges kainic acid51–54. Comparable results were obtained when inhibition and accurate flux assessments are urgently
establish resistance to
subsequent exposure to lethal
rat cerebellar granule neurons were administered with awaited to elucidate whether proficient autophagic
hypoxia, and which is known to an Atg7‑targeting siRNA51, and when rat cortical neu- responses limit or support the detrimental effects of
robustly activate autophagy. rons were transfected with a lentiviral vector coding for excitotoxicity.

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Pharmacological modulation of autophagy in models of acute brain injury


Agent Target Phase Blood–brain barrier Comments
permeability
Inducers (and molecules associated with increased levels of autophagic markers)
Curcumin Unknown Unknown Permeant Reported to inhibit autophagy in some settings
Cystatin C Unknown Unknown Impermeant Endogenous inhibitor of cysteine proteases
Deferoxamine Fe ions*
2+
Elongation* Permeant Expected to inhibit autophagy as a result of limited ROS
availability
Kaempferol AMPK Initiation Unknown AMPK activation secondary to bioenergetic impairment
Lithium IMPA1 and Nucleation Permeant Relatively nonspecific agent, inhibits several other enzymes
GSK3β
Melatonin ROS* Elongation* Permeant Expected to suppress autophagy upon ATG4 inhibition
N‑acetyl-cysteine ROS* Elongation* Permeant Expected to suppress autophagy upon ATG4 inhibition
Pyruvate DAPK1 Nucleation Permeant Promotes BECN1 release from inhibitory interactions with BCL‑2
Rapamycin MTORC1 Initiation Permeant Potent MTORC1 inhibitor with clinical applications
Retinoic acid RARα* Transcription* Impermeant Reported to specifically inhibit CMA in some settings
and elongation
SB216763 GSK3β Nucleation Permeant Mixed inhibitor of GSK3α and GSK3β
Simvastatin HMGCR Initiation Permeant Inhibits AKT1–MTORC1 signalling
Trehalose Unknown Unknown Permeant Disaccharide that stimulates MTORC1‑independent autophagy
Trichostatin A Unknown Transcription* Impermeant Mixed inhibitor of class I and class II (but not class III) HDACs
Valproate GSK3β and Transcription* Permeant Relatively nonspecific agent, inhibits several other enzymes
HDACs and nucleation
Inhibitors (and molecules associated with decreased levels of autophagic markers)
3‑methyladenine VPS34 Nucleation Permeant Considerably nonspecific class III PI3K inhibitor
β‑asarone JNK1 Nucleation Permeant Limits BECN1 release from inhibitory interactions with BCL‑2
Apelin-derived APLNR Initiation Permeant Stimulates AKT1–MTORC1 signalling
peptide
Apocynin NADPH Elongation* Permeant Expected to limit ROS availability, hence blocking ATG4
oxidase* activation
Bafilomycin A1 Vacuolar H+ Lysosomal Permeant Nonspecific inhibitor of lysosomal functions
ATPase acidification
Caffeine Unknown Unknown Permeant Mostly reported to promote MTORC1‑dependent autophagy
Carnosine ROS* Elongation* Permeant Expected to limit ROS availability, hence blocking ATG4
activation
Ceftriaxone Unknown Unknown Permeant β-lactam antibiotic approved for use in humans
Chloroquine H+ ions Lysosomal Permeant Nonspecific inhibitor of lysosomal functions
acidification
E64d CAPN1 and Lysosomal Unknown Nonspecific inhibitor of lysosomal functions
CTSB proteolysis
GCEE ROS* Elongation* Unknown Expected to limit ROS availability, hence blocking ATG4
activation
GDNF GFRA1 Initiation Impermeant Stimulates AKT1–MTORC1 signalling
HGF MET Initiation Permeant Stimulates AKT1–MTORC1 signalling
Hydrogen sulfide Unknown Unknown Permeant Also reported to promote MTORC1‑dependent autophagy
IGF1 IGF1R Initiation Permeant Stimulates AKT1–MTORC1 signalling
KU55933 VPS34 Nucleation Unknown Exhibits improved selectivity for VPS34
l‑asparagine Unknown Fusion Impermeant May block autophagy more selectively than lysosomal inhibitors
LY294002 VPS34 Nucleation Permeant Exhibits improved selectivity for VPS34
Mdivi‑1 DNML1 Mitophagy Permeant Specific inhibitor of mitochondrial fragmentation
Melatonin ROS* Elongation* Permeant Expected to limit ROS availability, hence blocking ATG4
activation

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 (cont.) | Pharmacological modulation of autophagy in models of acute brain injury


Agent Target Phase Blood–brain barrier Comments
permeability
N‑acetyl-serotonin ROS* Elongation* Permeant Precursor of melatonin, expected to reduce ROS availability
NAD+ SIRT1* Unknown Permeant Expected to promote autophagy through SIRT1
Necrostatin‑1 RIPK1* Unknown Permeant Potent inhibitor of some forms of necroptosis
Neriifolin Na+/K+ ATPase Elongation* Impermeant Cardiac glycoside that potently inhibits autosis
Pepstatin Aspartyl Lysosomal Unknown Generally used in combination with E64d
proteases proteolysis
Pifithrin‑α p53 Transcription Permeant May also activate autophagy in specific settings
Propofol ROS* Elongation* Permeant Expected to limit ROS availability, hence blocking ATG4
activation
Rosiglitazone PPARγ* Unknown Impermeant Antidiabetic drug expected to promote (not inhibit) autophagy
Salubrinal GADD34 Initiation Permeant Blocks autophagy initiation by ER stress
SN50 NF‑κB* General* Unknown Cell-permeant peptide that inhibits NF‑κB nuclear translocation
Taurine MTORC1 Initiation Permeant Endogenous sulfonic acid with multiple biological functions
Wortmannin VPS34 Nucleation Impermeant Considerably nonspecific class III PI3K inhibitor
AMPK, AMP-activated protein kinase; APLNR, apelin receptor; ATG4, autophagy-related 4 cysteine peptidase; BCL-2, B cell lymphoma 2; BECN1, beclin 1; CAPN1,
calpain 1; CMA, chaperone-mediated autophagy; CTSB, cathepsin B; DAPK1, death-associated protein kinase 1; DNM1L, dynamin 1‑like; ER, endoplasmic reticulum;
GADD34 (official name: PPP1R15A), protein phosphatase 1 regulatory subunit 15A; GCEE, γ‑glutamylcysteinyl ethyl ester; GDNF, glial cell-derived neurotrophic
factor; GFRA1, GDNF family receptor α1; GSK3α, glycogen synthase kinase 3α; GSK3β, glycogen synthase kinase 3β; HDAC, histone deacetylase; HGF, hepatocyte
growth factor; HMGCR, 3‑hydroxy‑3‑methylglutaryl-CoA reductase; IGF1, insulin-like growth factor 1; IGF1R, IGF1 receptor; IMPA1, inositol(myo)-1(or
4)-monophosphatase 1; JNK1 (official name: MAPK8), mitogen-activated protein kinase 8; MET, MET proto-oncogene, receptor tyrosine kinase (HGF receptor);
MTORC1, mechanistic target of rapamycin complex 1; NF‑κB, nuclear factor-κB; p53 (official name: TP53), tumour protein p53; PI3K, phosphoinositide 3‑kinase;
PPARγ, peroxisome proliferator-activated receptor-γ; RARα, retinoic acid receptor-α; RIPK1, receptor-interacting protein kinase 1; ROS, reactive oxygen species;
SIRT1, sirtuin 1; VPS34 (official name: PIK3C3), phosphatidylinositol 3‑kinase catalytic subunit type 3. *To be confirmed.

Autophagy in acute neurotoxic responses limited the acute neurotoxic effects of rotenone on
Neurotoxic responses can be subdivided into two groups SH‑SY5Y cells. In Mes 23.5 cells exposed to rotenone, the
according to their kinetics of manifestation. Acute neuro- depletion of sestrin 2 (SESN2), an upstream inhibitor of
toxicity manifests symptomatically over periods of hours MTORC1, compromised autophagy induction and aggra-
to days after a single exposure to neurotoxins, whereas vated neurotoxicity, whereas SESN2 overexpression acti-
the symptoms of chronic neurotoxicity become clinically vated a robust, AMPK-dependent autophagic response
evident years to decades later, and generally stem from while limiting RCD60. Similar results have been obtained
continued exposure11,27. In particular, chronic neuro- with rat H9C2 cardiomyocytes treated with an siRNA
toxic responses are associated with the progressive loss specific for glyceraldehyde‑3‑phosphate dehydrogenase
of specific neuronal populations, eventually resulting in (Gapdh)65, an enzyme that also exerts autophagy-regu-
degenerative disorders such as Parkinson disease11. The lating functions66. Conversely, the depletion of tyrosine
beneficial effects of autophagy in neurodegeneration have 3‑monooxygenase/tryptophan 5‑monooxygenase acti-
been convincingly established11, and will not be discussed vation protein-ε (YWHAE; also known as 14‑3‑3ε), a
in further detail here. However, some of the neurotoxins protein that inhibits autophagy by sequestering BECN1
used in pharmacological models of neurodegeneration, into inactive complexes67, exacerbated the acute neuro-
including rotenone, 1‑methyl‑4‑phenyl‑1,2,3,6‑tetrahy- toxic effects of rotenone on PC12 cells61 (Supplementary
dropyridine (MPTP), 6‑hydroxydopamine (6‑OHDA), information S1,S2 (tables)).
paraquat and lactacystin, also trigger acute neuro­toxicity MPTP is frequently used to establish a Parkinson
in association with autophagic responses, particularly disease‑like disorder in rodents owing to its severe and
when used in high doses. rather specific toxicity for dopaminergic neurons of the
Rotenone establishes a Parkinson disease‑like state substantia nigra11. There is some evidence suggesting
in rats and mice, and is also frequently used in experi- that autophagy may mitigate the neurotoxic effects of
mental settings to inhibit complex I of the mitochondrial acute MPTP exposure. Specifically, the siRNA-medi-
respiratory chain8. Acute exposure of neuronal cells lines ated depletion of ATG7 nearly abolished the demise
— including mouse hybrid Mes 23.5 cells60, rat PC12 of rat glioma C6 cells following acute MPTP exposure,
cells61 and human SH‑SY5Y cells62–64 — to rotenone as did the administration of melatonin, which also
resulted in rapid RCD accompanied by signs of auto- suppressed several manifestations of autophagy in rat
phagy. Supporting a cytoprotective role of autophagy, cells and interfered with the ability of chronic MPTP
pharmacological activators of autophagy, including rapa- exposure to establish a Parkinson disease‑like state in
mycin63, deferoxamine (a chelator of Fe2+ ions that triggers mice68. According to one study, acute MPTP exposure
autophagy through poorly characterized mechanisms)64 displaced BECN1 from inhibitory interactions with
and kaempferol (a phytoestrogen that activates AMPK)62, BCL‑2 and induced cell death in human neuroblastoma

