You are on page 1of 71

TEÓRICO AUTOFAGIA 1

REGULATION OF MAMMALIAN AUTOPHAGY IN PHYSIOLOGY AND PATHOPHYSIOLOGY

(Macro)autophagy is a bulk degradation term autophagy in 1963. He used the word


process that mediates the clearance of to describe the phenomenon associated
long-lived proteins and organelles. with single- or double-membraned vesicles
Autophagy is initiated by double- that contained cytoplasm, including
membraned structures, which engulf organelles, at various stages of digestion.
portions of cytoplasm. The resulting In this way, autophagy can be
distinguished from heterophagy, where
autophagosomes ultimately fuse with
cells degrade extracellular material. De
lysosomes, where their contents are
Duve's “autophagy” likely described
degraded. Although the macroautophagy, which we will call
term autophagy was first used in 1963, the autophagy, the focus of this review.
field has witnessed dramatic growth in the Autophagy, which is highly conserved from
last 5 years, partly as a consequence of the yeast to humans, is a bulk degradation
discovery of key components of its cellular process involved in the clearance of long-
machinery. In this review we focus on lived proteins and organelles. During
mammalian autophagy, and we give an autophagy, phagophores [also called pre-
overview of the understanding of its autophagosomal structures (PAS)]
machinery and the signaling cascades that elongate and fuse while engulfing a portion
regulate it. As recent studies have also of cytoplasm within double-membraned
vesicles, called autophagosomes. The
shown that autophagy is critical in a range
autophagosomes first fuse with
of normal human physiological processes,
endosomes to form hybrid organelles
and defective autophagy is associated with
called amphisomes that later fuse with
diverse diseases, including acidic lysosomes where the entrapped
neurodegeneration, lysosomal storage cytosolic contents are degraded (Fig. 1).
diseases, cancers, and Crohn's disease, we There are two other forms of autophagy,
discuss the roles of autophagy in health which we will not review, but will briefly
and disease, while trying to critically mention. The first, chaperone-mediated
evaluate if the coincidence between autophagy (CMA), involves selective
autophagy and these conditions is causal translocation of the cytosolic proteins that
or an epiphenomenon. Finally, we consider are marked by a pentapeptide motif with a
the possibility of autophagy upregulation consensus sequence similar to KFERQ
as a therapeutic approach for various across the lysosomal membrane. Cytosolic
chaperones aid in the target recognition
conditions.
and unfolding, and a multimer comprising
I. INTRODUCTION the lysosomal protein LAMP-2a
(lysosomal-associated membrane protein-
The word autophagy is derived from the 2a) subunits is thought to be rate-limiting
Greek roots “auto” (self) and “phagy” for target translocation into lysosomes.
(eating) and broadly refers to the cellular The second is microautophagy, a poorly
catabolic processes in which cytoplasmic understood phenomenon in mammalian
target material is transported to lysosomes cells, where the cytosolic contents are
for degradation. Christian de Duve, who engulfed by direct invagination of the
was awarded the Nobel Prize for his work lysosomal membranes into tubulovesicular
on lysosomes, first used the structures.
TEÓRICO AUTOFAGIA 2
TEÓRICO AUTOFAGIA 3

Fig. 1.Mammalian macroautophagy (autophagy). Autophagy involves the degradation of


cytosolic proteins and organelles in the lysosomes via double-membraned structures called
autophagosomes. Pre-autophagosomal structures (PAS) or phagophores elongate and fuse to
form autophagosomes. The membrane source involved in autophagosome biogenesis may
involve contributions from the ER, mitochondria, and the plasma membrane. Two
macromolecular complexes regulate the initiation of PAS formation: mTOR-Atg13-ULK1
complex and Beclin-Vps34 complex. 3-Methyladenine (3-MA) and wortmannin inhibit Vps34.
Several Beclin 1 binding proteins that regulate mammalian autophagy have been identified
(red, inhibit autophagy; green, induce autophagy). Phosphorylation of Bcl-2 results in its
dissociation from Beclin 1 to induce autophagy. The Beclin 1-Bcl-2 interaction was diminished
by the autophagy-inducing agent xestospongin B that however did not affect the IP3R-Bcl-2
interaction. NAF-1 interacts with Bcl-2 at the ER and stabilizes the Bcl-2-Beclin 1 interaction.
Two ubiquitin-like conjugation systems [Atg5-Atg12 conjugation and LC3-phosphatidyl
ethanolamine (PE) conjugation] are involved in the elongation of PAS. The Atg5-Atg12
congugation involves Atg7 (E1-like) and Atg10 (E2-like), while Atg7 and Atg3 act as the E1-like
and E2-like, respectively, in LC3-PE conjugation. The Atg12-Atg5 is noncovalently conjugated
to Atg16L1 (Atg12-Atg5.Atg16L1), resulting in an 800-kDa complex containing Atg12-
Atg5.Atg16L1 tetramers that are linked via the coiled-coil domain of Atg16L1. The Atg12-
Atg5.Atg16L1 complex exhibits an E3-like activity towards LC3-PE conjugation. Cross-talk
between the two ubiquitin-like systems has been implicated. Beclin 1 complex containing
Rubicon and UVRAG is involved in the fusion step of autophagy.
Until the early 1990s, autophagy was from macromolecules and by assisting the
predominantly studied using clearance of misfolded proteins and
morphological and biochemical methods. damaged organelles, autophagy is vital in a
In 1992, Ohsumi's group demonstrated range of physiological and pathological
that autophagy occurred in Saccharomyces situations, including during early
cerevisiae (baker's yeast). Soon thereafter, embryonic development and neonatal
Ohsumi's lab and others, like Klionsky and starvation, for the degradation of disease-
Thumm, published data using this tractable causing aggregate-prone proteins and in
organism to identify autophagy genes (Atg the clearance of pathogenic bacteria.
genes). The discovery of yeast autophagy Dysfunction in the autophagy pathway has
genes was followed by the identification of been implicated in an increasing number of
mammalian orthologs with similar roles human diseases, from infectious diseases
and provided a series of reagents for to cancer and neurodegeneration (341).
characterizing the molecular machinery of
the system as well as its roles in normal In this review, we aim to broadly outline
physiology and disease (240). the biology of mammalian autophagy and
the key genes involved in the process. We
Autophagy in mammalian systems occurs will consider how autophagy is important
under basal conditions and can be for normal mammalian physiology and its
stimulated by stresses like starvation, roles in a range of disease. Finally, we will
various pathologies, or by treatment with consider the possibility that autophagy
pharmacological agents like rapamycin. In upregulation may be beneficial for certain
addition to its roles in maintaining normal human diseases.
cellular homeostasis by liberating nutrients
TEÓRICO AUTOFAGIA 4

II. AUTOPHAGY MACHINERY membrane source to autophagosomes


during starvation (150a). Recently, we
More than 30 different genes regulating showed that plasma membrane is a key
autophagy have been identified in yeast, contributor for autophagosome precursors
and many of these have mammalian (415a). The formation of new
orthologs. In addition, many genes with autophagosomes requires the activity of
multiple functions, including roles in the class III phosphatidylinositol 3-kinase
autophagy that were not identified in the (PI3K), Vps34. Phosphatidylinositol-3-
relatively specific yeast screens, have been phosphate (PI-3-P), the product of Vps34
identified. These genes can be grouped activity, plays an essential role in the early
according to their functions at key stages stages of the autophagy pathway. Recent
of the autophagy pathway: initiation, studies have identified strong
elongation, maturation, and fusion with colocalization of early autophagosome
the lysosomes. markers in PI-3-P-enriched structures that
were formed upon starvation (8). The
A. Initiation of Autophagosome Formation precise role of PI-3-P is still unclear;
however, this raises the importance of PI-
The membrane source from which 3-P effector proteins in autophagosome
autophagosomes arise is still a matter of formation. Indeed, the recently identified
debate, and there may be multiple mammalian Atg18 homolog WIPI-1 binds
sources. It has been hypothesized that to PI-3-P and is recruited to
autophagosomes can either be generated autophagosomal membranes, especially
de novo from preexisting intracellular upon autophagy induction (401). The small
precursor molecules, or could arise from GTPase Rab5, an early endocytic protein,
other intracellular membrane structures interacts with and activates Vps34
like the endoplasmic reticulum (ER) (8). (58, 483). Inhibition of Vps34 activity by 3-
The latter hypothesis has recently been methyladenine (3-MA) or wortmannin
supported by more evidence suggesting leads to inhibition of autophagosome
that ER could contribute to formation (26, 391, 472).
autophagosome formation (168, 582).
Autophagosomes have frequently been Vps34 is part of the autophagy-regulating
reported to form in the vicinity of ER. macromolecular complex (PI3K complex)
Furthermore, rough ER was found to consisting of Beclin 1/Atg6, Atg14/barkor,
surround both the outer and inner surfaces and p150/Vps15 (Fig. 1) (189, 227, 500).
of the cup-shaped pre-autophagosomal The activity of Vps34 is enhanced by its
structures. Accumulation of these ER- interaction with Beclin 1 (123). Several
associated early autophagic structures was Beclin 1 binding proteins have been
seen in cells that had a block in maturation identified, and disruption of their
of pre-autophagosomal structures. ER interaction with Beclin 1 affects
association with the outer surface was, autophagosome formation. The Beclin 1
however, not observed when the pre- binding partners that induce autophagy
autophagosomal structures matured into include ambra-1 (110), UVRAG (298), and
autophagosomes. Thus the ER may be one bif-1 (506). On the other hand, the binding
of the membrane sources contributing to of the anti-apoptotic proteins Bcl-2 or Bcl-
the maturation of pre-autophagosomal XL to Beclin 1 inhibits autophagy (389).
structures (168, 582). It has recently been Furthermore, the inositol 1,4,5-
suggested that mitochondria may be a trisphosphate receptor (IP3R) that can bind
TEÓRICO AUTOFAGIA 5

to Bcl-2 can also interact with Beclin 1. The Two ubiquitin-like reactions are involved in
IP3R antagonist xestospongin B decreases the elongation of the pre-autophagosomal
this interaction and induces autophagy structures. In the first of the reactions, the
(552). The Beclin 1-Bcl-2 interaction was ubiquitin-like protein Atg12 is covalently
also diminished by xestospongin B, even tagged to Atg5 (342). Atg12 is first
though this drug does not affect the IP3R- activated by Atg7 (E1 ubiquitin activating
Bcl-2 interaction (67). Another component enzyme-like) and then transferred to Atg10
in the IP3R complex, NAF-1 (nutrient- (E2 ubiquitin conjugating enzyme-like).
deprivation autophagy factor-1), has Atg12 is finally linked by its COOH-terminal
recently been identified and acts at the ER glycine to an internal lysine (K130) residue
(45). NAF-1 interacts with Bcl-2 at the ER of Atg5 (342). The Atg12-Atg5 then forms a
and stabilizes the Bcl-2-Beclin 1 conjugate with Atg16L1 (Atg12-
interaction, while knockdown of NAF-1 Atg5.Atg16L1), resulting in an 800-kDa
decreases Bcl-2-Beclin 1 interaction and complex containing Atg12-Atg5.Atg16L1
promotes autophagy (45). Activation of c- tetramers that are linked via the coiled-coil
Jun NH2-terminal kinase-1 (JNK1) during domain of Atg16L1 (340). This complex is
starvation results in phosphorylation of essential for the elongation of the pre-
Bcl-2 and Bcl-XL that releases their binding autophagosomal membrane, but
to Beclin 1, thus inducing autophagosome dissociates from fully formed
formation (562). autophagosomes.

A second macromolecular complex The second ubiquitin-like reaction involves


implicated in the initiation step of the protein microtubule-associated
autophagosome formation is the FIP200 protein 1 light chain 3 (MAP1-
(163)-ULK1/Atg1 complex LC3/LC3/Atg8). LC3 is synthesized as a
(44, 46, 126, 178, 209). Atg13 binds ULK1 precursor form and is cleaved at its COOH
or its homolog ULK2 and mediates their terminus by the protease Atg4B (170, 516),
interaction with FIP200. Under nutrient resulting in the cytosolic isoform LC3-I.
deprivation conditions, Atg13 and ULK1/2 LC3-I is conjugated to
are dephosphorylated (see section V for phosphatidylethanolamine (PE) in a
details), thereby activating ULK1/2, which reaction involving Atg7 (E1-like) and Atg3
phosphorylates FIP200 to induce (E2-like) to form LC3-II (211, 518). LC3-II is
autophagosome formation specifically targeted to the elongating
(46, 126, 178, 209). autophagosome membrane and, unlike
the Atg12-Atg5.Atg16L1 complex, remains
B. Elongation on completed autophagosomes until
fusion with the lysosomes, after which LC3-
Elongation of phagophores probably II on the cytoplasmic face of
requires membrane input from other autolysosomes can be delipidated by Atg4
organelles. Atg9 is one protein that may and recycled (516). (LC3 is also found on
play this role. Atg9 is a transmembrane the internal surface of autophagosomes,
protein that cycles between the trans- and this is degraded in the autolysosomes.)
Golgi network and endosomes, probably The relatively specific association of LC3-II
carrying membrane for expansion of with autophagosomes makes it an
phagophore (584). Other components of excellent marker for studying autophagy
Atg9 sorting remain to be identified. (241).
TEÓRICO AUTOFAGIA 6

Cross-talk between the two ubiquitin-like (7, 109, 147, 199, 282, 428). Mutation or
systems has been reported. The Atg12- loss of proteins important for formation of
Atg5.Atg16L1 complex can function in an multivesicular bodies (MVBs) (e.g., ESCRT)
E3 ubiquitin ligase-like manner to facilitate leads to an inhibition of autophagosome
LC3-I conjugation to PE (157). The Atg16L1 maturation (109, 282, 428). UVRAG, a
complex is believed to bring LC3 to the site Beclin 1 interacting protein, is also involved
of lipidation for the final conjugation with in the maturation step by recruiting the
PE, but the mechanism by which LC3 is fusion machinery on the autophagosomes.
targeted to specific membranes remains to This function is independent of its
be identified (121). Atg10 can interact with interaction with Beclin 1 (299). UVRAG
LC3 and facilitate LC3 conjugation to PE recruits the class C Vps proteins and via this
(358). Similarly, Atg3 interaction activates Rab7, thereby
coimmunoprecipitates with Atg12, and promoting fusion with late endosomes and
overexpression of Atg3 increases Atg5- lysosomes (299). A recently identified
Atg12 conjugation (517). LC3 can mediate Beclin 1 interacting protein, Rubicon, also
membrane tethering and hemifusion, and functions in the maturation of
these functions may be crucial for the autophagosomes. Rubicon is thought to be
expansion and/or assisting the final fusion a part of a distinct Beclin 1 complex
of the double-membrane cups into fused containing hVps34, hVps15, and UVRAG
vesicles (355). Consistent with this that suppresses autophagosome
hypothesis, loss of Atg3 or a loss-of- maturation (329, 603). More work is
function mutation in Atg4B leads to required to exactly characterize the
decreased closure of isolation membranes different Beclin 1 complexes and where in
(120, 494). the autophagy pathway they act.

C. Maturation and Fusion In addition to the fusion machinery, proper


lysosomal function is also essential for
Unlike in yeast cells where fusion to be successful. Inhibition of the
autophagosomes are formed at the single lysosomal H+-ATPase by chemicals like
phagophore assembly site next to the bafilomycin A1 inhibits the fusion of
vacuole, mammalian autophagosomes are autophagosomes with
formed randomly in the cytoplasm. endosomes/lysosomes (577). This might
Autophagosomes move bidirectionally be because lysosomal acidification is
along microtubules with a bias towards the required for normal fusion. However, the
microtubule organising center (MTOC), possible mechanisms responsible are
where the lysosomes are enriched. This unclear.
transport towards MTOC requires the
function of dynein motor proteins A recent study has identified an alternative
(233, 413, 566). Depolymerization of autophagy pathway that seems to be
microtubules or inhibition of dynein- activated when mouse cells lacking Atg5 or
dependent transport results in inhibition of Atg7 were treated with stressors like
autophagy (245, 560). Autophagosomes etoposide. This Atg5/Atg7-independent
first fuse with endosomes and then with autophagy requires certain other key
lysosomes where the fate of the autophagy proteins, like Ulk1 and Beclin 1,
autophagosomes ends. The fusion step and the progression of this autophagy
involves proteins such as ESCRT, SNAREs, process occurs in a Rab9-dependent
Rab7, and the class C Vps proteins manner (362).
TEÓRICO AUTOFAGIA 7

III. SELECTIVE AUTOPHAGY pathogens (xenophagy) have also been


described as specific phenomena
Autophagy has long been considered to be (289, 563). Mitochondria are a major
a nonselective bulk degradation pathway. source of reactive oxygen species (ROS)
However, in the last few years, several and are therefore especially prone to ROS
forms of selective autophagy have been damage. Maintaining a healthy population
identified which lead to degradation of of mitochondria is essential for the well-
specific organelles, proteins, and being of cells. The major degradative
pathogens in yeast, flies, and mammals. pathway in mitochondrial turnover is
Whether nonselective autophagy really mitophagy (230). Mitophagy has been
occurs in a random manner is an directly visualized in hepatocytes from
interesting question. Recently, a study of transgenic mice expressing green
the dynamics of 1,500 proteins revealed fluorescent protein (GFP) fused with LC3
they were degraded in an ordered fashion (230, 343). GFP-LC3 formed small green
during starvation, implying selectivity dotted structures that became associated
under conditions typically associated with with mitochondria following starvation.
nonselective autophagy (260). Cytosolic These structures became crescent-like in
proteins were degraded rapidly, whereas shape and went on to completely
proteins annotated to various complexes sequester individual mitochondria.
and organelles were degraded later at
different time periods. Selective Mitophagy can be induced by nutrient
autophagy may be involved, possibly via deprivation and photodamage. Both these
positive selection for incorporation into stimuli can induce the mitochondrial
autophagosomes [e.g., by permeability transition (MPT) (286). Unlike
posttranslational modifications like in yeast cells, a common feature of
ubiquitination or acetylation (202, 238)] mammalian mitophagy is loss of
and/or negative selection (e.g., due to membrane potential. During MPT, the
decreased accessibility of certain permeability transition pore, a
proteins/organelles for autophagic nonselective large-conductance inner
engulfment). However, some of these mitochondrial membrane channel, opens,
observations may also be unrelated to any resulting in depolarization and
selection and may be the consequence of mitochondrial outer membrane rupture.
multiple different pathways contributing Cyclosporin A, an MPT blocker, can prevent
to degradation of a given substrate; many mitophagy occurring in mitochondria
proteins are degraded both by the undergoing MPT (230). In HeLa cells,
ubiquitin-proteasome system (where inhibitor factor 1 (IF1), a nuclear DNA
substrates are “primed” for degradation by encoded protein that regulates F1F0-ATP
E3 ligases) and autophagy. synthase, is involved in mitochondrial
clearance by autophagy (39). Mitophagy is
A variety of organelles are cleared by also induced during normal developmental
autophagy. Homeostatic levels of ER, processes like reticulocyte maturation or
peroxisomes, mitochondria, and development of sperm cells. Mitochondrial
ribosomes are maintained by ER-phagy, clearance in reticulocytes required the
pexophagy, mitophagy, and ribophagy, Bcl2-related protein Nix along with Ulk1
respectively (20, 259, 432, 530). Lipid activity (267, 469). Nix has recently been
degradation by autophagy shown to bind to LC3/GABARAP and thus
(macrolipophagy) and degradation of may target mitochondria to
TEÓRICO AUTOFAGIA 8

autophagosomes (366, 468). During and a reduced probability for a fusion


hypoxia, BNIP3, a protein whose event. This predicts the formation of a
expression is suppressed in many forms of subpopulation of nonfusing mitochondria
cancer, is involved in mitochondrial that also have reduced delta psi(m) and
clearance as a survival mechanism to decreased levels of the mitochondrial
control generation of ROS (13, 598). fusion protein OPA1. Inhibition of fission
decreases mitochondrial autophagy and
One way that mitophagy may be beneficial results in the accumulation of oxidized
would be if it could selectively enhance mitochondrial proteins, reduced
removal of dysfunctional mitochondria. respiration, and impaired insulin secretion.
This may occur via the protein Parkin, These studies suggest a fusion-fission-
which is mutated in certain cases of related process that enables selective
autosomal recessive Parkinson's disease. removal of damaged mitochondria by
Parkin is selectively recruited to autophagy (540).
dysfunctional mitochondria with low
membrane potential. After recruitment, Peroxisome abundance is increased in cells
Parkin facilitates the engulfment of in response to peroxisome proliferators, a
mitochondria by autophagosomes and process that is counterbalanced by
thus allows selective removal of damaged degradation of peroxisomes by pexophagy
mitochondria (357). The relocation of (432). Evidence for the contribution of the
Parkin to mitochondria relies on the autophagic machinery in peroxisomal
expression of phosphatase and tensin degradation in mammals comes from
homolog (PTEN)-induced putative kinase 1 conditional knockout mice for Atg7, an
(PINK1), another protein mutated in forms essential autophagy gene. In control
of autosomal recessive Parkinson's hepatocytes, excess peroxisomes were
disease. Overexpression of the wild-type, surrounded by autophagosomes,
but not the mutant, PINK1 causes suggesting a selective process. Atg7-
translocation of Parkin to mitochondria deficient hepatocytes had impaired
(554). Loss of PINK1 induces oxidative removal of excess peroxisomes, and these
stress and subsequently leads to the excess peroxisomes were not surrounded
clearance of depolarized mitochondrial by by autophagosomes (194). It is possible
autophagy requiring beclin-1 (55, 76). that one way that peroxisomes may be
targeted to autophagosomes is via
Another way that one may be able to ubiquitination of cytosolic-facing proteins.
mediate selective clearance of Such proteins may be recognized by the
mitochondria appears to be related to the ubiquitin-binding protein p62 that is
increased likelihood that a small targeted to autophagosomes by binding to
mitochondrion could be engulfed by an Atg8 (231).
autophagosome, compared with a larger
mitochondrion. Mitochondria undergo ER volume is increased in response to ER
multiple cycles of fusion and fission in a stress where an excess of unfolded
“kiss and run” pattern. Fission events often proteins is sensed by the unfolded protein
generate unlike daughter mitochondria: response (UPR). When the UPR becomes
one daughter has increased membrane inactivated, the ER reestablishes its
potential [delta psi(m)] and a high homeostatic volume by ER-phagy (20).
probability of subsequent fusion, while the Electron microscopic studies in yeast have
other has decreased membrane potential revealed large vesicles, bound by double
TEÓRICO AUTOFAGIA 9

membranes, densely filled with stacked postnatal hypoglycemia. Glucagon


membrane cisternae and no other elevates cAMP levels resulting in the
organelles, indicating a selective process formation of hepatic autophagic vacuoles,
(20). Autophagy may also play a role during activation of the glycogen-hydrolyzing acid
the UPR by eliminating areas of ER that are glucosidase, and degradation of glycogen
enriched in potentially dangerous unfolded to glucose (256).
proteins. In mammalian cells, autophagy
occurs in response to ER stress and ER IV. CROSS-TALK BETWEEN THE
protein aggregates (545), but whether or UBIQUITIN-PROTEASOME SYSTEM,
not this is a selective process is yet to be CHAPERONE-MEDIATED AUTOPHAGY,
demonstrated. AND AUTOPHAGY

