Вы находитесь на странице: 1из 12

Essays in Biochemistry (2017) 61 663–674

https://doi.org/10.1042/EBC20170057

Review Article

Molecular control of chaperone-mediated autophagy


Steve Catarino1,2 , Paulo Pereira1,2,3 and Henrique Girão1,2
1 Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; 2 CNC.IBILI, University of Coimbra, Coimbra, Portugal;
3 Chronic Diseases Research Center (CEDOC), NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
Correspondence: Henrique Girão (hmgirao@fmed.uc.pt)

Chaperone-mediated autophagy (CMA) is a selective form of autophagy in which cytoso-


lic proteins bearing a pentapeptide motif biochemically related to the KFERQ sequence,
are recognized by the heat shock protein family A member 8 (HSPA8) chaperone, delivered
to the lysomal membrane, and directly translocated across the lysosomal membrane by a
protein complex containing lysosomal associated membrane protein 2a (Lamp2a). Since its
discovery over two decades ago, the importance of this pathway in cell proteostasis has
been made increasingly apparent. Deregulation of this pathway has been implicated in a
variety of diseases and conditions, including lysosomal storage diseases, cancer, neurode-
generation and even aging. Here, we describe the main molecular features of the pathway,
its regulation, cross-talk with other degradation pathways and importance in disease.

Introduction
Autophagy is one of the major proteolytic pathways in the cell, in which cytoplasmic content is deliv-
ered to lysosomes for degradation. Three main mechanisms of autophagic degradation have been de-
scribed. In macroautophagy, the most studied and best characterized form of autophagy, portions of
the cytoplasm, including protein aggregates and organelles, are sequestered inside a double membrane
structure termed as the autophagosome. This structure, in turn, can fuse with the late endosome or di-
rectly with the lysosome, where its contents are eventually degraded. In microautophagy, the lysosome
itself directly engulfs portions of cytosolic content, through invaginations of the lysosomal membrane.
In contrast, chaperone-mediated autophagy (CMA) is a strictly selective protein degradation process in
which cytosolic proteins bearing a KFERQ motif are recognized by heat shock protein family A mem-
ber 8 (HSPA8), also known as Hsc70, and delivered to the surface of the lysosome where they bind to
lysosomal associated membrane protein 2a (Lamp2a), and are eventually transported into the lysosomal
lumen where they are degraded. Although macro and microautophagy were both initially described as
non-selective degradation pathways, degradation of proteins, nucleic acids and lipids by these pathways
can be achieved through the bulk non-selective engulfment of cytoplasmic content or through selective
targeting mechanisms. Multiple mechanisms targeting substrates for macro and microautophagy have al-
ready been identified, involving the post-translational modification of substrates and the recruitment of
receptor and adaptor proteins, demonstrating that these pathways are also tightly regulated and can be
highly selective. Thus, rather than substrate specificity, the major factor distinguishing CMA from macro
and microautophagy, is that substrates reach the lysosomal lumen directly without the need of an inter-
mediate vesicular structure [1].

The CMA pathway


Received: 17 August 2017 Substrate recognition
Revised: 23 October 2017 Targeting of substrates for CMA degradation relies on a pentapeptide motif biochemically related to the
Accepted: 01 November 2017 KFERQ sequence [2]. Exposure of this motif during protein unfolding, disassembly of protein complexes
Version of Record published: or the removal of proteins from membranes, leads to the recruitment of HSPA8 and subsequent target-
12 December 2017 ing of the lysosomal membrane. All bona fide CMA substrates either contain a KFERQ motif in their


c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 663
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

Figure 1. The CMA pathway


Soluble cytosolic substrates bearing a KFERQ motif are first recognized by HSPA8 and transported to the lysosomal membrane where they bind
to monomeric Lamp2a. With the assistance of Hsp90, Lamp2a oligomerizes into a functional translocation complex. Following substrate unfolding
mediated by a chaperone complex; the substrate is translocated into the lysosomal lumen with the aid of a lysosomal form of HSPA8. After the release
of the substrate from the translocation complex, cytosolic HSPA8 mediates the disassembly of the Lamp2a translocation complex in order to restart
the process.

amino acid sequence or undergo post-translational modifications that result in a KFERQ-like motif. For example,
acetylation of a lysine residue has been shown to replace the glutamine residue in some CMA substrates [3,4]. In
some cases, exposure of the KFERQ motif and HSPA8 binding is insufficient for the delivery of the substrate to the
Lamp2a CMA receptor, and an additional post-translational modification of the substrate is required, as is the case for
the phosphorylation of lipid droplet (LD) protein perilipin 2 (PLIN2) [5] and the ubiquitination of the transcription
factor hypoxia inducible factor 1α (HIF1α) [6].

Substrate translocation into the lysosome


The recognition of the KFERQ motif by the chaperone HSPA8 takes place in the cytosol, after which the substrate
is directed to the surface of the lysosome where it binds to Lamp2a. The mechanism whereby CMA substrates are
translocated into the lysosomal lumen after binding to Lamp2a is still poorly understood. However, several steps
of this process have already been elucidated. Salvador et al. [7], demonstrated that CMA substrates are unfolded
at the surface of the lysosome before translocation, through a mechanism that requires ATP and HSPA8. In addi-
tion, translocation of the substrate was also shown to require the presence of multiple other cytosolic chaperones
at the lysosomal membrane including Hsp40, Hsc70-interacting protein (Hip) and Hsp70-Hsp90 organizing protein
(Hop) [8]. Upon reaching the lysosome, CMA clients preferentially bind to monomeric Lamp2a, which induces the
oligomerization of the protein into a high molecular weight complex active for translocation. Hsp90 regulates Lamp2a
oligomerization and also stabilizes the protein at the lysosomal membrane [9]. Translocation of substrates is assisted
by a lysosomal form of HSPA8 [10], as lysosome populations that do not contain this protein are less competent at
degrading CMA substrates. Thus, only lysosomes containing HSPA8 in their lumen are considered to be active for
CMA. Furthermore, in the absence of substrate, HSPA8 also induces the disassembly of Lamp2a oligomers, mak-
ing Lamp2a available for substrate binding in order to initiate another round of translocation [9] (Figure 1). A more
comprehensive history of the discovery of the molecular mechanisms of CMA can be found elsewhere [11]. Although
canonically CMA substrates consist of soluble cytosolic proteins, the integral membrane protein ryanodine receptor
type 2 (RyR2) has been reported as a CMA substrate [12].