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

SK‑N‑SH cells69. However, the authors of this study The neurotoxic effects of methamphetamine were
did not test whether autophagy itself was modulated associated with morphological signs of autophagy as
in these conditions. Independent data demonstrated early as 1994 (REF. 82). It was only more than a decade
that the activation of autophagy by systemic adminis- later, however, that autophagy was suggested to mediate
tration of rapamycin or valproate plus lithium limited the neuroprotective effects in this setting, when 3‑MA
the ability of MPTP to cause long-term symptoms of and the expression of a dominant-negative form of
Parkinson disease in mice70,71; however, acute neuro- VPS34 were found to exacerbate the demise of rat PC12
toxicity was not investigated. Another study provided and human SH‑SY5Y cells exposed to low concentra-
correlative evidence in support of a cytoprotective role tions of methamphetamine83,84. Corroborating these
of autophagy in acute MPTP neurotoxicity in vivo 72. findings, the downregulation of PINK1 aggravated the
In this experimental scheme, investigators relied on demise of methamphetamine-stimulated PC12 cells,
the administration of mesenchymal stem cells (which whereas PINK1 overexpression exerted robust neuro-
were suggested to activate autophagy), but they did not protective effects that correlated with the activation of
attempt to modulate autophagy by pharmacological or mitophagy 85. Similar results were obtained in rat N27
genetic means, casting doubts on the actual relevance dopaminergic cells subjected to the siRNA-mediated
of their findings. downregulation of LC3 or treated with 3‑MA86, as well
Similarly, it is difficult to conclude whether autophagy as in human non-neuronal cells in which autophagy
limits or exacerbates the acute toxicity of 6‑OHDA, para- was inhibited with BECN1‑targeting siRNAs, 3‑MA
quat or lactacystin. The rat striatum was protected from or chloroquine87. It has also been proposed that caf-
6‑OHDA in the presence of 3‑MA73; by contrast, both feine may boost the neurotoxicity of methampheta-
3‑MA and the depletion of parkinson protein 7 (PARK7; mine88 by inhibiting autophagy 89. However, caffeine
a component of the molecular machinery for mitophagy triggers robust autophagic responses in neurons and
also known as DJ‑1) exacerbated the death of SH‑SY5Y many other cell types90, implying that the link between
cells acutely exposed to paraquat74,75, and rapamycin lim- caffeine and methamphetamine should be interpreted
ited the acute cytotoxic potential of lactacystin on PC12 with caution. Further substantiating the neuroprotec-
cells76 (Supplementary information S1,S2 (tables)). Taken tive activity of autophagy in this setting, rapamycin
together, these observations indicate that the relevance limited neurotoxicity in rats that received metham-
of autophagy in the acute neurotoxic effects of agents phetamine in combination with antiretroviral agents
commonly used to generate animal models of Parkinson and purified HIV‑1 envelope glycoproteins (which
disease remains to be determined. were used together as a model of drug addiction in
Accumulating evidence suggests that autophagy has patients with HIV‑1 treated with antiretroviral drugs)91
an important role in the response of the CNS to psycho- (Supplementary information S1 (table)).
active molecules, including cocaine, methamphetamine, Apparently at odds with these observations, data
ethanol and the atypical antipsychotic olanzapine, as well obtained in rat N27 cells92, rat PC12 cells93, human
as to experimental toxins such as trimethyltin. Cocaine SK‑N‑SH cells94 and rat primary cortical neurons95 sug-
has recently been reported to trigger conventional auto- gest that autophagy contributes to (rather than coun-
phagic responses in human SVGA astrocytes, mouse teracts) the acute neurotoxicity of methamphetamine.
immortalized BV‑2 microglial cells, primary rat micro- These findings, however, are mostly correlative (that
glial cells and primary mouse cortical neurons, as well is, no experimental modulation of the process was
as in the mouse brain in vivo77–79, but to robustly activate undertaken)92–94 or present other problematic aspects.
MTORC1 (and hence to inhibit autophagy) in neurons For example, both bafilomycin A1 and rapamycin were
from the nucleus accumbens80. Rapamycin markedly counter-intuitively found to aggravate the detrimen-
reduced locomotor sensitization (one of the neurobehav- tal effects of methamphetamine on neurite length, but
ioural symptoms of cocaine administration) in rats81, as autophagic flux was not investigated in this study 95
did the deletion of either Mtor or Raptor from specific (Supplementary information S2 (table)). Therefore,
neuronal populations in mice80. Apparently at odds with it seems that autophagy mainly counteracts the acute
these findings, however, inhibition of autophagy with neurotoxicity of methamphetamine.
wortmannin also decreased the ability of microglial Similar considerations can be made regarding the
cells to secrete neuroinflammatory factors in response impact of autophagy on the acute neurotoxic effects
to cocaine77, whereas 3‑MA and bafilomycin A1 limited of ethanol, olanzapine and trimethyltin. Indeed, both
Trimethyltin cocaine-induced RCD in human SVGA astrocytes78 and wortmannin and a BECN1‑targeting shRNA aggra-
A tin-containing organic in primary mouse cortical neurons79. Moreover, siRNAs vated the acute toxicity of ethanol in SH‑SY5Y cells96,
compound with prominent targeting Atg5, Becn1 or Gapdh markedly limited the and ischaemic preconditioning led to robust neuropro-
neurotoxic activity towards the
demise of primary mouse cortical neurons acutely tective effects in the same model97. Interestingly, chronic
hippocampus.
exposed to cocaine79. Taken together, these data indicate ethanol administration caused the downregulation of
Locomotor sensitization that some aspects of the neurotoxicity of cocaine, such as various components of the autophagic machinery in the
Long-lasting exacerbation of locomotor neuron sensitization, are attenuated by auto- cerebral cortex of wild-type mice, but not in mice lacking
a psychostimulant-induced phagy (Supplementary information S1 (table)), whereas the gene encoding Toll-like receptor 4 (Tlr4)98. Taken
locomotor response, elicited by
the repeated intermittent
other aspects, including neuroinflammation and RCD, together, these data suggest that TLR4 signalling may
administration of the same depend on the activation of autophagic responses promote the chronic neurotoxicity of ethanol by inhib-
psychoactive agent. (Supplementary information S2 (table)). iting autophagy. As the damage-associated molecular

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

pattern (DAMP) HMGB1 (REF. 99) is one of the main Taken together, these data provide limited experi-
endogenous ligands for TLR4, it is tempting to specu- mental support for the hypothesis that autophagy medi-
late that neurons succumbing to acute exposure to eth- ates bona fide neuroprotective effects in the course of
anol may be the source of TLR4‑activating HMGB1, neonatal hypoxia–ischaemia. Moreover, work from
which in turn limits the ability of remaining neurons several independent groups overtly argues against this
and glial cells to mount proficient autophagic responses interpretation. Indeed, 3‑MA limited the demise of pri-
and therefore survive chronic ethanol neurotoxicity; mary rat cortical neurons, rat hippocampal slices and
however, this remains to be formally demonstrated. SH‑SY5Y cells exposed to oxygen and glucose deprivation
Irrespectively, human SH‑SY5Y cells subjected to the (OGD) in vitro114,115. Similarly, newborn rats receiving an
depletion of BECN1 and LC3 exhibited increased sensi- intracerebro­ventricular injection of 3‑MA after ischaemia
tivity to olanzapine100, as did mice exposed to olanzapine (more than 4 hours after the ischaemic episode) exhibited
in the presence of chloroquine100. Similar results were considerably smaller lesions compared with control ani-
obtained in neuronal and astrocytic cultures exposed to mals116. Comparable results were obtained with newborn
trimethyltin alone or in combination with pharmaco- rats treated with the cardiac glycoside neriifolin, a bona fide
logical modulators of autophagy, including 3‑MA, bafi- inhibitor of autosis (BOX 2), immediately after carotid
lomycin A1, chloroquine, rapamycin and lithium101,102. artery occlusion14. Moreover, 7‑day-old rats receiving a
Overall, these observations favour the interpretation lentiviral vector encoding a Becn1‑specific shRNA in the
that autophagic responses limit the acute neurotoxic striatum exhibited twice the amount of intact striatal tis-
effects of various psychoactive agents (Supplementary sue 24 hours after a hypoxic–ischaemic challenge than did
information S1 (table)). rats receiving a control shRNA53. An even higher degree of
long-term protection was afforded to pyramidal neurons
Autophagy in neonatal asphyxia in the hippocampal CA regions of mice in which Atg7 was
Newborn babies who have died as a consequence of specifically deleted from neurons106. Extending these find-
hypoxic–ischaemic encephalopathy exhibit ultrastruc- ings, the neuron-specific deletion of Atg7 inhibited RCD
tural and biochemical manifestations of autophagy in in the hippocampus, thalamus, cortex and striatum of
various cerebral areas, encompassing the basal gan- newborn mice subjected to neonatal hypoxia–ischaemia,
glia, thalamus, cortex and hippocampus103,104. Such globally reducing tissue loss by approximately 40%104.
autophagic markers include cytoplasmic vacuoliza- Notably, the possibility that such neuroprotection would
tion, BECN1 upregulation, LC3 lipidation and p62 originate from a preconditioning effect, reflecting the
degradation103,104. Similar findings were observed in hormetic adaptation of neurons to the absence of ATG7,
the brains of mice and rats experimentally subjected was experimentally discarded104. Newborn mice lacking
to hypoxia–ischaemia in the perinatal period53,103–107. both copies of the gene encoding the mitophagy recep-
Generally, rodent neurons manifesting signs of auto- tor BCL‑2/adenovirus E1B 19 kDa interacting protein 3
phagy upon neonatal asphyxia also exhibit morpho- (Bnip3) exhibited much smaller infarcts than did their
logical and biochemical markers of RCD and ongoing wild-type counterparts at 7 and 28 days (but not at 1 and
cellular degeneration. These signs include axonal and/or 3 days) after hypoxia–ischaemia117. Indeed, infarct size
somatodendritic swelling, mitochondrial fragmenta- 1 day after the hypoxic–ischaemic challenge was much
tion, chromatin fragmentation and condensation, and larger in Bnip3−/− mice than in wild-type mice, perhaps
caspase 3 activation103,105,108. Based on these correlative indicating that the rapid mitophagic removal of dam-
observations alone, however, one cannot conclude aged mitochondria is a cytoprotective mechanism in the
whether autophagic responses in the neonatal brain first phases of neonatal hypoxia–ischaemia. Irrespective
mediate neuroprotective or neurotoxic effects. of this possibility, the observations discussed here lend
The first study to investigate the effects of pharma- solid support to the hypothesis that autophagy aetiolog-
cological modulators of autophagy in newborn rats ically contributes to the pathological effects of perinatal
subjected to hypoxia–ischaemia observed that post-­ asphyxia (Supplementary information S2 (table)).
ischaemic administration of rapamycin efficiently limited Interestingly, it has been shown that the midbrain
damage in this model109. Such a neuroprotective effect of releases endogenous cardiac glycosides (namely, ouabain
rapamycin could be blocked by the subsequent admin- or endobain) in response to hypoxia–ischaemia118. It is
istration of 3‑MA110, suggesting that this effect origi- therefore tempting to speculate that the brain has evolved
Caspase 3
A cysteine protease that
nated from an autophagic response. However, although a safeguard mechanism that inhibits autophagic responses
precipitates apoptotic variants the post-ischaemic administration of 3‑MA and wort- when they may have neurotoxic consequences (such as
of regulated cell death, mannin changed the molecular mechanism by which during hypoxic–ischaemic challenges), but not when they
irrespective of the subcellular neurons died (switching it from caspase 3‑dependent are in place to preserve neuronal homeostasis (such as
compartment in which they are
apoptosis to necrosis)109,111, neither of these inhibitors under baseline conditions or in response to neurotoxins).
initiated.
of autophagy truly limited damage in newborn rats sub- This hypothesis awaits urgent experimental verification.
Cardiac glycoside jected to a hypoxic–ischaemic episode109 (Supplementary
A natural compound, such as information S1 (table)). Further complicating the issue, Autophagy in adult stroke
digoxin or digitoxin, that exerts lithium (which normally triggers autophagy) was neuro­ Brains of adult mice119–121 and rats54,122–141 subjected to
positive inotropic effects and
suppresses autosis as it inhibits
protective in a rat model of neonatal hypoxia–ischaemia, an ischaemic challenge exhibit various ultrastructural
the plasma membrane Na+/K+ but reduced LC3 lipidation, which is consistent with and biochemical markers of autophagy, including
ATPase. autophagy inhibition, rather than activation112,113. cytoplasmic vacuolization, MTORC1 inhibition, LC3