Regulation of selective autophagy has A. Ubiquitin-Proteasome System and


been well studied in yeast where elegant Autophagy
genetic experiments have elucidated
genes that are specifically involved in the Besides autophagy, the other major route
different selective forms of autophagy, for for intracellular protein degradation is the
example, Atg19 and Atg20 in ER-phagy, ubiquitin-proteasome system (UPS). In
Uth1, Aup1, Atg32 in mitophagy, PEX3 and contrast to macroautophagy, this process
PEX14 in pexophagy, and Ubp3p/Bre5p in is highly specific. In mammalian cells,
ribophagy (216, 259, 376, 545). Much less about a thousand E3 enzymes are
is known about the molecular targets and responsible for the specific detection and
signaling pathways responsible for binding of their substrates, which include
selective pexophagy and ER-phagy in misfolded proteins and short half-life
mammals. Indeed, it is frequently very regulatory proteins. In this process, the
difficult to make a strong claim for ubiquitously expressed, 76-amino acid
selective autophagy without identifying protein ubiquitin is first activated by an E1
candidate modulators that can influence enzyme and subsequently transferred onto
autophagic degradation of the specific an E2 ubiquitin-conjugating enzyme. The
organelle/protein in question, without interaction between an E2 enzyme
influencing autophagic clearance of other (carrying the activated ubiquitin molecule)
substrates. and an E3 enzyme (carrying the
degradation substrate) leads to the
Selective autophagy is not limited to transfer of the ubiquitin onto the target
degradation of organelles like substrate. In this reaction, a covalent
mitochondria and the ER. Glycogen isopeptide bond is formed between the
autophagy occurs in mammals under COOH-terminal glycine of the ubiquitin
conditions of demand for the massive molecule and the lysine residues in the
hepatic production of glucose, such as in target protein (171). The ubiquitin
the postnatal period. Glycogen autophagy molecule itself contains seven internal
appears to be a selective process, since lysines at positions 6, 11, 27, 31, 33, 48,
autophagic vacuoles are predominantly and 63. In a second cycle, the cascade of
distributed at the borders of glycogen E1, -2, and -3 enzymes adds another
stores and contain various quantities of ubiquitin molecule to one of these seven
glycogen (256). Glycogen autophagy is internal lysines. Depending on which of
induced by glucagon, which is secreted these lysines link the ubiquitin molecules
after birth as a result of the normal to a chain, the tagged protein is destined
TEÓRICO AUTOFAGIA 10

for different pathways. A protein that conditions (117, 294), and long-lived
carries a chain of at least four K48-linked proteins can also be degraded by the UPS
ubiquitins can be recognized by the (116). It is now clear that a number of
machineries that allow transfer to the proteins can be degraded by both
proteasome. At least in some autophagy and the proteasome
circumstances, this appears to be (71, 81, 225, 559). Furthermore, mutations
mediated by ubiquitin-binding proteins, that interfere with the proteasomal
like p97 (152). degradation of a protein may increase the
dependency of such proteins on autophagy
The proteasome consists of a 20S central for their degradation, as this will then
complex and two 19S lid complexes. The become the default clearance route. This is
barrel-shaped central complex comprises especially true for mutated proteins with
four stacked rings. The two identical outer an increased aggregation tendency, as
rings are each made up of seven α-subunits oligomeric and higher-order structures will
(termed α 1–7), and the two identical inner become inaccessible to the narrow
rings each consist of seven β-subunits proteasome barrel.
(termed β 1–7). The proteolytic activity
resides in the inner two β-rings. The two Numerous studies in yeast and mammalian
19S lid complexes that bind to either side cells, including primary neurons, have
of the central complex restrict access to reported that proteasome inhibition leads
the proteolytic sites. Subunits of these lid to the upregulation of autophagy
complexes bind to ubiquitin chains on (49, 88, 89, 193, 418). This cross-talk may
degradation substrates, release the be mediated by JNK1 activation following
ubiquitin chains, and allow the substrate to proteasome inhibition, which would be
enter the central, proteolytic complex. predicted to induce autophagy via Bcl-2
Proteasomal degradation of target phosphorylation, thereby reducing its
substrates results in short polypeptides ability to bind and inhibit the function of
that are released into the cytoplasm or the Beclin 1 (337, 389, 562). In vivo data
nucleus, which are then further degraded further support these findings: impairment
into amino acids by peptidases (171). of the proteasome leads to
neurodegeneration in the fly eye. This
The UPS and autophagy have long been phenotype is enhanced when essential
regarded as two separate cellular autophagy genes are knocked down, and
pathways with distinct functions. It was rescued by autophagy induction with
previously thought that autophagy rapamycin (380).
predominantly degraded long half-life
proteins and that the clearance of the While proteasome inhibition may induce
short half-life proteins that were typical autophagy, the converse does not occur.
UPS substrates would not be affected by a Indeed, inhibition of autophagy leads to
compromise in autophagy. However, impaired degradation of substrates
recent findings suggest that the two destined for the proteasome (255). This is
systems do “communicate,” at least under dependent on the ubiquitin-binding
certain circumstances, and might even protein p62, which is an autophagy
have compensatory functions in some substrate that accumulates in autophagy-
cases. Indeed, short half-life proteins deficient cells (25, 250). The resulting
normally degraded by the UPS can be elevated levels of p62 appear to
degraded by autophagy under certain compromise the UPS by delaying the
TEÓRICO AUTOFAGIA 11

delivery of ubiquitinated substrates to the chain that is necessary to signal for the
proteasome, by sequestering them away degradation of proteasome clients (231).
from other ubiquitin-binding proteins (e.g., However, definitive data supporting this
p97). Therefore, prolonged inhibition of mechanism for the clearance of
autophagy would also result in the reduced endogenous proteins are still awaited. p62
flux through UPS; this may be relevant to is unlikely to be necessary for the clearance
the interpretation of studies examining the of most autophagic substrates, as there
effects of autophagy knockout in various does not appear to be a defect in bulk
tissues, since some of the effects may be autophagy in p62 knockout mice (250).
secondary consequences of UPS Another LC3 interactor with many
compromise (e.g., p53 elevation and functional similarities to p62 has recently
apoptosis) (306, 310). been identified [NBR1 protein (neighbor of
BRCA1 gene 1)], and it is possible that
An interesting coordinate regulation of there may be redundancy for their putative
autophagy and the UPS appears to occur roles in autophagy (236–238, 273). Also,
during muscle atrophy. In this condition, HDAC6, which also binds ubiquitin and
both proteasomal and autophagic protein interacts directly with microtubules, is
degradation are upregulated, and this involved in autophagic degradation of
upregulation is coordinated by the mutant proteins during proteasomal
transcription factor FoxO3 (600). In this inhibition (193, 380). A recent study
case, FoxO3 appears to mediate reported that degradation of
independent effects on autophagy and the polyubiquitinated proteins by the
UPS (600). proteasome or autophagy is mediated by
BAG1 and BAG3, respectively. A switch
Recently, a growing amount of data have from BAG1 to BAG3 in aged cells suggests
drawn attention to p62 and its possible that autophagy is used more extensively
role in connecting autophagy with the UPS. for the turnover of polyubiquitinated
p62 is cleared by both the UPS and proteins (125). Although ubiquitinated
autophagy (250, 255) and is commonly proteins are autophagy substrates, more
detected in ubiquitin-containing protein work is needed to understand the exact
aggregates associated with various role of ubiquitination as a signal for
neurodegenerative diseases (25, 91, 268– selective autophagy.
270, 352). p62 polymerizes via its NH2-
terminal PB1 domain and binds to B. ERAD and Autophagy
polyubiquitin chains via its COOH-terminal
UBA domain. In vitro data reveal that p62 Most secreted and transmembrane
interacts with LC3, and it has been proteins in eukaryotic cells are folded and
suggested that this may facilitate the modified in the ER before they are further
autophagic clearance of ubiquitin-positive transported towards their destination.
protein aggregates (25, 250, 381). The Proteins that fail to fold properly in the ER
strongest support for this hypothesis initially receive aid by an intricate system
comes from studies using an artificially of molecular chaperones. However,
ubquitinated peroxisomal integral proteins that do not fold at this stage are
membrane protein, where it was retrotranslocated to the cytoplasm and
demonstrated that a single ubiquitin targeted for proteasomal degradation via
molecule is sufficient as a degradation cytosolic polyubiquitination, a process
signal, in contrast to the tetra-ubiquitin called ER-associated degradation (ERAD).
TEÓRICO AUTOFAGIA 12

When misfolded proteins are not exported which induces autophagy by reducing
efficiently to the cytoplasm and Beclin 1 binding to Bcl-xL (142, 592). On the
accumulate in the ER, the UPR is often other hand, Kouroku et al. (257)
induced. Three main pathways are demonstrated that ER stress caused
activated to alleviate the accumulation of activation of autophagy via PERK1 and
proteins in the ER. 1) Inositol-requiring eIF2α. Calcium release due to ER stress has
protein-1 (IRE1) oligomerizes in the also been implicated in the pathway
stressed ER and via X-box binding protein- leading to autophagy induction (180, 433).
1 (XBP1) activates the transcription of UPR However, some of these calcium studies
genes, whose products are involved in lipid were based on experiments using
synthesis, or ERAD, or code for thapsigargin, which may have incorrectly
chaperones. 2) ER stress leads to the interpreted that the increased numbers of
processing of activating transcription autophagosomes occurring after
factor-6 (ATF6). The resulting ATF6 thapsigargin treatment were the result of
fragment translocates to the nucleus and increased autophagosome synthesis. It
activates UPR genes. 3) Protein kinase RNA appears that thapsigargin causes a calpain-
(PKR)-like ER kinase (PERK) oligomerizes dependent reduction in autophagosome
under ER stress and phosphorylates the α- synthesis along with a block in
subunit of eukaryotic translation initiation autophagome-lysosome fusion. So,
factor-2 (eIF2α), thus inhibiting translation, although this compound increases
thereby decreasing the ER protein load. autophagosome numbers (due the block in
Additionally, PERK activates UPR gene autophagosome-lysosome fusion), its
transcription (423, 548). overall effects are to impede autophagic
flux and the clearance of autophagic
Many studies have reported that ER substrates (566).
stressors, like DTT, tunicamycin, or
thapsigargin, or proteasome inhibitors C. CMA and Autophagy
induce the formation of autophagosomes
(19, 89, 180, 257, 367, 433, 583). The In addition to the relationships with the
activation of autophagy under ER stress UPS, (macro)autophagy is also modulated
may have a cytoprotective effect, as by the activity of CMA. Deletion of the
genetic or chemical inhibition of lysosomal receptor LAMP-2a, a key
autophagy leads to a higher susceptibility component of CMA, induces
towards cell death (19, 89, 367). The macroautophagy (321, 322). Whereas
molecular pathway that is responsible for macroautophagy is a relatively unspecific
the induction of autophagy after ER stress degradation pathway, CMA is highly
is not entirely clear. Different data suggest specific. Thus it is somewhat surprising
an involvement of IRE1, signaling via JNK-1 that autophagy-incompetent cells activate
but not XBP1, and independence of PERK1 CMA (221), as this pathway requires a
and ATF6 (89, 367). The involvement of specific pentapeptide signal in its
JNK-1 is particularly interesting, as substrates. While only 30% of the
phosphorylation of Beclin 1 by this kinase cytoplasmic proteins carry the required
is essential for the induction of autophagy signal (56), enhanced degradation of these
under starvation conditions (562). Another proteins may be sufficient to reduce the
mechanism may involve Death-associated overall load of proteins and thereby
protein kinase 1 (DAPK1) activation by ER alleviate cell stress.
stress leading to Beclin 1 phosphorylation,
TEÓRICO AUTOFAGIA 13

V. SIGNALING PATHWAYS REGULATING signaling molecule that regulates diverse


MAMMALIAN AUTOPHAGY cellular functions, such as initiation of
mRNA translation, cell growth and
Several signaling pathways seem to proliferation, ribosome biogenesis,
regulate autophagy in mammalian cells. transcription, cytoskeletal reorganization,
Similar to yeast, the classical pathway long-term potentiation, and autophagy
involves the serine/threonine kinase, (445). mTOR is the mammalian ortholog of
mammalian target of rapamycin (mTOR). the yeast protein kinase TOR that
The induction of autophagy by mTOR negatively regulates autophagy (291). The
inhibition under starvation conditions is a mTOR pathway involves two functional
well-known phenomenon in mammalian complexes: a rapamycin-sensitive mTOR
cells (509). Various pathways and small complex 1 (mTORC1), which regulates
molecules regulating autophagy via mTOR autophagy, consisting of the mTOR
and mTOR-independent mechanisms, catalytic subunit, raptor (regulatory
which have been identified in recent years, associated protein of mTOR), GβL (G
are described below. protein β-subunit-like protein; also known
as mLST8), and PRAS40 (proline-rich Akt
A. mTOR Pathway substrate of 40 kDa); and mTOR complex 2
(mTORC2) consisting of mTOR, rictor
The mTOR pathway is the most studied (rapamycin-sensitive companion of
pathway regulating mammalian autophagy mTOR), GβL, SIN1 (SAPK-interacting
(Fig. 2). mTOR [also known as rapamycin protein 1), and PROTOR (protein observed
and FKBP12 target-1 (RAFT1), rapamycin with rictor) that is not a direct autophagy
target-1 (RAPT1), and FKBP12-rapamycin regulator (145, 581).
associated protein (FRAP)] is an important
TEÓRICO AUTOFAGIA 14

Fig. 2.The phosphatidylinositol 3-kinase (PI3K)-mammalian target of rapamycin (mTOR)


pathway regulating autophagy. The PI3K pathway is triggered by the binding of insulin (or
growth factors) to insulin receptor (IR), thereby activating PI3K. Activated PI3K converts PIP 2 to
PIP3, which then recruits phosphoinositide-dependent kinase 1 (PDK1) and Akt to the cell
membrane. Akt is then phosphorylated and activated, which in turn phosphorylates and
inactivates tuberous sclerosis complex (TSC) 1/2, leading to activation of Rheb and
consequently mTORC1. Akt can also be phosphorylated and activated by mTORC2. AMPK, a
cellular energy sensor of changes in the intracellular ATP/AMP ratio, directly phosphorylates
TSC2, thereby providing the priming phosphorylation for subsequent phosphorylation of TSC2
by glycogen synthase kinase 3 (GSK3) to inhibit mTORC1 signaling. The mTOR pathway
involves two functional complexes: a rapamycin-sensitive mTORC1 that regulates autophagy,
consisting of the mTOR catalytic subunit, raptor, mLST8, and PRAS40; and mTORC2 comprising
of mTOR, rictor, mLST8, SIN1, and PROTOR. Amino acids activate mTORC1 via Rag GTPases
and suppress autophagy. The rate-limiting factor of amino acid signaling is L-glutamine, which
is initially taken up by its high-affinity transporter SLC1A5, followed by its efflux by the
SLC7A5/SLC3A2 bidirectional transporter in exchange for uptake of essential amino acids,
which subsequently activates mTORC1. The ULK1-Atg13-FIP200 complex acts as an integrator
of the autophagy signals downstream of mTORC1. Under nutrient-rich conditions, mTORC1
suppresses autophagy by interacting with this complex and mediating phosphorylation-
dependent inhibition of Atg13 and ULK1. Under starvation conditions or rapamycin treatment,
mTOR dissociates from the complex, resulting in dephosphorylation-dependent activation of
ULK1 and ULK1-mediated phosphorylations of Atg13, FIP200, and ULK1 itself, which triggers
autophagy. The mTORC1 pathway regulates cell growth mainly through 4E-BP1 and p70S6K.
Phosphorylation-dependent activation of p70S6K can also inhibit IRS1, thereby exerting a
feedback loop mechanism.

Many diverse signals, such as growth raptor and mTOR (229). Inhibition of
factors, amino acids, glucose, energy mTORC1 and induction of autophagy by
status, and different forms of stress, rapamycin is associated with reduced
regulate the raptor-mTOR (mTORC1) phosphorylation of two of its downstream
pathway (445). The activity of mTORC1 can effectors, ribosomal protein S6 kinase-1
be inhibited by rapamycin (sirolimus), a (S6K1, also known as p70S6K) and
lipophilic macrolide antibiotic first isolated translation initiation factor 4E binding
from Streptomyces protein-1 (4E-BP1, also known as PHAS-1)
hygroscopicus (145, 365). Rapamycin is a at Thr389/Thr421/Ser424 and
potent autophagy inducer in various cell Thr37/Thr46, respectively (242, 445, 464).
lines from yeast to mammalian cells, Interestingly, a study by Scott et al. (471)
including neurons (28, 365, 415, 416, 427). showed that p70S6K is a positive regulator
In mammalian cells, rapamycin forms a of autophagy. When TOR is inactivated
complex with the immunophilin FK506- during starvation-induced autophagy in
binding protein of 12 kDa (FKBP12), which the Drosophila fat body, p70S6K needs to
then stabilizes the raptor-mTOR be activated for some time to allow
association and inhibits the kinase activity maximal autophagy, after which loss of
of mTOR (228). The binding of GβL to p70S6K activity prevents excessive
mTOR stimulates its kinase activity, and autophagy in TOR-inactive state, probably
GβL is necessary for the formation of a by inhibiting the PI3K pathway through a
rapamycin-sensitive interaction between negative-feedback loop involving
TEÓRICO AUTOFAGIA 15

downregulation of the insulin receptor However, the role of glutamine may be


substrates (IRS) (243, 471). Furthermore, tissue specific, since this amino acid
p70S6K activation appears to be necessary stimulates autophagy in intestinal cells
but not sufficient to induce autophagy, as (434). The Rag GTPases mediate amino
it activates autophagy in TOR mutants but acid signaling to mTORC1 activation by
not in wild-type Drosophila (471). Whether binding to raptor and further redistributing
these effects of p70S6K on autophagy are mTOR to a subcellular compartment
seen in mammals is unclear. containing its activator rheb (440). A
recent report suggests that amino acids
A recent study indicates that rapamycin induce the translocation of mTORC1 to
does not inhibit mTORC1 completely, due lysosomal membranes where the Rag
to rapamycin-resistant functions of GTPases reside. This recruitment of
mTORC1. Instead, a selective ATP- mTORC1 requires a trimeric Ragulator
competitive small molecule mTOR complex containing the proteins encoded
inhibitor, Torin1, was found to induce by the MAPKSP1, ROBLD3, and c11orf59
autophagy to a much greater extent than genes. Thus, in the presence of amino
rapamycin (528). Although Torin1 inhibits acids, the Rag GTPases that are tethered to
both mTORC1 and mTORC2 directly, its lysosomal surface by the Ragulator act as a
effects on autophagy are mTORC2 docking site for mTORC1, which is then
independent but mTORC1 dependent due activated by Rheb (439).
to suppression of rapamycin-resistant
functions of mTORC1 that are required for The other mTOR complex, mTORC2, which
inhibition of autophagy (528). phosphorylates (Ser-473) and activates
Akt, is known to be rapamycin insensitive
Other conditions, such as nutrient (195, 444). However, recent studies have
deprivation, also stabilize the raptor-mTOR shown that prolonged treatment with
complex, thereby inhibiting mTORC1 and rapamycin inhibits mTORC2 activity and
inducing autophagy (228, 242, 332). Under Akt signaling in certain mammalian cell
conditions of limited extracellular amino types (446, 595). Rapamycin derivatives
acids, autophagy recycles intracellular such as temsirolimus (CCI-779) and
constituents to provide an alternative everolimus (RAD001) inhibit the activity of
source of amino acids (242). Conversely, mTORC1, thereby reducing the
diverse extracellular signals, such as amino phosphorylation of p70S6K and 4E-BP1
acids and growth factors, activate mTORC1 (595).
and suppress autophagy (145, 242, 332).
Recently, the rate-limiting factor that Many of the molecular mechanisms linking
enables essential amino acids to inhibit TOR signaling to the autophagic machinery
mTORC1 has been identified as L- have been dissected in yeast, where the
glutamine (360). The influx of L-glutamine Atg1 kinase and the Atg1-Atg13-Atg17
by its high-affinity transporter SLC1A5 complex are thought to act downstream of
initially increases its intracellular TOR regulating autophagosome formation
concentration. The heterodimeric (212, 573). Recent studies have identified
SLC7A5/SLC3A2 bidirectional transporter some of the molecular components
then uses L-glutamine as an efflux downstream of mTORC1 in mammalian
substrate in exchange for the cellular autophagy. Atg13 binds to the mammalian
uptake of essential amino acids, which Atg1 homologs ULK1 or ULK2 and mediates
subsequently activates mTORC1 (360). the interaction of ULK1/2 with FIP200,
TEÓRICO AUTOFAGIA 16

thereby forming a ULK1/2-Atg13-FIP200 rictor-mTOR complex phosphorylates Akt


stable complex that signals to the on Ser-473, which may facilitate the
autophagic machinery downstream of phosphorylation by PDK1 of the activation
mTOR (126, 178, 209). Under nutrient-rich loop of Akt on Thr-308. Akt has been
conditions, mTORC1 suppresses shown to modulate autophagic activity
autophagy through direct interaction with regulated by the PI3K pathway. Mutations
this complex and mediates causing Akt activation or inactivation cause
phosphorylation-dependent inhibition of suppression and induction of autophagy,
the kinase activities of Atg13 and ULK1. respectively (80).
Under starvation conditions or rapamycin
treatment, mTOR dissociates from the The different classes of PI3K have distinct
complex, resulting in the inhibition of effects on autophagy (334). Increases in
mTOR-mediated phosphorylation of Atg13 the class I PI3K product, PIP3, inhibit
and ULK1. This leads to autophagy by inducing Akt activation
dephosphorylation-dependent activation (391), whereas hyperactivity of the
of ULK1 (and ULK2) and ULK1-mediated phosphoinositide phosphatase PTEN
phosphorylations of Atg13, FIP200, and (phosphatase and tensin homologue
ULK1 itself, which triggers autophagy deleted from chromosome 10), a tumor
(126, 178, 209). Therefore, the ULK1- suppressor, stimulates autophagy by
Atg13-FIP200 complex acts as an inhibiting the PI3K pathway (6). In contrast,
integrator of the autophagy signals increasing the levels of the class III PI3K
downstream of mTORC1. However, it is not product, PI-3-P, stimulates autophagy
clear yet how phosphorylation of these (391). Autophagy can be downregulated by
proteins regulates their activities. inhibitors of PI3K activity, such as 3-MA,
wortmannin. and 2-(4-morpholinyl)-8-
B. PI3K Pathway phenylchromone (LY294002)
(26, 391, 472).
A major signaling cascade controlling
mTORC1 is the PI3K pathway (Fig. 2) (242). The phosphorylation-dependent Akt
The binding of growth factors or insulin to activation results in the phosphorylation of
cell surface receptors activates the class Ia a host of other proteins, including the
PI3K, wherein its regulatory subunit tumor suppressor proteins mutated in
mediates activation of its p110 catalytic tuberous sclerosis which form a complex
subunit by the direct interaction with called the tuberous sclerosis complex
phosphotyrosine residues of the activated (TSC), consisting of a heterodimer of TSC1
receptors or adaptor proteins. Activated (hamartin) and TSC2 (tuberin). TSC1/2
PI3K then converts the plasma membrane affect cell growth and survival (40, 445).
lipid phosphatidylinositol-4,5- TSC1/2, an upstream integrator of various
bisphosphate (PIP2) to signals regulating the mTORC1 pathway, is
phosphatidylinositol-3,4,5-trisphosphate the GTPase-activating protein (GAP) for
(PIP3), which in turn recruits pleckstrin the Ras family GTP-binding protein rheb,
homology (PH) domain proteins, such as which directly binds and activates the
the serine/threonine kinases raptor-mTOR complex (130, 315, 521).
phosphoinositide-dependent kinase 1 Several other kinases, including AMP-
(PDK1) and Akt/PKB, to the plasma activated kinase (AMPK), ribosomal S6
membrane (40). Akt is activated by its kinase 1 (RSK1), and extracellular signal
phosphorylation on two different sites. The regulated kinase 1/2 (ERK1/2), signal to
TEÓRICO AUTOFAGIA 17