Regulation of Lamp2a
Levels of Lamp2a at the lysosomal membrane, as well as efficient assembly and disassembly of the Lamp2a transloca-
tion complex are considered the rate limiting steps of the CMA pathway. As such, up-regulation of CMA degradation

664 
c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

in response to prolonged starvation, oxidative stress or inhibition of other proteolytic pathways is accompanied by an
increase in Lamp2a levels and Lamp2a-positive lysosomes. Conversely, reducing Lamp2a levels results in a decrease
in CMA activity.
Lamp2a can be found both at the lysosomal membrane or in the lysosomal lumen associated with lipids, existing
in a dynamic equilibrium between the two locations [13]. Studies performed using isolated lysosomes showed that in
the presence of substrate, Lamp2a membrane levels initially decrease until stabilizing after 20 min, with a correspond-
ing increase in the luminal levels of the protein. When the substrate was removed, part of the luminal Lamp2a was
trafficked back to the membrane to restore the initial levels of the protein, but only in CMA-active lysosomes isolated
from starved rats [10]. This trafficking of Lamp2a may partly explain the increase in Lamp2a lysosomal membrane
levels, with the concomitant decrease in luminal levels, observed during prolonged starvation when CMA is activated
[13].
Cathepsin A, a lysosomal protease also known as protective protein/cathepsin A (PPCA), has been shown to reg-
ulate CMA [14]. Indeed, cathepsin A was shown to interact with Lamp2a, preferentially with the monomeric form,
an interaction that was reduced in the presence of CMA substrates. Likewise, up-regulation of CMA activity through
prolonged starvation also reduced the interaction of cathepsin A with Lamp2a. Cathepsin A was shown to be respon-
sible for the cleavage of Lamp2a between the transmembrane and luminal regions, inducing the removal of the protein
from the lysosomal membrane and its degradation in the lysosomal lumen. Mutation of the cathepsin A cleavage site
on Lamp2a, prevented its cleavage and increased the clearance of CMA substrates. Thus, cathepsin A was shown to
regulate the levels of Lamp2a in basal conditions, to maintain a steady level of the protein on the lysosomal mem-
brane. In conditions where CMA is up-regulated, binding of cathepsin A to Lamp2a is reduced, presumably due to
altered conditions of the lysosomal lumen, and Lamp2a levels increase to allow for increased CMA degradation [14].
CMA activity can also be regulated through the distribution of Lamp2a in discrete microdomains of the lysosomal
membrane [15]. A subset of the Lamp2a protein present on the lysosomal membrane was found to accumulate in
detergent-resistant regions enriched in cholesterol. However, activation of CMA induces the mobilization of Lamp2a
out of these domains. Moreover, disruption of these microdomains through cholesterol depletion enhances the degra-
dation of substrates by CMA, while loading of cells with cholesterol had the opposite effect. As mentioned above,
following substrate binding, Lamp2a oligomerizes into a complex that promotes substrate translocation into the lyso-
somal lumen. Accordingly, it was found that Lamp2a present in the detergent-resistant regions was in a monomeric
state, while Lamp2a outside these microdomains could be found forming oligomeric complexes. In addition, Lamp2a
present in these microdomains was also found to be more susceptible to cleavage by cathepsin A [15], suggesting that
these lipid microdomains are the degradation site of Lamp2a.
Thus, Lamp2a at the lysosomal membrane, and by corollary CMA activity, are controlled by several dynamic equi-
libria between pools of Lamp2a present in the lysosomal lumen and membrane, and also between pools of Lamp2a
present in or outside discrete lipid microdomains on the lysosomal membrane. In basal conditions, a significant part
of Lamp2a is stored in the lysosomal lumen or in cholesterol and glycosphingolipid-rich regions of the membrane
where it is susceptible to cathepsin A cleavage and degradation. In times of need, these pools of Lamp2a are mobilized
to other regions of the lysosomal membrane, where Lamp2a can oligomerize into translocation competent complexes
upon substrate binding.
Several molecular effectors have been shown to modulate this assembly/disassembly cycle of the Lamp2a transloca-
tion complex. Glial fibrillary acidic protein (GFAP) binding to Lamp2a stabilizes the multimeric complex by prevent-
ing its disassembly by HSPA8. In the presence of GTP, elongation factor 1 α (EF1α) is released from its complex with
p-GFAP, which allows the self-assembly of p-GFAP with the pool of GFAP associated with Lamp2a. Release of GFAP
from Lamp2a allows the disassembly of the translocation complex by HSPA8 [16]. p-GFAP levels are controlled by
the activity of the kinase Akt which, in turn, is controlled by target of rapamycin complex 2 (TORC2) and pleckstrin
homology domain and leucine-rich repeat protein phosphatase 1 (PHLPP1). TORC2 acts as a CMA inhibitor by acti-
vating Akt, while PHLPP1 activates CMA by dephosphorylating Akt. Targeting PHLPP1 to the lysosomal membrane
was shown to depend on the Rho GTPase Rac1, whose binding to the lysosome is, in turn, regulated by GTP levels
[17]. Thus, GTP reduces CMA activity both by releasing p-GFAP from its inhibitory complex with EF1α, and also by
inhibiting Rac1-mediated recruitment of PHLPP1 to the lysosome and subsequent Akt dephosphorylation (Figure
2).
In addition to the mechanisms described above, where the levels of Lamp2a available for substrate binding at the
lysosomal membrane are regulated through protein degradation or the partition of the protein, in specific cases,
Lamp2a levels can also be regulated at the transcriptional level. For example, the increase in CMA-mediated protein
degradation in response to oxidative stress is not only due to an enhanced susceptibility of oxidized substrates to
degradation by CMA, but also owing to the up-regulation of Lamp2a levels. However, contrary to CMA activation


c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 665
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

Figure 2. Regulation of Lamp2a at the lysosomal membrane


Soluble cytosolic substrates bearing a KFERQ motif are first recognized by HSPA8 and transported to the lysosomal membrane where they bind
to monomeric Lamp2a. With the assistance of Hsp90, Lamp2a oligomerizes into a functional translocation complex. Following substrate unfolding
mediated by a chaperone complex; the substrate is translocated into the lysosomal lumen with the aid of a lysosomal form of HSPA8. After the release
of the substrate from the translocation complex, cytosolic HSPA8 mediates the disassembly of the Lamp2a translocation complex in order to restart
the process.
GFAP stabilizes the Lamp2a translocation complex. A signalling cascade consisting of TORC2 and Akt phosphorylates GFAP, promoting its dimeriza-
tion with the GFAP pool bound to Lamp2a. Release of GFAP from Lamp2a allows the recruitment of HSPA8 and the disassembly of the translocation
complex. Monomeric Lamp2a then migrates to lipid microdomains where it is susceptible to cathepsin A cleavage and lysosomal degradation. GTP
levels modulate Lamp2a. High GTP levels inhibit CMA by inducing the disassociation of EF1α from p-GFAP, allowing it to dimerize with Lamp2a-bound
GFAP, while low GTP levels up-regulate CMA by fostering the recruitment of Rac1 and PHLPP1 to the lysosomal membrane, where PHLPP1 dephos-
phorylates Akt, preventing GFAP phosphorylation.

in response to prolonged starvation, where the increase in Lamp2a available for substrate translocation is associated
with a relocation of the protein and a decrease in its degradation rate, during oxidative stress, the increase in Lamp2a
levels at the lysosomal membrane was shown to be mainly a consequence of increased transcription of the protein
[18].
Alternative splicing of the Lamp2 gene gives rise to three isoforms: Lamp2a which functions as a receptor and
translocation complex in CMA; Lamp2b which has been implicated in macroautophagy and Danon’s disease [19], and
Lamp2c, which is involved in the degradation of nucleic acids by autophagy [20,21]. These three variants of the Lamp2
gene differ mainly in their C-terminal, which is exposed on the cytosolic side of the lysosomal membrane. Recently, a
novel function of Lamp2c in negatively regulating CMA has been reported [22]. CMA regulates antigen presentation
through the targeted degradation of negative regulators of T-cell activation, such as the ubiquitin ligase Itch and the
calcineurin inhibitor RCAN1 [23]. Overexpression of Lamp2a and HSPA8 has also been shown to increase antigen
presentation through MHC class II (MHCII) [24]. Overexpression of Lamp2c was shown to reduce MHCII antigen
presentation of proteins that rely on CMA for epitope delivery to MHCII while no differences were observed for
proteins and peptides that rely on endocytosis and macroautophagy for antigen presentation [22]. Overexpression of
Lamp2c increased the cellular levels of two well-characterized CMA substrates, p-IκBα and RNAse A. The association
of HSPA8 with the CMA substrate glutamate decarboxylase (GAD) was also diminished by Lamp2c overexpression,
suggesting that Lamp2c inhibits CMA by obstructing chaperone association and the translocation of CMA substrates
into lysosomes.

