ArticlePDF AvailableLiterature Review

Targeting Autophagy In Disease: Recent Advances In Drug Discovery

Authors:

Abstract and Figures

Introduction Small molecules targeting autophagy have been highly implicated as new therapeutic agents to treat diseases of interest. With the increasing demand for autophagy-targeting drugs, this review attempts to provide an efficient strategy to explore major autophagy-based human disease interventions with newly explored mechanisms using small molecules and promising therapeutic approaches. Areas covered Introduced in this review are direct links and applications among autophagy pathways, their modulators, and phenotypic diseases, along with recent approaches. Autophagy-related diseases, machinery, and compounds are introduced to guide the appropriate investigation of autophagy in the pharmaceutical industry. The authors then provide their expert perspectives on the subject. Expert opinion The self-catabolic intracellular process autophagy occurs in organisms throughout their lifetime, supporting its critical role in organismal health across life stages. Because of the detrimental influence of dysfunctional cells to an organism and their etiology in numerous diseases, maintaining cellular quality control by recycling components through autophagy is essential to prevent health decline.
Autophagy-targeting strategies are established in various diseases including cancer, proteopathies, vascular disease, and other human diseases. (i) Applying autophagy for cancer treatment must be carefully considered based on its complex role in cancer. Characteristics of cancer such as protein abnormalities, morphology, and progressive growth can be modulated by the autophagy pathways depicted in Section 3.1. In brief, targeting autophagy in cancer can be divided into two strategies: autophagy activation and inhibition. In the autophagy activation approach, the purpose is to prevent cells from malignant transformation by maintaining cellular protection. The autophagy inhibition approach includes targeting autophagy initiation and its ability to ultimately degrade its contents. The goal of this approach is to inhibit cancer cells from exploiting autophagy for their propagation. (ii) Proteopathies are protein conformational disorders caused by the accumulation of abnormal protein aggregates such as amyloid β plaques, tau neurofibrillary tangles (NFTs), α-synuclein aggregates, and prions. The strategy for 'autophagy activation' could ameliorate pathological conditions derived from these conformational abnormality through promoting degradation of the pathological substrates as depicted in Section 3.2. (iii) In vascular disease including atherosclerosis, restenosis, intraocular neovascularization, and ischemia-reperfusion injury, autophagy plays a fundamental role in maintaining physiological homeostasis as depicted in Section 3.3. Targeting autophagy signaling such as mTOR-S6 K could improve atherosclerosis and restenosis by attenuating abnormal endothelial cell proliferation and protecting the blood brain barrier (BBB) against ROS-induced injury. ROS could negatively regulate autophagic flux, implying its role as a therapeutic target for regulating autophagy in vascular diseases. In addition, despite few clarified mechanisms, autophagy inhibition reduces VEGF protein levels, leading to an anti-intraocular neovascularization effect. (iv) Therapeutic approaches for targeting other human diseases including viral or bacterial infectious diseases and lysosomal storage diseases. Targeting strategies over autophagy pathways using specific modulators or combinational approaches with other medications are depicted in Section 3.4. In lysosomal storage disorders and bacterial infectious diseases, molecular regulators elevating autophagy turnover are promising therapeutics. In viral infections, however, this approach should be carefully determined based on how autophagy positively or negatively affects the host-virus relationship.
… 
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=iedc20
Expert Opinion on Drug Discovery
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iedc20
Targeting Autophagy In Disease: Recent Advances
In Drug Discovery
Dasol Kim , Hui-Yun Hwang & Ho Jeong Kwon
To cite this article: Dasol Kim , Hui-Yun Hwang & Ho Jeong Kwon (2020): Targeting Autophagy
In Disease: Recent Advances In Drug Discovery, Expert Opinion on Drug Discovery, DOI:
10.1080/17460441.2020.1773429
To link to this article: https://doi.org/10.1080/17460441.2020.1773429
Published online: 16 Jun 2020.
Submit your article to this journal
Article views: 61
View related articles
View Crossmark data
REVIEW
Targeting Autophagy In Disease: Recent Advances In Drug Discovery
Dasol Kim
a
*, Hui-Yun Hwang
a
* and Ho Jeong Kwon
a,b
a
Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University,
Seoul, Republic of Korea;
b
Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
ABSTRACT
Introduction: Small molecules targeting autophagy have been highly implicated as new therapeutic
agents to treat diseases of interest. With the increasing demand for autophagy-targeting drugs, this review
attempts to provide an efficient strategy to explore major autophagy-based human disease interventions
with newly explored mechanisms using small molecules and promising therapeutic approaches.
Areas covered: Introduced in this review are direct links and applications among autophagy pathways,
their modulators, and phenotypic diseases, along with recent approaches. Autophagy-related diseases,
machinery, and compounds are introduced to guide the appropriate investigation of autophagy in the
pharmaceutical industry. The authors then provide their expert perspectives on the subject.
Expert opinion: The self-catabolic intracellular process autophagy occurs in organisms throughout their
lifetime, supporting its critical role in organismal health across life stages. Because of the detrimental
influence of dysfunctional cells to an organism and their etiology in numerous diseases, maintaining cellular
quality control by recycling components through autophagy is essential to prevent health decline.
ARTICLE HISTORY
Received 9 December 2019
Accepted 20 May 2020
KEYWORDS
Autophagy; autophagy-
related diseases; autophagy-
targeting strategy; cancer;
proteopathies; vascular
diseases
1. Introduction
‘Autophagy’ indicates a ‘self-eating’ process wherein a cell
degrades its cytoplasmic material by lysosomal digestion [13].
This self-digestion removes damaged organic substrates and
exogenous microorganisms as a cellular defense strategy.
Based on these homeostatic roles that maintain vital cellular
functions, many recent studies have highlighted links between
autophagy modulation and various phenotypic diseases such as
cancer and vascular, neurodegenerative, and metabolic diseases
[47]. Therefore, discovering autophagy-regulating drugs has
been emphasized to provide potential disease-alleviating
approaches. However, the therapeutic application of autophagy
modulators without an understanding of their mechanism of
action could lead to undesirable adverse effects, since these
drugs trigger different responses based on the specific autop-
hagy pathways involved in each disease [4,8]. Therefore, eluci-
dating the molecular mechanisms of autophagy-regulating small
molecules in a disease-specific manner is crucial for their use in
treating autophagy-related diseases.
This review covers major autophagy-related diseases
along with emerging protein factors in autophagy pathways
and the small molecules that target them. To highlight
these diseases, 11 of the most deadly autophagy-related
maladies and symptoms are ranked based on the number
of cases reported during 2000–2009 and 2010–2019. During
both periods, cancer ranked highest, followed by aging,
proteopathies, diabetes, vascular disease, obesity, viral infec-
tion, microbial infection, myopathy, lysosomal storage dis-
orders (LSDs), and sepsis. Among them, aging, diabetes, and
obesity were excluded, as they are considered ‘certain
symptoms or metabolic statuses’ of the human body rather
than ‘diseases’. Thus, in this review, we primarily focus on
the three highest-ranking diseases (cancer, proteopathies,
and vascular disease), followed by other infectious and
lysosomal diseases. Because the autophagic process is
linked to diverse signaling networks in the cell, disease
pathogenesis is differentially affected by autophagy, based
on the signaling pathways involved. Therefore, it is critical
to clearly understand the implications of targeting autop-
hagy-regulating pathways and their roles in disease before
building a therapeutic approach. To support this, we review
small molecules that target autophagy-related pathways
and/or newly emerging factors and their roles in each dis-
ease. In addition, we cover therapeutic drugs that are cur-
rently used or undergoing clinical trials. This exploration
into the pharmacological intervention of autophagy-related
diseases and regulatory factors offers promising opportu-
nities for the strategic development of therapeutic
approaches against autophagy-related diseases.
2. Regulatory factors in the autophagy process
2.1. Regulatory factors in autophagy: from phagophore
to autolysosome
When autophagy is induced upon certain stress stimuli, cells
promote the dynamic interaction of several proteins that play
a part in the autophagy process. This comprises multiple steps,
including initiation/nucleation, elongation/expansion, and
CONTACT Ho Jeong Kwon kwonhj@yonsei.ac.kr Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and
Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
*These authors contributed equally to this work
EXPERT OPINION ON DRUG DISCOVERY
https://doi.org/10.1080/17460441.2020.1773429
© 2020 Informa UK Limited, trading as Taylor & Francis Group
maturation/degradation [9]. During autophagic ‘flux’ or turnover,
the first two steps, considered early-stage, relate to the formation
of the autophagosome, while the last step, considered late-stage,
relates to autophagosomal degradation [10].
At the beginning of initiation/nucleation, autophagy-related
gene (ATG) proteins assemble at the phagophore assembly site
(PAS) to form a small cup-shaped membrane precursor. These
ATGs include E1-like ligase ATG7, ATG6 homolog Beclin-1,
cysteine protease ATG4A-D, and microtubule-associated protein
1 light-chain 3 (LC3) [11]. The precursor phagophores eventually
close to form double- or multi-membraned vacuoles called
autophagosomes in the later elongation/expansion step [12].
During this step, LC3 undergoes lipidation through covalent
binding with phosphatidylethanolamine (PE), thereby resulting
in the conversion of LC3-I to LC3-II. This has been used as
a quantitative biomarker in the assessment of autophagic
activity.
In the final maturation/degradation step, autophagosomes
traffic toward the microtubule-organizing center (MTOC) to
fuse with lysosomes and degrade cargo through lysosomal
hydrolases. Two essential processes are pivotal for proper
autophagy turnover: (1) autophagosome-lysosome fusion,
and (2) lysosomal hydrolase activation. Although the mechan-
ism underlying autophagosome-lysosome fusion is currently
unclear, it is under intense investigation. Factors involved in
fusion include the endosomal sorting complexes required for
transport (ESCRTs), soluble N-ethylmaleimide-sensitive factor
attachment protein receptors (SNARE), ultraviolent radiation
resistance-associated gene (UVRAG), Rubicon (RUBCN), small
GTPase of the Ras-related protein 7 (RAB7), lysosomal asso-
ciated membrane protein 2 (LAMP2), ion channels, and other
tethering factors [1317]. Because lysosomal enzymes are acid
hydrolases (including proteases, glycosidases, nucleases,
phosphatases, and lipases) that are active at acidic pH (~5)
but not neutral pH, maintaining low pH is essential for ‘active
lysosomes’ to induce sequential autophagic turnover.
Accordingly, lysosomal ion channel proteins such as vacuolar-
type H
+
-ATPase (V-ATPase) and transient receptor potential
mucolipin 1 (TRPML1) play essential roles to maintain lysoso-
mal ionic homeostasis and membrane potential, resulting in
lysosome activation by hydrogen cation influx [18].
2.2. Regulatory factors in the expression of autophagy
genes
Several transcription factors act as master regulators during
signal transfer in the autophagic process [19]. Under nutrient-
rich conditions, these are sequestered in the cytoplasm in the
inactive state. However, upon certain stimuli to the cells, they
are modified by cytoplasmic enzymes and translocate into the
nucleus to participate in autophagy regulation at the tran-
scriptional level [19]. The primary master transcription factors
are portrayed below with a description of the mechanism by
which they are regulated to facilitate the expression of genes
encoding autophagy-related proteins (Figure 1).
2.2.1. Microphthalmia/transcription factor e (mit/tfe)
MiT/TFE family members (MITF, TFEB, TFE3, and TFEC) are
the most recognized master genes of autophagy, as they
play a pivotal role in autophagosomal and lysosomal bio-
genesis [20]. They share a common structure in the basic
helix-loop-helix (bHLH) leucine zipper (LZ) dimerization
motif, except for TFEC (transcription inhibition rather than
activation) [21]. MITF, TFEB, and TFE3 directly bind to the
coordinated lysosomal expression and regulation (CLEAR)
element [22,23], which is a common feature in the promoter
regions of lysosome genes. These proteins are primarily
phosphorylated by kinases such as mTORC1 and bound to
14-3-3 protein, which masks their nuclear localization signal
(NLS) to maintain them in the cytoplasm [24]. Upon autop-
hagy initiation, the repressive phosphorylation is removed
by phosphatases such as calcineurin, resulting in their
nuclear translocation for transcription of autophagy/lysoso-
mal genes. Indeed, several studies have revealed that mod-
ulation of Mit/TFE family members is a pivotal step in
a variety of pathologic phenotypes [25].
2.2.2. Nuclear factor erythroid-derived 2-like 2 (nfe2l2/
nrf2)
NFE2L2/NRF2 is considered a master regulator of cellular
homeostasis, as it controls the expression of protective
genes [26]. The protective genes, including antioxidant
genes, share a common cis-acting antioxidant response ele-
ment (ARE) in their upstream promoter region [26,27]. Under
benign conditions, KEAP1 directly binds to NRF2, leading to its
ubiquitination and degradation by the ubiquitin-proteasome
system (UPS) [28]. Upon oxidative stress, cysteine thiols on
KEAP1 are modified, releasing NRF2 to translocate into the
nucleus to control target gene expression [29]. Furthermore,
NRF2 is reported to regulate autophagy and vice versa. NRF2
modulates the expression of autophagy genes, including
SQSTM/p62 and LAMP2A [26,27]. However, increased SQSTM/
Article highlights
Autophagy is a fundamental catabolic process to maintain cellular
homeostasis in response to cell proliferation, hypoxia, stress gener-
ated by reactive oxygen species, various pathological invasions,
nutrient starvation, and aggregation of cytoplasmic components
leading to human pathological conditions. Recently, many reports
have highlighted autophagy-targeting strategies for therapeutic
purposes.
Many small molecule modulators perturbing autophagy have been
used to elucidate the interplay between specific autophagy pathways
and autophagy-related diseases.
In cancer, there have been significant disputes over autophagy-
targeting therapies due to their complex functions. Targeting autop-
hagy in cancer depends either on its tumor-suppressive or tumori-
genic actions. Targeting strategies using autophagy modulators can
be classified into autophagy activation and autophagy inhibition.
Maintaining protein homeostasis through autophagy degradation in
proteopathies can restore or remove toxic cellular protein aggregates
to restore normal physiological conditions. Targeting strategies with
pharmacological interventions will be emphasized through the reg-
ulation of various autophagy pathways in diverse protein conforma-
tional disorders such as amyloidosis, tauopathies, synucleinopathies,
and prion diseases.
Pharmacological perturbation through the cannibalizing effects of
autophagy can be considered a robust targeting strategy due to its
fundamental roles in mediating physiological processes across
diverse vascular diseases such as atherosclerosis, restenosis, intrao-
cular neovascularization, and ischemia-reperfusion injury.
2D. KIM ET AL.
p62 sequesters KEAP1 and promotes its degradation as
SQSTM/p62 itself is an autophagy substrate, thereby creating
a positive feedback loop for active NRF2 signaling [29]. This
master autophagy regulator is highlighted due to its preven-
tive role against human diseases.
2.2.3. Forkhead box o (foxo)
FoxO transcription factor family members (FoxO1, FoxO3,
FoxO4, and FoxO6) have established roles in diverse cellular
functions such as aging, longevity, and development [30].
Similar to the MiT/TFE family, the FoxO transcription factor
family is mostly controlled by phosphorylation [31]. AKT/JNK
kinase signaling is a major upstream regulator for FoxO1,
FoxO3, and FoxO4, but less for FoxO6 due to its lack of
a C-terminal AKT-dependent site [30,32]. Among these,
FoxO3 [33] and FoxO1 [34] were reported to be autophagy
gene transcription factors. A recent study exhibited the rele-
vance of FoxO4 to autophagy gene expression through
a triple genetic deletion of FoxO1, FoxO3, and FoxO4 in mus-
cle [35]. However, it is currently unclear whether FoxO4 binds
directly to the promoter region of autophagy genes. The FoxO
family is regarded as another pivotal modulator for cellular
homeostasis through autophagy, as these members have cen-
tral roles in diverse diseases.