mTORC1 by phosphorylating TSC2 and kinases that regulate stress-induced


inhibiting the activity of the TSC1/2 translational control programs, are also
heterodimer (445). involved in the regulation of stress-induced
autophagy. The mammalian eIF2α kinase
Apart from amino acids, mTORC1 can be a signaling pathway is essential for both
sensor of changes in the cellular energy virus- and starvation-induced autophagy,
state via AMPK (334). AMPK, which senses which is antagonized by the herpes simplex
changes in the intracellular ATP/AMP ratio, virus (HSV)-encoded neurovirulence gene
directly phosphorylates TSC2, thereby product ICP34.5 (511). Eukaryotic
providing the priming phosphorylation for elongation factor-2 (eEF-2) kinase, also
subsequent phosphorylation of TSC2 by referred to as Ca2+/calmodulin-dependent
glycogen synthase kinase 3 (GSK-3) to kinase III, also regulates autophagy via the
inhibit mTOR signaling (186). We have mTOR pathway. Inhibition of eEF-2 kinase
recently shown that inhibition of GSK-3β inhibits autophagy. The activity of eEF-2
suppress autophagy by activating mTOR kinase is enhanced in glioblastoma and
(449). While AMPK activation inhibits TOR other malignancies. Nutrient deprivation,
and stimulates autophagy in yeast, the which decreases the activity of S6 kinase,
AMPK activator AICAR inhibits autophagy was shown to enhance the activity of eEF-
in mammalian cells, but this is probably not 2 kinase activity (569).
due to direct effects on AMPK (333).
Recent evidence indicates that AMPK is The mitogen-activated protein kinase
required for mammalian autophagy, as (MAPK)/extracellular signal-regulated
inhibition of AMPK activity by compound C kinase (ERK) pathway, which is commonly
or a dominant negative construct strongly activated in cancers, regulates autophagy.
suppresses autophagy (333, 336). AMPK Carcinogens such as lindane, which
also mediates hypoxia-induced autophagy activates MAPK/ERK pathway, impair
by downregulating mTOR (382). Both LKB1 autophagy (62). However, starvation-
and Ca2+/calmodulin-dependent protein induced autophagy activates the ERK1/2
kinase kinase-β (CaMKKβ) play important MAPK pathway, and inhibition of the
roles in the phosphorylation and activation pathway by a MAPK kinase (MAPKK)
of AMPK. In addition, AMPK can be MEK1/2 inhibitor (PD-98059) suppresses
activated by other upstream kinases, but starvation-induced autophagy in human
the physiological significance of this is colon cancer HT-29 cell line (371). A recent
currently unclear. study shows that sustained MEK/ERK
activation results in complete disassembly
C. Other Protein Kinases of both mTORC1 and mTORC2, strongly
enhancing Beclin 1 activity, resulting in
Apart from mTOR and certain protein cytodestruction, while moderately
kinases like AMPK discussed above, several enhanced Beclin 1 activity, resulting in
other kinases have been reported to cytoprotection, is mediated via transient
regulate autophagy. Physiological or activation of MEK/ERK (556). Codogno and
pharmacological stimulation of IKK (IκB colleagues (369) have shown that a
kinase) induces autophagy via AMPK cytoplasmic heterotrimeric Gi3 protein
activation and mTOR inhibition, but in an regulates autophagy in a human colon
NFκB-independent manner (68). cancer HT-29 cell line. Autophagy is
Eukaryotic translation initiation factor 2α minimal when the Gαi3 protein is in the
(eIF2α) kinases, which are serine threonine GTP-bound form and becomes stimulated
TEÓRICO AUTOFAGIA 18

when GDP is bound to the Gαi3 protein inhibitor (SB203580) that prevented
(370). A similar function of Gαi3 protein in autophagy inhibition, implicating a role of
the regulation of the anti-autophagic p38 MAPK in autophagy (167, 555).
activity of insulin in the liver has also been Accumulation of mutant glial fibrillary
reported (138). Likewise, Gα-interacting acidic protein (GFAP) in Alexander disease
protein (GAIP), which is a regulator of G induces autophagy by activating p38 MAPK
protein signaling (RGS) protein and and inhibiting mTOR signaling pathways
activates the hydrolysis of GTP by the (514). Recent studies show that the
Gαi3 protein, increases autophagy (372). negative regulation of autophagy by p38α
Activation of ERK1/2 stimulates autophagy was due to a direct competition with
by phosphorylating GAIP, which stimulates mAtg9 for binding to p38-interacting
its GTPase activity towards the GTP-bound protein (p38IP) (561).
conformation of the Gαi3 protein.
Phosphorylation of GAIP by the ERK1/2 Recently, the stress-activated signaling
MAPK-dependent pathway is sensitive to molecule, c-Jun NH2-terminal kinase 1
amino acids, since the inhibition of (JNK1), but not JNK2, was shown to be an
autophagy by amino acids correlates with important mediator of starvation-induced
the inhibition of the ERK1/2 MAP kinases autophagy in mammalian cells. Starvation
and a low level of GAIP phosphorylation signal activates JNK1, which in turn
(371). Amino acids interfere with ERK1/2- phosphorylates Bcl-2 at multiple sites (Thr-
dependent autophagy by inhibiting the 69, Ser-70, and Ser-87) in the unstructured
activation of the kinase Raf-1 (388). loop, thereby disrupting the Bcl-2-Beclin 1
complex and triggering autophagy (562).
Autophagy is also modulated by cell
hydration, which is sensitive to a p38 MAPK

THE ROLE OF AUTOPHAGY IN NEURODEGENERATIVE DISEASE

Autophagy is a lysosomal degradative neurodegenerative diseases across the age


process used to recycle obsolete cellular spectrum with exceptional frequency. In
constituents and eliminate damaged late-onset disorders such as Alzheimer’s
organelles and protein aggregates. These disease, amyotrophic lateral sclerosis and
substrates reach lysosomes by several familial Parkinson’s disease, defects arise
distinct mechanisms, including delivery at different stages of the autophagy
within endosomes as well as pathway and have different implications
autophagosomes. Completion of digestion for pathogenesis and therapy. This Review
involves dynamic interactions among provides an overview of the role of
compartments of the autophagic and autophagy in neurodegenerative disease,
endocytic pathways. Neurons are focusing particularly on less frequently
particularly vulnerable to disruptions of considered lysosomal clearance
these interactions, especially as the brain mechanisms and their considerable impact
ages. Not surprisingly, mutations of genes on disease. Various therapeutic strategies
regulating autophagy cause for modulating specific stages of
TEÓRICO AUTOFAGIA 19

autophagy and the current state of drug recycling of basic metabolites for new
development for this purpose are also synthesis and energy stores and the
evaluated. reporting of information about the cell’s
health status and external environment.
Close on the heels of his discovery of The heavy reliance of neurons on
lysosomes in 1956, Christian DeDuve and autophagy is evident from observations
his colleagues described a process by that the brain is often the most severely
which cells digest their own cytoplasmic affected organ in primary lysosomal
constituents within lysosomes, which they disorders and that mutations in genes
termed autophagy (auto-self; phagy- involved in the global lysosomal network
eating)1. Subtypes of autophagy have (autophagy and endosomal pathway) are
since been identified, each involving causatively linked to neurodegenerative
different mechanisms of substrate delivery disorders with exceptional frequency.
to the lysosome, but lysosomal digestion Because neurons have unusually large
remains the common cardinal feature. expanses of dendritic and axonal
Until recently, lysosomes were viewed cytoplasm, they face particular hurdles in
merely as chambers for the terminal preventing dysfunctional organelles and
degradation of nonessential cell cellular waste from accumulating over a
components, but they are now also lifetime without the aid of cell division,
becoming appreciated as vital biosensors which mitotic cells can rely on to dilute
of cell homeostasis that report on the these waste burdens. Young neurons
general nutritional status of a cell through achieve this task by clearing autophagic
the release of its digestion products. substrates very efficiently2, despite the
Lysosomes serve as a docking platform for long distances that the many autophagic
an amino acid–sensing protein complex vacuoles generated in axons must travel to
and for mammalian target of rapamycin reach lysosomes, which are concentrated
(mTOR) kinase, a master regulator of mainly near the cell body3. Not
autophagy induction, and thus lysosome surprisingly, neurons are particularly
activity influences early and late events in vulnerable to slowdowns in the proteolytic
autophagy by modulating rates of clearance of autolysosomal substrates3.
substrate sequestration and delivery to Without competent autophagy, neurons
lysosomes as well as the transcription of accumulate ubiquitinated protein
genes involved in autophagosome and aggregates and degenerate 4,5. In
lysosome biogenesis (Fig. 1). Lysosomes neurodegenerative diseases, autophagy
also receive plasma membrane goes awry at various points along the
components and internalized materials via pathway, giving rise to distinct pathologic
endocytosis. Endosomes carrying these patterns with different implications for
cargoes frequently reach the lysosome by modulating autophagy as a therapy. To
fusing first with autophagosomes during review these issues, I will consider
autophagy (Fig. 1). The active autophagy as three major stages—
communication between endocytic and selection, sequestration and lysosomal
autophagic degradative compartments digestion of substrates—and discuss how a
and the numerous regulatory mechanisms defect in one or more stages contributes to
shared between these pathways pathogenesis in different
constitutes the greater ‘lysosomal neurodegenerative disorders. I then
network’ that controls the elimination of consider the merits and caveats of
obsolete cellular constituents, the modulating autophagy
TEÓRICO AUTOFAGIA 20

Figure 1 An overview of
macroautophagy.
Macroautophagy is responsible
for the bulk turnover of
cytoplasm and is the cell’s only
mechanism for organelle
turnover, which can occur
selectively or nonselectively.
Other subtypes of autophagy
deliver intracellular substrates
into lysosomes by different
mechanisms (see Fig. 3). The
principal stages of
macroautophagy include highly
regulated formation of an
isolation membrane (or
phagophore) under direction of
multiple signaling and protein
modification assemblies;
elongation of the isolation
membrane around a region of
cytoplasm or selected
substrate; closure of the inner
and outer bilayers of the
isolation membrane to form a
double membraned
autophagosome; fusion of the
autophagosome with a
lysosome, or, alternatively, sequential fusion of the autophagosome with an endosome
followed by a lysosome to form an autolysosome; and digestion of the autophagosome
contents by hydrolytic enzymes, yielding basic metabolites that are released into the
cytoplasm for new synthesis or as sources for energy. The process is depicted as a cycle,
emphasizing that amino acids released during lysosomal digestion are one mechanism by
which TORC1 on lysosomes may be modulated and influence signaling pathways controlling
autophagy induction. MVB, multivesicular body; PAS, preautophagosomal structure.

at a specific stage to treat these diseases membranelimited vacuole, the digestion of


and review the current state of drug the substrate by lysosomal hydrolases and
development for these purposes. the release of digestion products, some of
Substrate sequestration and which then influence signaling
autophagosome formation mechanisms that control autophagy
Macroautophagy, the major subtype of induction (Fig. 1). Macroautophagy is
autophagy, is a cycle involving the initiated through changes in the
enwrapping of selected substrates within a phosphorylation states of individual
TEÓRICO AUTOFAGIA 21

components of a stable autophagy-related of autophagy, influences neuronal


2 (Atg1)–Unc51-like kinase (ULK) complex functions as diverse as synaptic
(ULK1, ULK2, Atg13, FIP200 and plasticity10, dendritic arborization11 and
Atg101)6,7. ULK1 phosphorylation is myelination12. The phosphorylation of
mainly regulated by the mTOR complex 1 ULK1 triggers translocation of a
(TORC1) but may be activated by multiprotein complex containing beclin-1
AMPactivated protein kinase (AMPK) (also known as Atg6) and class III
independently of TORC1 (ref. 8), which phosphoinositide 3-kinase (PI3K CIII, also
therefore suggests at least two potential known as Vps34) from the
routes for modulating autophagy induction cytoskeleton13,14 to a
therapeutically. Various cellular stress preautophagosomal structure15 to initiate
signals, including lowered concentrations autophagosome formation (Fig. 2).
of specific amino acids, growth factors or Elongation of the isolation membrane and
ATP, hypoxia, certain types of protein its closure around a cytoplasmic cargo
aggregates, and endoplasmic reticulum requires membrane components from
stress9, suppress TORC1, thus activating several possible sources as well as two
ULK1 and ULK2 and turning on autophagy. complexes containing ubiquitin-like
In neurons, toggling between inactivation proteins and their respective conjugation
of TORC1 and activation of this complex, machineries17 and several SNARE
which promotes protein synthesis instead proteins18 (Fig. 2).

Figure 2 Autophagy
induction and
autophagosome
biogenesis.
Autophagosome
formation can be
initiated via mTOR
inhibition or AMPK
activation. This results in
the phosphorylation of
ULK1 at sites that
activate it and catalyze
phosphorylation of
other components of the
Atg1–ULK complex,
composed of ULK1,
ULK2, Atg13, FIP200 and
Atg101. ULK1 also
phosphorylates AMBRA,
a component of the PI3K CIII complex I (Vps34, Vps15, Atg14, and beclin-1), enabling it to
relocate from the cytoskeleton to the isolation membrane. Phosphatidylinositol 3-phosphate
(PI3P), generated by Vps34 activity, specifically binds the PI3P effectors WD repeat domain
phosphoinositide-interacting 1 (WIPI1) and WIPI2 and catalyzes the first of two types of
ubiquitination-like reactions that regulate isolation membrane elongation. In this first
TEÓRICO AUTOFAGIA 22

reaction, Atg5 and Atg12 are conjugated to each other in the presence of Atg7 and Atg10.
Attachment of the fully formed complex containing Atg5, Atg12 and Atg16L on the isolation
membrane induces the second complex to covalently conjugate phosphatidylethanolamine to
LC3 (ref. 183), which facilitates closure of the isolation membrane184. Atg9 (the Atg9–Atg2–
Atg18 complex), another factor essential for this event, cycles between endosomes, the Golgi
and the phagophore, possibly carrying lipid components for membrane expansion. Atg4
removes LC3-II from the outer surface of newly formed autophagosomes, and LC3 on the inner
surface is eventually degraded when the autophagosome fuses with lysosomes. The steps
known to be affected in neurodegenerative diseases are indicated in red. HD, Huntington’s
disease; PD, Parkinson’s disease; PE, phosphatidylethanolamine.

Figure 3 Substrate
recognition and
selective autophagy.
Misfolded proteins and
protein aggregates are
selectively targeted for
macroautophagy by
specific autophagy
receptors. Initial
recognition by a
chaperone complex
(Bag3, HspB8 and
Hsc70) and E3 ligase–
mediated
ubiquitination recruits
p62, resulting in the
assembly of the
ubiquitin (Ub)- labeled
proteins into p62
bodies. Autophagic
degradation occurs
when p62 and adaptor
proteins (for example,
NBR1 and ALFY)
interact with LC3 on
the forming
autophagosome.
Mitophagy refers to
the selective targeting
of a damaged
mitochondrion for
macroautophagy.
Reduced PINK1
turnover on the injured depolarized mitochondrion increases the abundance of PINK1, which
recruits and phosphorylates the E3 ubiquitin ligase parkin. Voltage-dependent anion channel
TEÓRICO AUTOFAGIA 23

1 (VDAC1) is polyubiquitinated by parkin, recruiting p62, which binds LC3 to initiate


mitochondrion sequestration in the forming autophagosome. In CMA, the Hsc70 chaperone
complex delivers cytosolic proteins containing a KFERQ motif to the lysosomal lumen for
degradation through the binding to a LAMP2-containing protein complex located on the
lysosomal membrane. Microautophagy (not shown) delivers small quantities of proteins or
organelles directly into late endosomes and possibly lysosomes by invagination of the surface
membrane of the vesicle. HD, Huntington’s disease; MFN2, mitofusin-2; PD, Parkinson’s
disease; PDB, Paget’s disease of bone.

It is worth noting for the later discussion its depletion, which then interferes with
of drug discovery that unconventional autophagosome membrane nucleation
forms of autophagy under certain stress and sequestration. In Lafora’s disease, a
conditions may occur independently of form of myoclonus epilepsy associated
some components of this machinery19 and with progressive neurodegeneration, loss-
that some Atg proteins have functions of-function mutations in the protein
unrelated to autophagy20. Despite its phosphatase laforin abnormally activate
complexity, autophagosome formation has mTOR24 and thereby reduce the rate,
only infrequently been reported to be although not necessarily the competence,
impaired in chronic neurodegenerative of autophagosome formation, leading to
diseases and, in some like Alzheimer’s the cytoplasmic accumulation of
disease and amyotrophic lateral sclerosis ubiquitinated proteins. Substrate
(ALS), autophagy induction may actually be recognition and selective autophagy
higher than normal, as discussed later. Autophagy was originally recognized as a
Autophagy is disrupted at multiple stages nonselective process, but several forms of
in Parkinson’s disease, a disorder selective autophagy that target specific
characterized by inclusions containing α- subgroups of substrates have now been
synuclein (‘Lewy bodies’) in affected identified (Fig. 3). Newly discovered
neurons. One effect of characteristically autophagic receptors respond to focal
high intracellular α-synuclein cellular insults, such as organelle injury,
concentrations is inhibition of the GTPase pathogen invasion or abnormally altered
Rab1A, believed to interfere with or aggregated proteins, and then target
omegasome formation through Atg9 these altered structures for selective
mislocalization21 (Fig. 2). In models of autophagy. Autophagic receptors and
Huntington’s disease, autophagosomes other selectivity adaptor proteins tether
form properly and are cleared despite the targeted substrate to core autophagic
slower-than-normal macroautophagic machinery, such as microtubule-associated
protein turnover. A possible explanation protein light chain 3 (LC3) and Atg12– Atg5,
lies in the observation that effecting elimination of the substrate but
autophagosomes seem to be unusually minimally altering overall
devoid of cargoes in these models and in macroautophagy25. The autophagic
brain samples from people with receptors p62 (also known as SQSTM1),
Huntington’s disease, suggesting that NBR1, NDP52, optineurin (OPTN), histone
autophagosome membranes fail to engage deacetylase 6 (HDAC6) and NIX26
substrates properly during sequestration. recognize and facilitate elimination
The underlying mechanism may involve ofubiquitinated proteins, and some of
adventitious binding of beclin-1 by these receptors also target peroxisomes
aggregates of mutant huntingtin leading to and internalized pathogens27,28.
TEÓRICO AUTOFAGIA 24

Additional molecular chaperones and inclusionbody myopathy, Paget’s disease


adaptor proteins with a predilection for of the bone and frontotemporal dementia
misfolded proteins aid this process25,28. (IBMPFD). VCP regulates endolysosomal
Parkin (also known as PARK2) and PINK1 sorting of endocytosed ubiquitinated
(also known as PARK6), two proteins cargoes38, and its depletion or the
involved in mitochondrial fusionfission expression of IBMPFD-linked mutants in
events, target damaged mitochondria for cell models causes immature
selective autophagy (‘mitophagy’). PINK1, autophagosomes containing strongly
a serine-threonine kinase on the outer ubiquitin-positive substrates to
mitochondrial membrane, is activated accumulate, which suggests a role for VCP
when mitochondrial function is in selective clearance of ubiquitinated
compromised and the membrane substrates and possibly autophagosome
depolarized. This enables parkin, a maturation39,40. Mutations in the genes
cytoplasmic E3 ubiquitin ligase, to bind and encoding PINK1 and parkin, which account
ubiquitinate exposed membrane proteins, for the majority of autosomal-recessive
thereby recruiting p62, LC3 and HDAC6, cases of Parkinson’s disease, impede
which initiate mitophagy29–31. The mitophagy, causing damaged
recognition of cargoes for sequestration or mitochondria to accumulate and possibly
delivery into the lysosomal lumen is initiate apoptosis through the intrinsic
relatively susceptible to disruption in pathway or via reactive oxygen species–
neurodegenerative diseases. Mutations in mediated oxidation of membrane lipids
SQSTM1 initially identified in patients with and lysosome membrane
Paget’s disease of bone have now been destabilization41. Interestingly, another
found in familial and sporadic ALS32, function of VCP, which is abrogated by
disorders that typically feature p62- pathogenic mutations, is the clearance of
positive inclusions in affected neurons. damaged mitochondria via the PINK1–
Mutant p62 may promote pathogenesis in parkin pathway42. Chaperone-mediated
several ways, including overactivating autophagy (CMA), another form of
nuclear factor-κB (NF-κB)33 and disrupting selective (and constitutive) autophagy,
autophagic clearance of ubiquitinated mediates the lysosomal degradation of a
proteins and protein aggregates34, sizeable family of proteins containing a
although the latter mechanism has not consensus pentapeptide motif, (for
been shown directly. Rare ALS-associated example, KFERQ) in their primary
mutations of another autophagy receptor, sequence, which includes a substantial
OPTN, also impair NF-κB signaling35 while number of disease-causing aggregate-
causing the protein to accumulate in TAR prone proteins. In CMA, a cytosolic
DNA-binding protein 43–positive neuronal chaperone protein, Hsc70, escorts
inclusions36. OPTN may also have substrate proteins to lysosome-associated
ubiquitin-independent roles in protein membrane protein type 2a (LAMP2A), the
aggregate clearance by autophagy37, CMA receptor on a subpopulation of
although, again, disruption of this function lysosomes. The protein substrate is then
by disease mutations has not been directly unfolded, translocated across the
demonstrated. Selective autophagy is also lysosomal membrane and degraded in
disrupted by mutations in valosin- lysosomes43. As might be imagined, CMA
containing protein (VCP, also known as is especially vulnerable in
p97), an endosomal member of the AAA+ neurodegenerative diseases involving
family of ATPase proteins that lead to misfolded and aggregated pathogenic
TEÓRICO AUTOFAGIA 25

proteins (for example, tau in stage of autophagy because the many


frontotemporal lobar dementia with autophagosomes generated in axons and
ubiquitin-positive inclusions (FTLD-U) and nerve terminals depend heavily on
leucine-rich repeat kinase 2 (LRRK2) and α- retrograde transport to complete the
synuclein in Parkinson’s disease)44–46. digestion process. During this retrograde
These altered proteins bind abnormally journey, nearly all autophagosomes first
tightly to LAMP2A, which blocks their fuse with late endosomes abundant in the
translocation across the lysosomal neuritic processes3,52,53. The formation
membrane. As importantly, the loss of of these amphisomes requires some of the
function of LAMP2A, believed to be the same proteins that direct formation of late
rate-limiting factor for CMA, impedes endosomes (also referred to as
access of other CMA substrates to the multivesicular bodies), including Rab11,
lysosomal lumen46. Possibly relevant to the HOPS complex and components of
various late-age onset neurodegenerative multiple endosomal sorting complex
diseases, CMA activity declines with age required for transport (ESCRT) complexes
owing to falling LAMP2A amounts on that mediate cargo sorting into
lysosomes47. The decrease in the amounts intraluminal vesicles of the multivesicular
of LAMP2A and Hsc70 in nigral bodies25,54,55. Autolysosome formation
dopaminergic neurons of people with also requires late endosome proteins,
Parkinson’s disease outpaces the decline including notably Rab7 (ref. 56) and
during normal brain aging and is also seen syntaxin 5 (ref. 57), and is strongly
in transgenic mice expressing mutant α- influenced by changes in intracellular lipid
synuclein48,49. Lysosomal digestion of content58. Rab7 and LC3 form a complex
autophagic substrates Autophagosome- with FYVE and coiled-coil domain
lysosome fusion. Autophagosomes fuse containing 1 (FYCO1) that promotes
either with lysosomes generating an microtubule plus end-directed transport of
autolysosome or initially with late autophagic vacuoles59. Interactions of
endosomes to form amphisomes that later Rab7 with other endolysosome proteins
fuse with lysosomes (Fig. 4a). Cells increase influence lysosome positioning60,61.
opportunities for lysosomal fusion by These interrelationships closely link
directing autophagosomes to move along endocytic and autophagic dysfunction in
microtubules toward the perinuclear various neurodegenerative disorders62.
microtubule-organizing center (MTOC) of The dynein–dynactin complex is the target
the neuron, where lysosomes are most of mutations causing lower motor neuron
abundant3. Under conditions of stress, disease (with mutations in the dynactin
lysosomes also move toward the MTOC to p150 subunit) and axonal Charcot-Marie-
engage autophagosomes even more Tooth hereditary neuropathy type 2
quickly50. Disassembling microtubules (CMT2, with mutations in the dynein motor
with vinblastine or deleting dynein, the complex), and its dysregulation may also
motor component of the dynein–dynactin underlie the motor neuron disease spinal
complex mediating retrograde transport, and bulbar muscular atrophy63. Decreased
provokes a massive buildup of activity of the dynein complex as a result of
autophagosomes in neuronal dynactin mutations is also believed to
processes2,51, which highlights the cause autophagosomes to appear in motor
importance of microtubule-based vesicle neurons of people with ALS before clinical
transport for autophagy. Neurons are symptoms, as well as autolysosomes and
particularly vulnerable to disruption at this p62-positive inclusions to accumulate as
TEÓRICO AUTOFAGIA 26

these neurons degenerate at clinical stages several protein-misfolding diseases65.


of disease64. Loss of dynein complex Amphisome formation may be impaired in
function increases autophagosome familial ALS and frontotemporal dementia
number and the amounts of aggregation- linked to chromosome 3.
prone proteins in models of ALS and