CMA cross-talk with other proteolytic pathways


To ensure a healthy proteome, proteolytic pathways in cells are tightly controlled. Thus, when one major pathway
is blocked or is overloaded with substrate, alternative compensatory pathways are up-regulated to avoid substrate

666 
c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

accumulation and aggregation and maintain homoeostasis. During starvation, the first response of the cell to en-
sure its metabolic needs is to activate macroautophagy. However, during prolonged starvation, macroautophagy is
redirected from protein degradation to the preferential breakdown of lipids, with protein breakdown being at least
partially ensured by the CMA pathway instead [25,26]. Blockage of macroautophagy has been shown to constitutively
up-regulate CMA by increasing the number of CMA competent lysosomes containing HSPA8 in their lumen [27].
Conversely, in a mouse model with a hepatic deletion of Lamp2a, impaired CMA activity can be compensated for
by macroautophagy and proteasomal degradation, however this compensation mechanism is compromised during
stress conditions or with aging [28]. Although the mechanisms and players mediating this cross-talk are not fully
elucidated, it has been suggested that transcription factor EB (TFEB), which controls the expression of autophagy
and lysosomal genes [29], can play an important role in this process. TFEB was identified as a CMA substrate and
displayed increased cytosolic and nuclear levels in the liver of Lamp2a knockout mice [28], suggesting its possible role
in controlling compensation mechanisms between CMA and macroautophagy. Similarly, several components of the
20S proteasome are CMA substrates [30], which may account for the increased proteasomal degradation observed in
Lamp2a knockout mice [28].
CMA has also been demonstrated to regulate the degradation of lipids contained within LDs [31]. LDs are cellular
organelles responsible for the storage of lipids, which consist of a lipid ester core surrounded by a phospholipid mono-
layer and proteins, which protect them from the cytosol [32]. Degradation of LD content can occur both through the
action of cytosolic lipases or through a specialized form of macroautophagy termed as lipophagy. Inhibition of CMA
through the knockout of Lamp2a was shown to lead to the accumulation of triglycerides and LD. As CMA is only
responsible for the degradation of proteins and not lipids, a protein intermediate was needed to mediate the effect of
CMA upon LD lipolysis. This was shown to be PLIN2 and 3, perilipin proteins that are present on the surface of LD.
Inhibition of CMA or mutation of the KFERQ motif of PLIN2 led to the accumulation of the protein on the surface
of LD, this, in turn, reduced the interaction of LD with the cytosolic lipase adipose triglyceride lipase (ATGL) and
also with several components of the lipophagy machinery, providing the mechanistic link between CMA and lipolysis
[31].

HSPA8 in other degradation pathways


HSPA8 is a multipurpose chaperone involved in a variety of processes in the cell, functioning as a nexus for the
interplay of several pathways. This ability of HSPA8 is tied to the presence of a variety of interacting domains in the
structure of the protein [33,34]. As such, depending on its molecular interactors, HSPA8 is involved in the delivery
of substrates to both the proteasome and lysosome.
Indeed, HSPA8 can assist in the targeting of unfolded proteins to the proteasome by recruiting the E3 ubiquitin
ligase C-terminus of Hsc70-interacting protein (CHIP), which is responsible for attaching ubiquitin moieties to the
substrate, leading to their recognition and degradation by the 26S proteasome [35].
In chaperone-assisted selective autophagy (CASA), the co-chaperone BCL2-associated athanogene 3 (BAG-3) in-
duces the formation of a multichaperone complex that includes HSPA8 and CHIP. In this case, CHIP is responsible
for the ubiquitination of client proteins, leading to their recognition by the macroautophagy receptor p62, the re-
cruitment of phagophore membranes, autophagosome formation and, ultimately, their degradation in the lysosome
[36].
Importantly, the interaction of HSPA8 with KFERQ-like motifs is also utilized to signal the selective degradation of
cytosolic substrates in late endosomes, through a process termed as endosomal microautophagy [37]. In this process,
cytosolic substrates bearing a KFERQ-like motif are recognized by HSPA8 and transported to the surface of late en-
dosomes, where HSPA8 binds to phosphatidylserine present in the late endosomal membrane [33,37]. Unlike CMA,
Lamp2a plays no role in endosomal microautophagy and the substrate does not need to be unfolded prior to internal-
ization into the late endosome. Instead substrates are internalized through invaginations of the endosomal membrane
with the aid of the endosomal sorting complexes required for transport (ESCRT) I and III [37]. It is still unclear what
determines whether substrates containing KFERQ-like motifs are degraded by CMA or endosomal microautophagy.

Ubiquitination in CMA
The different proteolytic pathways of the cell are also interconnected by the targeting mechanisms employed for mark-
ing substrates for degradation. Canonically, protein ubiquitination is considered a targeting signal for the degradation
of substrates by the 26S proteasome [38], however, protein ubiquitination is also used to target membrane proteins for
degradation through the endolysosomal pathway by interaction with the ESCRT complexes [39,40] and also in forms
of selective macroautophagy where ubiquitinated proteins are recognized by macroautophagy receptors such as p62


c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 667
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

[41], which connect substrates to the nascent phagophore through their interaction with microtubule-associated pro-
tein 1 light chain 3 (MAPLC3) family members [38]. More recently, ubiquitination was shown to target the delivery
of substrates to CMA [6]. The ability of ubiquitin to function as a targeting motif for several different processes is
due to the plasticity of the ubiquitin signal. In broad terms, ubiquitin is conjugated to the lysine amino group of a
substrate through the sequential action of an E1 ubiquitin-activating enzyme, an E2 ubiquitin-conjugating enzyme
and an E3 ubiquitin ligase. Ubiquitin itself can be ubiquitinated on any of its lysine residues or N-terminal, forming
polyubiquitin chains with different topologies. Chains formed by lysine (K) 48 linked ubiquitins generally function
as a signal for proteasomal degradation, while K63 chains have been associated with the endolysosomal and selective
macroautophagy pathways. Studies by Ferreira et al. [6] have also implicated K63 ubiquitin chains in CMA degrada-
tion using HIF1α as a substrate. HIF1α is a component of the transcription factor heterodimer HIF1 that controls
the expression of genes that ensure cell survival in response to hypoxic conditions. In normoxic conditions, HIF1α
is continuously targeted for degradation by its modification with K48 chains, catalysed by the von Hippel–Lindau
(VHL) E3 ubiquitin ligase [42]. However, in hypoxic conditions, HIF1α no longer interacts with VHL and its degra-
dation by the proteasome is instead controlled by other E3 ligases such as CHIP [43]. HIF1α is also a substrate for
CMA, bearing a KFERQ-like motif necessary for its interaction with HSPA8 and Lamp2a. In addition to its role in
promoting the degradation of HIF1α by the proteasome, CHIP was also shown to be required for the interaction of
HIF1α with Lamp2a [44]. Furthermore, CHIP catalyses the conjugation of K63 ubiquitin chains to HIF1α, required
for directing the protein for degradation in the lysosome through CMA. Inhibiting the formation of K63 chains did
not negatively affect the interaction of HIF1α with HSPA8, but decreased its interaction with Lamp2a [6]. The poten-
tial of CHIP to direct substrates for proteasomal or CMA degradation is likely to be related to its ability to catalyse the
conjugation of both K48 or K63 chains, dependent on the E2-conjugating enzymes it is associated with [45]. Whether
this ubiquitin targeting mechanism is specific for HIF1α or applies to more CMA substrates is still unclear.