2.2.4. CCAAT/enhancer-binding protein (c/ebp)
C/EBP family members (C/EBPα, C/EBPβ, C/EBPγ, C/EBPδ, C/EBPε,
and C/EBPζ) are characterized by a conserved b-Zip domain at
the C-terminus that mediates dimerization and DNA binding
[36]. C/EBP family members are known to regulate cell prolifera-
tion and differentiation at the transcriptional level [37]. Recent
studies have revealed that they also have a role in autophagic
regulation. Among them, C/EBPβ was reported to control the
expression of autophagy genes, including Beclin-1, ATG5, and
ATG4 [38,39]. More recently, C/EBPδ (CEBPD) was found to
activate transcription of the autophagy genes LC3B and ATG3
in hepatocellular carcinoma (HCC) [40]. Another transcription
factor, C/EBPζ (C/EBP-homologous protein/CHOP), alone or in
combination with ATF4, was revealed to bind the promoter
regions of a set of autophagy genes through eIF2α/ATF4 path-
way activation under ER stress or starvation conditions, thereby
regulating autophagy [41]. Thus, the C/EBP family can regulate
cellular homeostasis as autophagy master genes, supporting
their utility as molecular targets for related diseases.
2.2.5. GATA transcription factor
Transcription factors in the GATA family (GATA1, GATA2, GATA3,
GATA4, GATA5, and GATA6) are characterized by their ability to
bind the (A/T)GATA(A/G) sequence in DNA [42]. Although GATA
Figure 1. Master genes in the transcriptional regulation of autophagy. (i) MiT/TFE family members are attenuated by phosphorylation in the cytoplasm. When they
are modified by phosphatases such as calcineurin, they translocate into the nucleus and bind the CLEAR domain in DNA to promote transcription of autophagy-
related genes. (ii) NFE2 L2/NRF2 are inhibited by KEAP1 binding in the cytoplasm. Upon oxidative stress, a disulfide bond forms in KEAP1, thereby releasing NFE2L2/
NRF2 for nuclear translocation. The transcription factors bind to ARE domains in DNA and promote target gene expression. (iii) FoxO family members remain inactive
in the cytoplasm by phosphorylation. Upon signaling stimuli, phosphatase PP2A dephosphorylates them, leading to their nuclear translocation to enhance target
gene expression. (iv) C/EBP family members undergo posttranslational modifications in the cytoplasm. Cellular kinases such as ERK1/2 modify them by
phosphorylation to promote their nuclear translocation and transcriptional activity. (V) GATA family members are cell type-specifically regulated. GATA1 is mainly
known as a transcription factor for autophagy genes. GATA1 is activated upon acetyl modification by the acetyltransferase CBP, which promotes autophagy gene
transcription.
EXPERT OPINION ON DRUG DISCOVERY 3
family members were originally considered to play roles in hema-
topoietic cells and cardiac tissue [43], it was found that GATA1
directly activates the transcription of pivotal autophagy genes
such as LC3B and its homologs, and genes involved in lysosomal
biogenesis and function [44]. Because GATA1 is a cell type-specific
master regulator in hematopoietic development, it can be lever-
aged due to its specificity and suggests a new strategy for target-
ing related diseases.
3. Autophagy and disease targeting
Numerous studies have revealed an interplay between autop-
hagy and a variety of human diseases. In this section, major
diseases such as cancer, proteopathies, vascular disease, and
others are discussed (Figure 2), followed by recent chemother-
apeutic strategies that target autophagy (Table 1).
3.1. Cancer
Autophagy-targeting strategies for cancer treatment have been
considered promising therapeutic approaches but are controver-
sial due to duals roles for autophagic function in cancer therapy.
The potential complexity of autophagy in cancer is likely depen-
dent on cancer type, location, genetic variation, stage, and tumor
microenvironment [46,47]. Some investigations present autop-
hagy activation as tumor suppressive function, while the others
suggest its tumorigenic characteristics according to specific autop-
hagy pathways. Hence, understanding these differences and the
exact biological roles autophagy plays in cancer is critical for
therapeutic importance. Notably, recent studies have identified
numerous modulators of autophagy as cancer targets.
3.1.1. Autophagy activation in cancer
Autophagy was originally considered a tumor inhibitory
mechanism to reduce cancer progression or increase cell
ProteopathiesCancer
Other diseases
Microbial
infection
Viral
infection
Targeting
Autophagy
Vascular diseases
Restenosis
Atherosclerosis
BBB disruption
Aβ
plaque
α-syn
body
Tau
NFT
Prion
Aggregates
degradation
PI3K-Akt-mTOR
pathway
Tubulin
AMPK-mTOR
pathway
Rhes
Autophagy
cycle
Abnormal
growth
Malignant
transformation
Stress
UVRAG
SNARE
PRDX1
ROS
Lipophagy
TFEB/Nrf2
Neovascularization
Akt-mTOR
pathway
Autophagy
VEGF
Dysfunctional
lysosome
Lysosomal
hydrolysis
AMPK
pathway
mTOR-NLRP3
pathway
Autophagy
Inflammation
Replication
Figure 2. Autophagy-targeting strategies are established in various diseases including cancer, proteopathies, vascular disease, and other human diseases. (i)
Applying autophagy for cancer treatment must be carefully considered based on its complex role in cancer. Characteristics of cancer such as protein abnormalities,
morphology, and progressive growth can be modulated by the autophagy pathways depicted in Section 3.1. In brief, targeting autophagy in cancer can be divided
into two strategies: autophagy activation and inhibition. In the autophagy activation approach, the purpose is to prevent cells from malignant transformation by
maintaining cellular protection. The autophagy inhibition approach includes targeting autophagy initiation and its ability to ultimately degrade its contents. The
goal of this approach is to inhibit cancer cells from exploiting autophagy for their propagation. (ii) Proteopathies are protein conformational disorders caused by the
accumulation of abnormal protein aggregates such as amyloid β plaques, tau neurofibrillary tangles (NFTs), α-synuclein aggregates, and prions. The strategy for
‘autophagy activation’ could ameliorate pathological conditions derived from these conformational abnormality through promoting degradation of the pathological
substrates as depicted in Section 3.2. (iii) In vascular disease including atherosclerosis, restenosis, intraocular neovascularization, and ischemia-reperfusion injury,
autophagy plays a fundamental role in maintaining physiological homeostasis as depicted in Section 3.3. Targeting autophagy signaling such as mTOR-S6 K could
improve atherosclerosis and restenosis by attenuating abnormal endothelial cell proliferation and protecting the blood brain barrier (BBB) against ROS-induced
injury. ROS could negatively regulate autophagic flux, implying its role as a therapeutic target for regulating autophagy in vascular diseases. In addition, despite few
clarified mechanisms, autophagy inhibition reduces VEGF protein levels, leading to an anti-intraocular neovascularization effect. (iv) Therapeutic approaches for
targeting other human diseases including viral or bacterial infectious diseases and lysosomal storage diseases. Targeting strategies over autophagy pathways using
specific modulators or combinational approaches with other medications are depicted in Section 3.4. In lysosomal storage disorders and bacterial infectious diseases,
molecular regulators elevating autophagy turnover are promising therapeutics. In viral infections, however, this approach should be carefully determined based on
how autophagy positively or negatively affects the host-virus relationship.
4D. KIM ET AL.
mortality. The dual roles of autophagy can be partially attrib-
uted to different actions from the same factors. Although
increasing some biological elements can be beneficial to
tumor progression in the initial steps of tumorigenesis, such
as ROS generation, accumulation of p62/SQSTM1-mediated
misfolded aggregates, damaged mitochondria, and inflamma-
tion, their accumulation during later stages of tumor growth
can cause deleterious effects such as metabolism dysfunction,
genetic impairment, and eventual tumor suppression [48,49].
For autophagy activation strategies to target cancer progres-
sion, ATG family members like ATG2, ATG5, ATG7, ATG9, and
ATG12, and Beclin-1 and VPS34 are upstream factors necessary
for phagophore formation [50]. The impact of depleting or
mutating these factors has been studied in cancer cell lines
and mouse models, revealing that reduction of autophagy
increases cellular proliferation, supporting a role for autopha-
gic activation against tumorigenesis. In other studies, various
signaling players, such as AKT (curcumin, AZD7328) [51,52],
mTOR (rapamycin, resveratrol, torin 2) [53,54], AMPK (metfor-
min, kaempferol, aspirin, nilotinib) [3,5557], and MAPK (bor-
tezomib, p38-MAPK-JUNK pathway activator) [46], have been
shown as valuable targets both for inhibition of cancer
Table 1. Representative autophagy-targeting agents in autophagy-related diseases.
Agents Structure Targeting autophagy
Autophagy
pathways Ref.
Cancer Vinblastine Autophagy inhibition and
cancer cell death
Disrupts polymerization of
both non-acetylated or
acetylated microtubules,
leading to inhibition of
fusion between
autophagosomes
and lysosomes
[76]
Berbamine Autophagy inhibition and
cancer cell death
Inhibits SNARE complex
formation, leading to
autophagy inhibition by
disrupting the interaction
between BNIP3 and SNAP29
[77]
Proteopathies Alborixin Autophagy activation and amyloid β
(Aβ) plaque clearance in
amyloidosis
Induces autophagy by
targeting the PTEN/PI3-K/
AKT signaling axis
[94]
Lonafarnib Autophagy activation and tau
aggregate clearance in tauopathies
Induces autophagy by
suppressing Rhes
farnesylation followed by
increased lysosomal activity
[122]
Trehalose and rapamycin Additive autophagy activation and
protection of dopaminergic
neurons in Parkinson’s disease
Induces autophagy to improve
dopaminergic deficits
through the AMPK-mTOR
signaling axis
[134]
AR-12 Autophagy activation and
combinatorial effects with cellulose
esters, a prion conversion inhibitor,
in prion diseases
Induces pro-autophagic
activity that significantly
extends the survival of
prion-infected mice by
targeting the mTOR
pathway
[145]
(Continued )
EXPERT OPINION ON DRUG DISCOVERY 5
progression and autophagy activation in various cancer cell
lines and mouse models. Some of these autophagy activators
have been applied as clinical FDA-approved treatments such
as rapamycin derivatives (renal cell carcinoma, pancreatic or
breast cancer), nilotinib (chronic myelogenous leukemia), and
bortezomib (myeloma/lymphoma) after verifying their effec-
tiveness. However, recent studies demonstrated that autop-
hagy can promote both cell death or cell survival in cancer
according to the stage of progression. For example, elevating
autophagy through an acidic pH microenvironment contri-
butes to cancer progression or survival during initial stages
[58]. It is responsible for the poor response to autophagy
activation in clinical uses, such as with the mTOR inhibitor
rapamycin, and should be considered in detail when targeting
autophagy activation in cancer therapy.
3.1.2. Autophagy suppression in cancer
Autophagy inhibition, resulting in the inability to remove
targeted protein cargo, is also considered a suppressive
approach against tumorigenesis. For example, key biomarkers
such as LC3 and p62 are used to monitor autophagy levels, as
they are associated with the formation of autophagosomal
membranes that envelop protein cargo for subsequent degra-
dation. Autophagic machinery activated by these factors is
correlated with cancer phenotypes such as cancer survival
and metastasis, vessel invasion, and tube formation at the
early stages of progression [59]. When autophagy cannot
occur after phagophore formation, which means blocking
the initiation of forming autophagosomes by inhibiting targets
involved in these pathways, it may support tumor growth in
multiple cells and tumor types, as well as trigger resistance to
Table 1. (Continued).
Agents Structure Targeting autophagy
Autophagy
pathways Ref.
Vascular
diseases
PRDX1 mimetics
(ebselen, gliotoxin)
Maintaining lipophagic flux and
cholesterol efflux in atherosclerosis
Rescues impaired lipophagy
under pathologic conditions
by reducing produced ROS
[154]
Indatraline Autophagy activation and inhibition
of the rapid proliferation of
smooth muscle cells in restenosis
Induces autophagy without
apoptotic cellular toxicity
by targeting the AMPK-
mTOR-S6 K signaling axis
[45]
3-methyladenine or
chloroquine
Autophagy inhibition in initiation or
degradation steps, respectively,
and suppressed neo-
vascularization in ocular vascular
diseases
Inhibits autophagy leading to
regulation of the
angiogenic factors VEGF
and PEDF
[157]
Sirolimus
(rapamycin)
Autophagy activation leading to
maintaining the endothelial barrier
in ischemia-reperfusion injury
Induces autophagy by
targeting mTOR and
regulates the localization of
tight junction protein Cldn5
[159]
Viral infection 3-methyladenine
or
wortmannin
Autophagy inhibition at initiation and
suppressed viral infection and
replication in cells
Inhibits autophagy by
targeting the PI3-K/AKT
signaling axis
[171]
Microbiome-
mediated
diseases
Alpinetin Autophagy activation to ameliorate
the intestinal barrier and colitis
Induces autophagy by
targeting the TSC2/mTOR
signaling axis and
suppressing apoptotic cell
death signaling
[187]
6D. KIM ET AL.
various therapies. Autophagy initiation can also be suppressed
by inhibitors upstream of the phagophore. For instance, phos-
phatidylinositol 3-kinase (PI3-K) is a protein kinase that acti-
vates autophagy through PI3-K-AKT-mTOR, and through ERK
signaling crosstalk is involved in cancer progression and sur-
vival. In several cancers, including glioblastoma, cutaneous
melanoma, and HCC, this signaling cascade is highly activated,
so their inhibition is considered a therapeutic strategy to
suppress cancer [60]. Inhibitors of PI3-K (idelalisib: the class
I inhibitor, 3-methyladenine: the class I and class III dual
inhibitor, wortmannin: the class I inhibitor) [6163] or MEK/
ERK (PD98059) [64] suppress autophagy, increasing cancer cell
death. This early-stage inhibitory strategy has potential for
clinical applications, as idelalisib has already been developed
as a treatment for chronic lymphocytic leukemia, follicular
B-cell non-Hodgkin’s lymphoma, and small lymphocytic lym-
phoma (FDA approved).
Another emerging cancer target is the UVRAG protein,
which regulates Beclin-1-class III phosphatidylinositol 3-kinase
(PI3-KC3)-mediated autophagic and tumor-suppressive activ-
ity. Beclin-1 is mono-allelically deleted or mutated in several
cancers, including breast, colon, and ovarian cancers [65].
UVRAG positively acts in the Beclin-1-PI3-KC3-dependent acti-
vation of autophagy and therefore inhibits tumorigenesis [66],
making it an interesting target for further investigation. As
a case study for developing small molecules that specifically
target autophagy, autophagy-activating rapamycin and its
derivatives showed antitumor effects in HCC patients by inhi-
biting the mTOR pathway and cancer progression in clinical
trials [67,68]. Although some did not show an antitumor effect
in clinical trials [69], introducing rapamycin as autophagy
activator opened a gateway to take advantage of mTOR-
related autophagy and its practical application to cancer. To
date, combinatorial approaches using different autophagic
pathway modulators with mTOR inhibitors have been
a promising therapeutic strategy in HCC treatment [70].
During autophagosome formation and recycling, LC3
undergoes processing by some ATG proteins (ATG4, ATG7,
ATG3, and the 12-5-16L1 complex, sequentially). Among
them, ATG4 plays roles in priming pro-LC3 into LC3-I and
cleaving LC3-II into LC3-I to maintain the autophagic process
[71]. Recent articles reviewed small molecules targeting
ATG4B, such as FMK-9a, tioconazole, and Z-FG-FMK, suggest-
ing that ATG4B could be a molecular target in cancer through
an autophagy inhibition approach [71]. Inhibiting autophagic
flux in the later stages of the autophagic process has been
increasingly considered a key strategy for promoting cancer
cell death through apoptosis, necrosis, and ferroptosis [7274].
Many cancers, including glioblastoma, breast cancer, and non-
small cell lung cancer, have the ability to sustain or intensify
autophagic degradation following radiation, playing a key role
in cancer cell resistance to radiotherapy and antitumor drugs
[75]. Therefore, the development of autophagic degradation
inhibitors in combination with other agents has been promis-
ing. The accumulation of cellular cargo proteins in autophago-
somes can be induced by either interrupting their fusion with
lysosomes or inhibiting lysosomal proteolysis. Fusion between
autophagosomes and lysosomes to form autolysosomes can
be suppressed by interrupting a variety of factors such as
microtubules and SNARE complexes. Microtubules are
required for trafficking and fusion of mature autophagosomes
with lysosomes. During this process, microtubule acetylation is
required. Vinblastine disrupts microtubule polymerization
both in non-acetylated and acetylated microtubules and
impairs LC3-associated fusion between autophagosomes and
lysosomes [76]. SNARE complexes comprise cytoplasmic
SNAP29, autophagosomal syntaxin 17, and lysosomal VAMP8,
all of which interact with each other and are required for
autolysosome fusion [77]. Berbamine is an autophagic flux
inhibitor that potently increases BCL2 interacting protein 3
(BNIP3) expression and the interaction between BNIP3 and
cytoplasmic SNAP29, blocking the interaction between
SNAP29 and lysosomal VAMP8 [77]. Berbamine-mediated inhi-
bition of SNARE complex formation and autophagic flux has
shown chemotherapeutic effects in cancer.