Figure 4 Clearance of autophagic substrates. (a) Fusion of an autophagosome with an


endosome or a lysosome is shown. LC3-II– associated autophagosomes move along
microtubule tracks, the principal binding sites for LC3. Autophagosomes generated actively in
axons fuse quickly with Rab7-positive late endosomes, and the amphisomes move nearly
exclusively in the retrograde direction via the dynein–dynactin complex. Microtubule
acetylation, regulated in part by HDAC6, recruits dynein and kinesin-1 to microtubules and is
essential for fusion of autophagosomes with lysosomes. Amphisome formation requires some
of the same proteins that direct formation and maturation of multivesicular bodies (MVBs)
and late endosomes, including Rab11, the HOPS complex, and components of multiple ESCRT
complexes that mediate cargo sorting into intraluminal vesicles of the multivesicular bodies.
Autolysosome formation also requires late endosome proteins, including Rab7, syntaxin 5,
several SNARES, LAMP1 and LAMP2 and endosomal Vps34 PI3K CIII complex. Following
TEÓRICO AUTOFAGIA 27

autophagosome-lysosome fusion, the inner membrane LC3-II is degraded and the outer
membrane LC3-II is deconjugated and recycled by Atg4. (b) For lysosomal digestion, lysosomal
acidification depends on vATPase, a multimeric enzyme complex that moves protons across
the lysosome membrane. The electrical gradient created is counterbalanced by parallel influx
of anions mediated by chloride proton antiporters. Cations, including calcium, can efflux
through distinct channels or transporters, including TPC2 and TRPML (mucolipin), which may
also influence pH. Proton leak pathways (dashed lines) require continued vATPase activity to
maintain a steady-state pH. Acidification kinetics are also contingent on the luminal buffering
power (not depicted). The more than 60 intralumenal acid hydrolases and the major lysosomal
integral membrane proteins are not depicted. The functions of many of the 25 lysosomal
transmembrane proteins are not yet clarified. Following substrate digestion, autophagy
regulation continues through interactions of the generated amino acids and a protein
complex, the ragulator, composed of multiple amino acid–sensing small GTPases, including
Rheb and Rag, which reversibly dock TORC1 and TFEB, depending on the amino acid
concentration. At low amino acid concentrations, as occurs during starvation, TORC1 activity
and phosphorylation of its substrate TFEB are inhibited, which releases TFEB to translocate to
the nucleus and induce5 transcription of genes that support increased autophagy. In a final
mTOR-dependent event, membranes from the maturing autolysosome are recovered for
lysosome biogenesis by a process involving the pinching off of evaginated surface membrane
(bottom drawing). AD, Alzheimer’s disease; FTD3, frontotemporal dementia 3; MND, motor
neuron disease; ML IV, mucolipidosis type IV; MPS IIIA, mucopolysaccharidosis type IIIA; MSD,
multiple sulfatase deficiency; NCL, neuronal ceroid lipofuscinosis; PD, Parkinson’s disease;
SBMA, spinal-bulbar muscular atrophy

In both disorders, mutations of CHMP2B superoxide dismutase (SOD1), the rate of


(loss of function charged multivesicular lysosome protein degradation slows,
body protein 2B) disrupt the ESCRT inducing autophagosome accumulation
machinery and cause ubiquitin-positive accompanied by neurodegeneration73.
aggregates to accumulate in neurons66,67. Lysosomal mechanisms. Completion of
Rab7, another effector of endosome autophagy requires lysosomal digestion of
maturation essential for complete autophagic cargoes and the release of
autophagy flux68, is mutated in forms of metabolites for reuse and signaling
CMT2, resulting in Rab7 functions (Fig. 4b). Upon fusion with an
overactivation69,70. Rab7 affects autophagosome, lysosomes introduce the
functions of another lateendosome necessary degradative machinery,
protein PLEKHM1, and mutations in the including a proton pump—the vacuolar
gene encoding this protein cause ATPase (vATPase)—and several dozen
osteopetrosis71, a disease primarily of hydrolytic enzymes of all types (for
lysosomes in osteoclasts but that example, proteases, lipases and
frequently has a neurodegenerative nucleases). These hydrolases, most of
component (Table 1). A mutant form of which have acidic pH optima, are activated
ALS2, an activator of Rab5, impedes ALS2- when the autolysosomal lumen becomes
mediated Rab5 activation on newly acidified to pH ~4.5–5.0 by the vATPase, a
forming amphisomes in this familial form large protein complex on the lysosomal
of ALS72, and, when it is expressed in a membrane that imports hydrogen ions74.
mouse model of ALS that expresses The ionic milieu of the lysosome is
TEÓRICO AUTOFAGIA 28

controlled by anion and cation channels nuclear translocation of dephosphorylated


that regulate ion fluxes (Ca2+, Cl−, K+ and TFEB and the increased expression of
others) and may also modulate H+ content lysosomal network genes. A sustained
(that is, lysosomal pH) and possibly period of induction reactivates TORC1 and
interactions of lysosomes with the plasma returns it to the lysosome, thus
membrane and other vesicles74. Heavily terminating autophagy50,76. Lysosomal
glycosylated membrane proteins, such as membrane, but not intralumenal contents,
LAMP1 and LAMP2, protect lysosomal is recaptured from expanded
membranes from self-digestion74, but autolysosomes by pinching off regions of
lysosome integrity is susceptible to the autolysosomal membrane76, which is
disruption in disease states by a host of also a TORC1-dependent process. These
modified products of lysosomal digestion lysosome–TORC1 inter-relationships have
or by internalized exogenous agents, and thus far been mainly demonstrated under
the stability of these lysosomal nutrient starvation conditions that are
membranes may be a potential normally not seen in neurons in vivo
therapeutic target75. Although crucial for because the brain has unique mechanisms
proteolytic clearance during autophagy, to buffer against such extreme nutritional
lysosomes also influence the initial stages challenges82. Nevertheless, it seems
of autophagy. Notably, this occurs through reasonable to suspect that the
a signaling complex anchored to the pathophysiological conditions in
vATPase, which reversibly docks TORC1 neurodegenerative disease states can elicit
and the transcription factor EB (TFEB) (Fig. similar lysosome–TORC1 cross talk. For
4b). When translocated to the nucleus, example, loss of the lysosomal proton
TFEB coordinately upregulates expression gradient caused by energy depletion or
of most genes involved in lysosome pathological conditions may depress
biogenesis and additional genes required TORC1 activity by blocking proton-coupled
for autophagosome formation. TORC1 exchange of nutrients or by directly
prevents this nuclear translocation76,77 affecting the vATPase83. Similarly,
and preserves its tether to protein 14-3-3 lysosomal membrane permeabilization
on lysosomes. Negative regulation of TFEB could promote nutrient leakage and
by TORC1 is mediated through the amino suppression of TORC1 (refs. 84,85). The
acid–Rag GTPase pathway78,79, a very close connection between lysosomal
component of a multiprotein signaling dysfunction and neurodegeneration is
complex (the ‘ragulator’) containing illustrated by the lysosomal storage
several small GTPases, including RHEB, that disorders (LSDs), the majority of which are
serve as amino acid–sensing activators of characterized by severe
TORC1 (ref. 80). TORC1 activation and TFEB neurodegenerative phenotypes. In many
phosphorylation on lysosomes are LSDs, a defect in any one of several
maintained in part by amino acids functionally dissimilar lysosomal enzymes
accumulating within the lysosome or leads to impaired autophagic turnover of
released during lysosomal proteolysis and proteins, causing particularly extensive
by a coupled glutamine–leucine shuttle autophagy pathology in the brain86. For
system regulating intracellular leucine81. example, the lysosomal protease cathepsin
When amino acid supplies are lowered, as D (CatD) is ubiquitously expressed, yet
occurs during starvation, the inhibited loss-of-function mutations of CatD cause
TORC1 relocates away from lysosomes, neuronal ceroid lipofuscinoses (NCLs; also
which induces autophagy and allows known as Batten’s disease), a severe
TEÓRICO AUTOFAGIA 29

neurodegenerative disorder associated that in the LSDs and more robust than in
with congenital mental retardation, or a any other late-onset brain disease.
juvenile neurodegenerative NCL subtype Autophagic vacuoles and especially
associated with dementia in humans and autolysosomes are the principal organelles
other mammalian species87. Deleting CatD contained within characteristic giant
or CatB and CatL causes similar phenotypes neuritic swellings97, which suggests that
in mice88. In Niemann-Pick type C disease autophagosomes can form and fuse with
(NPC), defective trafficking of cholesterol lysosomes but that elimination of
and other lipids caused by mutations in substrates from these autolysosomes is
either of two related endosomal genes, defective. Inhibiting lysosomal proteolysis
NPC1 and NPC2, leads to robust produces similar neuropathology in wild-
autophagic vacuole accumulation type mice and exacerbates amyloid and
preceding neuronal loss89. Notably, NPC is autophagy pathology in mouse models of
one of a very few disorders in which Alzheimer’s disease98. Key genetic risk
neuropathological lesions typical of factors and proteins involved in
Alzheimer’s disease, such as neurofibrillary Alzheimer’s disease pathogenesis directly
tangles, endosome anomalies and impair lysosome function98. The
intracellular amyloid β-peptide (Aβ42), Alzheimer’s disease–linked protein
develop in the absence of mutations in presenilin 1 is required for lysosome
Alzheimer’s-related genes90. Dystrophic acidification and thus for protease
neurites filled mainly with autophagic activation99. Presenilin 1 mutations, the
vacuoles, another identifying feature of most common cause of early-onset familial
Alzheimer’s disease pathologies that is also Alzheimer’s disease, disrupt these
seen in NPC, are caused by an inhibition of lysosomal functions99 and markedly
lysosomal proteases caused by stored accelerate disease onset and
lipids and exacerbated by excessive neuropathological severity100. The
autophagy induction89. Defective strongest genetic risk factor for late-onset
autophagy may also be the basis for Alzheimer’s disease is an allele of APOE
neurodegeneration in mucolipidosis type (which encodes a neuronal cholesterol
IV, a congenital disorder that transport protein) that encodes a protein
disproportionately affects the brain. Loss- variant called ApoE4 that destabilizes
of-function mutations in TRPML1, an ion lysosomal membranes101 in an allele-
channel on late endosomes and lysosomes, specific manner. Other factors contributing
abnormally acidifies lysosomal pH91 and to Alzheimer’s disease pathogenesis such
delays the clearance but not the fusion of as reactive oxygen species and
autophagic vacuoles92. Autophagosome accumulated Aβ peptide, oxidized lipids
clearance may be impaired in and lipoproteins similarly impede
osteopetrosis associated with lysosomal proteolysis, damage lysosomal
neurodegeneration93,94. In one form of membranes and disrupt lysosomal
the disease, the vATPase A3 subunit is integrity, releasing proteases that can
mutated, leading to defective lysosomal mediate the mixed necrosis and apoptosis
acidification95. Autophagosome-lysosome pattern of neuronal cell death seen in this
fusion is also impaired in several additional disease102–104. Abnormal upregulation
lysosomal storage disorders95,96 (Table of Rab5 and associated endocytosis
1). Disruption of autolysosomal proteolysis effectors, one of the earliest disease-
in Alzheimer’s disease causes a striking specific responses of neurons in
neuronal autophagy pathology similar to Alzheimer’s disease brains105, interferes
TEÓRICO AUTOFAGIA 30

with lysosome proteolysis by markedly related mutations has been linked to


accelerating endocytosis and backing up defects in endosomal-lysosomal
protein and lipid cargoes in swollen late trafficking116, lysosomal pH and calcium
endosomes106. Many factors that increase regulation117 and CMA45. A mouse model
risk of Alzheimer’s disease also promote of Parkinson’s disease resulting from
Rab5 upregulation, including mutant forms exposure to the neurotoxin 1-methyl-4-
of β-amyloid precursor protein (APP) found phenyl-1,2,3,6-tetrahydropyridine (MPTP)
in familial Alzheimer’s disease, APP shows accumulation of autophagosomes
duplication, the ApoE4 variant of and an early decrease in lysosome number
apolipoprotein and elevated amounts of in dopaminergic neurons as a result of
dietary cholesterol98. In sporadic and lysosomal membrane destabilization and
familial Parkinson’s disease, the affected cytosolic release of cathepsins84. Finally,
neurons develop inclusions containing α- mutations in lysosomal acid β-glucosidase
synuclein. Elevated amounts of this (GCase) that cause Gaucher’s disease also
protein, such as those resulting from increase the risk of Parkinson’s disease in
triplication of the gene encoding α- carriers who are not affected develop-
synuclein, are sufficient to cause mentally by Gaucher’s disease118. The
Parkinson’s disease107. Much of the α- compromised lysosomal degradation and
synuclein, especially overexpressed and neurodegeneration seen after deletion of
misfolded aggregate-prone forms, is the gene encoding GCase in mice can be
degraded by macroautophagy, which is largely prevented by overexpressing wild-
upregulated as CMA becomes type, but not mutant, forms of GCase119.
compromised in this disease108–110. Elevated autophagy induction in
Autophagic vacuoles accumulate when neurodegenerative states Increased
mutant or even wild-type α-synuclein is induction of autophagy is relatively
overexpressed in transgenic mice109,110, frequent in neurodegenerative diseases,
and, in rat PC12 cells, expression of mutant including in some disorders in which
α-synuclein decreases lysosomal autolysosome clearance mechanisms are
acidification111 and slows lysosomal impaired. What this greater induction rate
protein turnover46. Lysosomal pH is also signifies for disease development or
high in cells from patients with Parkinson’s implies for autophagy-related therapies,
disease caused by loss-of-function however, is not straightforward. In
mutations of lysosomal ATPase, ATP13A2, experimental models in which a disease-
leading to proteolytic failure112. The most related protein (for example, α-synuclein,
common autosomal-dominant form of or SOD1) is overexpressed120–124, the
Parkinson’s disease and a familial variant observed autophagy induction may in part
that closely resembles sporadic be a response to increased protein burden
Parkinson’s disease clinically113 involves independently of disease-specific factors,
LRRK2 mutation. Specific inhibitors of the and assessing this contribution may
kinase encoded by this gene exist114, require future investigations of additional
making it a particularly interesting model systems (such as knock-in mice or
therapeutic target. The range of LRRK2 fibroblasts and stem cells from patients).
functions in different diseases is Autophagy induction in response to
expanding115, and roles in autophagy and mutant or damaged proteins and
endosomal trafficking (via GTPase aggregates could be compensatory and
regulation) have been proposed. Altered neuroprotective at early stages of disease,
LRRK2 activity due to Parkinson’s disease– becoming counterproductive only if
TEÓRICO AUTOFAGIA 31

lysosomal clearance becomes amounts of toxic protein aggregates and


compromised as in some overexpression incompletely digested autolysosomal
models120–123. Compromised metabolites, more effective responses to
autolysosome function could also stress from redeploying non-essential
secondarily elevate autophagy induction constituents for energy and adaptive
when the concentrations of amino acids protein synthesis, and suppression of
are decreased and TORC1 activity is apoptotic cascades or necrosis by
suppressed. A pathological level of prevention of lysosomal membrane
autophagy induction has been inferred in a destabilization103,123. Stimulating the
few neuropathological states from the induction of autophagy has received the
cytoprotection that is seen when greatest attention so far, although
autophagy is blocked genetically or preventing or reversing impairments in
pharmacologically, as in hypoxic ischemic autolysosomal clearance of substrates is an
brain injury125 and two Parkinson’s equally promising therapeutic approach
disease models: 6-hydroxydopamine– and may prove to be the strategy of choice
injured nigral neurons and N-methyl-4- or necessity for certain neurodegenerative
phenyl pyridium (MPP) neurotoxicity126. disorders (Table 2). At least in cell
Although autophagy induction responses models2,89,128, when lysosomal
are generally viewed as neuroprotective, clearance is impaired, inducing autophagy
possibly in these acute injury situations the exacerbates pathology, which implies that,
response may be overexuberant. In NPC1- in a disease such as Alzheimer’s disease,
null cells89 and possibly in the mutant the success of any autophagy induction
ESCRT-III mouse model of frontotemporal intervention may depend on first relieving
dementia54, in both of which inhibition of the block in lysosomal clearance or
autophagy had ameliorative effects, the possibly preempting this block by
observed benefit may reflect a lowered risk stimulating autophagy induction very early
for lysosomal membrane destabilization in the disease. Enhancing autophagy
due to reduced delivery of autophagic induction and sequestration. Substantial
substrates to compromised lysosomes95. benefits of therapy with autophagy-
Therapeutic modulation in such situations inducing agents have been observed in
is clearly complex; however, in certain various mouse models of different
disorders in which autophagy clearance neurodegenerative diseases. Rapamycin, a
mechanisms are not known to be relatively selective inhibitor of TORC1,
substantially compromised, such as ameliorates neuropathology and
Huntington’s disease, the further neurodegeneration in transgenic mouse
promotion of autophagy induction might models of Huntington’s disease (mutant
be a logical interventional strategy. huntingtin)129, Alzheimer’s disease
Autophagy modulation as a therapy (mutant APP)130,131, prion disease132,
Autophagy is an attractive therapeutic spinocerebellar ataxia type 3 (SCA type 3;
target for a diverse range of diseases127. ref. 133) and Parkinson’s disease (mutant
For most of the neurodegenerative α-synuclein)134. Notably, with a few
disorders discussed earlier, the available exceptions, these studies have relied on
evidence favors a strategy of enhancing models in which a mutant pathogenic
the efficacy of autophagy by targeting the protein is overexpressed, and how an
stages that are specifically disrupted in increased protein burden contributes to
each disease. The expected benefits of the observed phenotype or its rescue by
such modulation might include lowered stimulation of autophagy is currently
TEÓRICO AUTOFAGIA 32

unclear. Moreover, in nearly all of these mediated tau phosphorylation, lithium has
studies, contributions of possible been studied in relatively small numbers of
autophagyindependent salutary effects of individuals with prodromal Alzheimer’s
mTOR inhibition on neural cells and mouse disease (mild cognitive impairment), and
behavior135 have not been excluded. Also, positive outcomes on some cognitive and
in a few models— SOD1 (G93A) transgenic tau measures have been observed145.
mice136 and MPTP neurotoxin models of Negative results have been reported in
Parkinson’s disease137— rapamycin several small short-term trials of lithium in
worsens autophagy functions and people with Alzheimer’s disease, although
neurotoxicity, possibly for reasons these may reflect trial design
discussed in the previous section. TORC1 limitations145 as well as the lower efficacy
inhibitors with greater specificity and expected of therapeutics applied late in
fewer pharmacological limitations or side this disease. Among a group of other US
effects are now under investigation, Food and Drug Administration–approved
including agents that inhibit both TORC1 compounds that also inhibit the
and TORC2 (ref. 138). For all of these phosphoinositol cycle139, the
agents, a key caveat to their use is the hypertensive agent rilmenidine showed
possibility of off-target effects given the promising effects in a study of a transgenic
diverse physiological roles of mTOR mouse model of Huntington’s disease that
signaling. Autophagy induction can be expresses mutant huntingtin, and it is in
upregulated independently of mTOR by clinical trials for this disease127. Trehalose,
activating the ULK1 kinase AMPK, through a disaccharide with pharmacological
the stimulation of cAMP– inositol 1,4,5- chaperone activity, which possibly acts
trisphosphate (IP3) or calpain–G- through AMPK activation, enhances
stimulatory protein α (Gsα) pathways139. clearance of mutant forms of huntingtin, α-
Not surprisingly, drugs influencing these synuclein and tau while conferring
signaling pathways affect multiple cytoprotective effects in cell and
processes besides autophagy, some of transgenic mouse models146. Plant-based
which may amplify or limit therapeutic polyphenols have a wide range of
efficacy140. The mood stabilizer lithium is molecular actions, which for some include
one of a group of clinically used agents (for activation of AMPK. Polyphenols, such as
example, valproate and carbamazepine) resveratrol, quercetin and catechins, and
that induce mTOR-independent autophagy plant alkaloids, such as berberine and
by inhibiting inositol monophosphatase curcumin, share this property and a range
and the phosphoinositol cycle141. Lithium of neuroprotective actions in models of
enhances the cellular clearance of neurodegenerative disease, particularly in
aggregate-prone forms of huntingtin and models of Alzheimer’s disease—a disorder
α-synuclein141. It has similar benefits in in which the link to altered AMPK signaling
SOD1 G93A–expressing mice that respond is relatively strong140. That some of these
poorly to rapamycin142, although, compounds extend lifespan in part by
interestingly, lithium yields mixed results in promoting autophagy via sirtuin 1 (SIRT1),
several ALS models that do respond to a molecule that influences longevity147,
rapamycin143,144, which suggests that indicates how closely cellular aging
the disease mechanisms in the different mechanisms are tied to autophagy failure
ALS models may not be identical. Mainly in late-onset neurodegenerative diseases.
because of its additional inhibitory effect Combining an agent that lowers IP3
on glycogen synthase kinase 3β (GSK-3β)- concentrations (for example, lithium,
TEÓRICO AUTOFAGIA 33

trehalose, small-molecule enhancers of the substrate target. For example,


rapamycin, L-690,330 and calpastatin) with acetylation of mutant huntingtin
rapamycin may have additive selectively targets it to
benefits143,144 but may also possibly autophagosomes153 and might be
multiply off-target effects—good and bad. accomplished using histone deacetylase
Several compounds, such as the natural inhibitors, already known to be
flavone finsetin, have dual actions on neuroprotective in Huntington’s disease
TORC1 and AMPK activities, both favoring models. Aging-related declines in LAMP2A
autophagy induction, and are function could be targeted to enhance
neuroprotective in several disease CMA, as suggested by the increased
models148. Protein phosphatase 2A huntingtin clearance seen in cells
(PP2A) agonists, which are currently in overexpressing LAMP2A or Hsc70 (ref.
clinical trials for Alzheimer’s disease 154). Membrane-penetrable synthetic
because of their inhibitory effects on tau ‘minichaperones’ mimicking the actions of
hyperphosphorylation, also promote Hsc70 or other co-chaperones might be
induction via TORC1 and AMPK149. modeled after earlier prototypes155.
Interestingly, the clinically used Enhancing lysosome efficiency.
antidiabetic agent metformin, which Autolysosomal dysfunction is relatively
attenuates disease development in some unexplored as a therapeutic target given its
but not all Alzheimer’s disease mouse frequent occurrence in brain diseases.
studies140, is a strong AMPK activator and Numerous therapeutic directions are
also increases PP2A activity among other possible, and those investigated so far
metabolic effects. Targeting a different show promise in disease models, strongly
early stage of autophagy, the beclin-1–PI3K supporting this approach and underscoring
CIII complex, when modulated, can the pathogenic importance of lysosomal
enhance autophagosome formation. dysfunction in Alzheimer’s disease,
Intracerebral delivery of virally packaged Parkinson’s disease and possibly other
Becn1 accelerates autophagy, reduces neurodegenerative disorders. Stimulation
accumulation of pathogenic proteins and of sluggish cathepsin activities, achieved by
slows the development of genetically deleting the endogenous
neurodegenerative changes in transgenic lysosomal cysteine protease inhibitor
mouse models of Parkinson’s disease110 cystatin B, substantially accentuated
and SCA type 3 (ref. 150). Smallmolecule autophagy and attenuated β-amyloid
beclin-1 mimetics151 could, therefore, pathology as well as cognitive decline in a
hold clinical promise for transgenic mouse model of mutant APP
neurodegenerative disease. Enhancing expression156. Similar functional benefits
selective autophagy. Selectively and improved synaptic functions have
eliminating specific autophagic targets been seen in other APP mouse models
while preserving basic housekeeping after either deleting cystatin C157,
functions of macroautophagy has overexpressing CatB158 or
considerable conceptual appeal as a pharmacologically raising the activities of
therapeutic intervention. cysteine proteases159. The latter strategy,
Pharmacologically mobilizing an which involves the chronic administration
autophagic receptor that targets specific of a cysteine protease inhibitor, suggests a
substrates could conceivably be achieved possible chaperone mechanism by which
by altering the receptor’s expression or more active protease is stabilized and
phosphorylation state152 or by modifying delivered from the endoplasmic reticulum
TEÓRICO AUTOFAGIA 34

to lysosomes. Indeed, reversible cell- limitations are imposed by the blood-brain