CMA in disease
Aging
It has been shown that CMA function declines with age, impairing the ability of cells/organisms to cope with insults
[11]. This decline in CMA was first observed in cells from the liver of aged rats and was correlated with a decrease
in substrate binding and translocation into the lysosomal lumen due to a decrease in Lamp2a levels at the lysosomal
membrane. In these cells, the number of CMA-competent lysosomes, those containing HSPA8 in their lumen, was
increased, possibly in an attempt to compensate for the decreased CMA activity [46]. This decrease in Lamp2a does
not stem from transcriptional down-regulation of Lamp2a, but rather from altered lysosomal dynamics of the protein
[47]. Indeed, lysosomes isolated from the livers of older rats displayed reduced rates in the mobilization of Lamp2a
from the lysosomal lumen to the membrane. Interestingly, while the degradation rate of luminal Lamp2a was in-
creased, Lamp2a present in the membrane was stabilized. These effects were attributed to changes in the composition
of the lysosomal membrane with age, where it was found that CMA-competent lysosomes contained less cholesterol
[47], one of the main components of the microdomains where membrane Lamp2a that is subjected to cathepsin A
cleavage localizes [15]. However, a later report by the same group challenged this notion as lysosomes isolated from
the liver of aged mice were shown to contain slightly higher levels of cholesterol [48]. The different animal models
used in each study may explain these differences. Nevertheless, in mice, feeding animals with a diet rich in cholesterol
was shown to have similar effects to aging, with an increase in lysosomal membrane cholesterol levels, reduction in
lysosomal Lamp2a levels, and a decrease in CMA activity, suggesting a common mechanism [48].
Further highlighting the importance of CMA decline in the pathology of aging, preserving CMA function in the
liver of aging mice was shown to improve cell homoeostasis and liver function [49]. Using a double transgenic mouse
model in which the levels of Lamp2a expression can be controlled, it was shown that preventing the decline in CMA
activity with age decreased the accumulation of oxidized proteins and aggregates, preserved mitochondrial activ-
ity, and maintained liver function. Interestingly, the transgenic mice also displayed preserved macroautophagic and
ubiquitin-proteasome function, pathways whose activity are known to decline with age. As mentioned above, block-
age of CMA has been demonstrated to induce the up-regulation of autophagy and the ubiquitin–proteasome system
[28], however these systems eventually fail due to their incapability to deal with the increased substrate load. It is
possible that by preserving CMA function, the up-regulation, and eventual age-dependent exhaustion, of the other
proteolytic pathways does not occur, thus contributing to the preservation of the activity of all three pathways [49].

668 
c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

Lysosomal storage diseases


The lysosomal storage disease cystinosis is caused by mutations in the cystinosin (CTNS) gene, which encodes the
lysosomal cystine transporter. Deficiencies in CTNS lead to the abnormal accumulation of cystine in lysosomes,
which in turn causes cell malfunction in organs such as the brain, eyes, kidneys, liver and muscle. Cystine-depletion
therapies are only moderately effective, indicating that cystine accumulation is not responsible for all symptoms of
the disease. Indeed, cystinosis has been linked to defects in CMA [50]. In accordance, not only were Lamp2a levels
decreased in cystinotic cells, but also the distribution of the protein was shown to be defective in CTNS knockout
mice, with Lamp2a failing to take residence in lysosomes. In turn, this was associated with defects in CMA substrate
translocation into lysosomes. Overexpression of CTNS in cells, but not cystine depletion, was shown to rescue Lamp2a
mislocalization [50]. This result was corroborated using CTNSK280R , a mutant with impaired transporter activity,
which was shown to be capable of rescuing Lamp2a localization [51]. Rescue of Lamp2a distribution in cystinotic
cells was shown to depend on Rab11, Rab7 and its effector the late endosome trafficking regulator Rab-interacting
lysosomal protein (RILP), proteins that are involved in endosomal and lysosomal transport mechanisms, identifying
these proteins as regulators of Lamp2a trafficking. Activation of the transcription factor TFEB was also shown to
rescue Lamp2a distribution in cystinotic cells, presumably through the up-regulation of RILP [51]. These studies
suggest that therapies that aim to improve CMA function, in addition to cystine depletion, may be a valuable strategy
for the treatment of cystinosis.

Cancer
CMA up-regulation has been associated with many types of cancer cells and tumours, as a strategy to promote cell
survival [52]. Blockage of CMA has been shown to reduce the proliferative and metastatic ability of malignant cells,
thus establishing the potential of CMA inhibitors as anticancer drugs. The role of CMA in promoting cancer cell sur-
vival has been linked to several mechanisms, ranging from providing general protection against oxidative stress [53],
degradation of specific anti-oncogenic targets [54] and also the protection of pro-oncogenic proteins from degrada-
tion by other pathways [55]. CMA has also been implicated in tumour resistance to therapy [56]. Macrophages, one
of the main cell types present in the tumour microenvironment, have been reported to activate CMA in other tumour
resident cells, through the release of IL-17, promoting cell survival and chemoresistance [57].
Although CMA activation is more commonly associated with tumour cell survival, CMA has also been proposed
to have a negative effect on some cancer cell types, through the degradation of oncogenic proteins such as p53 [58] or
hexokinase 2 [59]. A recent report has further highlighted the tumour suppressive role of CMA in myelocytomatosis
viral oncogene homologue (MYC) mediated fibroblast malignant transformation [60]. In this case, inhibition of CMA
through the depletion of Lamp2a in mouse fibroblasts was shown to increase MYC protein levels and cell proliferation
rates which results in higher cellular transformation capacity. However, despite the presence of two KFERQ-like motifs
on MYC, the protein was shown not to be a direct target for lysosomal degradation [60]. MYC is a known proteasomal
substrate, whose ubiquitination is regulated by the phosphorylation state of residue serine (S)62, which prevents MYC
ubiquitination. Cancerous inhibitor of PP2A (CIP2A) prevents the dephosphorylation of S62 catalysed by protein
phosphatase 2 (PPP2/PP2A), thus contributing to stabilize MYC [61,62]. The tumour suppressive role of CMA was
linked to the degradation of CIP2A. Indeed, CMA was demonstrated to be required for the lysosomal degradation
of CIP2A, with the consequent dephosphorylation, ubiquitination and degradation of MYC by the proteasome [60].
This study provides another example of the cross-talk that exists between autophagy and proteasomal degradation.