As another strategy for inhibiting autophagic flux in cancer,
lysosomotropic reagents such as chloroquine (CQ, under clin-
ical trial) inhibit lysosomal uptake, resulting in increased lyso-
somal pH and impaired autophagy degradation [78]. Either
direct or indirect inhibition of lysosomal proteins can be
a method for disrupting autophagic flux and cancer, as in
the following examples: (1) targeting lysosomal enzymes
with the combinatorial treatment of pepstatin A and E64d
completely blocks lysosomal functionality by inactivating
aspartic/thiol proteases and cathepsin B, D, H, and L in lyso-
somes [79]; (2) targeting the lysosomal membrane protein
V-ATPase, which acidifies lysosomes as an electrogenic H
+
pump, using bafilomycin A1 to inhibit V-ATPase, elevate
vacuolar pH, and inhibit lysosomal uptake [2]; and (3) target-
ing cytoplasmic interactors such as Hsp70 chaperone and
ubiquitin-like protein 4A (UBL4A). Hsp70 stabilizes lysosomes
through a direct interaction with the endolysosomal anionic
phospholipid bis(monoacylglycerol)phosphate (BMP), which
leads to direct binding between lysosomal sphingomyelin
and acid sphingomyelinase (ASM) to maintain lysosomal
homeostasis [80,81]. Pifithrin-μ (PES) directly inhibits Hsp70
activity to suppress tumorigenesis [82]. Alternatively, UBL4A
maintains protein metabolism and cellular homeostasis.
UBL4A causes dysfunction in lysosomal degradation through
direct binding with lysosomal-associated membrane protein 1
(LAMP1), causing an antitumor effect [83]. Therefore, UBL4A
can be a promising target for disrupting autophagy degrada-
tion in cancer therapy.
3.1.3. Crosstalk between autophagy and mechanism of
cell death in cancer
Although autophagy is a cell survival process by recycling
deleterious organic substrates, overactive autophagy can
lead to cellular suicide. Crosstalk between autophagy and
mechanisms of cell death such as apoptosis has been investi-
gated for their crucial roles in cellular homeostasis. Because
autophagy can act as both a tumor-suppressive mechanism by
degrading factors that contribute to cancer as well as an
apoptotic driver for cancer cell death, its impairment can
lead to favorable environments for cancer. However, autop-
hagy also facilitates the survival of cancer cells under stressful
EXPERT OPINION ON DRUG DISCOVERY 7
conditions such as the acidic tumor microenvironment or
hypoxia. Recently, targeting interactions between autophagic
and apoptotic factors has been implicated in cancer therapeu-
tics. Autophagy and apoptosis are interconnected through
several factors, including a Beclin-1-BCL2 interaction, caspase-
and calpain-mediated Beclin-1 cleavage, increased B-cell lym-
phoma 2-interacting mediator of cell death (BIM) expression
by mTOR inhibition, interactions between UVRAG and BCL2-
associated X protein (BAX), and interactions between FAS-
associated protein with death domain (FADD) and ATG5
[43,84]. Overall, the interconnectivity between autophagy
and apoptosis requires an integrative approach for developing
autophagy-targeting small molecules against specific diseases.
3.1.4. Brief discussion about autophagy in cancer
Due to the dual role of autophagy in the pathophysiological
phenotypes of malignant cancers, it is important to determine
whether autophagy activators or inhibitors are required to treat
a specific cancer type based on tumorigenic characteristics and
autophagy-related pathways. To date, multi-dimensional knowl-
edge of autophagy-modulating small molecules in preclinical stu-
dies have given valuable results in cancer research [85]. Therefore,
making use of autophagic modulators without genetic modifica-
tions will contribute to the discovery and control of autophagic
signaling molecules that play key roles in specific autophagy-
related cancer pathways.
3.2. Proteopathies
Proteopathies, also known as proteinopathies or protein con-
formational disorders (PCDs), refer to a class of diseases in
which certain proteins can aggregate in non-native, structu-
rally abnormal conformations, or become more susceptible to
aggregation even in their native conformation. These aggre-
gates become toxic and disrupt the function of cells, tissues,
and organs [86]. Recent studies have reported that inducing
autophagy flux significantly ameliorates these pathologies by
directly degrading the abnormal aggregates [87]. Therefore,
pharmacologically modulating autophagy is regarded as
a promising strategy against these diseases.
3.2.1. Amyloidopathies
Amyloidopathies (also called as β-amyloidopathies) consist of
amyloid β (Aβ) plaque deposition in the brain, which is a key
feature in neurodegenerative disorders including Alzheimer’s dis-
ease (AD) [88], Parkinson’s disease dementia (PDD) [89], and
dementia with Lewy bodies (DLB) [90]. The formation of Aβ aggre-
gates has been well established. The amyloid precursor protein
(APP, a transmembrane protein) undergoes two different cleavage
processes via three secretase enzymes: α-secretase, β-secretase
(BACE1), and γ-secretase [91]. In non-amyloidogenic processes,
APP is cleaved by α-secretase to leave the C-terminal fragment
83 (αCTF) in the cell membrane, which is then cleaved by γ-
secretase and released as the soluble P3 peptide fragment. In the
amyloidogenic process, APP is cleaved by β-secretase that leaves
the C-terminal fragment 99 (βCTF) in the cell membrane, which is
then cleaved by γ-secretase and released as
1-40
or the less
soluble Aβ
1-42
. These Aβ peptides are prone to aggregate into
oligomers, fibrils, and plaques, hallmarks of neurodegenerative
disorders and AD in particular [92]. Autophagy is a key regulator
of Aβ metabolic activity such as clearance and secretion [93],
implying that balanced regulation of autophagy is critical for
neuronal homeostasis.
As described in Section 3.1, the PI3-K-AKT-mTOR pathway is
a conventional signal for autophagy induction. Recent studies
revealed that several modulators promoted clearance
through autophagy induction by targeting this pathway,
including alborixin [94], rapamycin [95], AR12 (an analog of
the COX-2 inhibitor celecoxib) [96], and ginsenoside com-
pound K [97]. AMPK is also known to induce autophagy by
directly phosphorylating ULK1/Beclin-1 to form the phago-
phore or through attenuation of the autophagy inhibitory
factor mTOR [98]. Hence, AMPK activators such as resveratrol
[99] and its analogs (RSVA314, RSVA405) [100] and ginseno-
side Rg2 [101] exhibited reduced levels by activating
autophagy. However, another indirect AMPK activator, metfor-
min, is currently controversial for its effect on amyloid pathol-
ogy, as it exhibited gender-specific responses in an APP mouse
model [102] and showed contradictory results for the regula-
tion of Aβ formation [103]. This indicates that delicate mod-
ulation is required for AMPK targeting approaches.
Additionally, regulation of autophagy by master transcrip-
tion factors has also been considered for amyloid-maladies.
For instance, resveratrol exhibited fAβ degradation by promot-
ing TFEB deacetylation through Sirtuin 1 (SIRT1) [104].
Tacrolimus (FK506) also reduced amyloid plaques resulting in
neuroprotection [105], in which TFEB activation could occur
through interactions with another protein target [2].
3.2.2. Tauopathies
Tauopathies are a group of neurodegenerative disorders
including Alzheimer’s disease (AD), chronic traumatic ence-
phalopathy (CTE), and corticobasal degeneration (CBD) [106].
It is well established that tauopathies feature the pathological
accumulation of abnormal forms of the microtubule-
associated protein tau (MAPT) in neurons and glia, which are
susceptible to forming insoluble aggregates through misfold-
ing [107] or hyper-phosphorylation [108]. These aggregates
lead to neuronal cell death by overcoming the entire neuron
and forming neurofibrillary tangles (NFTs), a major hallmark of
neurodegenerative diseases together with Aβ plaques [109].
Several studies have revealed that autophagy dysregulation
occurs in tauopathies [110]. It is therefore pivotal to rescue
and augment autophagy in order to control pathogenic tau in
tau-mediated neuronal disorders. Currently, studies have
focused on the AKT/mTOR axis to regulate autophagy. For
example, autophagy enhancement with AKT/mTOR axis regu-
lators such as rapamycin [111], selenomethionine [112], and
PP242 [113] reduced tau aggregates or mitigated cognitive
deficits in tauopathy models.
Moreover, the indirect AMPK activator metformin amelio-
rated tauopathy through autophagy induction [114] and also
reduced tau phosphorylation through AMPK-mediated activa-
tion of protein phosphatase 2A (PP2A) [115]. However, the
relevance of PP2A and AMPK is currently controversial [116],
since AMPK is known to induce autophagy through phosphor-
ylation and reportedly elevates tau phosphorylation toward
tau-mediated toxicity [117]. Consequently, several studies
8D. KIM ET AL.
have focused on an alternative approach of promoting autop-
hagy gene master transcription factors or autophagic flux
machinery. According to these reports, tau pathology was
reduced by activating TFEB with trehalose [118], fisetin [119],
flubendazole [120], and activating NRF2 with fisetin [119] and
sulforaphane [121]. This drove the development of trehalose,
fisetin, and sulforaphane dietary supplements for neuronal
health. Furthermore, a recent study has indicated the farnesyl-
transferase inhibitor lonafarnib as a new therapeutic agent
against tau-maladies by reducing Rhes farnesylation and
increasing lysosomal activity and tau protein turnover [122].
These results support the therapeutic potential of targeting
key autophagic pathway components to treat tauopathies.
3.2.3. Synucleinopathies
Synucleinopathies (also called as α-synucleinopathies) are neu-
rodegenerative disorders characterized by abnormal deposits of
α-synuclein (α-Syn) aggregates in the brain [123]. Α-Syn aggre-
gates, which are mainly triggered by genetic mutations promot-
ing misfolding [124] or overexpression [125], disrupt the
dopamine transporter in dopaminergic neurons, leading to
synaptic dysfunction, cell death, and the progression of neuro-
degenerative diseases [126]. The three major types of synuclei-
nopathies are Parkinson’s disease (PD), dementia with Lewy
bodies (DLB), and multiple system atrophy (MSA) [123]. Since
homeostatic autophagy is impaired in synucleinopathies, thera-
peutic approaches to restore autophagy have emerged as clin-
ical strategies against them [127]. Studies have revealed the
amelioration of pathological synuclein features in vitro and
in vivo through pharmacological treatment with autophagy
modulators targeting mTOR such as rapamycin [128], and those
targeting AMPK such as metformin, 5-aminoimidazole-4-carbox-
amide ribonucleotide (AICAR) [129,130], resveratrol [131], and
trehalose [132,133]. This supports the benefit of autophagy path-
way regulation in synuclein-related disorders. Furthermore,
a recent combinatorial approach revealed significant additive
improvements to dopaminergic deficits through dual targeting
of AMPK and mTOR by trehalose and rapamycin, respectively
[134]. Several studies also pursued the pharmacological activa-
tion of autophagy through other pathways. Lithium, which is
known to inhibit both inositol monophosphatase (IMPase) and
glycogen synthase kinase-3β (GSK3β), induced autophagy by
reducing intracellular 1,4,5-inositol trisphosphate (IP3) levels
[135,136] and prevented α-Syn accumulation and neurotoxicity
[137]. Additionally, tyrosine kinase inhibitors, including nilotinib
[138], saracatinib (under clinical trials), and masitinib [139], also
promoted α-Syn clearance and prevented its accumulation
through activation of the AKT/mTOR pathway [140]. In conclu-
sion, numerous studies have reported new autophagic factors
and their modulators as relevant to α-synucleinopathies, provid-
ing beneficial tools for understanding the pathologic process
and offering opportunities for therapeutic development against
to α-Syn-related proteopathies.
3.2.4. Prion diseases
Prion diseases (also known as transmissible spongiform ence-
phalopathies (TSEs)) are a group of progressive neurodegen-
erative disorders caused by the conversion of normal prion
proteins (PrP
C
, cellular form, α-helical structure) to misfolded
prions (PrP
Sc
, scrapie form, β-pleated sheet structure) with
abnormal structure, which have the ability to transmit their
misfolded shape to normal prions [141]. Although the
mechanism of how prion aggregates kill nerve cells remains
unclear, it is reported that these abnormal prions are toxic to
neurons [142], leading to neuronal cell death that leaves
microscopic fluid-filled spaces behind and turns brains into
sponge-like shapes that causes progressive neuronal dete-
rioration [143]. The ‘gain of toxicity’ in prion proteins occurs
as sporadic (spontaneous), genetic (familial), and acquired
(infectious/transmissible), leading to human prion diseases
such as Kuru, Creutzfeldt–Jakob disease (CJD), Gerstmann-
Sträussler-Scheinker syndrome (GSS), and familial insomnia
(FI) [144]. Several studies have suggested a role for autophagy
in the pathogenesis of prion diseases. A recent study utilized
the mTOR regulators rapamycin and AR12 to examine their
combinatorial effect with cellulose ethers (CEs, prion conver-
sion inhibitor) in prion diseases [145]. Although there was
only a slight improvement without additive effects in vivo,
the trial itself is significant and implies the application of
mTOR-mediated autophagy in prion disease. In addition to
factors related to autophagosome formation, others con-
nected with autophagic turnover have received considerable
attention. Spermine facilitates autolysosome fusion and
autophagic degradation by inducing the acetylation of struc-
tural tubulin machinery [146]. Tacrolimus exhibited prion
degradation in an autophagy-dependent manner by enhan-
cing autophagy-related gene expression [147], which might
have relevance with autophagy master transcription fac-
tors [2].
3.2.5. Brief discussion about autophagy in proteopathies
Various proteopathies in which certain proteins become struc-
turally abnormal are introduced in Section 3.2. Autophagic
turnover becomes defective by protein aggregates which
aggravates these conditions, suggesting that reactivating
autophagy is required to ameliorate these diseases.
Targeting mTOR signaling has been implicated as an impor-
tant therapeutic approach for conventionally elevating autop-
hagy. However, due to its roles in cellular metabolism,
especially during neuronal development [148], chronic regula-
tion of mTOR should be carefully regulated. Therefore, instead
of targeting essential pathways related to broad energy meta-
bolism, other factors involved in autophagy should be regu-
lated such as autolysosome fusion and lysosomal activity.
3.3. Vascular diseases
Increasing studies have revealed that balanced autophagy
plays an essential role in mediating vascular physiological
processes, including: (1) as a protective cellular mechanism
in vessel walls; (2) in smooth muscle cell survival and prolif-
eration; (3) in the preservation of endothelial function; (4) in
the degradation of abnormal plaques in vessel walls; and (5) in
effective efferocytosis [149151]. Therefore, pharmacological
intervention with modulators that stimulate the pro-survival
effects of autophagy in the vasculature have been implicated
to target diverse vascular diseases.
EXPERT OPINION ON DRUG DISCOVERY 9
3.3.1. Atherosclerosis
Atherosclerosis is the blockage of blood vessels caused by
plaque deposits. It is classified as an inflammatory pathology
occurring in blood vessels with several risk factors including
sustained diabetes, aging, obesity, arterial hypertension, and
the accumulation of oxidized low-density lipoprotein (oxLDL)
cholesterol. Recent reports have focused on acquired defects
in autophagy that aggravate atherosclerosis. Lipophagy, or the
autophagy-mediated degradation of lipids, has been under
investigation as a targeting strategy for atherosclerosis.