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

lipidation, p62 degradation and BECN1 and LAMP2 Interestingly, specific cerebral areas may benefit in
upregulation. At least two lines of evidence support a preferential manner from the activation of autophagy
the tenet that autophagy mediates considerable neuro­ in the course of, or after, an ischaemic insult. The post-
protective effects in adult rodents exposed to acute ischaemic administration of a chemical inhibitor of gly-
ischaemic episodes such as those caused by transient cogen synthase kinase 3β (GSK3β) activated autophagy
middle carotid artery occlusion (tMCAO; usual dura- in the microglia of rats subjected to pMCAO148. This
tion: 30 minutes to 3 hours) or permanent MCAO effect was associated with neuroprotection coupled with
(pMCAO) (Supplementary information S1 (table)). an autophagy-dependent anti-inflammatory effect 148.
First, genetic interventions that limit autophagic Methylene blue and pinocembrin (two antioxidants)
proficiency — including the deletion of mitochondrial administered after tMCAO also conferred remarkable
superoxide dismutase 2 (Sod2)121 or S‑antigen, retina and neuroprotection in rats, and this protective effect was
pineal gland (arrestin) (Sag)142, or the administration of a linked to the specific activation of autophagy in the
viral vector encoding a Tsc1‑targeting shRNA143 — aggra- ischaemic penumbra140,141. These latter observations suggest
vated neuronal loss in rodents subjected to tMCAO or that the beneficial effects of autophagy activators admin-
pMCAO. Similarly, the stereotaxic delivery of a viral vec- istered after ischaemia may reflect an improved salvage of
tor overexpressing nicotinamide phosphoribosyltrans- the ischaemic penumbra (Supplementary information S1
ferase (NAMPT)134,144, a manipulation that efficiently (table)). However, additional studies measuring auto-
upregulates autophagy, had remarkable neuroprotective phagy in specific cerebral areas are required to validate
effects in adult rats subsequently subjected to tMCAO. this hypothesis.
Moreover, inhibition of autophagy — caused by either the Activation of autophagy with rapamycin limited infarct
homozygous deletion of protein kinase AMP-activated size in mouse models of tMCAO and pMCAO; however,
α2 catalytic subunit (Prkaa2) or sirtuin 1 (Sirt1), or by similar data were obtained with the autophagy inhibitor
the downregulation of ATG7 or TSC1 — exacerbated chloroquine (although neuroprotection was slightly less
the neurotoxic effects of OGD in primary mouse144 remarkable)149. Moreover, both 3‑MA and mdivi‑1 (an
or rat 143,145 cortical neurons. Interestingly, the siRNA- inhibitor of mitophagy) exacerbated the loss of neurons
mediated downregulation of LAMP2 (which is an essen- associated with tMCAO in mice, yet exerted a neuro-
tial mediator of CMA), but not depletion of ATG5 (which protective or null effect, respectively, in mice subjected
is specifically involved in macroautophagy), increased to pMCAO145. These latter observations cast doubts on
the susceptibility of mouse Neuro2a cells to stan- the hypothesis that autophagy always exerts neuroprotec-
dalone hypoxia in vitro, whereas the activation of CMA tive activity in the course of adult stroke (Supplementary
with mycophenolic acid led to robust neuroprotective information S1,S2 (tables)).
effects131. These findings perhaps indicate that distinct Indeed, additional data support the tenet that auto-
forms of autophagy differentially contribute to situations phagy contributes to neurotoxicity in rodent models of
as multifactorial as an ischaemic insult in vivo. adult stroke. In particular, the VPS34 inhibitor 3‑MA
Second, the robust neuroprotective effects of pre- and the lysosomal inhibitor bafilomycin A1 limited
Metformin
conditioning, elicited by hypoxia or other interventions the infarct area in mice subjected to tMCAO150, in rats
An oral medication currently
licensed for the treatment of (including hyperbaric oxygen, the anaesthetic isoflu- subjected to pMCAO123,151 or tMCAO129, and in rats sub-
specific forms of diabetes. rane, metformin, thapsigargin and tunicamycin), appear to jected to global ischaemia in the four-vessel occlusion
Metformin activates rely on the establishment of sublethal ER stress138 cou- model124. Similarly, the expression of a Becn1‑targeting
AMP-activated protein kinase pled with the activation of hormetic autophagy (BOX 1). shRNA152 and the downregulation of eva‑1 homologue A
and inhibits respiratory
complex I.
Accordingly, mice lacking sphingosine kinase 2 (SPHK2; (EVA1A), a lysosomal protein involved in autophagic
a protein that activates BECN1) were insensitive to iso- responses122, lowered the sensitivity of rats to tMCAO.
Thapsigargin flurane-induced preconditioning 41,42, and Park2+/− mice, Bafilomycin A1 and 3‑MA and also limited the demise
A natural compound that which exhibit a selective defect in mitophagy, could of rat astrocytes caused by pMCAO in vivo or OGD
promotes endoplasmic
not establish normal preconditioning in response to in vitro128. Furthermore, several molecules that protect
reticulum stress by operating
as a non-competitive inhibitor thapsigargin and tunicamycin146. The ability of isoflu- neurons in rodents subjected to tMCAO, pMCAO or
of the sarco/endoplasmic rane, hypoxia, thapsigargin and tunicamycin to protect global ischaemia also inhibited autophagy. These mol-
reticulum Ca2+ ATPase. mouse cortical neurons against OGD was also abolished ecules include melatonin and its precursor N‑acetyl-
when neurons were previously transfected with a Becn1- serotonin127,130, propofol (an intravenous anaesthetic)129,
Tunicamycin
A mixture of homologous
or an Atg7‑targeting siRNA42,146. Moreover, autophagy carnosine54, β‑asarone (an ether found in some plants)125,
nucleoside antibiotics that activation (with rapamycin) or suppression (with 3‑MA, edavarone (a free radical scavenger)120, 2‑methoxye-
induce endoplasmic reticulum bafilomycin A1, mdivi‑1, KU55933 or AMPK inhibitors) stradiol (an endogenous metabolite of oestrogen that
stress by blocking N‑linked enhanced or blunted, respectively, neuroprotection in inhibits hypoxia-inducible factor 1)124, NAD+ (REF. 150),
glycosylation.
various experimental models of preconditioning. These glial cell-derived neurotrophic factor (GDNF) and
Ischaemic penumbra models include the following: mice pretreated with hepatocyte growth factor (HGF)126. Finally, the brains of
Ischaemic tissue potentially thapsigargin or tunicamycin and subsequently subjected mice lacking nuclear factor of κ-light poly­peptide gene
destined to infarction but not to tMCAO146; rats subjected to ischaemic precondition- enhancer in B cells 1 (Nfkb1) manifested exacerbated
irreversibly injured in the ing followed by pMCAO132,135,139,147; rats administered autophagic responses and were more sensitive to tMCAO
course of stroke. The ischaemic
penumbra is the main target of
with metformin and then exposed to pMCAO133; and than the brains of wild-type mice119 (Supplementary
acute therapeutic interventions rats placed in hyperbaric oxygen and then subjected to information S2 (table)). However, no causal relation-
in stroke patients. tMCAO137 (Supplementary information S1 (table)). ship between the neuroprotective activity of all these