Neurodegenerative diseases
It is widely accepted that deregulation of the proteolytic pathways is implicated in the onset and/or development of
neurodegenerative diseases (ND). The accumulation of potentially toxic misfolded protein aggregates is an important
feature of the neurodegenerative process, highlighting the importance of having fully functional degradation path-
ways to ensure a healthy proteome and cell homoeostasis. Thus, it is unsurprising that CMA has been implicated in
neurodegeneration.
One of the key features of Parkinson’s disease (PD) neurons is the accumulation of Lewey bodies, characterized
by the presence of aggregates of α-synuclein (ASN). Inhibition of CMA, through the targeted depletion of Lamp2a
in cultured neurons, has been shown to lead to the accumulation of ubiquitinated ASN [63], while boosting CMA
activity through the overexpression of Lamp2a was shown to counteract the detrimental effects of elevated ASN levels
[64]. Targeted depletion of Lamp2a in the rat substantia nigra pars compacta was shown to lead to the accumulation
of ubiquitinated ASN and the progressive loss of dopaminergic neurons, both cardinal features of PD [65].


c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 669
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

Several protein mutations associated with hereditary forms of PD, have been shown to block CMA [66]. Some
mutant forms of ASN have been shown to affect CMA through their increased binding capacity to Lamp2a. In these
circumstances, not only is ASN translocation and degradation in lysosomes impaired, but also the access of other
substrates to Lamp2a is impeded [67]. Similarly, mutant forms of leucine-rich repeat kinase 2 (LRRK2) and ubiq-
uitin C-terminal hydrolase L1 (UCH-L1), proteins whose mutation is often associated with PD, also block CMA by
inhibiting the translocation complex [68,69].
Besides the damage induced by the accumulation of protein aggregates, inhibition of CMA may also result in
the impairment of cellular protection pathways due to the accumulation of key CMA substrates. For example, the
ASN-mediated blockage of CMA was shown to disrupt the degradation of inactive MEF2D, a transcription factor
required for neuronal survival [70]. Moreover, improper clearance of damaged and non-functional protein deglycase
DJ-1 by CMA was suggested to make PD neurons more susceptible to mitochondrial dysfunction and cell death in
response to oxidative stress [71]. More recent studies have identified the F-box protein Fbw7β as a CMA substrate
[72]. Fbw7β is an F-box protein family member that provides substrate specificity to skp, cullin, F-box containing
complex (SCF) ubiquitin ligases and has been reported to be a substrate of Parkin, an ubiquitin ligase whose muta-
tion is in the origin of several hereditary forms of PD. Parkin targets Fbw7β for proteasomal degradation, resulting
in the stabilization of the SCF target Mcl-1, a mitochondrial prosurvival factor. In agreement, neurons depleted of
Parkin were shown to be more sensitive to oxidative stress, due to an inability to maintain adequate levels of Mcl-1
[73]. It was reported that 6-hydroxydopamine (6-OHDA), which is used in animal models of PD, induces the oxi-
dization of Fbw7β and its subsequent degradation through CMA in a neuronal cell line. Curiously, although analysis
of post-mortem tissues from the striata of PD patients showed that the oxidation levels of Fbw7β were increased,
the total levels of the protein were unaltered [72], suggesting that oxidized forms of Fbw7β accumulate due to the
impairment of both the proteasome and CMA in PD. Thus, CMA blockage can contribute to the pathogenesis of PD
as a consequence of the accumulation of misfolded proteins, and the deregulation of pathways that protect against
oxidative damage.
In addition to PD, CMA has also been implicated in Huntington’s disease (HD). This disease is caused by a mutation
that generates an expansion of the polyglutamine (polyQ) sequence present in the N-terminal of the Huntington (Htt)
protein [74]. This mutation causes the aggregation of the protein in the nucleus and cytoplasm, inducing cellular
toxicity. One of the mechanisms of protein aggregate clearance in cells is macroautophagy, which has been reported
to help clear mutant Htt aggregates from cells [75]. However, mutant Htt also contributes to disrupt macroautophagy
[76]. Indeed, the expression of mutant Htt was shown to impair the ability of nascent phagophores to encapsulate
organelles such as LDs and mitochondria, without affecting later steps of the macroautophagy pathway. As with other
conditions in which macroautophagy is inhibited, a compensatory up-regulation of the CMA pathway has also been
associated with HD [77]. Neurons overexpressing mutant Htt present higher levels of Lamp2a, both through the
stabilization of the protein at the lysosomal membrane and also due to the transcriptional up-regulation of the Lamp2a
splice variant of the Lamp2 gene. HSPA8 was also up-regulated in HD cells, with increased levels of HSPA8 present
in the lumen of lysosomes. A diminished efficiency of this compensatory up-regulation of CMA has been associated
with the exacerbation of HD symptoms with age [77].
Although Htt had been previously shown to be a CMA substrate [4], the degradation of full-length and mu-
tant Htt by CMA in basal conditions was less efficient when compared with the bona fide CMA substrate
glyceraldehyde-3-phosphate dehydrogenase (GAPDH) [77]. Nevertheless, a method to selectively enhance the degra-
dation of mutant Htt by harnessing the CMA targeting machinery has been described [78]. QBP1 is a synthetic pep-
tide that specifically binds to expanded polyQ tracts in several proteins and which has been shown to suppress polyQ
aggregation and toxicity [79]. QBP1 binds to the expanded polyQ of mutant Htt, but does not bind to the regular
16 residue long polyQ of wild-type Htt. To target this peptide for CMA degradation, Bauer et al. [78] constructed
a chimera in which two sequential QBP1 proteins were fused to the KFERQ-like sequence of ASN and a canonical
KFERQ sequence. Expression of this chimera was shown to reduce the aggregation and toxicity of mutant Htt in cells,
without affecting the levels of wild-type Htt. Moreover, expression of this chimera was shown to induce the degra-
dation of mutant Htt by CMA, both in vitro and in mouse animal models of HD. These experiments were suggested
as a proof of concept that peptides containing functional KFERQ motifs, capable of binding to HSPA8, fused to a
sequence that recognizes unfolded proteins, may be used as a general tool for targeting mutant proteins to the CMA
pathway [78].

670 
c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

Future perspectives
The role of CMA in cell, tissue and organ homoeostasis and function has been made increasingly apparent throughout
the years, with changes in CMA being linked to several diseases and conditions such as lysosomal storage diseases,
NDs, cancer and even aging. Thus, understanding the mechanisms that govern this pathway will be important for
understanding the pathology of these conditions and provide new molecular targets for intervention. Although sev-
eral aspects of the CMA pathway have been elucidated, there are still several outstanding questions in the field. Some
of the molecular players responsible for substrate translocation across the lysosomal membrane have been described,
however, the exact mechanism by which this translocation takes place is still obscure. The number of Lamp2a pro-
teins available for oligomerizing into the translocation complex is the rate-limiting step of CMA. The pool of Lamp2a
proteins ready for receiving substrate is in a dynamic equilibrium with pools of Lamp2a present in cholesterol and
glycosphingolipid-rich lipid domains and the lysosomal lumen. How Lamp2a is mobilized from these pools to the
active form available for substrate binding and translocation is still unknown. Given the multitude of roles played
by HSPA8 in the cell, and its involvement in multiple degradation pathways, it is still unclear what directs discrete
HSPA8 interacting proteins to CMA over other destinations. This question is particularly intriguing when HSPA8
plays a role in the delivery of KFERQ-containing substrates to both the surface of the lysosome for CMA and the
late endosome for endosomal microautophagy. Can all CMA substrates be degraded by endosomal microautophagy?
The report that conjugation to K63 ubiquitin chains is required for the CMA degradation of HIF1α also raises some
pertinent questions. Are there ubiquitin receptors at the surface of the lysosome that mediate the interaction of these
ubiquitinated substrates with the Lamp2a translocation complex? Are the ubiquitin moieties removed from the sub-
strate prior to translocation, similar to the ubiquitin chain recycling that takes place during proteasomal degradation
and endolysosomal sorting? Or are they degraded alongside the substrate? Given the affinity of the ESCRT complexes
for interacting with K63 ubiquitin chains, is this targeting mechanism also used in endosomal microautophagy? Clar-
ifying these and other questions will provide new insights into one of the major proteolysis pathways of the cell.