penetrant small-molecule inhibitors that barrier in treating central nervous system
act as pharmacological chaperones for disease. Additional lysosomal clearance
deficient or defective lysosomal hydrolases enhancement approaches exploit TFEB-
have proven effective in treating models of mediated upregulation of the gene
several LSDs160 and are in early-stage transcription that supports
clinical trials for Gaucher’s disease161. autophagosome formation and lysosomal
Pharmacological chaperones might be biogenesis, which can be modulated by
used to promote folding and delivery of phosphorylation. TFEB expression
other lysosomal components essential for attenuates storage phenotypes in
proteolysis, such as vATPase in Alzheimer’s neuronal stem cells isolated from the
disease99 and ATPase ATP13A2 in cerebral cortex of mice modeling multiple
Parkinson’s disease112, or to clear sulfatase deficiency and
accumulated lipids that impede mucopolysaccharidosis type IIIA and in
proteolysis89,162,163. Unclogging fibroblasts from people with Batten’s
lysosomes by clearing lipids is how 2- disease by a mechanism that also involves
hydroxypropylβ-cyclodextrin is suspected the activation of lysosome exocytosis169.
to work. This molecule is currently in early In this regard, non-neuronal cells are
clinical trials for NPC on the basis of its known to jettison some incompletely
efficacy in lowering autophagic substrate digested lysosomal substrates via calcium-
burden and rescuing neurological deficits regulated lysosomal exocytosis, a property
in an NPC mouse model164. Accumulation shared by glial cells and negatively
of the lipopigment lipofuscin during brain regulated by neutral sphingomyelinase 2
aging and in disease may also lower and neuraminidase170,171. This process is
lysosomal system efficiency165, and its impaired in mucolipidosis type IV172.
removal in several neuropathological Mature neurons are not known to
settings, including clinical macular exocytose lysosomal contents, but they
degeneration—a disorder with prominent can release cargoes via exosomes—a
lipofuscinosis166—is cytoprotective. The regulated and potentially druggable exit
venerable lipofuscinolytic agent pathway from multivesicular bodies. It has
centrophenoxine (meclofenoxate), a well- also been proposed that amphisome
tolerated ‘antiaging’ antioxidant that had cargoes such as neuronal Aβ, might be
equivocal effects for late-stage dementia disposed of in part by this alternative
in small early clinical trials167, may serve route173. A potential caveat to this
as a prototype for more effective approach is the recent discovery that the
lipofuscinolytic agents. Additional release and reuptake of pathogenic
approaches to clear storage materials or proteins such as tau and synuclein may
other interfering factors from lysosomes promote neuron-to-neuron propagation of
by pharmacologically reducing the disease via a prion-like templating of the
synthesis of precursors to the offending misfolded state of the protein174.
substrates or by replacing the missing or Lysosomal stability. The neuronal cell
defective hydrolase using direct death that is tightly linked to autophagy
administration or viral or stem cell delivery clearance failure involves lysosomal
are in clinical use for LSDs or are being membrane permeabilization (LMP) as a
actively investigated in clinical major triggering or amplifying factor103.
trials161,168. Although this is an area of Despite the knowledge that many
active and promising investigation, obvious pathogenic factors in neurodegenerative
TEÓRICO AUTOFAGIA 35

disease promote LMP as previously identification of over 400 candidate


discussed, there have been few attempts interacting proteins that support the basal
to mitigate this process pharmacologically autophagy network180 underscores the
so far. Calpains, which are abnormally complexity of autophagy but, more
activated in Alzheimer’s disease and importantly, hints at the potential for
several other major neuropathological manipulating a range of new control points
states, negatively regulate autophagy along the autophagy pathway in
induction and are important mediators of increasingly selective ways. These more
LMP in neuronal cell death. They act in part strategic interventions of the autophagic
by cleaving Hsp70, an established pathway may ultimately replace the
protectant against LMP175. Synthetic current first generation of drug targets
calpain inhibitors that may potentially act such as mTOR with its many possible off-
against varied targets have striking target effects on cell signaling. Concurrent
therapeutic effects in mouse models of programs to identify autophagy inhibitors
Alzheimer’s disease and other or enhancers for cancer, infectious disease
tauopathies176. Their action in both and other major disorders, which
promoting autophagy induction and complement the drug discovery programs
protecting against LMP further strengthens for neurodegenerative diseases, should
a previously strong rationale to develop yield a steady pipeline of candidate
highly specific calpain inhibitors to treat compounds. A major roadblock in moving
neurodegenerative disease. Lipids that these agents closer to the clinic, however,
accumulate abnormally in lysosomes in is the dearth of tools to measure
Alzheimer’s disease and some LSDs, autophagy activity in tissues and to
including gangliosides and sphingosine, establish target engagement or drug
promote LMP162,163,177 and provide efficacy in vivo. Available histological and
another rationale for pharmacologically biochemical readouts in tissues mainly
reducing their amounts using approaches assess autophagy induction and not overall
previously mentioned. Finally, elevated autophagic substrate turnover, which is
lysosomal pH has been linked to autophagy often blocked by disease at sites
failure and disease pathogenesis in a downstream of autophagy induction.
growing number of neurodegenerative Methods are urgently needed to monitor
disorders, and its correction is an obvious autophagic turnover rates in vivo directly
therapeutic target and could be potentially or through surrogate biomarkers and
druggable through the multiple pathways ideally to quantify changes in activity at
that regulate lysosomal pH178,179. each stage of the autophagy pathway,
Concluding considerations The evidence including the later lysosomal proteolytic
supporting a pathogenic role for stages, where static measurements of
autophagy impairment in several major protease activities in tissues are
neurodegenerative diseases is becoming notoriously uninformative of actual
persuasive and provides a strong rationale activities in situ. Evaluating the in vivo
for developing therapeutics to modulate efficacy of autophagy drugs in the brain is
autophagy in these disorders. As promising further challenged by obstacles such as
as these beginnings are, the surface has blood-brain barrier penetration of the
barely been cracked on the understanding agents and cellular heterogeneity of brain
of regulation of autophagy, especially in tissue. How neurons and glia contribute
the broader context of proteostasis and differentially to the overall autophagy
general metabolic regulation. The recent activity in brain and the possibility that
TEÓRICO AUTOFAGIA 36

different, or even reciprocal, responses to disease suggest that early drug


a drug may occur in different neural cell administration and thus very high margins
types are a few of the issues that will need of drug safety are necessary. In this regard,
to be considered in these in vivo therapies involving dietary products or
evaluations. Functional assays that enable manipulations represent a promising
autophagy fluxes in specific brain cell direction for future investigation given that
populations to be quantified in situ are autophagy is, above all, a homeostatic
essential for current drug validation response to changing supplies of nutrients,
studies in mouse models and for later use growth factors or energy. More complete
in clinical trials. Fluorescent reporters of characterization of nutrientsensing
CMA, autophagosome maturation and mechanisms and their regulation by
lysosomal proteolysis have recently been dietary factors and caloric intake may
developed for cell studies, and the ultimately enable basal autophagy to be
expression of such constructs in neuronal enhanced safely over long periods of time.
populations in mice would be an important Indeed, lifestyle practices, including diets
first step forward in preclinical testing of and exercise regimens to prevent the
new compounds. The recent major clinical metabolic syndrome of obesity, diabetes
trials of Alzheimer’s disease drugs have and cardiovascular dysfunction are already
emphasized the importance of establishing linked to a lowered risk for Alzheimer’s
target engagement to interpret outcomes, disease181, and failure of autophagy is
and the ongoing trials now rely heavily on strongly suspected to have an important
surrogate biomarkers measured in role in promoting this syndrome182. The
cerebrospinal fluid or by neuroimaging in diversity of possible ways that autophagy
addition to cognitive measures. Clinically might be targeted therapeutically provides
useful biomarkers of autophagy, a optimism that autophagy modulation may
particularly high priority, have yet to be ultimately prove to be an effective
investigated. Successes at preventing therapeutic approach for neuro-
disease onset in mouse models with degenerative diseases.
autophagy inducers and the more limited
success so far in reversing preexisting

MECHANISMS AND REGULATION OF AUTOPHAGOSOME FORMATION

Autophagy is an intracellular pathway for mechanism of action of these proteins


the bulk degradation of cytoplasmic during autophagosome biogenesis. In this
substances such as cytosol, protein review we briefly discuss recent findings on
aggregates and organelles. Autophagy is the molecular players and mechanisms
characterized by the formation of double- involved in autophagosome formation. In
membrane bound vesicles called particular, we will focus on the
autophagosomes, which engulf the cargo mechanisms regulating membrane
and transport it to the vacuole/lysosome recruitment as well as membrane
for breakdown and recycling. Even though remodeling during autophagosome
several proteins in this pathway have been formation.
identified, little is known about the
Introduction
TEÓRICO AUTOFAGIA 37

Macroautophagy (hereafter autophagy) is targeting (Cvt) pathway exists in yeast


an important intracellular degradation cells. This pathway fulfilling a biosynthetic
pathway conserved among eukaryotes. It function selectively delivers three cytosolic
mediates intracellular homeostasis by the enzymes – aminopeptidase 1 (Ape1), a-
degradation of long-lived proteins and mannosidase (Ams1), and aspartyl
entire organelles. Autophagy also aminopeptidase (Ape4) – to the vacuole
contributes to the adaptive response to [8–10]. In the following sections we will
starvation and various other extrinsic and briefly discuss recent progress in our
intrinsic stresses. Not surprisingly, defects understanding of autophagy with
in autophagy have been associated with emphasis on the regulation and
several human diseases, including mechanisms of autophagosome
increased susceptibility to infectious formation.
diseases, neurodegenerative disorders and
Autophagy proceeds through a series of
cancer. Despite its various important roles,
steps
the regulation and mechanisms of
autophagy induction and autophagosome Autophagy pathways can be broken down
formation remain poorly understood. into the following series of steps:
During autophagy cytoplasmic cargo is Induction, cargo recognition,
engulfed by double-membrane organelles autophagosome nucleation,
called autophagosomes. After their autophagosome expansion and
formation autophagosomes fuse with completion, autophagosome fusion with
lysosomes (or the vacuole in yeast), within the vacuole/lysosome and breakdown, and
which the inner membrane and the cargo recycling of the resulting macromolecules
are degraded. Both non-selective ‘bulk’ (Figure 1, [11]). Genetic analyses in yeast
autophagy as well as the selective have identified over 35 genes, termed Atg1
autophagy of specific proteins or to Atg35 that are required at one or more
organelles have been described [1]. During of these steps during autophagy [12]. A
non-selective autophagy, bulk cytosol and subset of these Atg proteins is required for
other cytoplasmic components are autophagosome formation in all types of
thought to be non-specifically sequestered autophagy. These proteins are referred to
into autophagosomes. By contrast, during as the ‘core’ autophagic machinery [13]
selective autophagy cargo is selectively and can be divided into three different
sequestered into autophagosomes. These functional groups: Firstly, the Atg1-ULK1
latter structures tend to contain little kinase complex is required for the
cytoplasm and their shape reflects that of induction of autophagy and regulates
their cargo. Selective types of autophagy together with Atg2 and Atg18 the cycling of
mediate the degradation of mitochondria Atg9 to an from the site of autophagosome
(mitophagy) [2,3], the elimination of formation. Secondly, Atg6 and Atg14 are
peroxisomes (pexophagy) [4], the selective part of a phosphatidylinositol 3-kinase
removal of ribosomes (ribophagy) [5] as complex required for vesicle nucleation.
well as the elimination of invading bacteria Finally, two conjugation systems that are
and viruses (xenophagy) [6,7]. In addition, built around the ubiquitin-like proteins
another selective and highly related Atg8 and Atg12 are essential for
pathway called the cytoplasm-to-vacuole autophagosome formation. The first
TEÓRICO AUTOFAGIA 38

conjugation system entails the conjugation phosphatidylethanolamine (PE) [14]. Atg8–


of Atg8 (or its orthologs in complex PE conjugation is mediated by Atg3 and
eukaryotes) to the membrane lipid Atg7.

The second conjugation system entails the Signaling in autophagy


conjugation of Atg12 to an internal lysine
The level of autophagy in a cell must be
of Atg5 [15]. The Atg5–Atg12 conjugate
finely balanced, as too little can be
subsequently forms a non-covalent
deleterious, but excessive levels of
complex with the coiled-coil protein Atg16
autophagy may also be harmful. The
[16]. Orthologs of most of these yeast Atg
cellular signaling events inducing or
proteins exist in complex eukaryotes [13].
restricting autophagy, as well as the
mechanisms of membrane formation and
TEÓRICO AUTOFAGIA 39

cargo selection, remain poorly understood. important for autophagy function remain
Epistasis analyses in yeast revealed that to be investigated in more detail.
the conserved Atg1/ULK1 kinase functions
Atg9: the membrane-recruiting factor?
upstream of cargo selection and
autophagosome induction. Atg1 is a In yeast autophagosomes are generated at
conserved serine-/threonine protein the pre-autophagosomal structure (PAS),
kinase and a key regulator of autophagy which is localized close to the vacuole.
and the Cvt pathway [17]. It is found in a Most Atg proteins are cytosolic and only
large complex composed of several transiently associate with the PAS [24].
proteins including Atg13, another Atg Atg9 is the only conserved transmembrane
protein essential for autophagy function protein essential for autophagosome
[18]. Atg1/ULK1 and Atg13 are highly formation [25]. In yeast, Atg9 is one of the
phosphorylated. Their phosphorylation first components associating with the PAS
status changes depending on the nutrient and most other Atg proteins require Atg9
availability, suggesting a phospho- for their localization at this perivacuolar
dependent regulatory mechanism for site [24], suggesting a role for Atg9 in
autophagy induction [18–20]. Indeed, assembling the core autophagic
Atg1/ULK1 and Atg13 were found to be machinery. Atg9 (mAtg9 in mammals)
phosphorylated by the target of rapamycin forms a conserved complex together with
(TOR) complex 1 under nutrient-rich Atg2 and Atg18 (WIPI-1 in mammals).
conditions in mammals and yeast, and Owing to its early localization to the PAS
additional regulated phosphorylation and it being an integral membrane protein,
events were observed for yeast Atg1 [21], Atg9 has been the focus of many studies
indicating that extra kinases are involved in aimed at understanding the nature of the
regulating the Atg1 complex. Atg13 PAS and the origin of the autophagosomal
phosphorylation in yeast is believed to membranes. Autophagosomes have been
reduce its binding affinity for Atg1, thereby proposed to either form from preexisting
preventing Atg1 kinase activation under membranes or to assemble de novo by
nutrient rich conditions. In flies and vesicular transport. Various membrane
mammals, conversely Atg1/ULK1 and sources have been suggested to supply
Atg13 constitutively bind to each other lipids during autophagosome formation
independent of the nutrient availability. (see below). In contrast to other Atg
This indicates mmthat the Atg1 complex in proteins, Atg9 in yeast is found in
these organisms is unlikely to be regulated peripheral cytoplasmic structures in
by its association with Atg13, but rather by addition to the PAS, and has been
phosphorylation or the nutrient- proposed to cycle between these two
dependent interaction of additional factors locations [26]. This cycling depends on the
[22,23]. While it seems likely that Atg1 kinase complex [26] as well as on the
Atg1/ULK1 plays an essential role in ability of Atg9 to multimerize [27]. Both
autophagy regulation from yeast to cycling and multimerization have been
humans, the precise regulation and suggested to contribute to the recruitment
components of the Atg1/ULK1 kinase of autophagosomal membranes, as
complex as well as Atg1 kinase targets mutants defective in cycling and
multimerization fail to support
TEÓRICO AUTOFAGIA 40

autophagosome formation [26,27]. In signaling might be involved in regulating


mammalian cells mAtg9 also shows a dual this process. Several factors involved in
localization. One pool of mAtg9 localizes to Atg9 trafficking have been described in
the perinuclear region where it colocalizes yeast and mammals and a picture is slowly
with proteins from the trans-Golgi emerging (Figure 2). Nevertheless, further
network. A second pool has been shown to analysis of thenature of the cycling events
partially overlap with endosomal markers taken by Atg9, as well as the regulation of
[28]. The endosomal pool increases the retrieval mechanisms by Atg1/ULK1
following nutrient starvation and ULK1 has and other factors, is required to
been found to regulate this redistribution understand the role of Atg9 in
event similar to yeast Atg1 [28]. The autophagosomal membrane recruitment.
precise role of Atg1/ULK1 in regulating the

localization of Atg9/mAtg9 remains to be Membrane dynamics during


further investigated. However, if Atg9 autophagosome formation
cycling results in a net transport of lipids to
In mammalian cells several sources for the
the autophagosomal membrane, the
autophagosomal membrane have been
density of Atg9 on the membrane
suggested. The endoplasmic reticulum
structures leaving the PAS must be higher
[29–31], the Golgi apparatus [32 ,33 ],
than on the structures reaching the PAS.
mitochondria [34] as well as the plasma
Importantly, Atg9 is believed to be absent
membrane [35,36 ] have been implicated
from the completed autophagosome [25],
in contributing lipids to isolation
implying that a retrieval mechanism from
membranes. As discussed in a number of
these structures must also exist. Atg1
recent reviews it now seems likely that
TEÓRICO AUTOFAGIA 41

mammalian cells have the ability to utilize Golgi tubulo-vesicular compartment


different compartments as a lipid source positive for Atg9 moves en bloc to the
for autophagosome formation [37,38]. vacuole [39].It was further proposed that
However, it is not clear whether a common this tubulo-vesicular compartment
mechanism exists that mediates the undergoes homotypic fusion and
formation of isolation membranes and remodeling in order to give rise to isolation
ultimately autophagosomes from these membranes [39]. It remains unclear how
various donor membranes. It has been this remodeling occurs. The mechanism by
observed in electron microscopy studies which vesicular precursors fuse to mediate
that isolation membranes are connected to isolation membrane formation is not fully
the ER by small necklike continuities understood. Yeast Atg8 when conjugated
[30,31]. Supporting evidence for continuity to PE has been proposed to mediate vesicle
between the isolation membrane and the tethering and hemifusion [40]. Hemifusion
ER comes from light microscopy is an intermediate step towards full fusion
experiments where it was observed that an where the contacting but not the distal
ER-resident transmembrane protein has monolayers of the membranes are fused
access to isolation membranes [29]. [41]. Recently the tethering activity of
Similarly, it has been postulated from yeast Atg8 has been confirmed; however,
advanced light microscopy studies that the hemifusion activity of Atg8 is only
isolation membranes grow out of the outer observed when very high PE
mitochondrial membrane [34]. The concentrations are present in the vesicles
mechanism by which the isolation [42]. These high PE concentrations are
membranes keep their unique identity unlikely to represent the physiological
while connected to their donor conditions in vivo. Thus, unless the tubulo-
membranes(ER and mitochondria, vesicular precursors have an unusual and
respectively) remains unclear. highly fusogenic lipid composition, Atg8 is
Alternatively, it has been proposed that unlikely to mediate fusion. In the same
isolation membranes are formed by the paper the authors presented evidence for
fusion of vesicular carriers that are derived the involvement of SNARE proteins in
from the plasma membrane [35,36 ] or the membrane fusion events that give rise to
Golgi apparatus [32 ,33 ]. The fusion of the Atg9-positive tubulovesicular
these vesicular carriers could mediate the precursors [42]. It therefore seems likely
formation of isolation membranes and that these SNAREs are localized on the
eventually autophagosomes.In these precursors when they undergo homotypic
vesicular fusion models there is no need fusion during isolation membrane
for any diffusion barrier separating the formation and could thus mediate their
isolation membrane from the donor fusion. As in yeast, the lipidated forms of
membrane, since autophagosome the mammalian Atg8 homologues LC3 and
formation occurs far from the donor GATE-16 have been shown to mediate
membrane. Recent evidence lends support membrane tethering [43 ]. In addition, in
to the vesicular precursor hypothesis. By the same study both proteins were shown
following Atg9 in Atg9 overexpressing cells to mediate full fusion of vesicles at
using electron microscopy, it was observed relatively low and thus physiologically
that after induction of autophagy a post- relevant PE concentrations [43 ]. The
TEÓRICO AUTOFAGIA 42

reason for the higher apparent fusogenic the core molecular machinery required for
activity of LC3 and GATE-16 compared to autophagy. The challenge for the future is
yeast Atg8 is unclear. However, in to understand the mechanisms of action as
agreement with findings in yeast where well as the regulation of the autophagic
SNAREs promote the fusion of protein machinery. For example, how does
autophagosomes [42], it has recently been Atg1 induce autophagosome formation
shown that SNARE-dependent homotypic (i.e. what are its relevant targets)? What is
fusion of Atg16L positive precursors is the role of Atg9 during autophagosome
required for autophagosome formation in formation? Does it indeed supply lipids to
mammalian cells [36 ]. Cells may therefore the isolation membrane? How are
employ the conventional fusion machinery membranes sculpted into isolation
composed of SNAREs and their accessory membranes and finally autophagosomes?
molecules to form autophagosomes. What are the roles of the Atg8 and Atg12
conjugation systems during these
Concluding remarks
processes? Answering these questions will
In recent years tremendous progress has bring us closer to a mechanistic
been made regarding the identification of understanding of the autophagic process.

MAMMALIAN AUTOPHAGY: CORE MOLECULAR MACHINERY AND SIGNALING


REGULATION
Autophagy, a cellular catabolic pathway, is machinery and signaling pathways that are
evolutionarily conserved from yeast to involved in mammalian autophagy.
mammals. Central to this process is the
Introduction
formation of autophagosomes, double-
membrane vesicles responsible for Autophagy, literally meaning ‘self-eating’,
delivering long-lived proteins and excess or embraces three major intracellular
damaged organelle into the lysosome for pathways in eukaryotic cells,
degradation and reuse of the resulting macroautophagy, microautophagy, and
macromolecules. In addition to the chaperonemediated autophagy (CMA),
hallmark discovery of core molecular which share a common destiny of
machinery components involved in lysosomal degradation, but are
autophagosome formation, complex mechanistically different from one another
signaling cascades controlling autophagy [1,2]. During macroautophagy, intact
have also begun to emerge, with mTOR as organelles (such as mitochondria) and
a central but far from exclusive player. portions of the cytosol are sequestered
Malfunction of autophagy has been linked into a double-membrane vesicle, termed
to a wide range of human pathologies, an autophagosome. Subsequently, the
including cancer, neurodegeneration, and completed autophagosome matures by
pathogen infection. Here we highlight the fusing with an endosome and/or lysosome,
recent advances in identifying and thereby forming an autolysosome. This
understanding the core molecular latter step exposes the cargo to lysosomal
TEÓRICO AUTOFAGIA 43

hydrolases to allow its breakdown, and the autophagy was first identified in
resulting macromolecules are transported mammalian cells approximately 50 years
back into the cytosol through membrane ago, our molecular understanding of it only
permeases for reuse (Figure 1). By started in the past decade, largely based on
contrast, microautophagy involves the the discovery of autophagy-related (ATG)
direct engulfment of cytoplasm at the genes initially in yeast followed by the
lysosome surface, whereas CMA identification of homologs in higher
translocates unfolded, soluble proteins eukaryotes [4]. Among these Atg proteins,
directly across the limiting membrane of one subset is essential for autophagosome
the lysosome. In this review, we will focus formation, and is referred to as the ‘core’
on mammalian macroautophagy molecular machinery [5]. These core Atg
(hereafter referred to as autophagy), proteins are composed of four subgroups:
which plays important physiological roles first, the Atg1/unc-51-like kinase (ULK)
in human health and disease. The basal, complex; second, two ubiquitin-like
constitutive level of autophagy plays an protein (Atg12 and Atg8/LC3) conjugation
important role in cellular homeostasis systems; third, the class III
through the elimination of damaged/old phosphatidylinositol 3-kinase
organelles as well as the turnover of long- (PtdIns3K)/Vps34 complex I; and fourth,
lived proteins and protein aggregates, and two transmembrane proteins, Atg9/mAtg9
thus maintains quality control of essential (and associated proteins involved in its
cellular components. On the other hand, movement such as Atg18/WIPI-1) and
when cells encounter environmental VMP1. The proposed site for
stresses, such as nutrient starvation, autophagosome formation, to which most
hypoxia, oxidative stress, pathogen of the core Atg proteins are recruited, is
infection, radiation, or anticancer drug termed the phagophore assembly site
treatment, the level of autophagy can be (PAS). In this review, we mainly highlight
dramatically augmented as a the recent advances in mammalian
cytoprotective response, resulting in autophagy in terms of the molecular
adaptation and survival; however, machinery involved in the formation and
dysregulated or excessive autophagy may maturation of autophagosomes and the
lead to cell death. Thus, defective signaling cascades needed for the
autophagy has been implicated in the regulation of autophagy. The clarification
pathogenesis of diverse diseases, such as of how autophagy is modulated in
certain types of neuronal degeneration response to intracellular and
and cancer, and also in aging [3]. Although
TEÓRICO AUTOFAGIA 44
TEÓRICO AUTOFAGIA 45