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

experimental interventions and autophagy inhibition has Pharmacological activation of autophagy with rapamy-
been established, implying that the latter may represent an cin, melatonin, simvastatin, trichostatin A or recombi-
epiphenomenon of the former. nant cystatin C (CST3; an endogenous cysteine protease
In summary, the current literature does not allow us inhibitor) also limited brain damage in rats subjected
to conclude whether autophagy mediates bona fide cyto- to SAH168–172. In this setting, inhibiting autophagy with
protective or cytotoxic effects in adult rodents subjected 3‑MA or wortmannin either aggravated SAH-dependent
to ischaemic episodes. As autophagy appears to mediate acute neurodegeneration168,170 or abolished neuroprotec-
neurotoxic effects in the course of neonatal hypoxia– tion conferred by autophagy-activating interventions172.
ischaemia, developmental stage may partially account Taken together, these observations suggest that autophagy
for the apparently contradictory findings reported here. efficiently counteracts acute neurodegeneration caused by
However, we fear that in many cases, drawing solid con- SCI and SAH (Supplementary information S1 (table)).
clusions is complicated by the use of pharmacological Accumulating evidence indicates that proficient auto-
agents with limited specificity and the lack of accurate phagic responses also protect the CNS from the delete-
flux determinations. It will be important to tackle the rious effects of TBI. Becn1+/− mice exhibited decreased
impact of autophagy on adult stroke with robust meth- neuronal survival and limited functional recovery
odological approaches that take these two points into upon hemicerebellectomy compared with wild-type
adequate consideration. mice, and such effects were insensitive to rapamycin
administration173. Conversely, rapamycin improved
Autophagy in neurotrauma the hemicerebellectomy-induced neurological deficits
Several observational studies have associated distinct in wild-type mice173, highlighting a mechanistic link
forms of neurotrauma — including spinal cord injury between the neuroprotective effects of rapamycin and
(SCI), subarachnoid haemorrhage (SAH) and traumatic autophagy. Rapamycin administration post-TBI also
brain injury (TBI) — with morphological and biochem- limited neurological damage in mice subjected to closed-
ical markers of autophagy (see above), both in experi- head injury174 and weight-drop damage175. Similar results
mental samples from rodents153–160 and in human biopsy were obtained in mice and rats receiving other phar-
samples153. Although only two of these studies attempted macological activators of autophagy, including mela-
to elucidate whether these biomarkers reflected the acti- tonin176, methylene blue177, luteolin (a plant flavonoid)178
vation of a proficient autophagic response or the blockade and 17‑allylamino-demethoxygeldanamycin (17‑AAG)179.
of autophagic degradation, both favoured the latter inter- Importantly, the neuroprotective effects of melatonin and
pretation over the former 154,159. Indeed, motor neurons 17‑AAG were abolished in the presence of 3‑MA176,179, and
of rats subjected to contusive SCI and cortical neurons of those of 17‑AAG were exacerbated by the concomitant
mice subjected to TBI exhibited an accumulation administration of rapamycin179. Similarly, 3‑MA blunted
of LC3‑containing autophagosomes and defective p62 the well-characterized beneficial effects of post-injury
degradation starting at day 1 post-trauma154,159. hypothermia in a fluid percussion rat model of TBI180,
Results from various interventional studies in ani- suggesting that at least part of the neuroprotective activity
mal models of neurotrauma support the tenet that of hypothermia may be associated with the activation of
autophagy exerts robust cytoprotective effects (but is autophagy (Supplementary information S1 (table)).
usually impaired) in the course of neurotrauma, and Nevertheless, various pharmacological inhibitors of
hence favours functional recovery. Rapamycin pre- autophagy also have neuroprotective activity in rodent
served neuronal survival, promoted axonal regeneration models of TBI. In particular, post-injury chloroquine
towards the lesion and improved the restoration of hind- reduced brain oedema and improved neurological recov-
limb function in rodents subjected to SCI161,162. Similar ery in rats subjected to controlled cortical impact 181.
effects were observed with other autophagy-activating Moreover, pretreatment with 3‑MA or bafilomycin A1
interventions, including the post-SCI administration of attenuated neuronal damage and improved behavioural
a splicing isoform of human vascular endothelial growth outcomes in a mouse weight-drop model of TBI182.
factor A (VEGFA165)163, retinoic acid (which also limited Similar observations were made in mice receiving (pre-
the disruption of the blood–spinal cord barrier)164 and TBI) an autophagy-blocking peptide derived from the
simvastatin (an anti-atherosclerotic drug)165. Notably, adipocytokine apelin183. In addition, the ability of various
the neuroprotection afforded by VEGFA165 and retinoic molecules to limit histological damage and behavioural
17‑Allylamino-
demethoxygeldanamycin
acid was abolished by the concomitant administration deficits in mice or rats subjected to TBI has been correl-
(17‑AAG). A derivative of of 3‑MA163 and chloroquine164, respectively, reinforcing atively linked to the inhibition of autophagy. Such agents
geldanamycin that targets heat the mechanistic link with autophagy in this setting. include hydrogen sulfide184, the chemical inhibitor of
shock protein 90 kDa-α family Rapamycin limited spinal cord damage and improved the necroptosis necrostatin-1 (REF. 185), the β-lactam anti­
class A member 1 (HSP90AA1;
recovery of locomotor activity post-SCI, even in diabetic biotic ceftriaxone186, the antioxidant γ‑glutamylcysteinyl
also known as HSP90).
rats, which are generally more sensitive to neurological ethyl ester 187 and the antidiabetic drug rosiglitazone188
Necroptosis damage than wild-type rats166. Moreover, both 3‑MA (Supplementary information S2 (table)). However, no
A specific type of regulated cell administration and the depletion of BECN1 exacerbated mechanistic connections have been established between
death that depends on the the percentage of rat primary spinal cord neurons suc- the neuroprotective activity of these molecules and
activity of receptor-interacting
serine/threonine kinase 3
cumbing to mechanical injury in vitro, whereas the over- autophagy inhibition, and some of these compounds
(RIPK3) and mixed lineage expression of BECN1 or the addition of rapamycin to are actually known to activate autophagy, at least in spe-
kinase domain-like (MLKL). the culture medium exerted neuroprotective effects167. cific settings. Thus, the precise role of autophagy in the

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Acute neurotoxicity Traumatic damage Adult stroke Excitotoxic challenge Neonatal asphyxia

mAMP SAH tMCAO Glutamate Hypoxic–ischaemic


Ethanol SCI pMCAO NMDA challenge
Cocaine TBI 4VO Kainic acid

Autophagy-dependent Ambiguous impact Autophagy-mediated


neuroprotection on acute brain damage neurotoxicity

Figure 3 | Impact of autophagy on acute brain injury. Neurons die in response to acuteNature injury while activating
Reviews | Neuroscience
a robust autophagic response. In some pathophysiologically relevant settings, including intoxication by various
psychoactive agents, spinal cord injury (SCI) and subarachnoid haemorrhage (SAH), autophagy mediates a clear
neuroprotective activity (green). In other scenarios, such as neonatal hypoxic–ischaemic encephalopathy, it is evident
that autophagic cell death by autosis actively contributes to neuronal demise (red). However, it remains to be
determined whether autophagy has a cytoprotective or a cytotoxic role in neurons subjected to various other forms of
acute injury, including cocaine intoxication, adult stroke and traumatic brain injury (TBI) (blue). This dichotomy reflects
the existence of a highly heterogeneous (and sometimes contradictory) literature based on experimental models and
biomarkers of autophagy that, at least in some cases, do not allow clear conclusions to be drawn. 4VO, four-vessel
occlusion; mAMP, methamphetamine; pMCAO, permanent middle cerebral artery occlusion; tMCAO, temporary
middle cerebral artery occlusion.

response of the CNS to TBI remains to be elucidated or with genetic approaches targeting a single component
(Supplementary information S1,S2 (tables)). Part of the of the autophagic machinery, even though each of these
apparent discrepancy between the results discussed here proteins is likely to participate in processes unrelated to
may be accounted for by context-dependent parameters, autophagy 189. Moreover, standard genetic approaches
including whether autophagy was modulated according involve the lifelong inhibition of autophagy in a specific
to a prophylactic or therapeutic paradigm. However, we tissue, which may be associated with a general rewir-
fear that method­ological issues concerning the specific- ing of cellular metabolism7. Fourth, at least part of the
ity of autophagy-modulating experimental interventions long-term functional impairment caused by acute brain
and the absence of accurate flux assessments may be the injury results from local inflammatory responses that
main cause of such heterogeneity. are initiated by microglia in response to danger signals
emitted by dying neurons190. It therefore remains possi-
Conclusions and perspectives ble that the effects attributed to activators or inhibitors
As discussed above, the impact of autophagy on certain of autophagy (at least partially) arise from: the impact of
forms of acute brain damage, including intoxication autophagy on the release of danger signals99; the extran-
with various psychoactive drugs, SCI, SAH and neona- euronal control of inflammation by autophagy 13; and/or
tal hypoxia–ischaemia has been clarified. However, how the ability of pharmacological modulators of autophagy to
autophagic responses modulate the neurotoxic effects of influence inflammatory processes as an off-target effect.
various other forms of acute brain damage — includ- One relevant example of a pharmacological modulator of
ing excitotoxicity, cocaine intoxication, adult stroke and autophagy that has an anti-inflammatory effect is chloro-
TBI — remains to be precisely elucidated (FIG. 3). Indeed, quine, as its anti-inflammatory activity is harnessed for
the data published in the past two decades on the topic the treatment of systemic lupus erythematosus191. It will be
are relatively heterogeneous and sometimes overtly important to determine to what extent the cytoprotective
contradictory. Although part of this apparent discrep- and anti-inflammatory functions of autophagy influence
ancy may stem from the heterogeneity of experimental the context-dependent impact of this process on acute
variables (including the type, intensity and duration of brain injury.
neuro­toxic challenges, and the sex and developmental Collectively, these caveats suggest that the current
stage of animals, among others), at least four important literature on this topic should be interpreted with cau-
points should also be taken into consideration. tion. Moreover, these considerations highlight the urgent
First, the intensity and duration of the stimulus may need for a novel wave of investigations that attempt to
determine whether autophagy contributes to the demise dissect the actual impact of autophagy on acute brain
of neurons exposed to acute stress (when neurotoxic injury with rigorous tools. Such tools include flux assays
conditions are intense or prolonged) or instead exerts that measure actual autophagic proficiency (as opposed
Systemic lupus
erythematosus prominent cytoprotective and hormetic effects (when to the accumulation of autophagic markers), pharma-
An autoimmune disease that neurotoxic conditions are weak or restricted in time)29 cological agents with improved specificity and genetic
has a complex aetiology, (BOX 1). Second, most of the studies presented above strategies that target at least two distinct components of
involves multiple organs, lacked accurate flux determinations25. Third, in many of the autophagic machinery in an inducible manner. We
progresses in an unpredictable
manner and is usually treated
the studies discussed above, autophagy was modulated surmise that such experimental approaches will even-
with systemic with pharmacological agents that exhibit a low degree tually clarify the actual effects of autophagy in various
immunosuppressants. of specificity (such as 3‑MA and lysosomal inhibitors)25, forms of acute brain injury.