Summary
• Chaperone-mediated autophagy (CMA) is one of the major proteolytic pathways in the cell, respon-
sible for the selective degradation of soluble cytosolic proteins in the lysosome.

• Substrates bearing a motif biochemically related to KFERQ are recognized by the chaperone HSPA8,
transported to the lysosomal membrane, and translocated into the lysosomal lumen with the assis-
tance of Lamp2a.

• Cross-talks among CMA, macroautophagy, and the ubiquitin–proteasome system exist, allowing the
shuttling of substrates between pathways and compensatory mechanisms.

• CMA has been implicated in the mechanism of several diseases and conditions, such as the lysoso-
mal storage disease cystinosis, Parkinson’s disease, Huntington’s disease, cancer and even aging.

Competing interests
The authors declare that there are no competing interests associated with the manuscript.

Funding
This work was supported by the European Regional Development Fund (FEDER) through the Operational Program for Competi-
tiveness Factors (COMPETE): HealthyAging2020 [grant number CENTRO-01-0145-FEDER-000012-N2323]; NETDIAMOND [grant
number POCI-01-0145-FEDER-016385] and grant POCI-01-0145-FEDER-007440 to CNC.IBILI. This work was also supported by
National Funds through the Portuguese Foundation for Science and Technology (FCT) [grant number FCT-UID/NEU/04539/2013]
and SFRH/BPD/68749/2010 (to S.C.)].


c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 671
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

Abbreviations
Akt, protein kinase B; ASN, α-synuclein; BCL2, B-cell lymphoma 2; CHIP, C-terminus of Hsc70-interacting protein; CMA,
chaperone-mediated autophagy; CTNS, cystinosin; DJ-1, protein deglycase DJ-1; EF1α, elongation factor 1 α; ESCRT, endo-
somal sorting complex required for transport; Fbw7β, F-box/WD repeat-containing protein 7; GFAP, glial fibrillary acidic protein;
HD, Huntington’s disease; HIF1α, hypoxia inducible factor 1α; HSPA8, heat shock protein family A member 8; Htt, Huntington;
IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; Lamp2a, lysosomal associated mem-
brane protein 2a; LD, lipid droplet; MEF2D, myocyte-specific enhancer factor 2D; Mcl-1, myeloid cell leukemia sequence 1;
MHCII, major histocompatibility complex class II; MYC, myelocytomatosis viral oncogene homologue; ND, neurodegenerative
disease; PD, Parkinson’s disease; PHLPP1, pleckstrin homology domain and leucine-rich repeat protein phosphatase 1; PLIN2,
perilipin 2; polyQ, polyglutamine; QBP1, polyglutamine binding peptide 1; Rac1, Ras-related C3 botulinum toxin substrate 1;
RCAN1, regulator of calcineurin 1; RILP, Rab-interacting lysosomal protein; SCF, skp, cullin, F-box containing complex; TFEB,
transcription factor EB; TORC2, target of rapamycin complex 2; VHL, von Hippel–Lindau.

References
1 Galluzzi, L., Baehrecke, E.H., Ballabio, A., Boya, P., Bravo-San Pedro, J.M., Cecconi, F. et al. (2017) Molecular definitions of autophagy and related
processes. EMBO J. 36, 1811–1836
2 Dice, J.F. (1990) Peptide sequences that target cytosolic proteins for lysosomal proteolysis. Trends Biochem. Sci. 15, 305–309
3 Lv, L., Li, D., Zhao, D., Lin, R., Chu, Y., Zhang, H. et al. (2011) Acetylation targets the M2 isoform of pyruvate kinase for degradation through
chaperone-mediated autophagy and promotes tumor growth. Mol. Cell 42, 719–730
4 Thompson, L.M., Aiken, C.T., Kaltenbach, L.S., Agrawal, N., Illes, K., Khoshnan, A. et al. (2009) IKK phosphorylates Huntingtin and targets it for
degradation by the proteasome and lysosome. J. Cell Biol. 187, 1083–1099
5 Kaushik, S. and Cuervo, A.M. (2016) AMPK-dependent phosphorylation of lipid droplet protein PLIN2 triggers its degradation by CMA. Autophagy 12,
432–438
6 Ferreira, J.V., Soares, A.R., Ramalho, J.S., Pereira, P. and Girao, H. (2015) K63 linked ubiquitin chain formation is a signal for HIF1A degradation by
chaperone-mediated autophagy. Sci. Rep. 5, 10210
7 Salvador, N., Aguado, C., Horst, M. and Knecht, E. (2000) Import of a cytosolic protein into lysosomes by chaperone-mediated autophagy depends on its
folding state. J. Biol. Chem. 275, 27447–27456
8 Agarraberes, F.A. and Dice, J.F. (2001) A molecular chaperone complex at the lysosomal membrane is required for protein translocation. J. Cell Sci.
114, 2491–2499
9 Bandyopadhyay, U., Kaushik, S., Varticovski, L. and Cuervo, A.M. (2008) The chaperone-mediated autophagy receptor organizes in dynamic protein
complexes at the lysosomal membrane. Mol. Cell. Biol. 28, 5747–5763
10 Cuervo, A.M., Dice, J.F. and Knecht, E. (1997) A population of rat liver lysosomes responsible for the selective uptake and degradation of cytosolic
proteins. J. Biol. Chem. 272, 5606–5615
11 Cuervo, A.M. and Wong, E. (2014) Chaperone-mediated autophagy: roles in disease and aging. Cell Res. 24, 92–104
12 Pedrozo, Z., Torrealba, N., Fernandez, C., Gatica, D., Toro, B., Quiroga, C. et al. (2013) Cardiomyocyte ryanodine receptor degradation by
chaperone-mediated autophagy. Cardiovasc. Res. 98, 277–285
13 Cuervo, A.M. and Dice, J.F. (2000) Regulation of lamp2a levels in the lysosomal membrane. Traffic 1, 570–583
14 Cuervo, A.M., Mann, L., Bonten, E.J., d’Azzo, A. and Dice, J.F. (2003) Cathepsin A regulates chaperone-mediated autophagy through cleavage of the
lysosomal receptor. EMBO J. 22, 47–59
15 Kaushik, S., Massey, A.C. and Cuervo, A.M. (2006) Lysosome membrane lipid microdomains: novel regulators of chaperone-mediated autophagy.
EMBO J. 25, 3921–3933
16 Bandyopadhyay, U., Sridhar, S., Kaushik, S., Kiffin, R. and Cuervo, A.M. (2010) Identification of regulators of chaperone-mediated autophagy. Mol. Cell
39, 535–547
17 Arias, E., Koga, H., Diaz, A., Mocholi, E., Patel, B. and Cuervo, A.M. (2015) Lysosomal mTORC2/PHLPP1/Akt regulate chaperone-mediated autophagy.
Mol. Cell 59, 270–284
18 Kiffin, R., Christian, C., Knecht, E. and Cuervo, A.M. (2004) Activation of chaperone-mediated autophagy during oxidative stress. Mol. Biol. Cell 15,
4829–4840
19 Nishino, I., Fu, J., Tanji, K., Yamada, T., Shimojo, S., Koori, T. et al. (2000) Primary LAMP-2 deficiency causes X-linked vacuolar cardiomyopathy and
myopathy (Danon disease). Nature 406, 906–910
20 Fujiwara, Y., Kikuchi, H., Aizawa, S., Furuta, A., Hatanaka, Y., Konya, C. et al. (2013) Direct uptake and degradation of DNA by lysosomes. Autophagy 9,
1167–1171
21 Fujiwara, Y., Furuta, A., Kikuchi, H., Aizawa, S., Hatanaka, Y., Konya, C. et al. (2013) Discovery of a novel type of autophagy targeting RNA. Autophagy
9, 403–409
22 Perez, L., McLetchie, S., Gardiner, G.J., Deffit, S.N., Zhou, D. and Blum, J.S. (2016) LAMP-2C inhibits MHC class II presentation of cytoplasmic antigens
by disrupting chaperone-mediated autophagy. J. Immunol. 196, 2457–2465
23 Valdor, R., Mocholi, E., Botbol, Y., Guerrero-Ros, I., Chandra, D., Koga, H. et al. (2014) Chaperone-mediated autophagy regulates T cell responses
through targeted degradation of negative regulators of T cell activation. Nat. Immunol. 15, 1046–1054
24 Zhou, D., Li, P., Lin, Y., Lott, J.M., Hislop, A.D., Canaday, D.H. et al. (2005) Lamp-2a facilitates MHC class II presentation of cytoplasmic antigens.
Immunity 22, 571–581