extracellular stresses relies largely on the autophagy and binds to ULK1 and ULK2
elucidation of the signaling network independent of nutrient status [12 ], in
upstream of the Atg machinery. Core contrast to the yeast Atg1–Atg17
molecular machinery ULK complexes The interaction. In addition, under nutrient-
yeast serine/threonine kinase Atg1 plays a rich conditions, the large ULK1–Atg13–
key role in the induction of autophagy, FIP200 complex contains mammalian
acting downstream of the target of TORC1 (mTORC1); conversely, following
rapamycin (TOR) complex 1 (TORC1). A nutrient deprivation, mTORC1 is quickly
family of mammalian Atg1 proteins has dissociated from the ULK1 complex [11 ].
been identified; among these, unc-51-like There are several phosphorylation events
kinases 1 (ULK1) and 2 have the highest within this complex, including
similarity with yeast Atg1 and appear to be phosphorylation of mAtg13 by ULK1, ULK2,
closely related. siRNA knockdown of ULK1 and mTORC1, phosphorylation of FIP200
or ULK2 blocks autophagy in HEK293 cells by ULK1 and ULK2, and phosphorylation of
[6 ]. However, ULK1/ mice display normal ULK1 and ULK2 by mTORC1 (Figure 1) [6 ,11
autophagy in response to nutrient ]. Under conditions that induce autophagy,
deprivation, but delay mitochondrial a decrease in mTORC1 activity leads to
clearance during reticulocyte maturation dephosphorylation of ULK1, ULK2, and
[7]. The basis for these differences is not mAtg13, activation of ULK1 and ULK2, and
known. It is possible that in some tissues, phosphorylation of mAtg13 and FIP200 by
ULK2 can compensate for the deficiency of ULK1 and ULK2 [6 ,11 ]. Further studies are
ULK1. Furthermore, a role of ULK3 in required to characterize the functional
autophagy induction in oncogene-induced significance of these phosphorylation
cell senescence has been described events. Recently, a new, mAtg13-
recently [8]. Thus, at least three ULKs are interacting protein, Atg101, was found to
involved in mammalian autophagy interact with ULK1 in a mAtg13-dependent
regulation and they have mechanistically manner, and is essential for autophagy
different roles in vivo. Yeast Atg1 exists in [13]. However, the role of the ULK1–
a complex with Atg13 and Atg17. Atg13 is Atg13–Atg101 complex in autophagy
phosphorylated in a TORC1-dependent regulation remains unclear.
manner and the phosphorylation state of
Two ubiquitin-like proteins, Atg12 and
Atg13 modulates its binding to Atg1 and
Atg8/LC3, and their conjugation systems
Atg17; inactivation of TORC1 leads to
dephosphorylation of Atg13, increasing Studies in yeast and mammals have
Atg1–Atg13– Atg17 complex formation identified two ubiquitin-like proteins,
and activating autophagy [4,9]. ULK1 and Atg12 and Atg8/LC3, and their respective,
ULK2 are also in a large complex that partially overlapping, conjugation systems,
includes the mammalian homolog of Atg13 which are proposed to act during
(mAtg13) and the scaffold protein FIP200 elongation and expansion of the
(an ortholog of yeast Atg17) [6 ,10,11 ]. phagophore membrane. Atg12 is
mAtg13 is essential for autophagy, and it conjugated to Atg5 in a reaction that
directly interacts with ULK1, ULK2, and requires Atg7 and Atg10 (E1 and E2-like
FIP200 independent of its phosphorylation enzymes, respectively). The Atg12–Atg5
state [6 ,11 ]. FIP200 is also required for conjugate then interacts noncovalently
TEÓRICO AUTOFAGIA 46

with Atg16L, which oligomerizes to form a complexes I and II. Complex I, consisting of
large multimeric complex called the Atg16L Vps34, Vps15, Atg6, and Atg14, is required
complex. Atg8/LC3 is cleaved at its C for the induction of autophagy, and the
terminus by Atg4 to generate the cytosolic lipid kinase activity of Vps34 is essential for
LC3-I with a Cterminal glycine residue, generating phosphatidylinositol (3)-
which is conjugated to phosphate (PtdIns(3)P) at the PAS to allow
phosphatidylethanolamine (PE) in a the recruitment of other Atg proteins.
reaction that requires Atg7 and the E2-like Complex II, consisting of Vps34, Vps15,
enzyme Atg3. The lipidated form of LC3 Atg6, and Vps38, is required for the
(LC3-II) is attached to both faces of the vacuolar sorting of carboxypeptidase Y. In
phagophore membrane, but is ultimately mammals, there are two types of PtdIns3K:
removed from the autophagosome outer classes I and III. Formation of the
membrane, which is followed by fusion of mammalian class III PtdIns3K complex,
the autophagosome with a late including hVps34, Beclin 1 (a homolog of
endosome/lysosome [4]. Recent work Atg6), and p150 (a homolog of Vps15), is
suggests that these two ubiquitination-like conserved. The orthologs of Atg14 and
systems are closely connected. On the one Vps38 have recently been identified and
hand, the Atg16L complex is localized to are called Atg14-like protein (Atg14L or
the phagophore and it can act as a novel Barkor) and ultraviolet irradiation
E3-like enzyme, determining the sites of resistance-associated gene (UVRAG),
Atg8/LC3 lipidation [14,15]. On the other respectively [18–20]. Atg14L plays an
hand, the Atg8/LC3 conjugation machinery important role in mammalian autophagy.
seems to be essential for the formation of Under nutrient-rich conditions, a
the Atg16L complex. In Atg3- deficient subpopulation of Atg14L localizes to the
mice, where no LC3-II can be detected, ER; upon starvation, Atg14L localizes to
Atg12–Atg5 conjugation is markedly Atg16L-positive and LC3-positive
reduced, and dissociation of the Atg16L structures, indicating the phagophore and
complex from the phagophore is delayed; autophagosome, respectively,
autophagosomes are smaller than in the independently of the interaction of Atg14L
wild type and appear either open-ended or with hVps34 and Beclin 1 [18,21].
multilamellar [16], indicating a role for the Importantly, depletion ofAtg14L reduces
Atg16L complex and LC3 lipidation for the Atg16L and LC3 puncta formation [21].
elongation and closure of the phagophore. Overexpression of Atg14L stimulates the
This hypothesis is further supported by the kinase activity of hVps34, and induces
observation that overexpression of an autophagy, whereas Atg14L knockdown
inactive mutant of Atg4 inhibits the reduces PtdIns(3)P production, and inhibits
lipidation of LC3, and in these cells a autophagy [19,22]. Thus, a possible role of
significant number of nearly complete Atg14L is to direct the class III PtdIns3K
autophagosomes are not closed [17]. complex to the phagophore to initiate the
recruitment of Atg machinery. Recent
Class III phosphatidylinositol 3-kinase
studies suggest that UVRAG participates in
complex
at least four different mechanisms to
In yeast, the only PtdIns3K is Vps34, and it regulate autophagy. First, UVRAG
exists in two different complexes, competes with Atg14L for binding to Beclin
TEÓRICO AUTOFAGIA 47

1; the interactions of Atg14L and UVRAG that are required for mammalian
with the Beclin 1–hVps34–p150 complex autophagy. mAtg9, with both the N and C
are mutually exclusive [18,19]. Second, termini in the cytosol, spans the
UVRAG interacts with Bif-1 (Baxinteracting membrane six times. It is located in the
factor 1); Bif-1 is required for autophagy transGolgi network and late endosomes,
and colocalizes with Atg5, LC3, and mAtg9 and upon starvation or rapamycin
during starvation [23]. It is proposed that treatment, redistributes to peripheral
the recruitment of Bif-1 via UVRAG may sites, overlapping with GFP-LC3-positive
provide the machinery to deform autophagosomes. The cycling of mAtg9
membranes, as Bif-1 has an N-BAR domain after starvation is ULK1-dependent, and
and shows membrane binding and bending also requires the kinase activity of hVps34
activities [24]. Third, UVRAG interacts with [27], which is similar to the yeast protein
the class C Vps/HOPS proteins, promoting [28]. Although its functions remain
autophagosome fusion with the late unclear, based on the existing data from
endosome/lysosome, thereby accelerating yeast Atg9, mAtg9 potentially contributes
delivery and degradation of autophagic to the delivery of membrane to the
cargo [25 ]. Fourth, the recently identified forming autophagosome, an attractive
Rubicon (RUN domain and cysteine-rich model that needs to be experimentally
domain containing, Beclin 1-interacting) tested in mammalian cells. In contrast to
protein forms a complex with UVRAG– mAtg9, VMP1 has no known homologs in
Beclin 1–hVps34–p150; this complex yeast. The localization of VMP1 is
localizes to the late endosome/lysosome controversial: in mammalian cells it is
and negatively regulates autophagosome localized to the plasma membrane and also
maturation [21,22]. Rubicon reduces colocalizes with LC3 and Beclin 1 upon
hVps34 activity and inhibits autophagy. In autophagy induction [29], whereas the
addition to hVps34, Atg14L, and UVRAG, VMP1 homolog in Dictyostelium
Beclin 1 also interacts with Ambra1 discoideum localizes to the ER [30]. In
(activating molecule in Beclin 1- regulated mammalian cells, ectopical overexpression
autophagy). Ambra1 functions as a positive of VMP1 triggers autophagy even under
regulator of autophagy and the mechanism nutrient-rich conditions, whereas
remains unclear [26]. Collectively, there depletion of VMP1 blocks starvation-
exist multiple mammalian hVps34–Beclin 1 induced and rapamycin-induced
complexes that may participate in distinct autophagy [29]. Importantly, VMP1
steps of autophagy regulation (Figure 1), interacts with Beclin 1, and this interaction
either at the early stage to promote is essential for autophagy induced by
autophagosome formation or at the later VMP1 overexpression [29]. VMP1 might
stage to promote autophagosome function as a transmembrane protein that
maturation. recruits Beclin 1 and other components in
the class III PtdIns3K complex to the
Transmembrane proteins in mammalian
phagophore. This is supported by a recent
autophagy
finding that a novel VMP1-interacting
Mammalian Atg9 (mAtg9) and vacuole protein, TP53INP2 (tumor protein 53-
membrane protein 1 (VMP1) are the two induced nuclear protein 2), is essential for
transmembrane proteins so far identified the translocation of Beclin 1 and LC3 to
TEÓRICO AUTOFAGIA 48

autophagosomes upon autophagy also sensitive to rapamycin, which in many


stimulation, potentially through its settings stimulates autophagy. However, a
interaction with VMP1 [31 ]. TP53INP2 is recent report challenged this view by
essential for autophagy. It translocates showing that rapamycin and siRNA
from the nucleus to autophagosomes upon knockdown of one of the key downstream
autophagy induction, where it interacts effectors of mTORC1, S6 kinase 1 (S6K1),
with LC3 as well as VMP1, but not Beclin 1. inhibit autophagy in cancer cells [33], and
a more recent finding shows that mTORC1
SIGNALING PATHWAYS REGULATING
regulates autophagy through an unknown
AUTOPHAGY
mechanism that is essentially insensitive to
PtdIns3K–Akt–mTORC1 rapamycin [34]. mTORC1 integrates
upstream activating signals that inhibit
TOR is a highly conserved serine/threonine
autophagy through the class I PtdIns3K-
protein kinase that acts as a central sensor
protein kinase B (PKB, also known as Akt)
of growth factors, nutrient signals, and
pathway (Figure 2). Upon association with
energy status. TOR serves as a master
growth factor, receptor tyrosine kinases
regulator of autophagy [32]. TOR exists in
undergo autophosphorylation and become
two distinct complexes, TORC1 and TORC2
activated, leading to the stimulation of two
that are conserved from yeast to
key signal-transducing components: the
mammals, and TORC1 has a primary
small GTPase Ras and class I PtdIns3K. Class
function in regulating autophagy. In yeast,
I PtdIns3K catalyzes the production of
inhibiting the TORC1 complex during
PtdIns(3)P at the plasma membrane, which
nitrogen starvation or by rapamycin
increases
stimulates autophagy [4]. The mTORC1 is
TEÓRICO AUTOFAGIA 49

membrane recruitment of both PKB and its downstream protein complex, the
activator PDK1 (phosphoinositide- tuberous sclerosis complex 1/2
dependent protein kinase 1), leading to the (TSC1/TSC2). The TSC1/TSC2 heterodimer,
activation of PKB. PtdIns3K activity can be which is a stable complex, senses the
opposed by PTEN, a 30 -phosphoinositide upstream inputs from various kinases,
phosphatase, subsequently decreasing including PKB and ERK1/2 [35,36].
PKB activity, and inhibiting mTOR. Phosphorylation of TSC2 by PKB or ERK1/2
PtdIns3K–PKB activation suppresses leads to the disruption of its complex with
autophagy in mammalian cells. PKB further TSC1, and results in mTOR activation.
activates mTORC1 through inhibiting a TSC1/TSC2 acts as the GTPase-activating
TEÓRICO AUTOFAGIA 50

protein for Rheb, a small GTP-binding pathway phosphorylates and stabilizes


protein that binds to and activates mTOR p27kip1, a cell cycle inhibitor, and
in its GTP-bound form. Ras has opposing stabilized p27kip1 induces autophagy [42].
roles in autophagy regulation: it inhibits An increase in the cytosolic free Ca2+
autophagy by activating the PtdIns3K– concentration and cytokines (such as
PKB–mTORC1 pathway, and at the same TRAIL) activates AMPK via activation of the
time, it may induce autophagy via the Raf- Ca2+/ calmodulin-dependent kinase
1–MEK1/2–ERK1/2 pathway [37,38]. kinase-b (CaMKKb) and transforming
Finally, the mTORC2 complex is also growth factor-b-activating kinase 1 (TAK1),
involved in autophagy regulation. Full respectively, and these pathways are
activation of PKB requires mTORC2 [39], required for Ca2+- induced or TRAIL-
and inhibition of PKB, caused by mTORC2 induced autophagy [43,44 ]. Moreover,
depletion, reduces the phosphorylation of, AMPK activity contributes to the induction
and therefore activates, the forkhead box of autophagy during hypoxia [45].
O (FoxO3) transcription factor, which
p53
stimulates autophagy in muscle cells
independent of the activity of mTORC1 The p53 tumor suppressor, the ‘guardian
[40]. of the cellular genome’, has dual positive
and negative regulatory roles in autophagy
AMPK
induction (Figure 2) [46]. Upon genotoxic
The AMP-activated protein kinase (AMPK) stress or oncogenic activation, the
is another sensor of cellular bioenergetics, activation of p53 induces autophagy; p53
specifically in response to energy stress. activates AMPK, which in turn, activates
During nutrient and energy depletion, the TSC1/TSC2 complex, leading to the
AMPK is activated by a decreased inhibition of the mTORC1 pathway [47].
ATP/AMP ratio through the upstream LKB1 p53 can also induce autophagy through
kinase (encoded by the Peutz–Jeghers upregulation of the damage-regulated
syndrome gene). Active AMPK leads to autophagy modulator (DRAM) [48].
phosphorylation and activation of Remarkably, chemical inhibition of p53,
TSC1/TSC2 and inhibition of mTORC1 knockdown of p53 with siRNA, or deletion
activity. Thus, the phosphorylation of of the p53 gene can trigger the onset of
TSC1/TSC2 by AMPK and PKB has opposite autophagy [49]. Several stimuli, including
effects on mTORC1 and connects mTORC1 starvation or ER stress, can induce HDM2-
with energy and growth factor signaling, dependent proteasomal degradation of
respectively (Figure 2). Recently, it is p53 to favor autophagy induction,
reported that AMPK regulates mTORC1 positioning p53 as a negative regulator of
signaling through an alternative autophagy. HDM2, the p53-specific E3
mechanism, whereby AMPK directly ubiquitin ligase, targets p53 to
phosphorylates Raptor, a subunit of proteasome-mediated destruction. The
mTORC1, and this Raptor phosphorylation inhibition of HDM2 blocks the depletion of
is important for the inhibition of mTORC1 p53 and also prevents the activation of
signaling by AMPK [41]. Thus, AMPK serves autophagy [49]. More importantly, it is the
as a positive regulator of autophagy. Under cytoplasmic p53 that exerts its inhibitory
stress conditions, the LKB1–AMPK function toward autophagy, in contrast to
TEÓRICO AUTOFAGIA 51

the transcriptionally active nuclear p53 dissociation of Bcl-2 from Beclin 1, and
that promotes autophagy. Upon inhibits autophagy; expression of a
reintroduction into p53/ cancer cells, constitutively active JNK1 results in
mutants of p53 that are restricted to the constitutive Bcl-2 multisite
cytosol effectively inhibit autophagy, phosphorylation, dissociation of Bcl-2 from
whereas mutants of p53 that accumulate Beclin 1 and stimulation of autophagy [53
within the nucleus fail to block autophagy ]. Third, a recent finding shows that the
[49]. The inhibitory role of cytosolic p53 in activation of Beclin 1 to induce autophagy
autophagy may contribute to the strong involves the phosphorylation of Beclin 1 by
oncogenic action of certain p53 mutants the death-associated protein kinase
that are preferentially localized to the (DAPK). DAPK physically interacts with
cytosol [50 ]. Beclin 1, and phosphorylates Beclin 1 on
Thr119 located at a crucial position within
Bcl-2 protein family
the BH3 domain of Beclin 1, and thus
In mammals, the Bcl-2 protein family plays promotes the dissociation of Beclin 1 from
a dual role in autophagy regulation. its inhibitor, Bcl-XL, and autophagy
Antiapoptotic proteins, such as Bcl2, Bcl- induction [54 ].
XL, Bcl-w, and Mcl-1, can inhibit
Concluding remarks
autophagy, whereas proapoptotic BH3-
only proteins, such as BNIP3, Bad, Bik, In the past decade there has been a
Noxa, Puma, and BimEL, can induce tremendous advance in our understanding
autophagy [51]. The binding of Bcl-2 to of the molecular machinery involved in
Beclin 1 disrupts the association of Beclin 1 mammalian autophagy. Nonetheless,
with hVps34, decreases Beclin 1- many outstanding questions remain to be
associated hVps34 PtdIns3K activity and answered, including the mystery of the
thereby inhibits autophagy. There are at membrane source for autophagosome
least three distinct mechanisms that may formation. By comparison, our knowledge
account for the release of Beclin 1 from its about the signaling regulation of
inhibitory interaction with Bcl-2/Bcl-XL autophagy is relatively limited, in
(Figure 2). One model depicts that the BH3 particular, with regard to the complex
domain of BH3-only proteins such as Bad, coordination between autophagy
may competitively disrupt the inhibitory machinery and signaling inputs. As an
interaction of Beclin 1 and Bcl-2/Bcl-XL [52 intracellular self-destructive system,
]. A second mechanism for the dissociation autophagy must be tightly regulated in
of Beclin 1 from its inhibitory interaction order to adapt to different intracellular and
with Bcl-2 involves the phosphorylation of extracellular stresses. This raises a
Bcl-2 by the stress-activated c-Jun N- fundamental question: How does the cell
terminal kinase 1 (JNK1). Starvation determine the specificity and magnitude of
induces the phosphorylation of Bcl-2 at autophagy based on the inputs from a
residues T69, S70, and S87 of the variety of signaling mechanisms?
nonstructured loop; expression of a Mammalian autophagy has gained
nonphosphorylatable Bcl-2 mutant (T69A, tremendous attention because of its
S70A, and S87A) or inhibition of JNK1 implications in a wide range of
abolishes the starvation-triggered physiological processes and diseases in
TEÓRICO AUTOFAGIA 52

humans. Our current understanding of this autophagy and its use as a therapeutic
process and continued examination of its intervention.
mechanism and regulation hold the
potential for practical modulation of

AUTOPHAGY AND THE INTEGRATED STRESS RESPONSE

Abstract signals such as contact-dependent signals,


hormones, cytokines and
Autophagy is a tightly regulated pathway neurotransmitters; and microbial
involving the lysosomal degradation of pathogens. Beyond a certain threshold,
cytoplasmic organelles or cytosolic such fluctuations are considered ‘stresses’,
components. This pathway can be meaning that the cell's response to this
stimulated by multiple forms of cellular stress determines whether it can function
stress, including nutrient or growth factor properly and survive.
deprivation, hypoxia, reactive oxygen
During the response to sublethal stress,
species, DNA damage, protein aggregates,
cells undergo rapid changes to adapt their
damaged organelles or intracellular
metabolism and protect themselves
pathogens. Both specific, stimulus-
against potential damage. This is
dependent and more general stimulus-
orchestrated through a multifaceted
independent signaling pathways are
cellular program, which involves the
activated to coordinate different phases of
concerted action of diverse stress response
autophagy. Autophagy can be integrated
pathways. One of the key pathways that
with other cellular stress responses
mediates stress-induced metabolic
through parallel stimulation of autophagy
adaptation and damage control is
and other stress responses by specific
macroautophagy, referred to simply as
stress stimuli, through dual regulation of
“autophagy”. During autophagy, cells form
autophagy and other stress responses by
double-membraned vesicles,
multi-functional stress signaling molecules,
autophagosomes, that sequester
and/or through mutual control of
organelles, proteins, or portions of the
autophagy and other stress responses.
cytoplasm, for delivery to the lysosome (He
Thus, autophagy is a cell biological process
and Klionsky, 2009). The sequestered
that is a central component of the
contents are degraded in the lysosome,
integrated stress response.
allowing cells to eliminate damaged or
Introduction harmful components through catabolism
and recycling to maintain nutrient and
Eukaryotic cells must adapt continuously energy homeostasis. Autophagy
to fluctuations in external conditions, constitutes a major protective mechanism
including physical parameters, such as that allows cells to survive in response to
temperature and ultraviolet light; chemical multiple stressors and that helps defend
cues such as ion concentrations, pH, organisms against degenerative,
oxygen tension, redox potentials and inflammatory, infectious, and neoplastic
metabolite concentrations; extracellular
TEÓRICO AUTOFAGIA 53

diseases (Levine and Kroemer, The Core Pathway of Mammalian


2008; Mizushima et al., 2008). Autophagy
Besides autophagy, the cellular response The core pathway of mammalian
to stress involves numerous other autophagy (Fig. 1) begins with the
pathways including those that regulate formation of an isolation membrane (also
nutrient uptake, intermediary metabolism, called a phagophore) and involves at least
cell cycle and growth control, cell fate and five molecular components, including: (1)
lineage decisions, and cellular the Atg1/unc-51-like kinase (ULK) complex;
survival/death programs. Therefore, it is (2) the Beclin 1/class III
not surprising that there is a close phosphatidylinositol 3-kinase (PI3K)
integration between signals that regulate complex; (3) two transmembrane proteins,
these cellular processes and those that Atg9 and vacuole membrane protein 1
regulate autophagy. In this review, we will (VMP1); (4) two ubiquitin-like protein
summarize recent advances in (Atg12 and Atg8/LC3) conjugation systems;
understanding how different cellular stress and (5) proteins that mediate fusion
signals and stress stimuli regulate between autophagosomes and lysosomes
autophagy. (Yang and Klionsky, 2010). Some of these
core autophagy pathway components are
directly controlled by cellular stress signals
(Fig. 1).
TEÓRICO AUTOFAGIA 54

FIG. 1 Overview of the major components of the core pathway of mammalian autophagy
Several key molecular components participate in the initiation, execution and completion of
autophagy. Autophagy inducers such as starvation modulate the inhibitory interaction of
TORC1 with the ULK1/2 complex. Through phosphorylation of Ambra1, and maybe through
other putative interactions, ULK1/2 complex (A) also regulates the activity of Beclin 1/ class III
phosphatidylinositol 3-kinase (PI3K) complex (B). Beclin 1 interacts with several enhancing
(blue) or inhibitory (grey) factors that modulate its binding to Vps34, the catalytic unit of the
PI3K, whose lipid kinase activity is essential for autophagy. In addition to these two complexes,
autophagosome formation requires the participation of two ubiquitin-like protein (Atg12 and
Atg8/LC3) conjugation systems and two transmembrane proteins (Atg9 and VMP-1) (C).
Whereas the roles of Atg9 and VMP-1 are currently not completely understood, both
conjugation systems are essential for the biogenesis of the isolation membrane, also called
‘phagophore’. In addition, the Atg8/LC3 system is required for autophagosome transport and
maturation, as well as for the selection of autophagic cargo. Fully mature autophagosomes
can fuse with Rab7-positive late endosomes to form amphisomes. Finally, autophagosomes or
amphisomes fuse their external membranes with those from acidic lysosomes to acquire
hydrolytic activity, degrade their cargo and recycle essential biomolecules to the
cytoplasm (D).