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

1. Kaur, J. & Debnath, J. Autophagy at the crossroads of 25. Klionsky, D. J. et al. Guidelines for the use and abnormalities in neural-specific Pten knock-out mice.
catabolism and anabolism. Nat. Rev. Mol. Cell Biol. interpretation of assays for monitoring autophagy J. Neurosci. 29, 1773–1783 (2009).
16, 461–472 (2015). (3rd edition). Autophagy 12, 1–222 (2016). 48. Zeng, L. H. et al. Tsc2 gene inactivation causes a more
2. Li, W. W., Li, J. & Bao, J. K. Microautophagy: lesser- This report provides a comprehensive and detailed severe epilepsy phenotype than Tsc1 inactivation in a
known self-eating. Cell. Mol. Life Sci. 69, 1125–1136 set of guidelines on how to select the most mouse model of tuberous sclerosis complex. Hum.
(2012). appropriate assays to monitor autophagic responses Mol. Genet. 20, 445–454 (2011).
3. Cuervo, A. M. & Wong, E. Chaperone-mediated in vitro and in vivo, and on how to correctly interpret 49. Pun, R. Y. et al. Excessive activation of mTOR in
autophagy: roles in disease and aging. Cell Res. 24, the results. postnatally generated granule cells is sufficient to
92–104 (2014). 26. Mehta, A., Prabhakar, M., Kumar, P., Deshmukh, R. & cause epilepsy. Neuron 75, 1022–1034 (2012).
4. Lamb, C. A., Yoshimori, T. & Tooze, S. A. The Sharma, P. L. Excitotoxicity: bridge to various triggers in 50. Dong, X. X. et al. p53 mediates autophagy activation
autophagosome: origins unknown, biogenesis neurodegenerative disorders. Eur. J. Pharmacol. 698, and mitochondria dysfunction in kainic acid-induced
complex. Nat. Rev. Mol. Cell Biol. 14, 759–774 6–18 (2013). excitotoxicity in primary striatal neurons.
(2013). 27. Galluzzi, L., Blomgren, K. & Kroemer, G. Mitochondrial Neuroscience 207, 52–64 (2012).
5. Galluzzi, L. et al. Autophagy in malignant membrane permeabilization in neuronal injury. Nat. 51. Sadasivan, S. et al. Acute NMDA toxicity in cultured
transformation and cancer progression. EMBO J. 34, Rev. Neurosci. 10, 481–494 (2009). rat cerebellar granule neurons is accompanied by
856–880 (2015). 28. Giorgi, F. S., Biagioni, F., Lenzi, P., Frati, A. & Fornai, F. autophagy induction and late onset autophagic cell
6. Komatsu, M. et al. Loss of autophagy in the central The role of autophagy in epileptogenesis and in epilepsy- death phenotype. BMC Neurosci. 11, 21 (2010).
nervous system causes neurodegeneration in mice. induced neuronal alterations. J. Neural Transm. (Vienna) 52. Wang, Y. et al. IGF‑1 alleviates NMDA-induced
Nature 441, 880–884 (2006). 122, 849–862 (2015). excitotoxicity in cultured hippocampal neurons against
This study elegantly demonstrates that the 29. Stamoula, E. et al. Low dose administration of autophagy via the NR2B/PI3K–AKT–mTOR pathway.
neuron-specific loss of a component of the glutamate triggers a non-apoptotic, autophagic J. Cell. Physiol. 229, 1618–1629 (2014).
autophagic machinery (ATG7) in mice causes an response in PC12 cells. Cell Physiol. Biochem. 37, 53. Ginet, V. et al. Involvement of autophagy in hypoxic–
accelerated neurodegenerative process associated 1750–1758 (2015). excitotoxic neuronal death. Autophagy 10, 846–860
with behavioural defects and early mortality. 30. Wang, Y. et al. p53 induction contributes to excitotoxic (2014).
7. Galluzzi, L., Pietrocola, F., Levine, B. & Kroemer, G. neuronal death in rat striatum through apoptotic and This study uses the stereotaxic intrastriatal
Metabolic control of autophagy. Cell 159, autophagic mechanisms. Eur. J. Neurosci. 30, delivery of a lentiviral vector encoding a
1263–1276 (2014). 2258–2270 (2009). Becn1‑specific shRNA to investigate the effect of
8. Galluzzi, L., Bravo-San Pedro, J. M. & Kroemer, G. 31. Borsello, T., Croquelois, K., Hornung, J. P. & Clarke, P. G. autophagic responses in newborn rats subjected to
Organelle-specific initiation of cell death. Nat. Cell N‑methyl-d‑aspartate-triggered neuronal death in severe hypoxia–ischaemia.
Biol. 16, 728–736 (2014). organotypic hippocampal cultures is endocytic, 54. Baek, S. H. et al. Modulation of mitochondrial function
9. Sica, V. et al. Organelle-specific initiation of autophagy. autophagic and mediated by the c‑Jun N‑terminal and autophagy mediates carnosine neuroprotection
Mol. Cell 59, 522–539 (2015). kinase pathway. Eur. J. Neurosci. 18, 473–485 (2003). against ischemic brain damage. Stroke 45,
10. Kroemer, G., Marino, G. & Levine, B. Autophagy and 32. Shacka, J. J. et al. Kainic acid induces early and 2438–2443 (2014).
the integrated stress response. Mol. Cell 40, transient autophagic stress in mouse hippocampus. 55. Martorell-Riera, A. et al. Mfn2 downregulation in
280–293 (2010). Neurosci. Lett. 414, 57–60 (2007). excitotoxicity causes mitochondrial dysfunction and
11. Menzies, F. M., Fleming, A. & Rubinsztein, D. C. 33. Wang, Y. et al. An autophagic mechanism is involved in delayed neuronal death. EMBO J. 33, 2388–2407
Compromised autophagy and neurodegenerative apoptotic death of rat striatal neurons induced by the (2014).
diseases. Nat. Rev. Neurosci. 16, 345–357 (2015). non‑N‑methyl-d‑aspartate receptor agonist kainic acid. 56. Wang, W. et al. MFN2 couples glutamate excitotoxicity
12. Choi, A. M., Ryter, S. W. & Levine, B. Autophagy in Autophagy 4, 214–226 (2008). and mitochondrial dysfunction in motor neurons.
human health and disease. N. Engl. J. Med. 368, 34. Chang, C. F. et al. Melatonin attenuates kainic acid- J. Biol. Chem. 290, 168–182 (2015).
651–662 (2013). induced neurotoxicity in mouse hippocampus via 57. Van Laar, V. S. et al. Glutamate excitotoxicity in
13. Deretic, V., Saitoh, T. & Akira, S. Autophagy in inhibition of autophagy and α-synuclein aggregation. neurons triggers mitochondrial and endoplasmic
infection, inflammation and immunity. Nat. Rev. J. Pineal Res. 52, 312–321 (2012). reticulum accumulation of Parkin, and, in the presence
Immunol. 13, 722–737 (2013). 35. Kulbe, J. R., Mulcahy Levy, J. M., Coultrap, S. J., of N‑acetyl cysteine, mitophagy. Neurobiol. Dis. 74,
14. Liu, Y. et al. Autosis is a Na+,K+-ATPase-regulated Thorburn, A. & Bayer, K. U. Excitotoxic glutamate 180–193 (2015).
form of cell death triggered by autophagy-inducing insults block autophagic flux in hippocampal neurons. 58. Alexander, A. et al. ATM signals to TSC2 in the
peptides, starvation, and hypoxia–ischemia. Proc. Brain Res. 1542, 12–19 (2014). cytoplasm to regulate mTORC1 in response to ROS.
Natl Acad. Sci. USA 110, 20364–20371 (2013). 36. Tian, J. et al. Protection of pyruvate against glutamate Proc. Natl Acad. Sci. USA 107, 4153–4158 (2010).
This study is the first morphological and excitotoxicity is mediated by regulating DAPK1 protein 59. Kim, T. S. et al. The ZFHX3 (ATBF1) transcription
biochemical characterization of autosis, and the complex. PLoS ONE 9, e95777 (2014). factor induces PDGFRB, which activates ATM in the
first demonstration of its pathophysiological 37. Caldero, J. et al. Lithium prevents excitotoxic cell death cytoplasm to protect cerebellar neurons from
relevance in the course of neonatal of motoneurons in organotypic slice cultures of spinal oxidative stress. Dis. Model. Mech. 3, 752–762
hypoxia–ischaemia. cord. Neuroscience 165, 1353–1369 (2010). (2010).
15. Galluzzi, L. et al. Essential versus accessory aspects of 38. Fulceri, F. et al. Autophagy activation in glutamate- 60. Hou, Y. S. et al. Sestrin2 protects dopaminergic cells
cell death: recommendations of the NCCD 2015. Cell induced motor neuron loss. Arch. Ital. Biol. 149, against rotenone toxicity through AMPK-dependent
Death Differ. 22, 58–73 (2015). 101–111 (2011). autophagy activation. Mol. Cell. Biol. 35, 2740–2751
16. Noda, N. N. & Inagaki, F. Mechanisms of autophagy. 39. Perez-Carrion, M. D. et al. Dendrimer-mediated siRNA (2015).
Annu. Rev. Biophys. 44, 101–122 (2015). delivery knocks down Beclin 1 and potentiates NMDA- 61. Sai, Y. et al. 14‑3‑3 proteins in the regulation of
17. Hoyer-Hansen, M. et al. Control of macroautophagy mediated toxicity in rat cortical neurons. J. Neurochem. rotenone-induced neurotoxicity might be via its
by calcium, calmodulin-dependent kinase kinase-β, 120, 259–268 (2012). isoform 14‑3‑3ε’s involvement in autophagy. Cell. Mol.
and Bcl‑2. Mol. Cell 25, 193–205 (2007). 40. Perez-Carrion, M. D. & Cena, V. Knocking down Neurobiol. 33, 1109–1121 (2013).
18. Inoki, K., Li, Y., Zhu, T., Wu, J. & Guan, K. L. TSC2 is HMGB1 using dendrimer-delivered siRNA unveils its 62. Filomeni, G. et al. Neuroprotection of kaempferol by
phosphorylated and inhibited by Akt and key role in NMDA-induced autophagy in rat cortical autophagy in models of rotenone-mediated acute
suppresses mTOR signalling. Nat. Cell Biol. 4, neurons. Pharm. Res. 30, 2584–2595 (2013). toxicity: possible implications for Parkinson’s disease.
648–657 (2002). 41. Yung, L. M. et al. Sphingosine kinase 2 mediates Neurobiol. Aging 33, 767–785 (2012).
19. Kim, J. et al. Differential regulation of distinct Vps34 cerebral preconditioning and protects the mouse 63. Pan, T. et al. Rapamycin protects against rotenone-
complexes by AMPK in nutrient stress and autophagy. brain against ischemic injury. Stroke 43, 199–204 induced apoptosis through autophagy induction.
Cell 152, 290–303 (2013). (2012). Neuroscience 164, 541–551 (2009).
20. Egan, D. F. et al. Phosphorylation of ULK1 (hATG1) by 42. Sheng, R. et al. Preconditioning stimuli induce 64. Wu, Y. et al. Neuroprotection of deferoxamine on
AMP-activated protein kinase connects energy sensing autophagy via sphingosine kinase 2 in mouse rotenone-induced injury via accumulation of HIF‑1α
to mitophagy. Science 331, 456–461 (2011). cortical neurons. J. Biol. Chem. 289, 20845–20857 and induction of autophagy in SH‑SY5Y cells.
These authors demonstrate a direct, (2014). Neurochem. Int. 57, 198–205 (2010).
MTORC1‑independent connection between the 43. Duran, J., Gruart, A., Garcia-Rocha, M., 65. Liang, S., Figtree, G., Aiqun, M. & Ping, Z. GAPDH-
regulator of autophagy, AMPK and one of the Delgado-Garcia, J. M. & Guinovart, J. J. Glycogen knockdown reduce rotenone-induced H9C2 cells death
upstream components of the molecular machinery accumulation underlies neurodegeneration and via autophagy and anti-oxidative stress pathway.
of autophagy, ULK1. autophagy impairment in Lafora disease. Hum. Mol. Toxicol. Lett. 234, 162–171 (2015).
21. Russell, R. C. et al. ULK1 induces autophagy by Genet. 23, 3147–3156 (2014). 66. Colell, A. et al. GAPDH and autophagy preserve
phosphorylating Beclin‑1 and activating VPS34 lipid 44. Sumanont, Y. et al. Effects of manganese complexes of survival after apoptotic cytochrome c release in the
kinase. Nat. Cell Biol. 15, 741–750 (2013). curcumin and diacetylcurcumin on kainic acid-induced absence of caspase activation. Cell 129, 983–997
22. Zalckvar, E. et al. DAP-kinase-mediated phosphorylation neurotoxic responses in the rat hippocampus. Biol. (2007).
on the BH3 domain of beclin 1 promotes dissociation of Pharm. Bull. 30, 1732–1739 (2007). 67. Wang, R. C. et al. Akt-mediated regulation of
beclin 1 from Bcl‑XL and induction of autophagy. EMBO 45. McMahon, J. et al. Impaired autophagy in neurons autophagy and tumorigenesis through Beclin 1
Rep. 10, 285–292 (2009). after disinhibition of mammalian target of rapamycin phosphorylation. Science 338, 956–959 (2012).
23. Tanaka, A. et al. Proteasome and p97 mediate and its contribution to epileptogenesis. J. Neurosci. These authors demonstrate that BECN1 can be
mitophagy and degradation of mitofusins induced by 32, 15704–15714 (2012). directly phosphorylated by AKT1, resulting in
Parkin. J. Cell Biol. 191, 1367–1380 (2010). 46. Zeng, L. H., Xu, L., Gutmann, D. H. & Wong, M. suppressed autophagic responses upon the
24. Narendra, D., Kane, L. A., Hauser, D. N., Fearnley, I. M. Rapamycin prevents epilepsy in a mouse model of sequestration of BECN1 by 14‑3‑3ε.
& Youle, R. J. p62/SQSTM1 is required for Parkin- tuberous sclerosis complex. Ann. Neurol. 63, 68. Su, L. Y. et al. Melatonin attenuates MPTP-induced
induced mitochondrial clustering but not mitophagy; 444–453 (2008). neurotoxicity via preventing CDK5‑mediated
VDAC1 is dispensable for both. Autophagy 6, 47. Zhou, J. et al. Pharmacological inhibition of mTORC1 autophagy and SNCA/α-synuclein aggregation.
1090–1106 (2010). suppresses anatomical, cellular, and behavioral Autophagy 11, 1745–1759 (2015).