672 
c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

25 Singh, R., Kaushik, S., Wang, Y., Xiang, Y., Novak, I., Komatsu, M. et al. (2009) Autophagy regulates lipid metabolism. Nature 458, 1131–1135
26 Cuervo, A.M., Knecht, E., Terlecky, S.R. and Dice, J.F. (1995) Activation of a selective pathway of lysosomal proteolysis in rat liver by prolonged
starvation. Am. J. Physiol. 269, C1200–C1208
27 Kaushik, S., Massey, A.C., Mizushima, N. and Cuervo, A.M. (2008) Constitutive activation of chaperone-mediated autophagy in cells with impaired
macroautophagy. Mol. Biol. Cell 19, 2179–2192
28 Schneider, J.L., Villarroya, J., Diaz-Carretero, A., Patel, B., Urbanska, A.M., Thi, M.M. et al. (2015) Loss of hepatic chaperone-mediated autophagy
accelerates proteostasis failure in aging. Aging Cell 14, 249–264
29 Settembre, C., Di Malta, C., Polito, V.A., Garcia Arencibia, M., Vetrini, F., Erdin, S. et al. (2011) TFEB links autophagy to lysosomal biogenesis. Science
332, 1429–1433
30 Cuervo, A.M., Palmer, A., Rivett, A.J. and Knecht, E. (1995) Degradation of proteasomes by lysosomes in rat liver. Eur. J. Biochem. 227, 792–800
31 Kaushik, S. and Cuervo, A.M. (2015) Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis. Nat. Cell
Biol. 17, 759–770
32 Fujimoto, T. and Parton, R.G. (2011) Not just fat: the structure and function of the lipid droplet. Cold Spring Harb. Perspect. Biol. 3,
https://doi.org/10.1101/cshperspect.a004838
33 Morozova, K., Clement, C.C., Kaushik, S., Stiller, B., Arias, E., Ahmad, A. et al. (2016) Structural and biological interaction of hsc-70 protein with
phosphatidylserine in endosomal microautophagy. J. Biol. Chem. 291, 18096–18106
34 Zuiderweg, E.R., Hightower, L.E. and Gestwicki, J.E. (2017) The remarkable multivalency of the Hsp70 chaperones. Cell Stress Chaperones 22,
173–189
35 Kriegenburg, F., Ellgaard, L. and Hartmann-Petersen, R. (2012) Molecular chaperones in targeting misfolded proteins for ubiquitin-dependent
degradation. FEBS J. 279, 532–542
36 Arndt, V., Dick, N., Tawo, R., Dreiseidler, M., Wenzel, D., Hesse, M. et al. (2010) Chaperone-assisted selective autophagy is essential for muscle
maintenance. Curr. Biol. 20, 143–148
37 Sahu, R., Kaushik, S., Clement, C.C., Cannizzo, E.S., Scharf, B., Follenzi, A. et al. (2011) Microautophagy of cytosolic proteins by late endosomes. Dev.
Cell 20, 131–139
38 Dikic, I. (2017) Proteasomal and autophagic degradation systems. Annu. Rev. Biochem. 86, 193–224
39 Girao, H., Catarino, S. and Pereira, P. (2009) Eps15 interacts with ubiquitinated Cx43 and mediates its internalization. Exp. Cell Res. 315, 3587–3597
40 Schuh, A.L. and Audhya, A. (2014) The ESCRT machinery: from the plasma membrane to endosomes and back again. Crit. Rev. Biochem. Mol. Biol. 49,
242–261
41 Bejarano, E., Girao, H., Yuste, A., Patel, B., Marques, C., Spray, D.C. et al. (2012) Autophagy modulates dynamics of connexins at the plasma membrane
in a ubiquitin-dependent manner. Mol. Biol. Cell 23, 2156–2169
42 Maxwell, P.H., Wiesener, M.S., Chang, G.W., Clifford, S.C., Vaux, E.C., Cockman, M.E. et al. (1999) The tumour suppressor protein VHL targets
hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399, 271–275
43 Bento, C.F., Fernandes, R., Ramalho, J., Marques, C., Shang, F., Taylor, A. et al. (2010) The chaperone-dependent ubiquitin ligase CHIP targets
HIF-1alpha for degradation in the presence of methylglyoxal. PLoS ONE 5, e15062
44 Ferreira, J.V., Fofo, H., Bejarano, E., Bento, C.F., Ramalho, J.S., Girao, H. et al. (2013) STUB1/CHIP is required for HIF1A degradation by
chaperone-mediated autophagy. Autophagy 9, 1349–1366
45 Paul, I. and Ghosh, M.K. (2014) The E3 ligase CHIP: insights into its structure and regulation. Biomed Res. Int. 2014, 918183
46 Cuervo, A.M. and Dice, J.F. (2000) Age-related decline in chaperone-mediated autophagy. J. Biol. Chem. 275, 31505–31513
47 Kiffin, R., Kaushik, S., Zeng, M., Bandyopadhyay, U., Zhang, C., Massey, A.C. et al. (2007) Altered dynamics of the lysosomal receptor for
chaperone-mediated autophagy with age. J. Cell Sci. 120, 782–791
48 Rodriguez-Navarro, J.A., Kaushik, S., Koga, H., Dall’Armi, C., Shui, G., Wenk, M.R. et al. (2012) Inhibitory effect of dietary lipids on chaperone-mediated
autophagy. Proc. Natl. Acad. Sci. U.S.A. 109, E705–E714
49 Zhang, C. and Cuervo, A.M. (2008) Restoration of chaperone-mediated autophagy in aging liver improves cellular maintenance and hepatic function.
Nat. Med. 14, 959–965
50 Napolitano, G., Johnson, J.L., He, J., Rocca, C.J., Monfregola, J., Pestonjamasp, K. et al. (2015) Impairment of chaperone-mediated autophagy leads to
selective lysosomal degradation defects in the lysosomal storage disease cystinosis. EMBO Mol. Med. 7, 158–174
51 Zhang, J., Johnson, J.L., He, J., Napolitano, G., Ramadass, M., Rocca, C. et al. (2017) Cystinosin, the small GTPase Rab11, and the Rab7 effector RILP
regulate intracellular trafficking of the chaperone-mediated autophagy receptor LAMP2A. J. Biol. Chem. 292, 10328–10346
52 Kon, M., Kiffin, R., Koga, H., Chapochnick, J., Macian, F., Varticovski, L. et al. (2011) Chaperone-mediated autophagy is required for tumor growth. Sci.
Transl. Med. 3, 109ra17
53 Saha, T. (2012) LAMP2A overexpression in breast tumors promotes cancer cell survival via chaperone-mediated autophagy. Autophagy 8, 1643–1656
54 Tang, Y., Wang, X.W., Liu, Z.H., Sun, Y.M., Tang, Y.X. and Zhou, D.H. (2017) Chaperone-mediated autophagy substrate proteins in cancer. Oncotarget 8,
51970–51985
55 Suzuki, J., Nakajima, W., Suzuki, H., Asano, Y. and Tanaka, N. (2017) Chaperone-mediated autophagy promotes lung cancer cell survival through
selective stabilization of the pro-survival protein, MCL1. Biochem. Biophys. Res. Commun. 482, 1334–1340
56 Hu, Y.L., DeLay, M., Jahangiri, A., Molinaro, A.M., Rose, S.D., Carbonell, W.S. et al. (2012) Hypoxia-induced autophagy promotes tumor cell survival and
adaptation to antiangiogenic treatment in glioblastoma. Cancer Res. 72, 1773–1783
57 Guo, B., Li, L., Guo, J., Liu, A., Wu, J., Wang, H. et al. (2017) M2 tumor-associated macrophages produce interleukin-17 to suppress oxaliplatin-induced
apoptosis in hepatocellular carcinoma. Oncotarget 8, 44465–44476