Oxidative stress triggered by excess ROS generation is
known as a main cause of atherosclerosis. The abnormal
occurrence of such stress results in vascular cell-induced
damage and excessive immune responses via direct oxidation
of molecular components and the accumulation of cell death-
induced debris, which is responsible for the occurrence and
aggravation of atherosclerosis [152]. To target autophagy in
atherosclerosis, a modulator of antioxidant proteins can be
used to scavenge ROS in macrophages because uncontrolled
ROS production results in lipophagic flux inhibition [153].
Peroxiredoxin 1 (PRDX1) is an antioxidant enzyme that main-
tains lipophagic flux by reducing the produced ROS. Therefore,
the delivery of PRDX1-mimetics (such as ebselen and glio-
toxin) or modulator excavation could rescue impaired lipo-
phagy and maintain cholesterol efflux [154].
3.3.2. Intraocular neovascularization
Intraocular neovascularization is a cause of ocular vascular
diseases and affects transparency in the eye. It results in
hemorrhage, multiple disorders of the eye, and contributes
to loss of eyesight and other serious complications associated
with these diseases, such as branch retinal vein occlusion, age-
related macular degeneration, and diabetic retinopathy [155].
In recent studies, autophagy has been considered a plausible
strategy to target vascular endothelial growth factor (VEGF)
and neovascularization. VEGF and pigment epithelium-derived
factor (PEDF) are intimately related to intraocular neovascular-
ization in retinal pigment epithelium (RPE) cells. A baseline
level of autophagy under normal conditions changes along
with age and pathological processes that can accompany
hypoxia, oxidative stress, and inflammation. Accordingly,
autophagy can be activated in RPE cells [156]. Activation of
autophagy upon hypoxia exposure promotes VEGF expression
and inhibits PEDF expression in RPE cells [157]. When pre-
treated with 3-MA (a PI3-K inhibitor) or CQ (a lysosomal inhi-
bitor), inhibitors of autophagic initiation and lysosomal
degradation, respectively, VEGF expression was reduced and
the expression of PEDF was increased, resulting in neovascular
disruption by inhibiting cell proliferation, tube formation, and
vascular endothelial cell migration [157]. Although the
detailed mechanisms of how autophagy regulates angiogenic
factor expression are yet to be elucidated, autophagy inhibi-
tors could be developed as effective therapeutic agents for
retinal neovascularization.
3.3.3. Ischemia-reperfusion injury
Ischemia-reperfusion injury (IRI) in brain microvascular
endothelial cells is the main cause of blood brain barrier
(BBB) integrity disruption, which can result in fatal ischemic
stroke in patients [158]. Maintaining the integrity of the BBB is
pivotal for homeostasis by exchanging essential nutrients and
ions in the brain. Damage to the BBB is primarily caused by
stress-induced ROS generation, dysfunction of endothelial
tight junctions, and elevated permeability [159]. Therefore,
protecting the structural integrity of the BBB has been
a focus for targeting various central nervous system diseases.
Autophagy is highly induced in ischemic brain injury with
therapeutic activity, including protection of endothelial cells
against stress or stimuli and recovery from abnormal BBB
permeability. In recent studies, autophagy stimulated by
serum starvation or AKT-mTOR-S6 K modulators such as rapa-
mycin and torin-1 was found to protect endothelial brain
barrier integrity by scavenging ROS, inhibiting the dysfunction
of tight junction proteins such as claudin-5 (Cldn5) and zonula
occludens-1 (ZO-1), and reducing elevated permeability in the
BBB [158160].
3.3.4. Brief discussion about autophagy in vascular
diseases
There is mounting evidence demonstrating that autophagy
plays an essential role in vascular disease through the mod-
ulation of autophagy-specific genes. Yet, how to correctly
induce beneficial autophagy without triggering the detrimen-
tal effects of abnormal immune responses and cell death
remains a challenge. It is therefore vital to identify pharmaco-
logical agents that modulate autophagy with greater specifi-
city toward select molecular factors in order to be a favorable
target for therapeutic development in vascular diseases.
3.4. Other therapeutic applications in human diseases
Autophagy principally serves a protective role to maintain
homeostasis against diverse pathologies, even beyond the
diseases mentioned above. In this section, additional diseases
are discussed in which autophagy has critical relevance but is
less reported, including viral and microbial diseases and lyso-
somal storage diseases (LSDs). In this context, pharmacological
modulation of autophagy in combination with other medica-
tions could be a beneficial strategy for their treatment.
3.4.1. Viral infection & microbiome diseases
Pathogenic viruses cause infectious diseases. Viral infections are
caused by the invasion of viruses and virions (viral particles) into
the host body, where they enter into cells for propagation [161].
Many antiviral medications act directly on virus-specific compo-
nents; however, these result in unfortunate side effects such as
drug-resistance by promoting mutations within the viruses them-
selves [162]. Numerous reports have implicated host cell autop-
hagy in viral diseases by protecting the intracellular machinery
from hijacking by the virus or restricting their growth [163]
(Table 2). Therefore, pharmacological agents targeting autophagy
factors in the host could be beneficial for treating viral infections.
A recent study reported that CALCOCO2/NDP52 and SQSTM1/p62,
receptors of virophagy (viral xenophagy), positively and negatively
regulated the production of coxsackievirus B3 (CVB3), respectively,
supporting their use as host targets for antiviral treatment [164].
Another report revealed that combined treatment with oseltamivir
10 D. KIM ET AL.
(Tamiflu) and rapamycin protected mice against lethal pH1N1
infection by not only blocking viral replication, but also suppres-
sing mTOR-NLRP3-IL-1β axis-mediated immune damage and indu-
cing autophagy [165], portraying autophagy targeting as
a beneficial strategy for viral drug development. In contrast,
other studies highlight that several viruses exploit autophagy to
stimulate their replication (Table 2). For instance, it was established
that after viruses enter into cells, they require ‘replication compart-
ments’ (RCs) for genome replication and final virion assembly [166].
Among cellular vesicular structures, autophagosomes can serve as
viral RCs [167], which could explain the interplay between autop-
hagy and viral prosperity. Hence, blocking autophagosome forma-
tion by inhibiting the PI3-K pathway with 3-MA or wortmannin can
be a strategy to reduce viral infections such as adenovirus, Zika,
Dengue, and Influenza A virus [168171]. Furthermore, recent
studies have reported reduced viral infections upon treatment
with autophagy flux inhibitors that target lysosomal activity or
autolysosomal fusion, such as CQ, (H
+
ion quencher) [171], bafilo-
mycin A1 (lysosomal V-ATPase/ER-calcium ATPase inhibitor) [172],
and U18666A (Niemann-Pick type C protein inhibitor) [173].
Alternatively, microbiome-mediated diseases are caused by
gut dysbiosis in which microbial balances collapse, altering
symbiotic relationships and perturbing host health [184]. The
symbiotic relationship between the gut microbiome and its
host is regulated through crosstalk using signaling molecules
such as microbial products and metabolites. However, when
this relationship is disrupted, deregulated molecular signaling
may be harmful to the host and can cause diverse disorders
such as immune-mediated/autoimmune diseases, metabolic/
cardiovascular diseases, infectious diseases, neuropsychiatric
disorders, and even cancer [185]. Recent reports have revealed
that autophagy regulation in host intestinal or colonic epithe-
lial cells changes with microbiome composition, and vice versa
[186]. These results indicate that host autophagy has func-
tional relevance with microbiome-mediated disorders, paving
the way for autophagy modulation to be a potential clinical
strategy for treatment. Using pharmacological approaches,
a recent study revealed alpinetin as a new therapeutic agent
for protecting the intestinal barrier by promoting autophagy
through regulation of the AhR/suv39h1/TSC2/mTOR axis [187],
indicating new bioactive molecules for targeting autophagy
upstream of mTOR. In one study, JWH-133 rescued autophagy
dysregulation in cultured inflammatory bowel disease (IBD)
biopsies by acting as an agonist of cannabinoid receptor 2
(CB2) [188], ligands of which are reported to induce autop-
hagy in an AMPK-dependent manner [189]. These results cor-
relate with another study portraying that AMPK activity is
pivotal for inducing host autophagy and clearing invading
bacteria through xenophagy (antibacterial autophagy), sug-
gesting a new approach for antibacterial medication develop-
ment [190].
3.4.2. Lysosomal storage disorders
LSDs belong to a group of rare inherited metabolic diseases
characterized by defects in lysosomal function, which occur
through mutations in lysosomal genes encoding hydrolases,
Table 2. Viruses that regulate autophagy for their propagation.
Virus Virus classification
Autophagy
regulation Targeting factors Ref.
Coxsackievirus B3 (CVB3) (+) ssRNA Naked Evasion of autophagic
degradation
SQSTM1/p62 (through expression of viral
proteinase 2A that cleaves SQSTM1)
[164]
Herpes simplex virus 1 (HSV-1) dsDNA
Enveloped
Evasion of autophagic
degradation
Beclin1 and TANK-binding kinase1 (TBK1)
(through expression of ICP34.5 that
inhibits beclin1 and TBK1)
[174]
Murine γ-herpesvirus 68 (MHV68)
Kaposi’s sarcoma-associated herpesvirus (KSHV)
dsDNA
Enveloped
Evasion of autophagic
degradation
Beclin1 (through expression of viral
homologue of BCL-2 that inhibit beclin1)
Rubicon-Beclin1 interaction and VPS34
(through expression of K7 protein that
promotes the interaction and inhibits
VPS34)
[175,176]
Human immunodeficiency virus type 1 (HIV-1) Dimer RNA
Enveloped
Evasion of autophagic
degradation
Beclin 1 and TFEB (through expression of
Nef that inhibit beclin1 and beclin1
mediated TFEB dephosphorylation)
[177]
Middle East respiratory syndrome coronavirus (MERS-CoV) (+) ssRNA
Enveloped
Evasion of autophagic
degradation
Akt1-SKP2-Beclin1 (through enhancing Akt1
phosphorylation, resulting in beclin1
degradation)
[178]
Sindbis virus (SINV) ssRNA
Enveloped
Vulnerable to autophagic
degradation
p62 (viral capsid protein binds to p62
resulting in virophagy)
[179]
Hepatitis C virus (HCV) ssRNA
Enveloped
Vulnerable to autophagic
degradation
SHISA5 (viral NS5A protein undergoes
transport to autophagosome by SHISA5)
[180]
Enhancing autophagy for
viral replication
Beclin1 (through expression of p7 ion
channel protein that directly binds to
beclin1)
[181]
Adenovirus dsDNA
Naked
Enhancing autophagy for
viral replication
Through induction of Atg12-Atg5 complex
formation
[168]
Zika virus (ZIKV) (+) ssRNA
Enveloped
Enhancing autophagy for
viral replication
Akt-mTOR pathway (through perturbing the
pathway by viral NS4A and NS4B)
[171]
Dengue virus (DV) (+) ssRNA
Enveloped
Enhancing autophagy for
viral replication
LC3 (through inducing LC3 cleavage by viral
NS4A to form autophagosome)
[169]
Influenza A virus (IAV) (-) ssRNA
Enveloped
Enhancing autophagy for
viral replication
Akt-TSC2-mTOR pathway or LC3 (through
modulation of the pathway or
relocalization of LC3 by viral M2 protein
[182]
Vesicular stomatitis virus (VSV) (-) ssRNA
Enveloped
Enhancing autophagy for
viral replication
Dependent on NLRX1
[183]
EXPERT OPINION ON DRUG DISCOVERY 11
enzyme cofactors, membrane proteins, and transporters [191].
The progressive accumulation of macromolecules within lyso-
somes due to defective degradation capacity leads to cellular
damage and affects various organs throughout the body [192].
Several studies have revealed possible therapeutic interven-
tions in some LSDs through pharmacological treatment with
autophagy inducers. At first, the mTOR inhibitor rapamycin
exhibited beneficial effects in a Gaucher disease (GD) droso-
phila model [193], and increased cellular viability in Niemann-
Pick disease type C1 disease model cells deficient in the NPC
intracellular cholesterol transporter 1 (NPC1) gene [194].
However, another study reported contradictory results about
the effects of rapamycin on NPC mice, in that lengthening life
span by rapamycin was dependent on the genetic background
[195]. Due to these controversial mTOR-targeting results,
several studies examined mTOR-independent autophagy indu-
cers to avoid perturbing the critical functions of mTOR [196].
The IMPase inhibitors lithium and L690,330 rescued cerebellar
cells from juvenile neuronal ceroid lipofuscinosis (JNCL)
pathology, in which the lysosomal transmembrane protein
CNL3 gene is defective, through reducing intracellular IP3
levels and enhancing autophagy [135,136]. One of the LSDs,
Vici syndrome, is caused by recessive mutations in the EPG5
gene, which encodes a crucial protein facilitating SNARE
assembly in the autolysosome fusion process. A recent report
revealed that under loss of EPG5 function, SNAP25 interferes
with the SNARE complex instead of SNAP29, resulting in
autophagy dysregulation. This suggests that SNAP25 could
be a potential therapeutic target for restoring autophagy in
LSDs [197].
Table 3. Autophagy-targeting therapeutic agents developed into pharmaceuticals/dietary supplements or under clinical trials.
Application state Agents Structure Targeting diseases Targeting factors/pathways Ref.
FDA approved Rapamycin derivatives Cancer mTOR
(Autophagy activation)
[67,68]
Nilotinib Cancer AMPK
(Autophagy activation)
[57]
Bortezomib Cancer 38-MAPK-JUNK pathway
(Autophagy activation)
[46]
Idelalisib Cancer PI3-K
(Autophagy inhibition)
[61]
Under clinical trial Chloroquine
/Hydroxy-chloroquine
Cancer
Viral infection
Lysosomal hydrogen
(Autophagy inhibition)
[78]
Saracatinib (AZD0530) Neurodegenerative disorders
- Alzheimer’s Disease
- Parkinson’s Disease
Tyrosine kinase Fyn
(Autophagy activation)
[139]
(Continued )
12 D. KIM ET AL.
4. Conclusion
Through a comprehensive understanding of autophagic
machinery, its fundamental importance to cellular health
emerges as an auspicious target for curing various phenotypic
diseases in humans. Pharmacological approaches have been
considered a major priority because of advantages such as
brief pharmacokinetics, easy treatment and delivery into
cells, and minimized immune responses. The development
and application of pharmacological agents are therefore pivo-
tal to providing new insight and exploration into autophagy-
related diseases and their underlying mechanisms. Careful
consideration is also required during this process, since autop-
hagy activation or suppression can yield defensive or destruc-
tive outcomes according to disease type, even at different
stages of the same disease. For instance, autophagy may be
utilized as a resistance mechanism for chemotherapy-treated
tumor cell survival, or alternatively, may induce unintended
death in important cells required for pathological recovery,
such as in proteopathies, vasculature abnormalities, metabolic
disorders, and aging. We have described both conventional
and recent concepts connecting specific autophagic path-
ways, targeting strategies using bioactive compounds, and
autophagy-related diseases. Collectively, the considerations
and recent advances highlighted in this review contributes
to a comprehensive understanding of autophagy signaling
control in human diseases.
5. Expert opinion
The self-catabolic intracellular process autophagy occurs in
organisms throughout their lifetime, supporting its critical
role in organismal health across life stages. Because of the
detrimental influence of dysfunctional cells to an organism
and their etiology in numerous diseases, maintaining cellular
quality control by recycling components through autophagy is
essential to defend against health decline. There is a history of
successful cases of small molecule development targeting
autophagy-related diseases (Table 3).
Because variations in signaling pathways or protein expres-
sion levels specific to each target diseases could change drug
efficacy and cause side effects, understanding autophagic
modulators and their molecular interactors in the cell are
critical components for therapeutic success. To utilize autop-
hagy-modulating compounds without serious side effects,
identification of their target proteins should be a focus during
the process. By clarifying these specific interactions, research-
ers can more comprehensively characterize a compound’s
effect and efficiently define their ideal spatial and temporal
Table 3. (Continued).
Application state Agents Structure Targeting diseases Targeting factors/pathways Ref.