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

69. Nopparat, C., Porter, J. E., Ebadi, M. & association with various degrees of synaptic and 112. Li, H. et al. Lithium-mediated long-term
Govitrapong, P. 1‑Methyl‑4‑phenylpyridinium-induced neuritic damage. Antimicrob. Agents Chemother. 60, neuroprotection in neonatal rat hypoxia–ischemia is
cell death via autophagy through a Bcl‑2/Beclin 1 168–179 (2015). associated with anti-inflammatory effects and
complex-dependent pathway. Neurochem. Res. 39, 92. Chandramani Shivalingappa, P., Jin, H., enhanced proliferation and survival of neural stem/
225–232 (2014). Anantharam, V., Kanthasamy, A. & Kanthasamy, A. progenitor cells. J. Cereb. Blood Flow Metab. 31,
70. Liu, K., Shi, N., Sun, Y., Zhang, T. & Sun, X. N‑acetyl cysteine protects against methamphetamine- 2106–2115 (2011).
Therapeutic effects of rapamycin on MPTP-induced induced dopaminergic neurodegeneration via 113. Li, Q. et al. Lithium reduces apoptosis and autophagy
Parkinsonism in mice. Neurochem. Res. 38, 201–207 modulation of redox status and autophagy in after neonatal hypoxia–ischemia. Cell Death Dis. 1,
(2013). dopaminergic cells. Parkinsons Dis. 2012, 424285 e56 (2010).
71. Li, X. Z. et al. Therapeutic effects of valproate (2012). 114. Shi, R. et al. Excessive autophagy contributes to
combined with lithium carbonate on MPTP-induced 93. Li, Y. et al. Taurine attenuates methamphetamine- neuron death in cerebral ischemia. CNS Neurosci.
parkinsonism in mice: possible mediation through induced autophagy and apoptosis in PC12 cells Ther. 18, 250–260 (2012).
enhanced autophagy. Int. J. Neurosci. 123, 73–79 through mTOR signaling pathway. Toxicol. Lett. 215, 115. Lu, Q., Harris, V. A., Kumar, S., Mansour, H. M. &
(2013). 1–7 (2012). Black, S. M. Autophagy in neonatal hypoxia ischemic
72. Park, H. J., Shin, J. Y., Kim, H. N., Oh, S. H. & 94. Nopparat, C., Porter, J. E., Ebadi, M. & brain is associated with oxidative stress. Redox Biol.
Lee, P. H. Neuroprotective effects of mesenchymal Govitrapong, P. The mechanism for the 6, 516–523 (2015).
stem cells through autophagy modulation in a neuroprotective effect of melatonin against 116. Puyal, J., Vaslin, A., Mottier, V. & Clarke, P. G.
parkinsonian model. Neurobiol. Aging 35, methamphetamine-induced autophagy. J. Pineal Res. Postischemic treatment of neonatal cerebral ischemia
1920–1928 (2014). 49, 382–389 (2010). should target autophagy. Ann. Neurol. 66, 378–389
73. Li, L. et al. Parkinson’s disease involves autophagy and 95. Li, I. H. et al. Autophagy activation is involved in (2009).
abnormal distribution of cathepsin L. Neurosci. Lett. 3,4‑methylenedioxymethamphetamine (‘ecstasy’)- 117. Shi, R. Y. et al. BNIP3 interacting with LC3 triggers
489, 62–67 (2011). induced neurotoxicity in cultured cortical neurons. excessive mitophagy in delayed neuronal death in
74. Gonzalez-Polo, R. A. et al. Inhibition of paraquat- PLoS ONE 9, e116565 (2014). stroke. CNS Neurosci. Ther. 20, 1045–1055 (2014).
induced autophagy accelerates the apoptotic cell 96. Chen, G. et al. Autophagy is a protective response to 118. De Angelis, C. & Haupert, G. T. Hypoxia triggers
death in neuroblastoma SH‑SY5Y cells. Toxicol. Sci. ethanol neurotoxicity. Autophagy 8, 1577–1589 release of an endogenous inhibitor of Na+-K+-ATPase
97, 448–458 (2007). (2012). from midbrain and adrenal. Am. J. Physiol. 274,
75. Gonzalez-Polo, R. et al. Silencing DJ‑1 reveals its 97. Wang, H., Bower, K. A., Frank, J. A., Xu, M. & Luo, J. F182–F188 (1998).
contribution in paraquat-induced autophagy. Hypoxic preconditioning alleviates ethanol 119. Li, W. L. et al. The regulatory role of NF‑κB in
J. Neurochem. 109, 889–898 (2009). neurotoxicity: the involvement of autophagy. autophagy-like cell death after focal cerebral ischemia
76. Pan, T. et al. Neuroprotection of rapamycin in Neurotox. Res. 24, 472–477 (2013). in mice. Neuroscience 244, 16–30 (2013).
lactacystin-induced neurodegeneration via autophagy 98. Pla, A., Pascual, M., Renau-Piqueras, J. & Guerri, C. 120. Liu, N., Shang, J., Tian, F., Nishi, H. & Abe, K. In vivo
enhancement. Neurobiol. Dis. 32, 16–25 (2008). TLR4 mediates the impairment of ubiquitin– optical imaging for evaluating the efficacy of
77. Guo, M. L. et al. Cocaine-mediated microglial proteasome and autophagy–lysosome pathways edaravone after transient cerebral ischemia in mice.
activation involves the ER stress–autophagy axis. induced by ethanol treatment in brain. Cell Death Dis. Brain Res. 1397, 66–75 (2011).
Autophagy 11, 995–1009 (2015). 5, e1066 (2014). 121. Mehta, S. L. et al. Manganese superoxide dismutase
78. Cao, L. et al. Cocaine-mediated autophagy in 99. Kroemer, G., Galluzzi, L., Kepp, O. & Zitvogel, L. deficiency exacerbates ischemic brain damage under
astrocytes involves sigma 1 receptor, PI3K, mTOR, Immunogenic cell death in cancer therapy. Annu. Rev. hyperglycemic conditions by altering autophagy.
Atg5/7, Beclin‑1 and induces type II programmed cell Immunol. 31, 51–72 (2013). Transl. Stroke Res. 2, 42–50 (2011).
death. Mol. Neurobiol. http://dx.doi.org/10.1007/ This is an exhaustive review of the molecular and 122. Li, L. et al. Transmembrane protein 166 regulates
s12035-015-9377-x (2015). cellular mechanisms whereby RCD can be perceived autophagic and apoptotic activities following focal
79. Guha, P., Harraz, M. M. & Snyder, S. H. Cocaine elicits as immunogenic by the immune system and hence cerebral ischemic injury in rats. Exp. Neurol. 234,
autophagic cytotoxicity via a nitric oxide–GAPDH initiate an adaptive immune response. 181–190 (2012).
signaling cascade. Proc. Natl Acad. Sci. USA 113, 100. Vucicevic, L. et al. Autophagy inhibition uncovers the 123. Wen, Y. D. et al. Neuronal injury in rat model of
1417–1422 (2016). neurotoxic action of the antipsychotic drug olanzapine. permanent focal cerebral ischemia is associated with
80. Sutton, L. P. & Caron, M. G. Essential role of D1R in Autophagy 10, 2362–2378 (2014). activation of autophagic and lysosomal pathways.
the regulation of mTOR complex1 signaling induced 101. Fabrizi, C. et al. Role of autophagy inhibitors and Autophagy 4, 762–769 (2008).
by cocaine. Neuropharmacology 99, 610–619 inducers in modulating the toxicity of trimethyltin in 124. Xin, X. Y. et al. 2‑Methoxyestradiol attenuates
(2015). neuronal cell cultures. J. Neural Transm. (Vienna) 119, autophagy activation after global ischemia. Can.
81. Wu, J., McCallum, S. E., Glick, S. D. & Huang, Y. 1295–1305 (2012). J. Neurol. Sci. 38, 631–638 (2011).
Inhibition of the mammalian target of rapamycin 102. Fabrizi, C. et al. Impairment of the autophagic flux in 125. Liu, L. et al. β-asarone attenuates ischemia–
pathway by rapamycin blocks cocaine-induced astrocytes intoxicated by trimethyltin. reperfusion-induced autophagy in rat brains via
locomotor sensitization. Neuroscience 172, 104–109 Neurotoxicology 52, 12–22 (2015). modulating JNK, p‑JNK, Bcl‑2 and Beclin 1. Eur.
(2011). 103. Ginet, V. et al. Dying neurons in thalamus of J. Pharmacol. 680, 34–40 (2012).
82. Cubells, J. F., Rayport, S., Rajendran, G. & Sulzer, D. asphyxiated term newborns and rats are autophagic. 126. Shang, J. et al. Antiapoptotic and antiautophagic
Methamphetamine neurotoxicity involves vacuolation Ann. Neurol. 76, 695–711 (2014). effects of glial cell line-derived neurotrophic factor and
of endocytic organelles and dopamine-dependent 104. Xie, C. et al. Neuroprotection by selective neuronal hepatocyte growth factor after transient middle
intracellular oxidative stress. J. Neurosci. 14, deletion of Atg7 in neonatal brain injury. Autophagy cerebral artery occlusion in rats. J. Neurosci. Res. 88,
2260–2271 (1994). 12, 410–423 (2016). 2197–2206 (2010).
83. Castino, R. et al. Suppression of autophagy 105. Ginet, V., Puyal, J., Clarke, P. G. & Truttmann, A. C. 127. Zheng, Y. et al. Inhibition of autophagy contributes to
precipitates neuronal cell death following low doses of Enhancement of autophagic flux after neonatal melatonin-mediated neuroprotection against transient
methamphetamine. J. Neurochem. 106, 1426–1439 cerebral hypoxia–ischemia and its region-specific focal cerebral ischemia in rats. J. Pharmacol. Sci. 124,
(2008). relationship to apoptotic mechanisms. Am. J. Pathol. 354–364 (2014).
84. Pasquali, L. et al. Role of autophagy during 175, 1962–1974 (2009). 128. Qin, A. P. et al. Autophagy was activated in injured
methamphetamine neurotoxicity. Ann. NY Acad. Sci. 106. Koike, M. et al. Inhibition of autophagy prevents astrocytes and mildly decreased cell survival following
1139, 191–196 (2008). hippocampal pyramidal neuron death after hypoxic– glucose and oxygen deprivation and focal cerebral
85. Lenzi, P. et al. A subcellular analysis of genetic ischemic injury. Am. J. Pathol. 172, 454–469 ischemia. Autophagy 6, 738–753 (2010).
modulation of PINK1 on mitochondrial alterations, (2008). 129. Cui, D. R. et al. Propofol prevents cerebral ischemia-
autophagy and cell death. Arch. Ital. Biol. 150, 107. Zhu, C. et al. The influence of age on apoptotic and triggered autophagy activation and cell death in the
194–217 (2012). other mechanisms of cell death after cerebral rat hippocampus through the NF‑κB/p53 signaling
86. Lin, M. et al. Methamphetamine-induced neurotoxicity hypoxia–ischemia. Cell Death Differ. 12, 162–176 pathway. Neuroscience 246, 117–132 (2013).
linked to ubiquitin–proteasome system dysfunction (2005). 130. Zhou, H. et al. N‑acetyl-serotonin offers
and autophagy-related changes that can be This study is the first to report the lipidation of LC3 neuroprotection through inhibiting mitochondrial
modulated by protein kinase Cδ in dopaminergic in the brains of mice subjected to unilateral death pathways and autophagic activation in
neuronal cells. Neuroscience 210, 308–332 (2012). hypoxia–ischaemia. experimental models of ischemic injury. J. Neurosci.
87. Ma, J. et al. Methamphetamine induces autophagy as 108. Takada, S. H. et al. Neonatal anoxia in rats: 34, 2967–2978 (2014).
a pro-survival response against apoptotic endothelial hippocampal cellular and subcellular changes related 131. Dohi, E. et al. Hypoxic stress activates chaperone-
cell death through the Kappa opioid receptor. Cell to cell death and spatial memory. Neuroscience 284, mediated autophagy and modulates neuronal cell
Death Dis. 5, e1099 (2014). 247–259 (2015). survival. Neurochem. Int. 60, 431–442 (2012).
88. Sinchai, T. et al. Caffeine potentiates 109. Carloni, S., Buonocore, G. & Balduini, W. Protective 132. Sheng, R. et al. Autophagy activation is associated
methamphetamine-induced toxicity both in vitro and role of autophagy in neonatal hypoxia–ischemia with neuroprotection in a rat model of focal cerebral
in vivo. Neurosci. Lett. 502, 65–69 (2011). induced brain injury. Neurobiol. Dis. 32, 329–339 ischemic preconditioning. Autophagy 6, 482–494
89. Pitaksalee, R. et al. Autophagy inhibition by caffeine (2008). (2010).
increases toxicity of methamphetamine in SH‑SY5Y 110. Carloni, S., Buonocore, G., Longini, M., Proietti, F. & 133. Jiang, T. et al. Acute metformin preconditioning
neuroblastoma cell line. Neurotox. Res. 27, 421–429 Balduini, W. Inhibition of rapamycin-induced confers neuroprotection against focal cerebral
(2015). autophagy causes necrotic cell death associated with ischaemia by pre-activation of AMPK-dependent
90. Moon, J. H. et al. Caffeine prevents human prion Bax/Bad mitochondrial translocation. Neuroscience autophagy. Br. J. Pharmacol. 171, 3146–3157
protein-mediated neurotoxicity through the induction 203, 160–169 (2012). (2014).
of autophagy. Int. J. Mol. Med. 34, 553–558 (2014). 111. Carloni, S. et al. Activation of autophagy and Akt/ 134. Wang, P. et al. Induction of autophagy contributes to
91. Sanchez, A. B. et al. Antiretrovirals, CREB signaling play an equivalent role in the the neuroprotection of nicotinamide
methamphetamine, and HIV‑1 envelope protein neuroprotective effect of rapamycin in neonatal phosphoribosyltransferase in cerebral ischemia.
gp120 compromise neuronal energy homeostasis in hypoxia–ischemia. Autophagy 6, 366–377 (2010). Autophagy 8, 77–87 (2012).