c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 673
Essays in Biochemistry (2017) 61 663–674
https://doi.org/10.1042/EBC20170057

58 Vakifahmetoglu-Norberg, H., Kim, M., Xia, H.G., Iwanicki, M.P., Ofengeim, D., Coloff, J.L. et al. (2013) Chaperone-mediated autophagy degrades mutant
p53. Genes Dev. 27, 1718–1730
59 Xia, H.G., Najafov, A., Geng, J., Galan-Acosta, L., Han, X., Guo, Y. et al. (2015) Degradation of HK2 by chaperone-mediated autophagy promotes
metabolic catastrophe and cell death. J. Cell Biol. 210, 705–716
60 Gomes, L.R., Menck, C.F.M. and Cuervo, A.M. (2017) Chaperone-mediated autophagy prevents cellular transformation by regulating MYC proteasomal
degradation. Autophagy 13, 928–940
61 Yada, M., Hatakeyama, S., Kamura, T., Nishiyama, M., Tsunematsu, R., Imaki, H. et al. (2004) Phosphorylation-dependent degradation of c-Myc is
mediated by the F-box protein Fbw7. EMBO J. 23, 2116–2125
62 Junttila, M.R., Puustinen, P., Niemela, M., Ahola, R., Arnold, H., Bottzauw, T. et al. (2007) CIP2A inhibits PP2A in human malignancies. Cell 130, 51–62
63 Vogiatzi, T., Xilouri, M., Vekrellis, K. and Stefanis, L. (2008) Wild type alpha-synuclein is degraded by chaperone-mediated autophagy and
macroautophagy in neuronal cells. J. Biol. Chem. 283, 23542–23556
64 Xilouri, M., Brekk, O.R., Landeck, N., Pitychoutis, P.M., Papasilekas, T., Papadopoulou-Daifoti, Z. et al. (2013) Boosting chaperone-mediated autophagy
in vivo mitigates alpha-synuclein-induced neurodegeneration. Brain 136, 2130–2146
65 Xilouri, M., Brekk, O.R., Polissidis, A., Chrysanthou-Piterou, M., Kloukina, I. and Stefanis, L. (2016) Impairment of chaperone-mediated autophagy
induces dopaminergic neurodegeneration in rats. Autophagy 12, 2230–2247
66 Sala, G., Marinig, D., Arosio, A. and Ferrarese, C. (2016) Role of chaperone-mediated autophagy dysfunctions in the pathogenesis of Parkinson’s
disease. Front. Mol. Neurosci. 9, 157
67 Cuervo, A.M., Stefanis, L., Fredenburg, R., Lansbury, P.T. and Sulzer, D. (2004) Impaired degradation of mutant alpha-synuclein by chaperone-mediated
autophagy. Science 305, 1292–1295
68 Orenstein, S.J., Kuo, S.H., Tasset, I., Arias, E., Koga, H., Fernandez-Carasa, I. et al. (2013) Interplay of LRRK2 with chaperone-mediated autophagy. Nat.
Neurosci. 16, 394–406
69 Kabuta, T., Furuta, A., Aoki, S., Furuta, K. and Wada, K. (2008) Aberrant interaction between Parkinson disease-associated mutant UCH-L1 and the
lysosomal receptor for chaperone-mediated autophagy. J. Biol. Chem. 283, 23731–23738
70 Yang, Q., She, H., Gearing, M., Colla, E., Lee, M., Shacka, J.J. et al. (2009) Regulation of neuronal survival factor MEF2D by chaperone-mediated
autophagy. Science 323, 124–127
71 Wang, B., Cai, Z., Tao, K., Zeng, W., Lu, F., Yang, R. et al. (2016) Essential control of mitochondrial morphology and function by chaperone-mediated
autophagy through degradation of PARK7. Autophagy 12, 1215–1228
72 Wang, X., Zhai, H. and Wang, F. (2017) 6-OHDA induces oxidation of F-box protein Fbw7beta by chaperone-mediated autophagy in Parkinson’s model.
Mol. Neurobiol., https://doi.org/10.1007/s12035-017-0686-0
73 Ekholm-Reed, S., Goldberg, M.S., Schlossmacher, M.G. and Reed, S.I. (2013) Parkin-dependent degradation of the F-box protein Fbw7beta promotes
neuronal survival in response to oxidative stress by stabilizing Mcl-1. Mol. Cell. Biol. 33, 3627–3643
74 Chen, S., Ferrone, F.A. and Wetzel, R. (2002) Huntington’s disease age-of-onset linked to polyglutamine aggregation nucleation. Proc. Natl. Acad. Sci.
U.S.A. 99, 11884–11889
75 Ravikumar, B., Vacher, C., Berger, Z., Davies, J.E., Luo, S., Oroz, L.G. et al. (2004) Inhibition of mTOR induces autophagy and reduces toxicity of
polyglutamine expansions in fly and mouse models of Huntington disease. Nat. Genet. 36, 585–595
76 Martinez-Vicente, M., Talloczy, Z., Wong, E., Tang, G., Koga, H., Kaushik, S. et al. (2010) Cargo recognition failure is responsible for inefficient
autophagy in Huntington’s disease. Nat. Neurosci. 13, 567–576
77 Koga, H., Martinez-Vicente, M., Arias, E., Kaushik, S., Sulzer, D. and Cuervo, A.M. (2011) Constitutive upregulation of chaperone-mediated autophagy in
Huntington’s disease. J. Neurosci. 31, 18492–18505
78 Bauer, P.O., Goswami, A., Wong, H.K., Okuno, M., Kurosawa, M., Yamada, M. et al. (2010) Harnessing chaperone-mediated autophagy for the selective
degradation of mutant huntingtin protein. Nat. Biotechnol. 28, 256–263
79 Nagai, Y., Fujikake, N., Ohno, K., Higashiyama, H., Popiel, H.A., Rahadian, J. et al. (2003) Prevention of polyglutamine oligomerization and
neurodegeneration by the peptide inhibitor QBP1 in Drosophila. Hum. Mol. Genet. 12, 1253–1259

674 
c 2017 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society

Вам также может понравиться