Dietary supplement Resveratrol Neuronal health AMPK pathway
SIRT1-TFEB pathway
(Autophagy activation)
[99,104]
Trehalose Neuronal health Autophagy master gene (TFEB)
(Autophagy activation)
[118]
Fisetin Neuronal health Autophagy master gene (NRF2)
(Autophagy activation)
[119]
Sulforaphane Neuronal health Autophagy master gene (NRF2)
(Autophagy activation)
[121]
Lithium Neuronal health IMPase
GSK3β
(Autophagy activation)
[135,136]
Ginsenoside Rg3 Neuronal health PI3-K pathway
(Autophagy activation)
[198]
EXPERT OPINION ON DRUG DISCOVERY 13
chemotherapeutic characteristics. As outlined in this review,
targeting strategies for autophagic pathways across several
diseases show strong potential as therapeutic strategies.
Several agents are introduced that have been developed
into clinical drugs, are under clinical trials, or are being used
as dietary supplements. For instance, rapamycin has been
developed into clinical anticancer medications, raising its
value in the mTOR inhibitor market which is expected to
significantly grow during 2019–2023 with CAGR over 3%,
size by 455 USD.85 million (Technavio, 2019). Other drugs
such as bortezomib, nilotinib (AMPK/mTOR/MAPK pathway
modulators), and idelalisib (PI3-K inhibitor) are also FDA
approved as anticancer therapeutics, supporting their efficacy
and safety. Other autophagy regulators like chloroquine, sar-
acatinib, and latrepirdine are also undergoing clinical trials,
and natural small molecules that induce autophagy have
been developed into dietary supplements for neuronal health
(resveratrol, trehalose, fisetin, sulforaphane, lithium, and gin-
senoside Rg3). These applications verify the utility and value of
autophagy-targeting strategies for drug development, with
further promise in treating currently undruggable pathologies
such as proteopathies. Instead of the current drugs on the
market for Alzheimer’s disease that merely delay patient
pathologies without removing the pathologic cause, autop-
hagy-targeting strategies offer promising therapeutic options
and new paradigms for future treatments.
Funding
This work was supported by grants from the National Research
Foundation of Korea, funded by the Korean government (MSIP;
2015K1A1A2028365, 2015M3A9B6027818, 2016K2A9A1A03904900,
2018M3A9C4076477), the Brain Korea 21Plus Project in the Republic of
Korea and ICONS (the Institute of Convergence Science), Yonsei University.
Declaration of interest
The authors have no other relevant affiliations or financial involvement
with any organization or entity with a financial interest in or financial
conflict with the subject matter or materials discussed in the manuscript
apart from those disclosed.
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
ORCID
Dasol Kim http://orcid.org/0000-0003-1314-2940
Hui-Yun Hwang http://orcid.org/0000-0001-9223-1428
Ho Jeong Kwon http://orcid.org/0000-0002-6919-833X
References
Papers of special note have been highlighted as either of interest (•) or of
considerable interest (••) to readers.
1. Kliosnky D. Guidelines for the use and interpretation of assays for
monitoring autophagy (3rd edition) (vol 12, pg 1, 2015).
Autophagy. 2016;12(2):443.
•• Useful review on investigating autophagy mechanisms and the
effective methods for the study of autophagy-modulating
strategies.
2. Kim D, Hwang HY, Kim JY, et al. FK506, an immunosuppressive
drug, induces autophagy by binding to the v-atpase catalytic sub-
unit a in neuronal cells. J Proteome Res. 2017 Jan 6;16(1):55–64.
3. Hwang HY, Cho SM, Kwon HJ. Approaches for discovering novel
bioactive small molecules targeting autophagy. Expert Opin Drug
Discov. 2017 Sep;12(9):909–923.
•• Useful review on discovery of autophagy modulators with
their therapeutic protein targets and related biological path-
ways with conventional and recent trends.
4. Thorburn A. Autophagy and disease. J Biol Chem. 2018 Apr 13;293
(15):5425–5430.
5. Jiang X, Overholtzer M, Thompson CB. Autophagy in cellular meta-
bolism and cancer. J Clin Invest. 2015 Jan;125(1):47–54.
6. Lunemann JD, Schmidt J, Schmid D, et al. Beta-amyloid is
a substrate of autophagy in sporadic inclusion body myositis.
Ann Neurol. 2007 May;61(5):476–483. .
7. Santos RX, Cardoso S, Correia S, et al. Targeting autophagy in the
brain: a promising approach? Cent Nerv Syst Agents Med Chem.
2010 Jun 1;10(2):158–168.
8. Towers CG, Thorburn A. Therapeutic targeting of autophagy.
EBioMedicine. 2016 Dec;14:15–23.
9. Kaur J, Debnath J. Autophagy at the crossroads of catabolism and
anabolism. Nat Rev Mol Cell Bio. 2015 Aug;16(8):461–472.
10. Yu L, Chen Y, Tooze SA. Autophagy pathway: cellular and molecular
mechanisms. Autophagy. 2018;14(2):207–215.
11. Galluzzi L, Green DR. Autophagy-independent functions of the
autophagy machinery. Cell. 2019 Jun 13;177(7):1682–1699.
12. Reggiori F, Ungermann C. Autophagosome maturation and fusion.
J Mol Biol. 2017 Feb 17;429(4):486–496.
13. Feng Q, Luo Y, Zhang XN, et al. MAPT/Tau accumulation represses
autophagy flux by disrupting IST1-regulated ESCRT-III complex
formation: a vicious cycle in Alzheimer neurodegeneration.
Autophagy. 2019;28:1–18.
14. Schmidt O, Teis D. The ESCRT machinery. Curr Biol. 2012 Feb 21;22
(4):R116–20.
15. Schaeffer V, Lavenir I, Ozcelik S, et al. Stimulation of autophagy
reduces neurodegeneration in a mouse model of human
tauopathy. Brain. 2012 Jul;135(Pt 7):2169–2177.
16. Cang C, Aranda K, Seo YJ, et al. TMEM175 Is an organelle K(+)
channel regulating lysosomal function. Cell. 2015 Aug 27;162
(5):1101–1112.
17. Zhu MX. A well-known potassium channel plays a critical role in
lysosomes. J Cell Biol. 2017 Jun 5;216(6):1513–1515.
18. Kolter T, Sandhoff K. Principles of lysosomal membrane digestion:
stimulation of sphingolipid degradation by sphingolipid activator
proteins and anionic lysosomal lipids. Annu Rev Cell Dev Biol.
2005;21(1):81–103.
19. Baek SH, Kim KI. Epigenetic control of autophagy: nuclear events
gain more attention. Mol Cell. 2017Mar2;65(5):781–785.
•• This article demonstrates the function of MiT/TFE family pro-
teins in autophagy pathway as the major master genes playing
autophagosomes and lysosome biogenesis.
20. Yang M, Liu E, Tang L, et al. Emerging roles and regulation of MiT/TFE
transcriptional factors. Cell Commun Signal. 2018 Jun 15;16(1):31.
21. Zhao GQ, Zhao Q, Zhou X, et al. TFEC, a basic helix-loop-helix
protein, forms heterodimers with TFE3 and inhibits
TFE3-dependent transcription activation. Mol Cell Biol. 1993
Aug;13(8):4505–4512.
22. Moller K, Sigurbjornsdottir S, Arnthorsson AO, et al. MITF has
a central role in regulating starvation-induced autophagy in
melanoma. Sci Rep. 2019 Jan 31;9(1):1055.
23. Pastore N, Brady OA, Diab HI, et al. TFEB and TFE3 cooperate in the
regulation of the innate immune response in activated
macrophages. Autophagy. 2016;12(8):1240–1258. .
24. Martina JA, Puertollano R. Rag GTPases mediate amino
acid-dependent recruitment of TFEB and MITF to lysosomes.
J Cell Biol. 2013 Feb 18;200(4):475–491.
14 D. KIM ET AL.
25. Lim H, Lim YM, Kim KH, et al. A novel autophagy enhancer as
a therapeutic agent against metabolic syndrome and diabetes.
Nat Commun. 2018 Apr 12;9(1):1438.
26. Pajares M, Jimenez-Moreno N, Garcia-Yague AJ, et al. Transcription
factor NFE2L2/NRF2 is a regulator of macroautophagy genes.
Autophagy. 2016 Oct 2;12(10):1902–1916.
•• This article demonstrates the function ofThe nuclear factor
erythroid-derived 2-like 2 (NFE2L2/NRF2) proteins in autop-
hagy pathway as the master genes and its target proteins.
27. Pajares M, Rojo AI, Arias E, et al. Transcription factor NFE2L2/NRF2
modulates chaperone-mediated autophagy through the regulation
of LAMP2A. Autophagy. 2018;14(8):1310–1322.
28. McMahon M, Itoh K, Yamamoto M, et al. Keap1-dependent protea-
somal degradation of transcription factor Nrf2 contributes to the
negative regulation of antioxidant response element-driven gene
expression. J Biol Chem. 2003 Jun 13;278(24):21592–21600.
29. Dodson M, Redmann M, Rajasekaran NS, et al. KEAP1-NRF2 signal-
ling and autophagy in protection against oxidative and reductive
proteotoxicity. Biochem J. 2015 Aug 1;469(3):347–355.
30. Salih DA, Brunet A. FoxO transcription factors in the maintenance
of cellular homeostasis during aging. Curr Opin Cell Biol. 2008
Apr;20(2):126–136.
•• This article demonstrates the diverse functional roles of
Forkhead box O (FoxO) transcriptional family proteins.
31. Wang XW, Hu SJ, Liu L. Phosphorylation and acetylation modifica-
tions of FOXO3a: independently or synergistically? Oncol Lett. 2017
May;13(5):2867–2872.
32. Jacobs FM, van der Heide LP, Wijchers PJ, et al. FoxO6, a novel
member of the FoxO class of transcription factors with distinct
shuttling dynamics. J Biol Chem. 2003 Sep 19;278(38):35959–35967.
33. Zhao J, Brault JJ, Schild A, et al. FoxO3 coordinately activates
protein degradation by the autophagic/lysosomal and proteasomal
pathways in atrophying muscle cells. Cell Metab. 2007 Dec;6
(6):472–483. .
34. Liu HY, Han J, Cao SY, et al. Hepatic autophagy is suppressed in the
presence of insulin resistance and hyperinsulinemia: inhibition of
FoxO1-dependent expression of key autophagy genes by insulin.
J Biol Chem. 2009 Nov 6;284(45):31484–31492.
35. O’Neill BT, Lee KY, Klaus K, et al. Insulin and IGF-1 receptors
regulate FoxO-mediated signaling in muscle proteostasis. J Clin
Invest. 2016 Sep 1;126(9):3433–3446.
36. Wedel A, Ziegler-Heitbrock HW. The C/EBP family of transcription
factors. Immunobiology. 1995 Jul;193(2–4):171–185.
37. Johnson PF. Molecular stop signs: regulation of cell-cycle arrest by C/
EBP transcription factors. J Cell Sci. 2005 Jun 15;118(Pt 12):2545–2555.
38. Yuk JM, Shin DM, Lee HM, et al. Vitamin D3 induces autophagy in
human monocytes/macrophages via cathelicidin. Cell Host
Microbe. 2009 Sep 17;6(3):231–243.
39. Guo L, Huang JX, Liu Y, et al. Transactivation of Atg4b by C/
EBPbeta promotes autophagy to facilitate adipogenesis. Mol Cell
Biol. 2013 Aug;33(16):3180–3190. .
40. Tsai HH, Lai HY, Chen YC, et al. Metformin promotes apoptosis in
hepatocellular carcinoma through the CEBPD-induced autophagy
pathway. Oncotarget. 2017 Feb 21;8(8):13832–13845.
•• This article demonstrates the function of CCAAT/enhancer-
binding protein D (CEBPD) transcriptional protein using its
modulator in autophagy pathway as one of the C/EBP family
members and its target proteins.
41. B’Chir W, Maurin AC, Carraro V, et al. The eIF2alpha/ATF4 pathway
is essential for stress-induced autophagy gene expression. Nucleic
Acids Res. 2013 Sep;41(16):7683–7699. .
42. Patient RK, McGhee JD. The GATA family (vertebrates and
invertebrates). Curr Opin Genet Dev. 2002 Aug;12(4):416–422.
43. Gordy C, He YW. The crosstalk between autophagy and apoptosis:
where does this lead? Protein Cell. 2012 Jan;3(1):17–27.
44. Kang YA, Sanalkumar R, O’Geen H, et al. Autophagy driven by a master
regulator of hematopoiesis. Mol Cell Biol. 2012 Jan;32(1):226–239.
•• This article demonstrates the functional roles of GATA1 tran-
scriptional protein in autophagy pathway as one of the GATA
family members and its target proteins.
45. Cho YS, Yen CN, Shim JS, et al. Antidepressant indatraline induces
autophagy and inhibits restenosis via suppression of mTOR/S6
kinase signaling pathway. Sci Rep. 2016 Oct 3;6(1):34655.
46. Wilde L, Tanson K, Curry J, et al. Autophagy in cancer: a complex
relationship. Biochem J. 2018Jun11;475(11):1939–1954.
•• Description on what considerations of the application of
autophagy machinery as targeting strategy in cancer.
47. Rabiee S, Tavakol S, Barati M, et al. Autophagic, apoptotic, and
necrotic cancer cell fates triggered by acidic pH microenvironment.
J Cell Physiol. 2019 Jul;234(7):12061–12069.
48. Yang ZJ, Chee CE, Huang S, et al. The role of autophagy in cancer:
therapeutic implications. Mol Cancer Ther. 2011 Sep;10
(9):1533–1541.
49. Poillet-Perez L, Despouy G, Delage-Mourroux R, et al. Interplay
between ROS and autophagy in cancer cells, from tumor initiation
to cancer therapy. Redox Biol. 2015;4:184–192.
50. Kang MR, Kim MS, Oh JE, et al. Frameshift mutations of
autophagy-related genes ATG2B, ATG5, ATG9B and ATG12 in gas-
tric and colorectal cancers with microsatellite instability. J Pathol.
2009 Apr;217(5):702–706. .
51. Guo S, Long M, Li X, et al. Curcumin activates autophagy and
attenuates oxidative damage in EA.hy926 cells via the Akt/mTOR
pathway. Mol Med Rep. 2016 Mar;13(3):2187–2193.
52. Butler DE, Marlein C, Walker HF, et al. Inhibition of the PI3K/AKT/
mTOR pathway activates autophagy and compensatory Ras/Raf/
MEK/ERK signalling in prostate cancer. Oncotarget. 2017 Aug 22;8
(34):56698–56713.
53. Simioni C, Cani A, Martelli AM, et al. Activity of the novel mTOR
inhibitor Torin-2 in B-precursor acute lymphoblastic leukemia and
its therapeutic potential to prevent Akt reactivation. Oncotarget.
2014 Oct 30;5(20):10034–10047.
54. Jung CH, Ro SH, Cao J, et al. mTOR regulation of autophagy. FEBS
Lett. 2010 Apr 2;584(7):1287–1295.
55. Din FV, Valanciute A, Houde VP, et al. Aspirin inhibits mTOR signaling,
activates AMP-activated protein kinase, and induces autophagy in
colorectal cancer cells. Gastroenterology. 2012 Jun;142(7):1504–15 e3.
•• Representative case study of autophagy inducer, aspirin as
a tumor suppresive autophagy mechanism. Cyclooxygenase
(COX) inhibitor, aspirin also induces autophagy in colorectal
cancer cells by activating AMP-activated protein kinase
(AMPK).
56. Huang WW, Tsai SC, Peng SF, et al. Kaempferol induces autophagy
through AMPK and AKT signaling molecules and causes G2/M
arrest via downregulation of CDK1/cyclin B in SK-HEP-1 human
hepatic cancer cells. Int J Oncol. 2013 Jun;42(6):2069–2077. .
57. Yu HC, Lin CS, Tai WT, et al. Nilotinib induces autophagy in hepa-
tocellular carcinoma through AMPK activation. J Biol Chem. 2013
Jun 21;288(25):18249–18259.
58. Rabiee Motmaen S, Tavakol S, Joghataei MT, et al. Acidic pH
derived from cancer cells as a double-edged knife modulates
wound healing through DNA repair genes and autophagy.
Int Wound J. 2020 Feb;17(1):137–148.
59. Masuda GO, Yashiro M, Kitayama K, et al. Clinicopathological cor-
relations of autophagy-related proteins LC3, beclin 1 and p62 in
gastric cancer. Anticancer Res. 2016 Jan;36(1):129–136.