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

135. Sheng, R. et al. Autophagy regulates endoplasmic 154. Sarkar, C. et al. Impaired autophagy flux is associated 177. Zhao, M., Liang, F., Xu, H., Yan, W. & Zhang, J.
reticulum stress in ischemic preconditioning. with neuronal cell death after traumatic brain injury. Methylene blue exerts a neuroprotective effect against
Autophagy 8, 310–325 (2012). Autophagy 10, 2208–2222 (2014). traumatic brain injury by promoting autophagy and
136. Fan, J. et al. Ischemic preconditioning enhances 155. Liu, C. L., Chen, S., Dietrich, D. & Hu, B. R. Changes in inhibiting microglial activation. Mol. Med. Rep. 13,
autophagy but suppresses autophagic cell death in rat autophagy after traumatic brain injury. J. Cereb. Blood 13–20 (2016).
spinal neurons following ischemia–reperfusion. Brain Flow Metab. 28, 674–683 (2008). 178. Xu, J. et al. Post-traumatic administration of luteolin
Res. 1562, 76–86 (2014). 156. Diskin, T. et al. Closed head injury induces protects mice from traumatic brain injury: implication
137. Yan, W. et al. Autophagy activation is involved in upregulation of Beclin 1 at the cortical site of injury. of autophagy and inflammation. Brain Res. 1582,
neuroprotection induced by hyperbaric oxygen J. Neurotrauma 22, 750–762 (2005). 237–246 (2014).
preconditioning against focal cerebral ischemia in rats. 157. Park, Y. et al. Chaperone-mediated autophagy after 179. Ma, L. et al. 17AAG improves histological and
Brain Res. 1402, 109–121 (2011). traumatic brain injury. J. Neurotrauma 32, functional outcomes in a rat CCI model through
138. Gao, B. et al. The endoplasmic reticulum stress 1449–1457 (2015). autophagy activation and apoptosis attenuation.
inhibitor salubrinal inhibits the activation of 158. Kanno, H., Ozawa, H., Sekiguchi, A. & Itoi, E. Spinal Neurosci. Lett. 599, 1–6 (2015).
autophagy and neuroprotection induced by brain cord injury induces upregulation of Beclin 1 and 180. Jin, Y., Lei, J., Lin, Y., Gao, G. Y. & Jiang, J. Y.
ischemic preconditioning. Acta Pharmacol. Sin. 34, promotes autophagic cell death. Neurobiol. Dis. 33, Autophagy inhibitor 3‑MA weakens neuroprotective
657–666 (2013). 143–148 (2009). effects of posttraumatic brain injury moderate
139. Jiang, T. et al. Ischemic preconditioning provides 159. Liu, S. et al. Disrupted autophagy after spinal cord hypothermia. World Neurosurg. 88, 433–446
neuroprotection by induction of AMP-activated injury is associated with ER stress and neuronal cell (2016).
protein kinase-dependent autophagy in a rat model of death. Cell Death Dis. 6, e1582 (2015). 181. Cui, C. M. et al. Chloroquine exerts neuroprotection
ischemic stroke. Mol. Neurobiol. 51, 220–229 160. Chen, S., Wu, H., Tang, J., Zhang, J. & Zhang, J. H. following traumatic brain injury via suppression of
(2015). Neurovascular events after subarachnoid hemorrhage: inflammation and neuronal autophagic death. Mol.
140. Jiang, Z. et al. The effects of methylene blue on focusing on subcellular organelles. Acta Neurochir. Med. Rep. 12, 2323–2328 (2015).
autophagy and apoptosis in MRI-defined normal Suppl. 120, 39–46 (2015). 182. Luo, C. L. et al. Autophagy is involved in traumatic
tissue, ischemic penumbra and ischemic core. PLoS 161. Wang, Z. Y., Liu, W. G., Muharram, A., Wu, Z. Y. & brain injury-induced cell death and contributes to
ONE 10, e0131929 (2015). Lin, J. H. Neuroprotective effects of autophagy functional outcome deficits in mice. Neuroscience
141. Zhao, G., Zhang, W., Li, L., Wu, S. & Du, G. induced by rapamycin in rat acute spinal cord injury 184, 54–63 (2011).
Pinocembrin protects the brain against ischemia– model. Neuroimmunomodulation 21, 257–267 183. Bao, H. J., Zhang, L., Han, W. C. & Dai, D. K.
reperfusion injury and reverses the autophagy (2014). Apelin‑13 attenuates traumatic brain injury-induced
dysfunction in the penumbra area. Molecules 19, 162. Goldshmit, Y. et al. Rapamycin increases neuronal damage by suppressing autophagy. Neurochem. Res.
15786–15798 (2014). survival, reduces inflammation and astrocyte 40, 89–97 (2015).
142. Wang, P. et al. ARRB1/β-arrestin‑1 mediates proliferation after spinal cord injury. Mol. Cell 184. Zhang, M. et al. Hydrogen sulfide offers
neuroprotection through coordination of Neurosci. 68, 82–91 (2015). neuroprotection on traumatic brain injury in parallel
BECN1‑dependent autophagy in cerebral ischemia. 163. Wang, H. et al. VEGF inhibits the inflammation in with reduced apoptosis and autophagy in mice. PLoS
Autophagy 10, 1535–1548 (2014). spinal cord injury through activation of autophagy. ONE 9, e87241 (2014).
143. Papadakis, M. et al. Tsc1 (hamartin) confers Biochem. Biophys. Res. Commun. 464, 453–458 185. Wang, Y. Q. et al. Necrostatin‑1 suppresses autophagy
neuroprotection against ischemia by inducing (2015). and apoptosis in mice traumatic brain injury model.
autophagy. Nat. Med. 19, 351–357 (2013). 164. Zhou, Y. et al. Retinoic acid prevents disruption of Neurochem. Res. 37, 1849–1858 (2012).
These authors elegantly demonstrate that the blood–spinal cord barrier by inducing autophagic flux 186. Cui, C. et al. Neuroprotective effect of ceftriaxone in a
inhibition of autophagy by the stereotaxic delivery after spinal cord injury. Neurochem. Res. 41, rat model of traumatic brain injury. Neurol. Sci. 35,
of a lentiviral vector encoding a Tsc1‑targeting 813–825 (2015). 695–700 (2014).
shRNA aggravates the neurotoxicity of tMCAO in 165. Gao, K. et al. Neuroprotective effect of simvastatin via 187. Lai, Y. et al. Autophagy is increased after traumatic
adult rats. inducing the autophagy on spinal cord injury in the rat brain injury in mice and is partially inhibited by the
144. Wang, P. et al. Nicotinamide model. Biomed. Res. Int. 2015, 260161 (2015). antioxidant γ-glutamylcysteinyl ethyl ester. J. Cereb.
phosphoribosyltransferase protects against ischemic 166. Zhou, K. L. et al. Stimulation of autophagy promotes Blood Flow Metab. 28, 540–550 (2008).
stroke through SIRT1‑dependent adenosine functional recovery in diabetic rats with spinal cord 188. Yao, J., Zheng, K. & Zhang, X. Rosiglitazone exerts
monophosphate-activated kinase pathway. Ann. injury. Sci. Rep. 5, 17130 (2015). neuroprotective effects via the suppression of
Neurol. 69, 360–374 (2011). 167. Wang, Z. Y., Lin, J. H., Muharram, A. & Liu, W. G. neuronal autophagy and apoptosis in the cortex
145. Zhang, X. et al. Cerebral ischemia–reperfusion- Beclin‑1‑mediated autophagy protects spinal cord following traumatic brain injury. Mol. Med. Rep. 12,
induced autophagy protects against neuronal injury by neurons against mechanical injury-induced apoptosis. 6591–6597 (2015).
mitochondrial clearance. Autophagy 9, 1321–1333 Apoptosis 19, 933–945 (2014). 189. Subramani, S. & Malhotra, V. Non-autophagic roles of
(2013). 168. Jing, C. H. et al. Autophagy activation is associated autophagy-related proteins. EMBO Rep. 14,
146. Zhang, X. et al. Endoplasmic reticulum stress induced with neuroprotection against apoptosis via a 143–151 (2013).
by tunicamycin and thapsigargin protects against mitochondrial pathway in a rat model of 190. Witcher, K. G., Eiferman, D. S. & Godbout, J. P.