Origin of the isolation membrane The Atg1/ULK Complex


The isolation membrane (also called The mammalian orthologs of yeast Atg1,
‘phagophore’) can be generated from ULK1 and ULK2, play a key role in
multiple sources that include the ER (Axe autophagy induction, acting downstream
et al., 2008; Hayashi-Nishino et al., of the mammalian target of rapamycin
2009; Yla-Anttila et al., 2009), the outer (mTOR) complex 1 (mTORC1, a polyprotein
mitochondrial membrane (Hailey et al., complex that contains mTOR, Raptor,
2010), and the plasma membrane mLST8/GßL, Deptor and PRAS40 (Efeyan
(Ravikumar et al., 2010). PI3P is required and Sabatini, 2010)). In normal (nutrient-
for the formation of ‘omegasomes’, the Ω- replete) conditions, mTORC1 possesses
shaped structures that bud from the ER kinase activity and interacts with a
during the initial steps of vesicle complex that contains ULK1, Atg13, FIP200
nucleation/autophagosome formation and Atg101 (Fig. 1A). Upon mTORC1
(Axe et al., 2008). ATG16L1 directly inhibition, for example by starvation (see
interacts with clathrin, which connects the below), mTORC1 dissociates from the ULK
endocytic pathway to autophagy complex, leading to dephosphorylation of
(Ravikumar et al., 2010). While it appears specific residues within ULK1 (or ULK2) and
that general autophagy inducers such as Atg13 (which are normally phosphorylated
starvation stimulate lipid recruitment from by mTORC1), the catalytic activation of
all known sources, it is not known whether ULK1 (or ULK2) and the ULK1 (or ULK2)-
specific stimuli (such as ER stress, mediated phosphorylation of other
mitochondrial damage or signals residues in Atg13 and FIP200 (Fig. 1A).
emanating from the plasma membrane) Thus, mTORC1 inhibition is probably
cause autophagosomes to be coupled to ULK1 (or ULK2) activation
preferentially formed from specific through a process that involves
membrane sources. dissociation of a large protein complex and
(de)phosphorylation events (Mizushima,
TEÓRICO AUTOFAGIA 55

2010). An intriguing, but not yet tested, Deptor, PRAS40 and ULK1/2, which are
possibility, is that the ULK1/ULK2 complex each inhibited by mTORC1-mediated
might also be positively regulated in low phosphorylation and, in turn, inhibit mTOR
energy conditions by its recently described activity. This may lead to amplification of
interactions with several subunits of the initially modest changes in TOR activity.
energy-sensing kinase, AMP-activated Negative feedback control is achieved via
protein kinase (AMPK) (Behrends et al., the products of autophagy including amino
2010). It is not entirely known how ULK1/2 acids as well as via inhibition of S6 kinase,
activates downstream components of the a protein that has mTORCI-dependent
autophagic machinery. However, ULK1 can activity and which may be necessary for
phosphorylate Ambra1 (Di Bartolomeo et sustained autophagy (Neufeld, 2010).
al., 2010), a component of the Beclin Thus, mTORC1 is part of a rheostat that is
1/Class III PI3K complex (He and Levine, either switched off (to inhibit autophagy at
2010) (Fig. 1A). the level of ULK1/2) or on (to induce
autophagy by ULK1/2 activation, as a result
There are multiple circuits of positive and
of a positive amplification loop). When on,
negative feedback in mTORC1-mediated
these effects are self-limited due to the
autophagy regulation (Neufeld, 2010) (Fig.
presence of negative feedback loops (Fig.
2A). Feed-forward mechanisms involve
2A).

FIGURE 2
Overview of selected signal transduction pathways that regulate autophagy components
that function in vesicle nucleation/phagophore formation
Selected signals that converge on ULK1/2 (A) and the Beclin 1 complex (B) are depicted. Note
the multiple positive and negative feedback loops depicted in A. For details see text.
phosphate (PI3P) which recruits effectors
Beclin 1 and its Interactome
such as the double FYVE domain-
The “Beclin 1 core complex” involves Beclin containing protein 1 (DFCP1) (Axe et al.,
1, Vps15, Vps34 and likely, Ambra1 (He and 2008) and WD-repeat protein interacting
Levine, 2010). This multiprotein complex with phosphoinositides (WIPI) family
must be formed for the allosteric proteins (Polson et al., 2010) to mediate
activation of the class III PI3K Vps34 to the initial stages of vesicle
generate phosphatidylinositol-3- nucleation/autophagosome formation.
TEÓRICO AUTOFAGIA 56

Numerous proteins that interact with in starvation conditions) (Wei et al., 2008)
Beclin 1 induce or inhibit autophagy (Fig. are required for autophagy induction in
1B). Atg14 (also called Atg14L or Barkor, response to specific forms of stress.
for Beclin 1-associated autophagy-related
The inositol-1,4,5 trisphosphate (IP3)
key regulator) is essential for PI3K activity
receptor, which is an IP3-activated calcium
and autophagy induction. UVRAG (UV
channel at the endoplasmic reticulum (ER),
radiation resistance-associated gene)
interacts with Beclin 1 indirectly, via Bcl-2
competes with Atg14 for binding to Beclin
(Fig. 2B). Upon cellular reduction of
1 and may promote PI3K activity in a cell
IP3 levels (or antagonist binding) and
type-specific fashion, and through
starvation, this interaction is disrupted
interactions with class C Vps/HOPS
(Vicencio et al., 2009). This mechanism
complexes, promotes autophagosome
may explain how agents that cause a
fusion with the late endosome/lysosome.
reduction of IP3 levels cause autophagy in
Bif-1/endophilin B1 interacts with Beclin 1
an mTOR-independent fashion (Sarkar et
via UVRAG to function as a positive
al., 2007). The toll-like receptor (TLR)
regulator of the PI3K complex, and has an
adaptor molecules MyD88 and TRIF also
N-BAR domain that may promote
may interact with Beclin 1, thereby
membrane curvature (Fig. 1B). Rubicon
reducing the binding of Beclin 1 to Bcl-2
(RUN domain protein as Beclin 1
and promoting autophagy (Shi and Kehrl,
interacting and cysteine-rich containing)
2008). During TLR4-induced autophagy,
negatively regulates autophagy (as well as
tumor necrosis factor receptor (TNFR)-
endocytic trafficking) through its
associated factor 6, TRAF6, interacts with
interaction with Beclin 1/PI3K complexes
Beclin 1 and mediates K63-linked
(Fig. 1B).
ubiquitination of Beclin 1, which enhances
Anti-apoptotic family members (such as Class III PI3K activity (Shi and Kehrl, 2010).
Bcl-2, Bcl-XL and Mcl-1) are also important The protein PINK1, which has been studied
negative regulators of autophagy through as a serine-threonine kinase that interacts
an inhibitory interaction of their BH3- with Parkin to stimulate mitophagy, also
binding groove with the BH3 domain of interacts with Beclin 1 (Michiorri et al.,
Beclin 1 (Maiuri et al., 2007; Pattingre et 2010). Although it is not clear how all these
al., 2005) (Fig. 2B). There are several proteins affect the overall composition and
distinct mechanisms through which function of the multiprotein Beclin 1-
autophagy-inducing signals can disrupt this containing complex, an attractive model is
inhibitory interaction, including that multiple proteins directly or indirectly
competition by pro-apoptotic BH3-only interact with Beclin 1 to relay extracellular
proteins (such as BNIP3, Bad, Bik, Noxa, and intracellular stress signals to the
Puma and BimEL) (Maiuri et al., 2007), autophagic machinery.
phosphorylation of the BH3 domain of
Beclin 1 by death-associated protein kinase Atg9 and VMP1
(DAPK) (Zalckvar et al., 2009) or
phosphorylation of Bcl-2 by c-Jun N- Atg9 may provide lipids to the phagophore
teminal kinase-1 (JNK1) (Pattingre et al., membrane by cycling between distinct
2009; Wei et al., 2008). Accordingly, BH3- subcellular compartments (Fig. 1C). The
only proteins (such as BAD in starvation or cycling of Atg9 requires Atg1/ULK1 and the
BNIP3 in hypoxia) (Bellot et al., kinase activity of Vps34. Another
2009; Maiuri et al., 2007) as well as Beclin possibility is that Atg9 cycling involves the
1/Bcl-2 dissociating kinases (such as JNK1 UVRAG/Bif-1-containing Beclin 1 complex
TEÓRICO AUTOFAGIA 57

since Bif-1 transiently associates with Atg9 associated with the autophagosome
after starvation (Orsi et al., 2010). VMP1, membrane, and its biochemical and
which interacts with Beclin 1, may function microscopic detection is widely used to
as a transmembrane protein that recruits measure cellular autophagy (Mizushima et
Beclin 1 (and other components of the al., 2010) (Fig. 1C). In mammals, four Atg4
Beclin 1 complex) to the phagophore (Fig. (Atg4A-B) and at least six orthologues of
1C). It also interacts with TP53INP2 (tumor Atg8 exist, among which LC3B (hereafter
protein 53-induced nuclear protein 2), referred to simply as LC3), GABARAP and
which like VMP1 is essential for autophagy GATE16 have been most studied
and the translocation of Beclin 1 and LC3 to (Weidberg et al., 2010).
autophagosomes (Yang and Klionsky,
Several stress signals regulate autophagy
2010).
at the level of the Atg8/LC3 conjugation
system. For example, the death-associated
Conjugation Systems
protein kinase, DAPK, positively regulates
Two ubiquitin-like conjugation systems are autophagy by associating with the LC3-
part of the vesicle elongation process (Fig. interacting cytoskeleton molecule MAP1B
1C). The first pathway involves the (Harrison et al., 2008) (in addition to
covalent conjugation of Atg12 to Atg5, phosphorylating Beclin 1, discussed above
with the help of the E1-like enzyme Atg7 (Zalckvar et al. 2009)). The cellular FLICE-
and the E2-like enzyme Atg10. This like inhibitor protein, c-FLIP, negatively
conjugate is organized into a complex by regulates autophagy by preventing Atg3
associating with Atg16 in a non-covalent from binding and activating LC3 (Lee et al.,
fashion to form the multimeric Atg12- 2009). Moreover, reactive oxygen species
Atg5-Atg16 complex, which functions as may regulate the active cysteine site of
the E3 ligase of LC3 (Yang and Klionsky, Atg4 (Fig. 1C) (Scherz-Shouval R et al.
2010). The abundance of Atg5 may be 2007). The enrichment of proteins with
regulated by calcium-dependent activation lipid kinase, WD40, and GTPase regulatory
of the cysteine protease, calpain, which domains in the mammalian Atg8-
cleaves and inactivates Atg5, at least in interaction network (Behrends et al., 2010)
cultured cells. Thus, the reduction of may provide additional clues as to how
cytosolic Ca2+ (or calpain inactivation) may stress signals interface with autophagy
prevent Atg5 cleavage (Fig. 1C), resulting in control at the level of Atg8/LC3 regulation.
increased cellular levels of full-length Atg5
Several proteins that possess an LC3-
and the pro-autophagic Atg12-Atg5
interacting region (LIR) and interact with
conjugate (Xia et al., 2010).
LC3 (and its paralogs) serve as adaptors to
The second pathway involves the target defined structures such as
conjugation of phosphatidylethanolamine ubiquitinated proteins or mitochondria to
(PE) to a glycine (Gly) residue of yeast the autophagic machinery (Fig. 1D). The
Atg8/mammalian LC3 by the sequential best-characterized examples are p62 (also
action of the protease Atg4, the E1-like known as sequestosome1, SQSTM1) and
enzyme Atg7, and the E2-like enzyme Atg3 NBR1 (Neighbor of BRCA1), which both
(Fig. 1B). This lipid conjugation leads to the recognize ubiquitinated proteins (Kirkin et
conversion of the soluble form of LC3 al., 2009) as well as BNIP3L (also known as
(named LC3-I) to the autophagic vesicle- NIX), which binds to mitochondrial
associated form, LC3-II (Yang and Klionsky, membranes (Novak et al., 2010). The
2010). The lipidated form of LC3 is stably regulation of these autophagy adaptor
TEÓRICO AUTOFAGIA 58

proteins is not yet well understood, but will Autophagy Induced by Nutrient Stress
likely be key to understanding how specific
Nutrient depletion is the most potent
stress stimuli trigger selective autophagy.
known physiological inducer of autophagy.
Go to: In the majority of cultured mammalian
cells, nutrient depletion induces
Autophagy, Stress Stimuli, and Stress autophagy within minutes, with maximal
Signals levels observed when cells are cultured in
the simultaneous absence of nutrients
Autophagy is induced by a variety of stress (such as amino acids and glucose) and
stimuli, including nutrient and energy growth factors (such as those contained in
stress, ER stress, pathogen-associated serum) (Boya et al., 2005). In mice,
molecular patterns (PAMPs) and danger- following starvation for 24-48 hours, most
associated molecular patterns (DAMPs), cells in most tissues display increased
hypoxia, redox stress, and mitochondrial numbers of autophagosomes (Mizushima,
damage. The stimulation of autophagy by 2010). Several critical molecules regulate
these stimuli involves diverse signals that starvation-induced autophagy (Fig. 3A); of
have overlapping functions in autophagy these, mTOR and AMPK have been best
and the control of other cellular stress characterized, and recent studies also
responses. suggest a crucial role for sirtuins.

FIGURE 3
Overview of the major signal transduction pathways that regulate autophagy in response
to starvation
TEÓRICO AUTOFAGIA 59

A summary of starvation-induced pro-autophagic signaling (A) is followed by a schematic


overview of the signaling cascades involving sirtuin-1 and Foxo 3a (B), AMPK (C) and mTORC1
(D).

Sirtuins and protein (de)acetylation ULK2, Beclin 1, VPS34, BNIP3 and BNIP3L,
ATG12, ATG4B, LC3,
Sirtuins are a family of NAD-dependent and GABARAPL1 (Mammucari et al., 2007).
deacetylases that sense environmental Another sirtuin, Sirt2, dissociates from
stress (Haigis and Sinclair, 2010). The FOXO1 upon serum starvation, thus
induction of autophagy by starvation (but causing the acetylation of FOXO1, favoring
not by mTORC1 inhibition or ER stress) its interaction with Atg7 in the cytoplasm
requires Sirt1 (Lee et al., 2008; Morselli et and stimulating autophagy (Zhao et al.,
al., 2010). Accordingly, Sirt1−/− mice display 2010). Thus, protein (de)acetylation
a phenotype consistent with impaired reactions influenced by sirtuins and other
autophagy, including increased levels of enzymes may control autophagy at
the autophagy substrate p62, multiple levels.
accumulation of damaged organelles,
disruption of energy homeostasis, and AMPK in starvation, energy depletion,
early perinatal lethality (Lee et al., 2008). and beyond
The transfection of cells with sirtuin 1
(Sirt1) with intact deacetylase activity is AMPK acts as a central node that
sufficient to stimulate autophagy in integrates several stress stimuli with
cultured mammalian cells (Lee et al., autophagy initiation (Fig. 3C). AMPK
2008). In this context, it is intriguing that monitors the energy status of the cell by
p300 acetyltransferase knockdown (Lee sensing its AMP:ATP ratio. Several
and Finkel, 2009), as well as histone upstream kinases, including liver kinase B1
acetylase inhibition by spermidine, a (LKB1, which is activated by energy
natural polyamine, potently induce depletion), calcium/calmodulin kinase
autophagy (Eisenberg et al., 2009), kinase-ß (CaMKKß, which is activated by
suggesting that protein acetylation may cytosolic Ca2+), and TGFß-activated kinase-
play a general role in autophagy 1 (TAK-1, which is also involved in IKK
regulation. activation), can activate AMPK by
phosphorylating a threonine residue on its
Sirt1 can deacetylate Atg5, Atg7 and LC3 catalytic α-subunit (Ruderman et al.,
(Lee et al., 2008) (Fig. 3A), while p300 can 2010). In addition, Sirt1 and AMPK engage
acetylate Atg5, Atg7, LC3 and Atg12 (Lee in a coordinated positive amplification
and Finkel, 2009). Furthermore, Sirt1 loop (the “Sirt1/AMPK cycle”) (Ruderman
deacetylates the transcription factor et al., 2010), that acts to initiate autophagy
forkhead box O3a, FOXO3a, yet another in nutrient deprivation conditions. Sirt1-
hub of autophagy regulation, leading to mediated deacetylation of LKB1 increases
enhanced expression of pro-autophagic its serine-threonine kinase activity and
Bnip3 (Kume et al., 2010). Akt inhibition stimulates its translocation from the
resulting from growth factor depletion also nucleus to the cytoplasm where it activates
causes FOXO3 activation, although via a AMPK. AMPK can also indirectly stimulate
different mechanism. Following its Sirt1 activation by the reduction of
dephosphorylation, FOXO3 translocates nicotinamide (NAM) and an increase in
into the nucleus (Fig. 3A) and upregulates NAD+ that may be catalyzed by NAM
multiple autophagy-related genes such as phosphoribosyltransferase upregulation,
TEÓRICO AUTOFAGIA 60

thus completing the feed forward circuitry Additional kinases involved in starvation-
(Lan et al., 2008; Ruderman et al., 2010).
induced autophagy
The best-studied mechanism by which
AMPK induces autophagy is through Several kinases besides AMPK function in
mTORC1 inhibition (see below), via starvation-induced upregulation of
phosphorylation of the tuberous sclerosis autophagy. This includes JNK1, which both
complex 2 (TSC2) and the regulatory phosphorylates Bcl-2, reducing its affinity
associated protein of mTOR, Raptor (Fig. for the BH3 domain of Beclin 1 (Wei et al.,
3D). The recent identification of an 2008), and phosphorylates Sirt1,
interaction between AMPK and ULK1 promoting its enzymatic activity (Nasrin et
(Behrends et al., 2010) raises the al., 2009) (Fig. 3A). Moreover, as noted
speculative possibility that AMPK may also below, the phosphorylation of eIF2α
act more directly on core components of (Kouroku et al., 2007; Talloczy et al., 2002)
the autophagy machinery to initiate and the activation of IKK (Criollo et al.,
autophagy. 2010) are essential for starvation-induced
autophagy. Other kinases such as p38α
mTORC1 inhibition in starvation-induced mitogen-activated protein kinase (p38α
MAPK, also known as MAPK14) potently
autophagy
inhibit starvation-induced autophagy.
Besides inhibition by AMPK, mTORC1 is p38α MAPK acts on p38IP, which is
also inhibited upon withdrawal of growth required for starvation-induced mAtg9
factors such as insulin or insulin-like trafficking and autophagosome formation
growth factor (Fig. 3C,D). In response to (Webber and Tooze, 2010). Thus, the
growth factors, Akt becomes catalytically coordinated activation (and inhibition) of
active (due to the sequential stimulation of several kinases is essential for the
growth factor-activated class I PI3K and induction of autophagy by nutrient
PI3K-dependent protein kinase 1, PDK1, depletion. The precise details of such
which phosphorylates Akt). Moreover, coordination, however, remain poorly
growth factors activate Ras, which understood.
stimulates a cascade involving Raf-1,
MEK1/2 and ERK1/2. Both Akt and ERK1/2 ER Stress and Autophagy
can phosphorylate one of two subunits of
The ER is not only involved in protein
the tuberous sclerosis complex 1/2
synthesis and maturation (including
(TSC1/TSC2), and Akt can phosphorylate
correct folding), but may also constitute a
Raptor, thus causing the activation of
major source/scaffold of the autophagic
mTOR (Fig. 3D) (Neufeld, 2010). Moreover,
isolation membrane (Hayashi-Nishino et
amino acids activate mTORC1
al., 2009; Yla-Anttila et al., 2009). The
independently of the Akt-TSC1/TSC2 axis,
unfolded protein response (UPR), the
through the Rag family of GTPases, which
major ER stress pathway (Buchberger et
directly interact with Raptor and recruit
al., 2010), is a potent stimulus of
mTORC1 to the lysosomal surface (Efeyan
autophagy. The three canonical branches
and Sabatini, 2010) (Fig. 3D). Thus, mTOR is
of the UPR are mediated by three ER
inhibited through multiple mechanisms
membrane-associated proteins, PERK
during starvation conditions (Sengupta et
(PKR-like eIF2α kinase, also known as
al., 2010).
EIF2AK3), ATF6 (activating transcription
factor-6), and IRE1 (inositol requiring
TEÓRICO AUTOFAGIA 61

enzyme 1) (Fig. 4A), all of which are PERK, IRE1 and ATF6 from their inhibition.
normally bound to and inactivated by the Among these, PERK and ATF6 acts as
chaperone BiP/GRP78. The occupancy of autophagy inducers, while IRE1 acts a
BiP/GRP78 by misfolded proteins releases negative regulator of autophagy.

FIGURE 4
Summary of the major signal transduction pathways that connect autophagy to ER stress
(A), eIF2α phosphorylation (B) and IKK activation (C)

For details see text.