60. Wu L, Yan B. Discovery of small molecules that target autophagy
for cancer treatment. Curr Med Chem. 2011;18(12):1866–1873.
61. Ikeda H, Hideshima T, Fulciniti M, et al. PI3K/p110 delta is a novel
therapeutic target in multiple myeloma. Blood. 2010 Sep 2;116
(9):1460–1468.
62. Seglen PO, Gordon PB. 3-methyladenine: specific inhibitor of
autophagic/lysosomal protein degradation in isolated rat
hepatocytes. Proc Natl Acad Sci U S A. 1982 Mar;79(6):1889–1892.
63. Abliz A, Deng W, Sun R, et al. Wortmannin, PI3K/Akt signaling pathway
inhibitor, attenuates thyroid injury associated with severe acute pan-
creatitis in rats. Int J Clin Exp Pathol. 2015;8(11):13821–13833.
64. Cerioni L, Palomba L, Cantoni O. The Raf/MEK inhibitor PD98059
enhances ERK1/2 phosphorylation mediated by peroxynitrite via
enforced mitochondrial formation of reactive oxygen species. FEBS
Lett. 2003 Jul 17;547(1–3):92–96.
EXPERT OPINION ON DRUG DISCOVERY 15
65. Liang XH, Jackson S, Seaman M, et al. Induction of autophagy and
inhibition of tumorigenesis by beclin 1. Nature. 1999 Dec 9;402
(6762):672–676.
66. Liang C, Feng P, Ku B, et al. UVRAG: a new player in autophagy and
tumor cell growth. Autophagy. 2007 Jan-Feb;3(1):69–71.
67. Decaens T, Luciani A, Itti E, et al. Phase II study of sirolimus in
treatment-naive patients with advanced hepatocellular carcinoma.
Dig Liver Dis. 2012 Jul;44(7):610–616. .
68. Huynh H, Chow KH, Soo KC, et al. RAD001 (everolimus) inhibits
tumour growth in xenograft models of human hepatocellular
carcinoma. J Cell Mol Med. 2009 Jul;13(7):1371–1380. .
69. Zhu AX, Kudo M, Assenat E, et al. Effect of everolimus on survival in
advanced hepatocellular carcinoma after failure of sorafenib: the
EVOLVE-1 randomized clinical trial. Jama. 2014 Jul 2;312(1):57–67.
70. Thomas HE, Mercer CA, Carnevalli LS, et al. mTOR inhibitors synergize on
regression, reversal of gene expression, and autophagy in hepatocellular
carcinoma. Sci Transl Med. 2012 Jun 20;4(139):139ra84.
71. Fu Y, Huang Z, Hong L, et al. Targeting ATG4 in cancer therapy.
Cancers (Basel). 2019 May 10;11(5):649.
72. Singh K, Sharma A, Mir MC, et al. Autophagic flux determines cell
death and survival in response to Apo2L/TRAIL (dulanermin). Mol
Cancer. 2014 Mar 23;13(1):70.
73. Gao M, Monian P, Pan Q, et al. Ferroptosis is an autophagic cell
death process. Cell Res. 2016 Sep;26(9):1021–1032.
74. Ashrafizadeh M, Mohammadinejad R, Tavakol S, et al. Autophagy,
anoikis, ferroptosis, necroptosis, and endoplasmic reticulum stress:
potential applications in melanoma therapy. J Cell Physiol. 2019
Nov;234(11):19471–19479.
75. Tam SY, Wu VW, Law HK. Influence of autophagy on the efficacy of
radiotherapy. Radiat Oncol. 2017 Mar 21;12(1):57.
76. Xie R, Nguyen S, McKeehan WL, et al. Acetylated microtubules are
required for fusion of autophagosomes with lysosomes. BMC Cell
Biol. 2010 Nov 22;11(1):89.
77. Fu R, Deng Q, Zhang H, et al. A novel autophagy inhibitor berbamine
blocks SNARE-mediated autophagosome-lysosome fusion through
upregulation of BNIP3. Cell Death Dis. 2018 Feb 14;9(2):243.
•• Representative case study of autophagy inhibitor, berbamine
as a tumorigenic autophagy mechanism. Protein-protein inter-
action (PPI) inhibitor of soluble N-ethylmaleimide-sensitive
factor attachment protein receptor (SNARE) complex forma-
tion, berbamine also inhibits autophagic flux in cancer by
inhibiting interactions among BCL2 interacting protein 3
(BNIP3) and Synaptosomal-associated protein
78. Palmeira Dos Santos C, Pereira GJ, Barbosa CM, et al. Comparative
study of autophagy inhibition by 3MA and CQ on cytarabinein-
duced death of leukaemia cells. J Cancer Res Clin Oncol. 2014
Jun;140(6):909–920.
79. Jung M, Lee J, Seo HY, et al. Cathepsin inhibition-induced lysoso-
mal dysfunction enhances pancreatic beta-cell apoptosis in high
glucose. PLoS One. 2015;10(1):e0116972.
80. Kirkegaard T, Roth AG, Petersen NH, et al. Hsp70 stabilizes lyso-
somes and reverts niemann-pick disease-associated lysosomal
pathology. Nature. 2010 Jan 28;463(7280):549–553.
81. Hwang HY, Cho YS, Kim JY, et al. Autophagic inhibition via lysoso-
mal integrity dysfunction leads to antitumor activity in glioma
treatment. Cancers (Basel). 2020 Feb 27;12(3):543.
82. Hsin IL, Sheu GT, Jan MS, et al. Inhibition of lysosome degradation
on autophagosome formation and responses to GMI, an immuno-
modulatory protein from Ganoderma microsporum. Br
J Pharmacol. 2012 Nov;167(6):1287–1300. .
83. Chen H, Li L, Hu J, et al. UBL4A inhibits autophagy-mediated
proliferation and metastasis of pancreatic ductal adenocarcinoma
via targeting LAMP1. J Exp Clin Cancer Res. 2019 Jul 9;38(1):297.
84. Su M, Mei Y, Sinha S. Role of the crosstalk between autophagy and
apoptosis in cancer. J Oncol. 2013;2013:102735.
85. Marinkovic M, Sprung M, Buljubasic M, et al. Autophagy modula-
tion in cancer: current knowledge on action and therapy. Oxid Med
Cell Longev. 2018;2018:8023821.
86. Carrell RW, Lomas DA. Conformational disease. Lancet. 1997 Jul
12;350(9071):134–138.
87. Thellung S, Corsaro A, Nizzari M, et al. Autophagy activator drugs:
a new opportunity in neuroprotection from misfolded protein
toxicity. Int J Mol Sci. 2019 Feb 19;20(4):901.
88. Regland B, McCaddon A, Garner B. Alzheimer’s amyloidopathy: an
alternative aspect. J Alzheimers Dis. 2019;68(2):483–488.
89. Allali G, Kern I, Laidet M, et al. Parkinsonism is a phenotypical
signature of amyloidopathy in patients with gait disorders.
J Alzheimers Dis. 2018;63(4):1373–1381.
90. Shimada H, Shinotoh H, Hirano S, et al. beta-amyloid in lewy body
disease is related to alzheimer’s disease-like atrophy. Mov Disord.
2013 Feb;28(2):169–175. .
91. Pajak B, Kania E, Orzechowski A. Killing me softly: connotations to
unfolded protein response and oxidative stress in alzheimer’s dis-
ease. Oxid Med Cell Longev. 2016;2016:1805304.
92. Murphy MP, LeVine H, Lovell MA. Alzheimer’s disease and the
amyloid-beta peptide. J Alzheimers Dis. 2010;19(1):311–323.
93. Nilsson P, Saido TC. Dual roles for autophagy: degradation and
secretion of alzheimer’s disease abeta peptide. Bioessays. 2014
Jun;36(6):570–578.
94. Wani A, Gupta M, Ahmad M, et al. Alborixin clears amyloid-beta by
inducing autophagy through PTEN-mediated inhibition of the AKT
pathway. Autophagy. 2019 Oct;15(10):1810–1828.
•• Representative case study of autophagy inducer, alborixin as
amyloidosis inhibitor. Alborixin also induces autophagy lead-
ing to clearance of amyloid β (Aβ) plaques in microglia and
neuronal cells by targeting PI3K/Akt signaling axis.
95. Spilman P, Podlutskaya N, Hart MJ, et al. Inhibition of mTOR by
rapamycin abolishes cognitive deficits and reduces amyloid-beta
levels in a mouse model of alzheimer’s disease. PLoS One. 2010 Apr
1;5(4):e9979.
96. Yang Y, Chen S, Zhang J, et al. Stimulation of autophagy prevents
amyloid-beta peptide-induced neuritic degeneration in PC12 cells.
J Alzheimers Dis. 2014;40(4):929–939. .
97. Guo J, Chang L, Zhang X, et al. Ginsenoside compound K promotes
beta-amyloid peptide clearance in primary astrocytes via autop-
hagy enhancement. Exp Ther Med. 2014 Oct;8(4):1271–1274.
98. Menon MB, Dhamija S. Beclin 1 phosphorylation - at the center of
autophagy regulation. Front Cell Dev Biol. 2018;6:137.
99. Vingtdeux V, Giliberto L, Zhao H, et al. AMP-activated protein
kinase signaling activation by resveratrol modulates amyloid-beta
peptide metabolism. J Biol Chem. 2010 Mar 19;285(12):9100–9113.
100. Vingtdeux V, Chandakkar P, Zhao HT, et al. Novel synthetic
small-molecule activators of AMPK as enhancers of autophagy
and amyloid-beta peptide degradation. Faseb J. 2011 Jan;25
(1):219–231.
101. Fan Y, Wang N, Rocchi A, et al. Identification of natural products
with neuronal and metabolic benefits through autophagy
induction. Autophagy. 2017 Jan 2;13(1):41–56.
102. Markowicz-Piasecka M, Sikora J, Szydlowska A, et al. Metformin -
a future therapy for neurodegenerative diseases: theme: drug dis-
covery, development and delivery in alzheimer’s disease guest
editor: davide brambilla. Pharm Res. 2017 Dec;34(12):2614–2627.
103. Son SM, Shin HJ, Byun J, et al. Metformin facilitates amyloid-beta
generation by beta- and gamma-secretases via autophagy
activation. J Alzheimers Dis. 2016;51(4):1197–1208. .
104. Bao J, Zheng L, Zhang Q, et al. Deacetylation of TFEB promotes
fibrillar abeta degradation by upregulating lysosomal biogenesis in
microglia. Protein Cell. 2016 Jun;7(6):417–433. .
105. Hong HS, Hwang JY, Son SM, et al. FK506 reduces amyloid plaque
burden and induces MMP-9 in AbetaPP/PS1 double transgenic
mice. J Alzheimers Dis. 2010;22(1):97–105.
106. Kneynsberg A, Combs B, Christensen K, et al. Axonal degeneration
in tauopathies: disease relevance and underlying mechanisms.
Front Neurosci. 2017;11:572.
107. Dujardin S, Begard S, Caillierez R, et al. Different tau species lead to
heterogeneous tau pathology propagation and misfolding. Acta
Neuropathol Commun. 2018Nov29;6:1–12.
108. Stoothoff WH, Johnson GV. Tau phosphorylation: physiological and
pathological consequences. Biochim Biophys Acta. 2005 Jan 3;1739
(2–3):280–297.
16 D. KIM ET AL.
109. He Z, Guo JL, McBride JD, et al. Amyloid-beta plaques enhance
alzheimer’s brain tau-seeded pathologies by facilitating neuritic
plaque tau aggregation. Nat Med. 2018 Jan;24(1):29–38. .
110. Piras A, Collin L, Gruninger F, et al. Autophagic and lysosomal
defects in human tauopathies: analysis of post-mortem brain
from patients with familial alzheimer disease, corticobasal degen-
eration and progressive supranuclear palsy. Acta Neuropathol
Commun. 2016 Mar 2;4(1):22.
111. Caccamo A, Magri A, Medina DX, et al. mTOR regulates tau phos-
phorylation and degradation: implications for Alzheimer’s disease
and other tauopathies. Aging Cell. 2013 Jun;12(3):370–380. .
112. Zhang ZH, Wu QY, Zheng R, et al. Selenomethionine mitigates
cognitive decline by targeting both tau hyperphosphorylation
and autophagic clearance in an alzheimer’s disease mouse model.
J Neurosci. 2017 Mar 1;37(9):2449–2462.
113. Salama M, Elhussiny M, Magdy A, et al. Dual mTORC1/mTORC2
blocker as a possible therapy for tauopathy in cellular model.
Metab Brain Dis. 2018 Apr;33(2):583–587. .
114. Chen JL, Luo C, Pu D, et al. Metformin attenuates diabetes-induced
tau hyperphosphorylation in vitro and in vivo by enhancing autop-
hagic clearance. Exp Neurol. 2019 Jan;311:44–56.
115. Barini E, Antico O, Zhao Y, et al. Metformin promotes tau aggrega-
tion and exacerbates abnormal behavior in a mouse model of
tauopathy. Mol Neurodegener. 2016 Feb 9;11(1):16.
116. Auger C, Knuth CM, Abdullahi A, et al. Metformin prevents the
pathological browning of subcutaneous white adipose tissue. Mol
Metab. 2019 Nov;29:12–23.
117. Domise M, Didier S, Marinangeli C, et al. AMP-activated protein
kinase modulates tau phosphorylation and tau pathology in vivo.
Sci Rep-Uk. 2016 May 27;6:26758.
118. Palmieri M, Pal R, Nelvagal HR, et al. mTORC1-independent TFEB
activation via Akt inhibition promotes cellular clearance in neuro-
degenerative storage diseases. Nat Commun. 2017 Feb 6;8
(1):14338.
•• This article describes autophagy targeting strategies including
specific autophagy pathways and their modulators in lysoso-
mal storage disorders (LSDs).
119. Kim S, Choi KJ, Cho SJ, et al. Fisetin stimulates autophagic degra-
dation of phosphorylated tau via the activation of TFEB and Nrf2
transcription factors. Sci Rep. 2016 Apr 26;6(1):24933.
120. Chauhan S, Ahmed Z, Bradfute SB, et al. Pharmaceutical screen
identifies novel target processes for activation of autophagy with
a broad translational potential. Nat Commun. 2015 Oct 27;6
(1):8620.
121. Jo C, Gundemir S, Pritchard S, et al. Nrf2 reduces levels of phos-
phorylated tau protein by inducing autophagy adaptor protein
NDP52. Nat Commun. 2014 Mar 25;5(1):3496.
122. Hernandez I, Luna G, Rauch JN, et al. A farnesyltransferase inhibitor
activates lysosomes and reduces tau pathology in mice with
tauopathy. Sci Transl Med. 2019 Mar 27;11(485):eaat3005.
•• Representative case study of autophagy inducer and farnesyl-
transferase inhibitor, lonafarnib as tauopathies inhibitor. lona-
farnib also induces autophagy leading to clearance of tau
aggregates by activating lysosomal function.
123. McCann H, Stevens CH, Cartwright H, et al. alpha-synucleinopathy
phenotypes. Parkinsonism Relat Disord. 2014 Jan;20(Suppl 1):S62–7.
124. Wang X, Becker K, Levine N, et al. Pathogenic alpha-synuclein
aggregates preferentially bind to mitochondria and affect cellular
respiration. Acta Neuropathol Commun. 2019 Mar 14;7(1):41.
125. Zhou W, Hurlbert MS, Schaack J, et al. Overexpression of human
alpha-synuclein causes dopamine neuron death in rat primary
culture and immortalized mesencephalon-derived cells. Brain Res.
2000 Jun 2;866(1–2):33–43.
126. Venda LL, Cragg SJ, Buchman VL, et al. alpha-Synuclein and dopa-
mine at the crossroads of Parkinson’s disease. Trends Neurosci.
2010 Dec;33(12):559–568.
127. Dehay B, Bove J, Rodriguez-Muela N, et al. Pathogenic lysosomal
depletion in Parkinson’s disease. J Neurosci. 2010 Sep 15;30
(37):12535–12544.
128. Ding K, Wang H, Wu Y, et al. Rapamycin protects against apoptotic
neuronal death and improves neurologic function after traumatic
brain injury in mice via modulation of the mTOR-p53-Bax axis.