transient ischemic brain injury: involvement of subarachnoid hemorrhage. Neuroscience 213, Priming the inflammatory pump of the CNS after
PARK2‑dependent mitophagy. Autophagy 10, 144–153 (2012). traumatic brain injury. Trends Neurosci. 38, 609–620
1801–1813 (2014). 169. Liu, Y. et al. Induction of autophagy by cystatin C: (2015).
147. Su, J., Zhang, T., Wang, K., Zhu, T. & Li, X. Autophagy a potential mechanism for prevention of cerebral 191. Lee, S. J., Silverman, E. & Bargman, J. M. The role of
activation contributes to the neuroprotection of vasospasm after experimental subarachnoid antimalarial agents in the treatment of SLE and lupus
remote ischemic perconditioning against focal cerebral hemorrhage. Eur. J. Med. Res. 18, 21 (2013). nephritis. Nat. Rev. Nephrol. 7, 718–729 (2011).
ischemia in rats. Neurochem. Res. 39, 2068–2077 170. Zhao, H. et al. Role of autophagy in early brain injury 192. Marino, G., Niso-Santano, M., Baehrecke, E. H. &
(2014). after subarachnoid hemorrhage in rats. Mol. Biol. Rep. Kroemer, G. Self-consumption: the interplay of
148. Zhou, X. et al. GSK‑3β inhibitors suppressed 40, 819–827 (2013). autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 15,
neuroinflammation in rat cortex by activating 171. Chen, J. et al. Melatonin-enhanced autophagy 81–94 (2014).
autophagy in ischemic brain injury. Biochem. Biophys. protects against neural apoptosis via a mitochondrial This article provides a comprehensive review of the
Res. Commun. 411, 271–275 (2011). pathway in early brain injury following a intricate crosstalk that exists between autophagy
149. Buckley, K. M. et al. Rapamycin up‑regulation of subarachnoid hemorrhage. J. Pineal Res. 56, 12–19 and various forms of RCD.
autophagy reduces infarct size and improves (2014). 193. Palikaras, K., Lionaki, E. & Tavernarakis, N.
outcomes in both permanent MCAL, and embolic 172. Shao, A. et al. Enhancement of autophagy by histone Coordination of mitophagy and mitochondrial
MCAO, murine models of stroke. Exp. Transl Stroke deacetylase inhibitor trichostatin A ameliorates biogenesis during ageing in C. elegans. Nature 521,
Med. 6, 8 (2014). neuronal apoptosis after subarachnoid hemorrhage in 525–528 (2015).
150. Zheng, C. et al. NAD+ administration decreases rats. Mol. Neurobiol. 53, 18–27 (2016). 194. Martins, I., Galluzzi, L. & Kroemer, G. Hormesis, cell
ischemic brain damage partially by blocking These authors provide robust pharmacological and death and aging. Aging (Albany NY) 3, 821–828
autophagy in a mouse model of brain ischemia. genetic evidence supporting the ability of (2011).
Neurosci. Lett. 512, 67–71 (2012). autophagy to limit neuronal loss following 195. Madeo, F., Pietrocola, F., Eisenberg, T. & Kroemer, G.
151. Kubota, C. et al. Constitutive reactive oxygen species hemicerebellectomy. Caloric restriction mimetics: towards a molecular
generation from autophagosome/lysosome in neuronal 173. Viscomi, M. T. et al. Stimulation of autophagy by definition. Nat. Rev. Drug Discov. 13, 727–740
oxidative toxicity. J. Biol. Chem. 285, 667–674 (2010). rapamycin protects neurons from remote (2014).
152. Zheng, Y. Q., Liu, J. X., Li, X. Z., Xu, L. & Xu, Y. G. degeneration after acute focal brain damage. 196. Madeo, F., Tavernarakis, N. & Kroemer, G. Can
RNA interference-mediated downregulation of Beclin1 Autophagy 8, 222–235 (2012). autophagy promote longevity? Nat. Cell Biol. 12,
attenuates cerebral ischemic injury in rats. Acta 174. Erlich, S., Alexandrovich, A., Shohami, E. & 842–846 (2010).
Pharmacol. Sin. 30, 919–927 (2009). Pinkas-Kramarski, R. Rapamycin is a neuroprotective 197. Kroemer, G. & Levine, B. Autophagic cell death:
This work demonstrates that a lentiviral vector treatment for traumatic brain injury. Neurobiol. Dis. the story of a misnomer. Nat. Rev. Mol. Cell Biol. 9,
encoding a Becn1‑specific shRNA stereotaxically 26, 86–93 (2007). 1004–1010 (2008).
injected into the ipsilateral lateral ventricle 175. Song, Q., Xie, D., Pan, S. & Xu, W. Rapamycin protects 198. Berry, D. L. & Baehrecke, E. H. Growth arrest and
decreases infarct volume, histological injury and neurons from brain contusion-induced inflammatory autophagy are required for salivary gland cell
neurological deficits in adult rats subjected to reaction via modulation of microglial activation. Mol. degradation in Drosophila. Cell 131, 1137–1148
tMCAO. Med. Rep. 12, 7203–7210 (2015). (2007).
153. Clark, R. S. et al. Autophagy is increased in mice after 176. Ding, K. et al. Melatonin protects the brain from This paper is one of the first to elegantly
traumatic brain injury and is detectable in human apoptosis by enhancement of autophagy after demonstrate the causal contribution of autophagy
brain after trauma and critical illness. Autophagy 4, traumatic brain injury in mice. Neurochem. Int. 91, to the demise of salivary gland cells in developing
88–90 (2008). 46–54 (2015). Drosophila melanogaster larvae.

NATURE REVIEWS | NEUROSCIENCE ADVANCE ONLINE PUBLICATION | 17


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

199. Menger, L. et al. Cardiac glycosides exert anticancer Recherche sur le Cancer (ARC); Cancéropôle Ile‑de‑France; the the Swedish Brain Foundation; the Swedish Board of Radiation
effects by inducing immunogenic cell death. Sci. Transl French Institut National du Cancer (INCa); the Fondation Safety; the Frimurare Barnhuset Foundation in Stockholm,
Med. 4, 143ra199 (2012). Bettencourt Schueller; the Fondation de France; the French Sweden; and governmental grants from the Swedish Agreement
200. Lin, M. H. & Akera, T. Increased (Na+,K+)-ATPase Fondation pour la Recherche Médicale (FRM); the European on Medical Education and Research (Avtal om läkarutbildning
concentrations in various tissues of rats caused by Commission (ArtForce); the European Research Council (ERC); och forskning (ALF)).
thyroid hormone treatment. J. Biol. Chem. 253, the LabEx Immuno-Oncology; the SIRIC (sites de recherche
723–726 (1978). intégrée sur le cancer) Stratified Oncology Cell DNA Repair and Competing interests statement
Tumor Immune Elimination (SOCRATE); the SIRIC Cancer The authors declare no competing interests.
Acknowledgements Research and Personalized Medicine (CARPEM); the Swiss
G.K. is supported by the following agencies and institutes: the Bridge Foundation; the Swiss Institute for Experimental Cancer
French Ligue Contre le Cancer (Équipe Labellisée); the French Research (ISREC); and the Paris Alliance of Cancer Research SUPPLEMENTARY INFORMATION
Agence National de la Recherche (ANR) — Projets Blancs; the Institutes (PACRI). K.B. is supported by the following agencies See online article: S1 (table) | S2 (table)
ANR under the framework of E‑Rare‑2 (the ERA-Net for and institutes: the Swedish Research Council; the Swedish ALL LINKS ARE ACTIVE IN THE ONLINE PDF
Research on Rare Diseases); the French Association pour la Childhood Cancer Foundation; the Swedish Cancer Foundation;

18 | ADVANCE ONLINE PUBLICATION www.nature.com/nrn


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like