PERK and eIF2α phosphorylation eIF2α phosphorylation initiates both a


general inhibition of protein translation, as
PERK mediates the transcriptional well as the selective translation of some
activation of the proteins LC3 and Atg5 in stress-responsive transcripts including that
hypoxic responses through the action of of the ATF4 transcription factor and certain
the transcription factors ATF4 and CHOP, autophagy genes (Hotamisligil, 2010).
respectively (Rouschop et al., 2010). PERK
may also reduce the translation of IkBα Cells that carry a non-phosphorylatable
(Deng et al., 2004), thereby activating NF- mutant of eIF2α (S51A) fail to induce
κB, which also could contribute to autophagy in response to starvation,
autophagy (Fig. 4B). PERK phosphorylates suggesting that eIF2α phosphorylation on
the eukaryotic initiation factor 2α (eIF2α) serine 51 (and by extension eIF2α kinases)
on residue serine 51, which then initiates a plays a major role in autophagy regulation
cascade of events that decreases the (Kouroku et al., 2007; Talloczy et al., 2002).
overload of misfolded proteins, thereby This phosphorylation event integrates
alleviating ER stress (Harding et al., 2003). various types of environmental and
TEÓRICO AUTOFAGIA 62

endogenous stress signals beyond ER IRE1


stress, such as amino acid deprivation,
exposure to double-stranded viral RNA, IRE1 is a serine/threonine kinase (which
osmotic stress, UV light exposure, heme activates JNK1) and also an
deficiency, hypoxia and oxidative stress endoribonuclease, which catalyzes the
(Harding et al., 2003). These divergent splicing of the mRNA encoding form of the
signals activate four different eIF2α kinases transcription factor XBP1, which then
including PERK (which is activated by ER transactivates UPR genes involved in ERAD,
stress, radiation or hypoxia), general protein folding and protein quality control
control non-derepressible-2 (GCN2, which (Hetz and Glimcher, 2009; Hotamisligil,
is activated by uncharged tRNAs in amino 2010). Perhaps unexpectedly, the
acid-starved cells), heme-regulated inhibition of IRE1 and its downstream
inhibitor (HRI, which is activated by heme effector XBP1 enhances autophagy
deficiency in erythroid precursor cells), and induction. Mice lacking XBP1 in neurons
PKR (which is activated by double-stranded exhibit increased levels of baseline
RNA and in some contexts, ER stress autophagy, which leads to increased
(Nakamura et al., 2010)) (Fig. 4B). Yeast turnover of an autophagic substrate,
GCN2 and mammalian PKR and PERK have mutant superoxide dismutase-1 (SOD1),
been shown to be required for autophagy and protection against mutant SOD1-
induced by starvation, viral infection, and induced amyotrophic lateral sclerosis (Hetz
ER stress, respectively (He and Klionsky, et al., 2009). Since the predominant
2009). These findings illustrate the outcome of ER stress is the induction
importance of eIF2α kinases in autophagy (rather than inhibition) of autophagy, it is
control, not only in the UPR but also in possible that IRE1/XBP1 dependent signals
other conditions of stress. have the function to dampen excessive
autophagy triggered via the PERK/eIF2α
How eIF2α phosphorylation contributes to pathway (Fig. 4A).
autophagy initiation is not known. One
highly speculative possibility is that eIF2α Immune Signals
phosphorylation might somehow affect
the ER in a manner that promotes the Infection (or exposure of cells to microbial
physical formation of the isolation products) constitutes a specialized form of
membrane. Another possibility is that cellular stress that, in many cases, results
eIF2α phosphorylation stimulates in autophagy induction (Sumpter and
autophagy via its effects on the Levine, 2010). Autophagy induction during
transactivation of autophagy genes. eIF2α infection is regulated by cytokines such as
phosphorylation stimulates the selective IFNγ (and downstream immunity-related
translation of the ATF4 transcription factor GTPases) as well as pathogen recognition
(although general translation is shut off), receptors (PRRs) that recognize conserved
which stimulates LC3 expression (which is components of pathogens or products of
necessary for sustained autophagy) (Milani their replication (PAMPs). The PRRs include
et al., 2009) (Fig. 4B). Moreover, there are families of Toll-like receptors (TLRs) that
direct interactions between eIF2α subunits recognize PAMPS to activate pro-
and core autophagy proteins, although it is inflammatory cytokines and type I
not yet known whether these interactions interferon production via NF-κB-, MAPK-
are biologically significant (Behrends et al., and interferon-regulatory pathways; NOD-
2010). like receptors (NLRs) that primarily activate
signaling via NF-κB and MAPK and
TEÓRICO AUTOFAGIA 63

components of the inflammasome; RIG-I However, the NF-κB family member


like receptors (RLRs) that recognize p65/RelA can upregulate beclin
cytoplasmic viral RNA and dA:dT rich 1 expression and autophagy during T cell
dsDNA; C-type lectins, and the double- activation (Copetti et al., 2009) and NF-κB-
stranded RNA binding protein kinase PKR incompetent cells are deficient in heat
(Sumpter and Levine, 2010). Since these shock-induced autophagy (Nivon et al.,
PRRs recognize not only PAMPS, but also 2009). Thus, there may be both IKK-
DAMPs (which include products of necrotic dependent, NF-κB-independent as well as
cells, hypoxia, abnormal intracellular ion NF-κB-dependent mechanisms for
gradients, reactive oxygen species, and autophagy induction. In addition, the p105
accumulation of misfolded proteins), this subunit of NF-κB can interact with certain
family of “immune signals” that regulates autophagy proteins, including Beclin 1
autophagy may represent a more (Behrends et al., 2010), but the significance
generalized system that cells use to elicit of these interactions has yet to be
autophagy in response to different forms explored.
of stress.
Aside from the regulation of autophagy by
TAK1, IKK and NF-κB NF-κB, autophagy may, in turn, regulate
NF-κB signaling. For example, NF-κB
The transcription factor, NF-κB, and some signaling may be negatively regulated by
of its upstream regulators, function to targeting IKK and the NF-κB inducing
integrate diverse stress signals including kinase, NIK, for autophagic degradation
immune signals with the autophagy (Qing et al., 2007). Furthermore, when
pathway. NF-κB is activated when the autophagy is inhibited, p62 accumulates
inhibitor of NF-κB (IκB) is phosphorylated and activates the NF-κB signaling pathway
by the IκB kinase (IKK), a kinase that is (Mathew et al., 2009), suggesting that
composed of one regulatory subunit (IKKγ, normal levels of autophagy may provide a
also known as NEMO) and two catalytic “break” on NF-κB signaling.
subunits (IKKα, IKKβ). The IKK complex is
activated in response to multiple different PKR and the “meta-inflammasome”
stressors such as reactive oxygen species,
An interesting recent study uncovers a
DNA damage, and ligation of death
potential relationship between an
receptors and PRRs. Frequently, this IKK
autophagy-regulatory eIF2α kinase, PKR,
activation is mediated by another
previously believed to respond primarily to
upstream kinase, TAK1 (Fig. 4C). (Baud and
virus infection (Talloczy et al.,
Karin, 2009). In murine and human cells,
2002; Talloczy et al., 2006), and the
knockout and/or knockdown of Tak1 or
coordinate regulation of JNK1 and insulin
any of the Ikk subunits (but not that of
signaling with ER stress and fatty acid
the Nf-κb subunit p65) prevents the
exposure. PKR was found to respond to ER
induction of autophagy in response to
stress and fatty acid exposure, and be
diverse stimuli including starvation,
required for the JNK1 phosphorylation,
rapamycin, p53 inhibition, or ER stress
serine phosphorylation of the insulin
(Criollo et al., 2010; Herrero-Martin et al.,
receptor substrate 1 (IRS1) and
2009). Conversely, expression of
consequent insulin resistance that occurs
constitutively active IKK subunits stimulate
in response to these stimuli (Nakamura et
autophagy through an NF-κB-independent
al., 2010). Based on these findings and a
pathway that relies on the activation of
previous report that PKR activates the
AMPK and JNK1 (Criollo et al., 2010).
TEÓRICO AUTOFAGIA 64

IKKβ-NF-κB pathway (Bonnet et al., 2000), to limit ER stress; this hypothesis would
Hotamisligil et al. (Hotamisligil, 2010) reconcile the speculated role of the meta-
proposed the existence of a ‘meta- inflammasome in autophagy activation
inflammasome’ composed of PKR, eIF2α, with the cytoprotective roles of autophagy.
JNK, IRS, IKK and other components that
link ER stress to more global stress Hypoxia and Anoxia
responses, including inflammatory
signaling and metabolic dysfunction. In the Hypoxia and anoxia (with oxygen
postulated “meta-inflammasome”, PKR concentrations <3% and <0.1%,
phosphorylates eIF2α, (which induces respectively) both cause autophagy
autophagy), activates the ‘inflammatory through a variety of different mechanisms.
kinases’ IKKß and JNK1 (which both induce Hypoxia-induced autophagy depends on
autophagy), and phosphorylates (and hypoxia-inducible factor, HIF, while anoxia-
inhibits) IRS1 (which also would induce induced autophagy is HIF-independent
autophagy) (Nakamura et al., 2010). Thus, (Majmundar et al., 2010; Mazure and
this hypothetical molecular platform might Pouyssegur, 2010). HIF is a heterodimer of
explain the functional overlap and a constitutive ß subunit and an oxygen-
crosstalk between the JNK1, IKK and eIF2α regulated α subunit that only becomes
pathways in the induction of autophagy. stabilized (and hence expressed) when
oxygen concentration declines below a
It is, however, unclear whether this threshold of ~5%. Upon moderate hypoxia
autophagy-promoting activity, rather than (1-3% oxygen), HIF activates the
other downstream effects of the signaling transcription of BNIP3 and BNIP3L(NIX),
molecules, would explain the pro- two BH3-only proteins that can disrupt the
inflammatory and adverse metabolic inhibitory interaction between Beclin 1 and
effects of the “metainflammasome”, since Bcl-2 (Bellot et al., 2009) (Fig. 5A).
genetic deletions of autophagy genes Moreover, BNIP3L, which often is present
generally activates inflammatory signaling at the outer surface of mitochondria,
(Sumpter and Levine, 2010; Virgin and possesses a WXXL motif that binds to LC3
Levine, 2009) and a recent study found and its homolog GABARAP (Novak et al.,
that hepatic suppression of 2010), thereby targeting mitochondria for
the Atg7 autophagy gene results in autophagic destruction. The transcription
increased ER stress and insulin resistance of BNIP3 is also upregulated by the
(Yang et al., 2010). Perhaps, the activation transcription factor FOXO3, provided that
of autophagy by the metainflammasome it is deacetylated by Sirt1 (Kume et al.,
serves as a negative feedback mechanism 2010).
TEÓRICO AUTOFAGIA 65

FIGURE 5
Overview of selected stress pathways that induce autophagy

The mechanisms involved in autophagy induction by hypoxia or anoxia (A); increased


oxidative damage (reactive oxygen species, ROS) (B); perturbation of the p53 system (C) or
mitochondrial dysfunction D) are represented schematically.
the transcription factors ATF4 and CHOP,
Additional pathways have been implicated respectively, which are both regulated by
in hypoxia-induced autophagy, especially PERK (Rouschop et al., 2010).
in cases of severe hypoxia or anoxia (Fig.
5A). These include the protein DJ-1 (also Redox Stress and p53
called CAP1/RS/PARK7, which regulates
autophagy by an unknown mechanism), Oxidative stress induces autophagy
the autocrine stimulation of a PDGFR- through multiple mechanisms (Fig. 5B).
dependent pathway, the stimulation of Exogenous hydrogen peroxide reportedly
AMPK through metabolic stress, and the can activate PERK (and thereby stimulate
UPR of the ER (Mazure and Pouyssegur, eIF2α phosphorylation) (Liu et al., 2008),
2010). Maximal induction of autophagy by directly oxidize and activate Atg4
hypoxia also requires PERK-mediated proteases (and thereby accelerate the
phosphorylation of eIF2α (Rouschop et al., production of proteolytically mature LC3)
2010), further underscoring the (Scherz-Shouval et al., 2007), and inhibit
importance of eIF2α phosphorylation as a mTOR either directly (Liu et al., 2008) or
general autophagy regulator. Hypoxia indirectly, via activation of PARP1 (as a
increases the transcription of the essential result of DNA damage) and/or via
autophagy genes LC3 and Atg5 through activation of a cytoplasmic pool of ATM
TEÓRICO AUTOFAGIA 66

and the subsequent activation of LKB1 and proliferation and cell death (Gan and Guan,
AMPK1(Alexander et al., 2010; Huang et 2008). Thus, an interesting question is
al., 2009). The cellular response to an whether p53 somehow negatively
increase in ROS often involves the regulates the ULK1 complex through its
activation of mitogen-activated protein interaction with FIP200.
kinases (MAPKs), including JNK1, which can
activate autophagy (Wong et al., 2010). Mitochondrial Damage
Finally, DNA damage also stimulates the
expression of pro-autophagic p53-induced Cells must remove damaged mitochondria
target genes. to prevent the accumulation of ROS. This
process of mitochondrial quality control is
The tumor suppressor protein p53, which mediated by mitophagy, the selective
can be activated by different types of stress autophagic removal of mitochondria.
including redox stresss, has a dual effect on Considerable advances have been made in
autophagy (Fig. 5C), acting as a positive understanding the mechanisms by which
regulator of autophagy via its damaged mitochondria are targeted for
transcriptional activity and as a negative autophagy, as well as the functional
regulator of autophagy via its cytoplasmic significance of mitochondrial quality
functions (Green and Kroemer, 2009). As a control in preventing aging,
transcription factor, p53 transactivates neurodegenerative diseases, and other
several autophagy inducers including pathologies.
DRAM1 (which may operate through JNK1
activation) and Sestrin2 (which binds to the In response to potentially lethal stress or
ternary complex TSC1/TSC2-AMPK, damage, mitochondrial membranes can be
inducing phosphorylation and activation of permeabilized through multiple distinct
TSC2 by AMPK). One of the few p53- biochemical routes. Indeed, mitochondrial
induced genes that inhibits autophagy is membrane permeabilization (MMP)
TIGAR, a fructose-2,6-bisphosphatase that constitutes one of the hallmarks of
stimulates the pentose phosphate shunt imminent apoptotic or necrotic cell death
and helps to lower intracellular ROS (Kroemer et al., 2007). However, if only a
(Bensaad et al., 2009). When present in the fraction of mitochondria are
cytoplasm, p53 acts as a tonic inhibitor of permeabilized, autophagic removal of the
autophagy. In several contexts of damaged mitochondria can rescue the cell.
autophagy induction, p53 must be The autophagic recognition of depolarized
depleted from the cytoplasm for optimal mitochondria is mediated by a refined
autophagy induction to ensue. This may voltage sensor, involving the mitochondrial
involve the activation of the p53-specific kinase, PINK1. Under normal
ubiquitin ligase, HDM2, which targets p53 circumstances, PINK1 is continuously
to proteasome-mediated destruction recruited to the mitochondrial outer
(Green and Kroemer, 2009). However, it is membrane, but subject to voltage-
not yet fully clear how cytoplasmic p53 dependent proteolysis, which leads to its
inhibits autophagy. The only essential removal from mitochondria and to
autophagy protein that is known to proteasome-mediated degradation
interact with p53 is FIP200, which is a (Narendra et al., 2010). Upon
multifunctional protein that is present in mitochondrial depolarization, PINK1
the ULK1 complex, yet also binds to Pyk2, rapidly accumulates on the mitochondrial
FAK, TSC2, ASK1 and TRAF2 and plays a role surface, facilitates the recruitment of the
in the control of cell adhesion, migration, E3 ubiquitin ligase Parkin (Narendra et al.,
TEÓRICO AUTOFAGIA 67

2010) which ubiquitinates mitochondrial be a key signal underlying distinct steps in


substrates including the outer membrane selective autophagy.
protein VDAC1, recruits the autophagy
adaptor molecule, p62/SQSTM1, and Autophagy Regulatory Pathways in the
thereby targets mitochondria for General Stress Response
autophagic removal (Geisler et al., 2010)
In the preceding section, we reviewed
(Fig. 5D). Both PINK1 and Parkin genes
specific stress stimuli and stress signals
were originally identified because loss-of-
that regulate autophagy. Certain general
function mutations affecting either of
concepts regarding the interrelationship
them cause familial Parkinson's disease in
between autophagy regulatory pathways
humans. Disease-causing mutations
and the general stress response are
in PINK1 and Parkin disrupt Parkin
beginning to emerge.
recruitment and Parkin-induced
mitophagy (Geisler et al., 2010; Narendra
et al., 2010). At least in some cell types Integration of Autophagy and other
(MEFs), depolarization-induced mitophagy Cellular Stress Responses
involves BNIP3L (also known as NIX) The integration of autophagy and other
(Novak et al., 2010) (Fig. 5D) and in mice, cellular stress responses can be
BNIP3L/Nix is required for the removal of conceptualized in the framework of three
mitochondria during erythroid maturation broad concepts. First, a single type of stress
(Sandoval et al., 2008). At present, the stimulus elicits a variety of signals that
functional relationship between NIX- trigger distinct cellular responses (one of
dependent mitophagy and the which is autophagy) that cooperate for the
PINK1/Parkin mitophagy pathway has not sake of optimal cellular repair and
been elucidated. adaptation. Second, distinct stress
Interestingly, the maturation of responses are often integrated through the
autophagosomes during mitophagy may ability of a single molecular event to
be controlled differently than during stimulate multiple adaptive pathways (one
starvation-induced autophagy. The of which is autophagy). Third, distinct
ubiquitin-binding deacetylase, histone cellular stress response pathways,
deacetylase-6 (HDAC6) promotes including autophagy, mutually control
autophagosomal maturation by recruiting other stress response pathways.
a cortactin-dependent, actin–remodelling Some representative examples of the first
machinery to ubiquitinated structures, concept (Fig. 6A) include (i) redox stress,
where assembly of the F-actin network which induces transcriptional
facilitates autophagosome-lysosome reprogramming through HIF, NF-κB and
fusion. (Lee et al., 2010). However, HDAC6 p53 activation, elicits the UPR, and
deficiency leads to a failure of stimulates both general and selective
autophagosomal maturation only in the autophagy; (ii) hypoxia, which induces
context of quality control autophagy adaptive responses including the
(mitophagy and protein aggregate transcriptional activation of angiogenic
removal), but not in starvation-induced and cytoprotective cytokines in parallel
autophagy (Lee et al., 2010). This specific with autophagy stimulation; and (iii) DNA
requirement for a ubiquitin-binding damage, which elicits nuclear p53
deacetylase in quality control autophagy translocation, cell cycle checkpoint
suggests that ubiquitin modification may activation, cell cycle arrest, and autophagy.
TEÓRICO AUTOFAGIA 68

FIGURE 6
Hypothetical models of key cellular stress response networks

A. Particular stress stimuli (e.g. oxidative damage, hypoxia or anoxia, nutrient starvation, ER
stress) can elicit different responses that cooperate to achieve optimal cellular repair and
adaptation. A diverse range of stressors activate interconnected cytoprotective mechanisms
able to modulate autophagy at different levels, such as transcriptional reprogramming,
protein modifications (phosphorylation, acetylation, etc.) or cell cycle modulation. B.
Autophagy inhibition and stress. Autophagy impairment leads to the accumulation of
damaged proteins and organelles, which in turn can elicit cellular stress. Moreover, disabled
autophagy can increase the abundance of p62, resulting in an enhanced activity of NF-κB,
which leads to enhanced inflammation. By contrast, p62 accumulation leads to the activation
of Nrf2 transcription factor and in a consequent increase in the expression of stress response
enzymes. C. Mutual exclusion between autophagy and apoptosis. Autophagy, as a
cytoprotective pathway, eliminates potential sources of pro-apoptotic stimuli such as
damaged mitochondria, thereby setting a higher threshold against apoptosis induction. By
contrast, the apoptosis-associated activation of proteases such as calpain and caspase-3 may
destroy autophagy-specific factors (Atg4D, Beclin 1 or Atg5), thereby suppressing autophagy.
Many of the signals discussed in preceding autophagy (Criollo et al., 2010; Herrero-
sections illustrate the second concept. For Martin et al., 2009). Activation of LKB1-
example, eIF2α phosphorylation AMPK stimulates autophagy, as well as the
participates in the UPR in stress granule phosphorylation and activation of p27kip1, a
formation, in general translational control, cyclin-dependent kinase inhibitor that
and in autophagy induction (Kimball et al., leads to cell cycle arrest (Liang et al., 2007).
2003; Talloczy et al., 2002). Similarly, mTOR inhibition results in autophagy
activation of TAK1 and IKK coordinates the induction, as well as in the inhibition of
activation of NF-κB signaling and that of anabolic reactions including IRES-
TEÓRICO AUTOFAGIA 69

independent mRNA translation. FOXO3 These examples underscore the mutual


activation induces the transcription of control that exists between autophagy and
genes whose products increase both inflammatory signaling pathways. Another
proteasomal and autophagic degradation important axis of mutual control of
(Mammucari et al., 2007). different stress responses pathways is
between autophagy and cell cycle
The control of other stress pathways by
regulation. Autophagy is blocked during G2
autophagy was discussed briefly in
phase and mitosis, and a recent study
previous sections. For example, as noted
indicates that this may be mediated by
above, deficient autophagy leads to p62
cyclin-dependent kinase-mediated
accumulation, which activates NF-κB
phosphorylation of Vps34, which
signaling (Mathew et al., 2009). In addition,
negatively regulates its interaction with
p62 accumulation in the setting of
Beclin 1 during mitosis (Furuya et al.,
deficient autophagy activates the stress
2010). Moreover, there is a strong
responsive transcription factor Nrf2
correlation between mitogenic signaling
(Komatsu et al., 2010). High levels of p62
and autophagy inhibition (Levine and
disrupt the interaction between the
Kroemer, 2008). This correlation is further
Cul3/Rbx1 ubiquitin ligase complex, Keap1
strengthened by a recent genome-wide
(which normally targets Nrf2 for
screen, which found that a large
proteasomal degradation) and Nrf2,
percentage of genes that negatively
resulting in enhanced Nrf2 activity and an
regulate autophagy are positively involved
increase in expression of Nrf2-regulated
in cell growth and proliferation (Lipinski et
stress response enzymes (Komatsu et al.,
al., 2010). We speculate that this mutually
2010) (Fig. 6B). In addition, several
exclusive regulation of autophagy and cell
different autophagy genes regulate
growth may constitute a fundamental
different aspects of inflammatory
mechanism that cells use to appropriately
signaling, although, in some instances, it is
adapt their metabolism to stressful
not known whether such regulation is a
environmental conditions.
consequence of autophagy or alternative
functions of the autophagy genes (Sumpter
Links among General Autophagy and
and Levine, 2010; Virgin and Levine, 2009).
Quality Control Mechanisms
Autophagy genes negatively regulate RLR-
mediated induction of type I interferon There is a growing consensus that
both via conjugation of Atg12-Atg5 to autophagy is stimulated either in response
CARD domains of RLR signaling molecules to a central “command” of the cell (for
and through elimination of dysfunctional example in response to nutrient
mitochondria. Atg16L1 and Atg7 negatively depletion), in which case it is non-selective
regulate inflammatory signaling, including and general, or as a result of ubiquitination
IL-1β and IL-18 secretion. Moreover, Atg9, reactions that specifically target ‘garbage’,
but not Atg7, negatively regulates i.e. protein aggregates or damaged
activation of STING, a recently discovered mitochondria or intracellular pathogens,
transmembrane protein that is required for autophagic degradation. Nonetheless,
for efficient activation of type I IFN and these two types – general versus selective
pro-inflammatory cytokine production in – of autophagy may not be completely
response to interferon stimulatory DNA separable from each other. Indeed,
(Saitoh et al., 2009). stimulation of the general autophagic
pathway, either by inhibiting mTOR or by
TEÓRICO AUTOFAGIA 70

stimulating mTOR-independent autophagy 2007). If MMP is limited to a fraction of


pathways, leads to reduced accumulation mitochondria, this will result in the
of mutated, aggregation-prone proteins selective autophagic removal of
(such as mutant α-synyclein- or huntingtin depolarized mitochondria and the
protein) and damaged mitochondria avoidance of cell death. Thus, the efficacy
(Sarkar et al., 2007; Williams et al., 2008). of autophagy may set a higher threshold
Similarly, starvation or mTOR inhibition for the ability of MMP to constitute an
leads to increased autophagic targeting of irreparable, lethal event. In addition, it is
intracellular bacteria (Gutierrez et al., plausible that the liberation of Bcl-2 and
2004). One possible interpretation of these FLIP from activated autophagy protein
findings is that a general increase in complexes may free up these molecules to
protein and organelle turnover may be block the intrinsic and extrinsic pathways
sufficient to avoid protein aggregation and of apoptosis, respectively (Lee et al.,
organellar degeneration. Another 2009; Pattingre et al., 2005).
attractive interpretation is that general
Given the predominantly cytoprotective
autophagy is not completely non-selective
role of autophagy, it seems (teleo)logical
and may preferentially degrade proteins
that induction of apoptosis would be
and structures that are on the “verge” of
coupled to the inactivation of autophagy
aggregation or damage. This yet unproven
(Fig. 6C). There are some examples of
concept predicts that an increase in
molecular events that support this
general autophagy would reset the
concept. Caspase-3 cleaves Beclin 1,
threshold of quality control so that
thereby destroying its pro-autophagic
organelles that exhibit only minor
activity. The C-terminal fragment of Beclin
alterations (and that normally would not
1 that results from this cleavage acquires a
be removed by autophagy) are subject to
new function and can amplify
autophagic turnover. This would explain
mitochondrion-mediated apoptosis
why an increase in general autophagy can
(Wirawan et al., 2010). Caspase-3
accelerate bacterial clearance, delay the
activation also cleaves and activates
development of multiple
Atg4D, an enzyme that catalyzes the
neurodegenerative diseases, and mediate
delipidation of the LC3 paralog GABARPL1.
a general anti-aging effect (Madeo et al.,
This proteolytic activation increases Atg4D
2010).
targeting to mitochondria via a putative
BH3 domain and enhances its cytotoxic
Mutual Exclusion between Autophagy
activity (Betin and Lane, 2009). Similarly,
and Apoptosis
the proteolytic activity of calpain can
Autophagy is usually a self-limited process destroy the pro-autophagic function of
that protects cells from cell death by Atg5 (Xia et al., 2010) while it generates an
multiple mechanisms that include, but are Atg5-derived pro-apoptotic
not limited to, maintainance of mitochondrion-permeabilizing fragment of
bioenergetic homeostasis, recycling of Atg5 (Yousefi et al., 2006). All these results
misfolded and aggregate-prone proteins, underscore that concept that autophagy
and removal of uncoupled or and apoptosis are antagonistic events that
permeabilized mitochondria. The intrinsic tend to inhibit each other.
pathway of apoptosis is initiated by
mitochondrial membrane
permeabilization (MMP) (Kroemer et al.,
TEÓRICO AUTOFAGIA 71

Concluding Remarks
In this review, we have described stimulus-
dependent, as well as common regulatory
and execution steps of autophagy, with a
focus on the close links that exist between
autophagy and different types of adaptive
and repair responses to stress. These links
consist of multiple and intricate
mechanisms that are intertwined in
complex intersecting pathways, which
often function in positive and negative
amplification loops, thus composing
molecular switches and homeostatic
devices. A more detailed molecular
comprehension of these regulatory
networks, as well as their biomathematical
integration using systems biology
approaches, may furnish testable models
for more refined experimental and
therapeutic manipulations of autophagy.
At present, the comprehension of
autophagic regulation has only just begun,
and its multiple links to cell growth,
proliferation, senescence and apoptosis
await further exploration. We expect that
additional major circuits of autophagy
regulation will emerge and perhaps
supersede in importance the pathways
that we currently know – or believe to
know.

You might also like