J Surg Res. 2015 Mar;194(1):239–247. .
129. Dulovic M, Jovanovic M, Xilouri M, et al. The protective role of
AMP-activated protein kinase in alpha-synuclein neurotoxicity in
vitro. Neurobiol Dis. 2014 Mar;63:1–11.
130. Perez-Revuelta BI, Hettich MM, Ciociaro A, et al. Metformin lowers
Ser-129 phosphorylated alpha-synuclein levels via
mTOR-dependent protein phosphatase 2A activation. Cell Death
Dis. 2014 May 8;5(5):e1209.
131. Ur Rasheed MS, Tripathi MK, Mishra AK, et al. Resveratrol protects
from toxin-induced Parkinsonism: plethora of proofs hitherto petty
translational value. Mol Neurobiol. 2016 Jul;53(5):2751–2760.
132. He Q, Koprich JB, Wang Y, et al. Treatment with trehalose prevents
behavioral and neurochemical deficits produced in an AAV alpha-
synuclein rat model of Parkinson’s disease. Mol Neurobiol. 2016
May;53(4):2258–2268. .
133. Yoon YS, Cho ED, Jung Ahn W, et al. Is trehalose an autophagic
inducer? Unraveling the roles of non-reducing disaccharides on
autophagic flux and alpha-synuclein aggregation. Cell Death Dis.
2017 Oct 5;8(10):e3091.
134. Pupyshev AB, Tikhonova MA, Akopyan AA, et al. Therapeutic acti-
vation of autophagy by combined treatment with rapamycin and
trehalose in a mouse MPTP-induced model of Parkinson’s disease.
Pharmacol Biochem Behav. 2019 Feb;177:1–11.
•• Case study for combinational approach of autophagy inducers
between trehalose and rapamycin. The simultaneous treat-
ment of trehalose with rapamycin shows considerable additive
effect on synuclein-related pathology by targeting AMPK-
mTOR signaling axis respectively.
135. Sarkar S, Floto RA, Berger Z, et al. Lithium induces autophagy by
inhibiting inositol monophosphatase. J Cell Biol. 2005 Sep 26;170
(7):1101–1111.
136. Criollo A, Maiuri MC, Tasdemir E, et al. Regulation of autophagy by
the inositol trisphosphate receptor. Cell Death Differ. 2007 May;14
(5):1029–1039. .
137. Hou L, Xiong N, Liu L, et al. Lithium protects dopaminergic cells
from rotenone toxicity via autophagy enhancement. BMC Neurosci.
2015 Nov 25;16(1):82.
138. Hebron ML, Lonskaya I, Moussa CE. Tyrosine kinase inhibition facil-
itates autophagic SNCA/alpha-synuclein clearance. Autophagy.
2013 Aug;9(8):1249–1250.
139. Hebron M, Moussa CE. Two sides of the same coin: tyrosine kinase
inhibition in cancer and neurodegeneration. Neural Regen Res.
2015 Nov;10(11):1767–1769.
140. Brahmachari S, Ge P, Lee SH, et al. Activation of tyrosine kinase
c-Abl contributes to alpha-synuclein-induced neurodegeneration.
J Clin Invest. 2016 Aug 1;126(8):2970–2988.
141. Wille H, Requena JR. The Structure of PrP(Sc) prions. Pathogens.
2018 Feb 7;7(1):20.
142. Fang C, Imberdis T, Garza MC, et al. A neuronal culture system to
detect prion synaptotoxicity. PLoS Pathog. 2016 May;12(5):
e1005623.
143. Geschwind MD. Prion diseases. Continuum (Minneap Minn). 2015
Dec;21(6 Neuroinfectious Disease):1612–1638.
144. Colby DW, Prusiner SB. Prions. Cold Spring Harb Perspect Biol. 2011
Jan 1;3(1):a006833.
145. Abdulrahman BA, Tahir W, Doh-Ura K, et al. Combining autophagy
stimulators and cellulose ethers for therapy against prion disease.
Prion. 2019 Jan;13(1):185–196.
•• Case study for combinational approach of mTOR-mediated
autophagy inducers including rapamycin or AR12 with cellu-
lose ethers (CEs), misfolded prions inhibitor.
146. Phadwal K, Kurian D, Salamat MKF, et al. Spermine increases acet-
ylation of tubulins and facilitates autophagic degradation of prion
aggregates. Sci Rep. 2018 Jul 3;8(1):10004.
147. Nakagaki T, Satoh K, Ishibashi D, et al. FK506 reduces abnormal
prion protein through the activation of autolysosomal degradation
EXPERT OPINION ON DRUG DISCOVERY 17
and prolongs survival in prion-infected mice. Autophagy. 2013
Sep;9(9):1386–1394. .
148. Perluigi M, Di Domenico F, Butterfield DA. mTOR signaling in aging
and neurodegeneration: at the crossroad between metabolism
dysfunction and impairment of autophagy. Neurobiol Dis. 2015
Dec;84:39–49.
149. Salabei JK, Hill BG. Autophagic regulation of smooth muscle cell
biology. Redox Biol. 2015;4:97–103.
150. De Meyer GR, Grootaert MO, Michiels CF, et al. Autophagy in
vascular disease. Circ Res. 2015 Jan 30;116(3):468–479.
151. Vindis C. Autophagy: an emerging therapeutic target in vascular
diseases. Br J Pharmacol. 2015 May;172(9):2167–2178.
152. Yang X, Li Y, Li Y, et al. Oxidative stress-mediated atherosclerosis:
mechanisms and therapies. Front Physiol. 2017;8:600.
153. Wang X, Sun D, Hu Y, et al. The roles of oxidative stress and
Beclin-1 in the autophagosome clearance impairment triggered
by cardiac arrest. Free Radic Biol Med. 2019 May 20;136:87–95. .
154. Jeong SJ, Kim S, Park JG, et al. Prdx1 (peroxiredoxin 1) deficiency
reduces cholesterol efflux via impaired macrophage lipophagic
flux. Autophagy. 2018;14(1): 120–133.
•• Representative recent case study of PRDX1-mimics, ebselen
and gliotoxin as lipophagy inducer in altherosclerosis. These
mimic agents rescues impaired lipophagy by reducing pro-
duced reactive oxigen specises (ROS).
155. Yoshida A, Yoshida S, Ishibashi T, et al. Intraocular
neovascularization. Histol Histopathol. 1999 Oct;14(4):1287–1294.
156. Zhu XR, Du JH. Autophagy: a potential target for the treatment of
intraocular neovascularization. Int J Ophthalmol. 2018;11
(4):695–698.
157. Li R, Du JH, Yao GM, et al. Autophagy: a new mechanism for
regulating VEGF and PEDF expression in retinal pigment epithe-
lium cells. Int J Ophthalmol. 2019;12(4):557–562.
•• Representative case study of targeting intraocular neovascu-
larization, 3-MA (PI3K inhibitor)- or chloroquine (lysosomal
inhibitor)- mediated inhibition of autophagy results in sup-
pressiing formation of ocular vasculature.
158. Li H, Gao A, Feng D, et al. Evaluation of the protective potential of
brain microvascular endothelial cell autophagy on blood-brain
barrier integrity during experimental cerebral
ischemia-reperfusion injury. Transl Stroke Res. 2014 Oct;5
(5):618–626. .
159. Yang Z, Huang C, Wu Y, et al. Autophagy protects the blood-brain
barrier through regulating the dynamic of claudin-5 in short-term
starvation. Front Physiol. 2019;10:2.
•• This article demonstrates small molecules that regulate clau-
din-5 leading to reducing blood brain barrier (BBB) permeabil-
ity to target ischemia-reperfusion injury (IRI).
160. Kim KA, Shin D, Kim JH, et al. Role of autophagy in endothelial
damage and blood-brain barrier disruption in ischemic stroke.
Stroke. 2018 Jun;49(6):1571–1579. .
161. Hemanth KT, Jitendra S, Surya PDV. A review on viral infection
diseases. Int J Res Pharm Pharm Sci. 2017 Nov;2(6):73–75.
162. Lin K, Gallay P. Curing a viral infection by targeting the host: the
example of cyclophilin inhibitors. Antiviral Res. 2013 Jul;99
(1):68–77.
163. Ahmad L, Mostowy S, Sancho-Shimizu V. Autophagy-virus inter-
play: from cell biology to human disease. Front Cell Dev Biol.
2018;6:155.
164. Mohamud Y, Qu J, Xue YC, et al. CALCOCO2/NDP52 and SQSTM1/
p62 differentially regulate coxsackievirus B3 propagation. Cell
Death Differ. 2019 Jun;26(6):1062–1076.
165. Jia X, Liu B, Bao L, et al. Delayed oseltamivir plus sirolimus treat-
ment attenuates H1N1 virus-induced severe lung injury correlated
with repressed NLRP3 inflammasome activation and inflammatory
cell infiltration. PLoS Pathog. 2018 Nov;14(11):e1007428. .
166. Schmid M, Speiseder T, Dobner T, et al. DNA virus replication
compartments. J Virol. 2014 Feb;88(3):1404–1420.
167. Furlong K, Hwang S. Autophagy and noroviruses. Viruses-Basel.
2019 Mar 12;11(3):244.
168. Rodriguez-Rocha H, Gomez-Gutierrez JG, Garcia-Garcia A, et al.
Adenoviruses induce autophagy to promote virus replication and
oncolysis. Virology. 2011 Jul-Aug;416(1–2):9–15. .
169. Lee YR, Hu HY, Kuo SH, et al. Dengue virus infection induces
autophagy: an in vivo study. J Biomed Sci. 2013 Sep 8;20(1):65.
170. Zhou Z, Jiang XJ, Liu D, et al. Autophagy is involved in influenza
A virus replication. Autophagy. 2009 Apr 1;5(3):321–328.
171. Cao B, Parnell LA, Diamond MS, et al. Inhibition of autophagy limits
vertical transmission of Zika virus in pregnant mice. J Exp Med.
2017Aug7;214(8):2303–2313.
•• Representative case study of targeting autophagy using
autophagy inhibitors, such as 3-methyladenine (3-MA) or wort-
mannin, in viral diseases. They can be straightforward strate-
gies to inhibit viral infection or replication in infected cells.
172. Mauvezin C, Neufeld TP. Bafilomycin A1 disrupts autophagic flux by
inhibiting both V-ATPase-dependent acidification and Ca-P60A/
SERCA-dependent autophagosome-lysosome fusion. Autophagy.
2015 Aug;11(8):1437–1438.
173. Cenedella RJ. Cholesterol synthesis inhibitor U18666A and the role
of sterol metabolism and trafficking in numerous pathophysiologi-
cal processes. Lipids. 2009 Jun;44(6):477–487.
174. Kanai R, Zaupa C, Sgubin D, et al. Effect of gamma34.5 deletions on
oncolytic herpes simplex virus activity in brain tumors. J Virol. 2012
Apr;86(8):4420–4431. .
175. Su M, Mei Y, Sanishvili R, et al. Targeting gamma-herpesvirus 68
Bcl-2-mediated down-regulation of autophagy. J Biol Chem. 2014
Mar 21;289(12):8029–8040.
176. Liang Q, Chang B, Brulois KF, et al. Kaposi’s sarcoma-associated
herpesvirus K7 modulates Rubicon-mediated inhibition of autop-
hagosome maturation. J Virol. 2013 Nov;87(22):12499–12503. .
177. Campbell GR, Rawat P, Bruckman RS, et al. Human immunodefi-
ciency virus type 1 nef inhibits autophagy through transcription
factor EB sequestration. PLoS Pathog. 2015 Jun;11(6):e1005018.
178. Gassen NC, Niemeyer D, Muth D, et al. SKP2 attenuates autophagy
through Beclin1-ubiquitination and its inhibition reduces
MERS-Coronavirus infection. Nat Commun. 2019 Dec 18;10(1):5770.
179. Orvedahl A, MacPherson S, Sumpter R Jr., et al. Autophagy protects
against Sindbis virus infection of the central nervous system. Cell
Host Microbe. 2010 Feb 18;7(2):115–127.
180. Kim N, Kim MJ, Sung PS, et al. Interferon-inducible protein SCOTIN
interferes with HCV replication through the autolysosomal degra-
dation of NS5A. Nat Commun. 2016 Feb 12;7(1):10631.
181. Aweya JJ, Mak TM, Lim SG, et al. The p7 protein of the hepatitis
C virus induces cell death differently from the influenza A virus
viroporin M2. Virus Res. 2013 Mar;172(1–2):24–34.
182. Choi Y, Bowman JW, Jung JU. Autophagy during viral infection - a
double-edged sword. Nat Rev Microbiol. 2018 Jun;16(6):341–354.
183. Lei Y, Wen H, Yu Y, et al. The mitochondrial proteins NLRX1 and
TUFM form a complex that regulates type I interferon and
autophagy. Immunity. 2012 Jun 29;36(6):933–946.
184. Shin C, Kim YK. Autoimmunity in microbiome-mediated diseases
and novel therapeutic approaches. Curr Opin Pharmacol. 2019
Dec;49:34–42.
185. Kho ZY, Lal SK. The human gut microbiome - a potential controller
of wellness and disease. Front Microbiol. 2018;9:1835.
186. Larabi A, Barnich N, Nguyen HTT. New insights into the interplay
between autophagy, gut microbiota and inflammatory responses
in IBD. Autophagy. 2020 Jan 2;16(1):38–51.
187. Miao Y, Lv Q, Qiao S, et al. Alpinetin improves intestinal barrier
homeostasis via regulating AhR/suv39h1/TSC2/mTORC1/autop-
hagy pathway. Toxicol Appl Pharmacol. 2019 Dec 1;384:114772.
•• Representative recent case study of autophagy inducing
agent, alpinetin, in microbial mediated disorders. alpinetin
rescues autophagy to protect intestinal barrier through AhR/
suv39h1/TSC2/mTOR signaling axis.
188. Tartakover Matalon S, Ringel Y, Konikoff F, et al. Cannabinoid
receptor 2 agonist promotes parameters implicated in mucosal
healing in patients with inflammatory bowel disease. United
European Gastroenterol J. 2019 Nov 14;8(3):271.
18 D. KIM ET AL.
189. Dando I, Donadelli M, Costanzo C, et al. Cannabinoids inhibit
energetic metabolism and induce AMPK-dependent autophagy in
pancreatic cancer cells. Cell Death Dis. 2013 Jun 13;4(6):e664.
190. Losier TT, Akuma M, McKee-Muir OC, et al. AMPK promotes xeno-
phagy through priming of autophagic kinases upon detection of
bacterial outer membrane vesicles. Cell Rep. 2019 Feb 19;26
(8):2150–65 e5.
•• An article emphasizes the importance of AMPK signaling as
xenophagy regulation for targeting bacterial mediated
disorders.
191. Platt FM, d’Azzo A, Davidson BL, et al. Lysosomal storage diseases.
Nat Rev Dis Primers. 2018 Oct 1;4:1–25.
192. Seranova E, Connolly KJ, Zatyka M, et al. Dysregulation of autop-
hagy as a common mechanism in lysosomal storage diseases.
Essays Biochem. 2017 Dec 12;61(6):733–749.
193. Kinghorn KJ, Gronke S, Castillo-Quan JI, et al. A drosophila model of
neuronopathic gaucher disease demonstrates
lysosomal-autophagic defects and altered mTOR signalling and is
functionally rescued by rapamycin. J Neurosci. 2016 Nov 16;36
(46):11654–11670.
194. Maetzel D, Sarkar S, Wang HY, et al. Genetic and chemical correc-
tion of cholesterol accumulation and impaired autophagy in hepa-
tic and neural cells derived from niemann-pick type c
patient-specific ips cells. Stem Cell Rep. 2014 Jun 3;2(6):866–880.
195. Calderon JF, Klein AD. Controversies on the potential therapeutic
use of rapamycin for treating a lysosomal cholesterol storage
disease. Mol Genet Metab Rep. 2018 Jun;15:135–136.
196. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism,
and disease (vol 168, pg 960, 2017). Cell. 2017 Apr 6;169(2):362.
197. Wang Z, Miao G, Xue X, et al. The vici syndrome protein EPG5 Is
a Rab7 effector that determines the fusion specificity of autopha-
gosomes with late endosomes/lysosomes. Mol Cell. 2016 Sep 1;63
(5):781–795.
•• This article describes autophagy targeting strategies including
specific autophagy pathways and their modulators in lysoso-
mal storage disorders (LSDs).
198. Moon JH, Lee JH, Lee YJ, et al. Autophagy flux induced by
ginsenoside-Rg3 attenuates human prion protein-mediated neuro-
toxicity and mitochondrial dysfunction. Oncotarget. 2016 Dec 27;7
(52):85697–85708.
EXPERT OPINION ON DRUG DISCOVERY 19
... It should be noted that each variant found in genes shared between autophagy and apoptosis (P53, BCL2, ATG5 and P14ARF) should be evaluated with caution as it could exert dual effect. Furthermore, we list genes encoding regulatory factors related to autophagy, such as transcription factors and non-coding RNA (Table 1) [66][67][68][69][70][71][72][73][74][75]. In addition, the interrelation between players of this complex network is depicted in Figure 2. Autophagy-related genes and those shared between autophagy and apoptosis could carry genetic variants that may modify the phenotypes of human disorders in which these two pathways play an important role. ...
... It should be noted that each variant found in genes shared between autophagy and apoptosis (P53, BCL2, ATG5 and P14ARF) should be evaluated with caution as it could exert dual effect. Furthermore, we list genes encoding regulatory factors related to autophagy, such as transcription factors and non-coding RNA (Table 1) [66][67][68][69][70][71][72][73][74][75]. In addition, the interrelation between players of this complex network is depicted in Figure 2. A recent study of Klaassen and co-workers described the SHANK gene family as a candidate modifier for a rare metabolic disease-phenylketonuria [76]. ...
... Microphthalmia/transcription factor E or MiT/TFE family members (MITF, TFEB, TFE3, and TFEC), nuclear factor erythroid-derived 2-like 2 (NFE2L2/NRF2), the forkhead box o (FoxO) family, the CCAAT/enhancer-binding protein (C/EBP) family, and the GATA transcription factor are the most recognized master genes of autophagy (for a comprehensive review, see Kim et al., 2020) [72]. Therefore, both variants in genes encoding for transcription factors as well as the promoter regions of genes regulated by them are worthy of investigation [73][74][75]. ...
Article
Full-text available
Glycogen storage diseases (GSDs) are rare metabolic monogenic disorders characterized by an excessive accumulation of glycogen in the cell. However, monogenic disorders are not simple regarding genotype–phenotype correlation. Genes outside the major disease-causing locus could have modulatory effect on GSDs, and thus explain the genotype–phenotype inconsistencies observed in these patients. Nowadays, when the sequencing of all clinically relevant genes, whole human exomes, and even whole human genomes is fast, easily available and affordable, we have a scientific obligation to holistically analyze data and draw smarter connections between genotype and phenotype. Recently, the importance of glycogen-selective autophagy for the pathophysiology of disorders of glycogen metabolism have been described. Therefore, in this manuscript, we review the potential role of genes involved in glycogen-selective autophagy as modifiers of GSDs. Given the small number of genes associated with glycogen-selective autophagy, we also include genes, transcription factors, and non-coding RNAs involved in autophagy. A cross-link with apoptosis is addressed. All these genes could be analyzed in GSD patients with unusual discrepancies between genotype and phenotype in order to discover genetic variants potentially modifying their phenotype. The discovery of modifier genes related to glycogen-selective autophagy and autophagy will start a new chapter in understanding of GSDs and enable the usage of autophagy-inducing drugs for the treatment of this group of rare-disease patients.
... For conserving cellular homeostasis, autophagy also controls cell survival pathways . To date, mammalian autophagy can be separated into three major types based on the cellular constituents that are delivered into the lysosome: macroautophagy (hereinafter referred to as autophagy), chaperone-mediated autophagy (CMA) and microautophagy (Hazari et al., 2020;Kim et al., 2020). The microautophagy pathway is the least characterized and involves the sequestration of cytoplasmic cargos directly at the surface of the lysosomal membrane; protrusion and/or membrane invagination followed by scission releases the cargo into the lysosomal lumen for subsequent degradation (Gatica et al., 2018;Lei and Klionsky, 2020). ...
Article
Full-text available
Macroautophagy (hereafter referred to as autophagy), a highly conserved metabolic process, regulates cellular homeostasis by degrading dysfunctional cytosolic constituents and invading pathogens via the lysosomal system. In addition, autophagy selectively recycles specific organelles such as damaged mitochondria (via mitophagy), and lipid droplets (LDs; via lipophagy) or eliminates specialized intracellular pathogenic microorganisms such as hepatitis B virus (HBV) and coronaviruses (via virophagy). Selective autophagy, particularly mitophagy, plays a key role in the preservation of healthy liver physiology, and its dysfunction is connected to the pathogenesis of a wide variety of liver diseases. For example, lipophagy has emerged as a defensive mechanism against chronic liver diseases. There is a prominent role for mitophagy and lipophagy in hepatic pathologies including non-alcoholic fatty liver disease (NAFLD), hepatocellular carcinoma (HCC), and drug-induced liver injury. Moreover, these selective autophagy pathways including virophagy are being investigated in the context of viral hepatitis and, more recently, the coronavirus disease 2019 (COVID-19)-associated hepatic pathologies. The interplay between diverse types of selective autophagy and its impact on liver diseases is briefly addressed. Thus, modulating selective autophagy (e.g., mitophagy) would seem to be effective in improving liver diseases. Considering the prominence of selective autophagy in liver physiology, this review summarizes the current understanding of the molecular mechanisms and functions of selective autophagy (mainly mitophagy and lipophagy) in liver physiology and pathophysiology. This may help in finding therapeutic interventions targeting hepatic diseases via manipulation of selective autophagy.
... Some examples of autophagy modulating drugs approved or in clinical trials for various diseases[335][336][337][338][339] ...
Thesis
Full-text available
Inflammatory bowel diseases (IBD) are chronic, idiopathic disorders of the gastrointestinal tract that affect mainly the young population. Disease-targeted, tolerable, cost-effective medications to treat IBD are still awaited. The discovery of the crucial involvement of the autophagy pathway in the genetic predisposition of IBD opened a novel avenue for exploration in IBD therapeutics. Autophagy is a vital process that plays a central role in maintaining cellular homeostasis and pharmacological regulation of autophagy has proven to be beneficial in several diseases. Hence, we evaluated the efficacy of P140 - a therapeutic phosphopeptide known to modulate autophagy processes in other autoimmune and inflammatory conditions - in murine models of IBD. We have demonstrated that the peptide exerts protective effects on colitis models and corrects the pathological dysfunctions in different autophagy pathways. Thus, after the era of drugs classified as "disease-modifying" therapeutics, emerging "mechanism-guided" pharmaceuticals seem to hold a lot of promises for treating inflammatory diseases.
... Thus, autophagy can also be regarded as a self-protection mechanism of cells [129]. Autophagy has been becoming a new target of breast cancer treatment, but the role of autophagy in cancer is quite complex, which acts as a double-edged sword in the tumor treatment [130][131][132]. On the one hand, it can increase tumor cell autophagy activity, which contributes to programmed forms of cell death. ...
Article
Full-text available
Triple-negative breast cancer (TNBC) is a subtype of human breast cancer with one of the worst prognoses, with no targeted therapeutic strategies currently available. Regulated cell death (RCD), also known as programmed cell death (PCD), has been widely reported to have numerous links to the progression and therapy of many types of human cancer. Of note, RCD can be divided into numerous different subroutines, including autophagy-dependent cell death, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis and anoikis. More recently, targeting the subroutines of RCD with small-molecule compounds has been emerging as a promising therapeutic strategy, which has rapidly progressed in the treatment of TNBC. Therefore, in this review, we focus on summarizing the molecular mechanisms of the above-mentioned seven major RCD subroutines related to TNBC and the latest progress of small-molecule compounds targeting different RCD subroutines. Moreover, we further discuss the combined strategies of one drug (e.g., narciclasine) or more drugs (e.g., torin-1 combined with chloroquine) to achieve the therapeutic potential on TNBC by regulating RCD subroutines. More importantly, we demonstrate several small-molecule compounds (e.g., ONC201 and NCT03733119) by targeting the subroutines of RCD in TNBC clinical trials. Taken together, these findings will provide a clue on illuminating more actionable low-hanging-fruit druggable targets and candidate small-molecule drugs for potential RCD-related TNBC therapies. Graphical abstract
... To date, there have been a number of drugs approved by the FDA for clinical use in cancer treatment (Rapamycin derivatives, Nilotinib, Bortezomib) through modulation of autophagy and a proportion of autophagy agonists are also in clinical trials for neurodegenerative diseases (130). A large number of clinical trials are associated with everolimus-eluting stents (NCT02389946, NCT00911976, NCT00783796, NCT02681016), whose use results in better hematological reconstitution and reduced inflammation and foreign body reactions. ...
Article
Full-text available
Increasing attention is now being paid to the important role played by autophagic flux in maintaining normal blood vessel walls. Endothelial cell dysfunction initiates the development of atherosclerosis. In the endothelium, a variety of critical triggers ranging from shear stress to circulating blood lipids promote autophagy. Furthermore, emerging evidence links autophagy to a range of important physiological functions such as redox homeostasis, lipid metabolism, and the secretion of vasomodulatory substances that determine the life and death of endothelial cells. Thus, the promotion of autophagy in endothelial cells may have the potential for treating atherosclerosis. This paper reviews the role of endothelial cells in the pathogenesis of atherosclerosis and explores the molecular mechanisms involved in atherosclerosis development.
Article
Fluorene was previously reported to have anticancer activity against human cancer cells. In this study, we examined the in vitro function of 9-methanesulfonylmethylene-2, 3-dimethoxy-9 H -fluorene (MSDF), a novel fluorene derivative, its anticancer potential in human hepatocellular carcinoma (HCC) cells and its underlying molecular mechanism. The disruption of cellular homeostasis caused by MSDF was found to promote reactive oxygen species (ROS) generation, leading to the activation of cellular apoptosis. As a survival strategy, cells undergo autophagy during oxidative stress. MSDF-induced apoptosis occurred through both receptor-mediated extrinsic and mitochondrial-mediated intrinsic routes. The development of acidic vesicular organelles and the accumulation of LC3-II protein suggest an increase in the autophagic process. Apoptosis was detected by double staining. The MAPK/ERK and PI3K/Akt signaling pathways were indeed suppressed during treatment. Along with elevated ROS generation and apoptosis, MSDF also caused anoikis and cell death by causing cells to lose contact with their extracellular matrix. ROS production was induced by MSDF and sustained by an NAC scavenger. MSDF-induced apoptosis led to increased autophagy, as shown by the suppression of apoptosis by Z-VAD-FMK. However, inhibition of autophagy by inhibitor 3-MA increased MSDF-induced apoptosis. More evidence shows that MSDF downregulated the expression of immune checkpoint proteins, suggesting that MSDF could be used in the future as an adjuvant to improve the effectiveness of HCC immunotherapy. Altogether, our results highlight the potential of MSDF as a multitarget drug for the treatment of HCC.
Article
Full-text available
The Arg/N-degron pathway, which is involved in the degradation of proteins bearing an N-terminal signal peptide, is connected to p62/SQSTM1-mediated autophagy. However, the impact of the molecular link between the N-degron and autophagy pathways is largely unknown in the context of systemic inflammation. Here, we show that chemical mimetics of the N-degron Nt-Arg pathway (p62 ligands) decreased mortality in sepsis and inhibited pathological inflammation by activating mitophagy and immunometabolic remodeling. The p62 ligands alleviated systemic inflammation in a mouse model of lipopolysaccharide (LPS)-induced septic shock and in the cecal ligation and puncture model of sepsis. In macrophages, the p62 ligand attenuated the production of proinflammatory cytokines and chemokines in response to various innate immune stimuli. Mechanistically, the p62 ligand augmented LPS-induced mitophagy and inhibited the production of mitochondrial reactive oxygen species in macrophages. The p62 ligand-mediated anti-inflammatory, antioxidative, and mitophagy-activating effects depended on p62. In parallel, the p62 ligand significantly downregulated the LPS-induced upregulation of aerobic glycolysis and lactate production. Together, our findings demonstrate that p62 ligands play a critical role in the regulation of inflammatory responses by orchestrating mitophagy and immunometabolic remodeling.
Article
Background Jujuboside A (JuA), as a main effective component of Jujubogenin, has long been known as a sedative-hypnotic drug. The aim of the current study was to investigate the potential effect of JuA on sepsis-induced cardiomyopathy (SIC) induced by lipopolysaccharide (LPS). Method Wide type C57BL/6 mice and neonatal rat cardiomyocytes (NRCMs) were exposed to LPS to establish myocardial toxicity models. Cardiac function of septic mice was detected by echocardiography. Moreover, the survival rate was calculated for 7 days. ELISA assays were used to analyze inflammatory factors in serum. Furthermore, western blotting, flow cytometry and TUNEL staining were performed to assess cell apoptosis and transmission electron microscopy detect the number of autophagosomes in myocardium. Finally, the expression of proteins related to pyroptosis, autophagy and oxidative stress was analyzed by western blotting and immunohistochemistry staining. Results Results showed that JuA pretreatment significantly improved the survival rate and cardiac function, and suppressed systemic inflammatory response in septic mice. Further study revealed that JuA could decrease cell apoptosis and pyroptosis; instead, it strengthened autophagy in SIC. Moreover, JuA also significantly decreased oxidative stress and nitrodative stress, as evidenced by suppressing the superoxide production and downregulating iNOS and gp91 expression in vivo. In addition, the autophagy inhibitor 3-MA significantly abolished the effect of JuA on autophagic activity in SIC. Conclusion In conclusion, the findings indicated that JuA attenuates cardiac function via blocking inflammasome-mediated apoptosis and pyroptosis, at the same time by enhancing autophagy in SIC, heralding JuA as a potential therapy for sepsis.
Article
Full-text available
Long noncoding RNAs (lncRNAs) are emerging regulators of vascular diseases, yet their role in diabetic vascular calcification/aging remains poorly understood. In this study, we identified a down-expressed lncRNA SNHG1 in high glucose (HG)-induced vascular smooth muscle cells (HA-VSMCs), which induced excessive autophagy and promoted HA-VSMCs calcification/senescence. Overexpression of SNHG1 alleviated HG-induced HA-VSMCs calcification/senescence. The molecular mechanisms of SNHG1 in HA-VSMCs calcification/senescence were explored by RNA pull-down, RNA immunoprecipitation, RNA stability assay, luciferase reporter assay, immunoprecipitation and Western blot assays. In one mechanism, SNHG1 directly interacted with Bhlhe40 mRNA 3'-untranslated region and increased Bhlhe40 mRNA stability and expression. In another mechanism, SNHG1 enhanced Bhlhe40 protein SUMOylation by serving as a scaffold to facilitate the binding of SUMO E3 ligase PIAS3 and Bhlhe40 protein, resulting in increased nuclear translocation of Bhlhe40 protein. Moreover, Bhlhe40 suppressed the expression of Atg10, which is involved in the process of autophagosome formation. Collectively, the protective effect of SNHG1 on HG-induced HA-VSMCs calcification/senescence is accomplished by stabilizing Bhlhe40 mRNA and promoting the nuclear translocation of Bhlhe40 protein. Our study could provide a novel approach for diabetic vascular calcification/aging.
Article
Autophagy, the process of lysosomal degradation of biological materials within cells, is often halted abnormally in proteopathies, such as tauopathy and amyloidopathy. Thus, autophagy regulators that rescue dysregulated autophagy have great potential to treat proteopathies. We previously reported that the natural small molecule kaempferide (Kaem) induces autophagy without perturbing mTOR signaling. Here, we report that Kaem promotes lysosomal degradation of microtubule-associated protein tau (MAPT) in inducible MAPT cells. Kaem enhanced autophagy flux by mitigating microtubule-associated protein 1 light chain 3 (LC3) accumulation when MAPT expression was induced. Kaem also promoted activation of transcription factor EB (TFEB) without inhibiting mTOR and without mTOR inhibition-mediated cytotoxicity. In addition, Kaem-induced MAPT degradation was abolished in the absence of mitochondrial elongation factor Tu (TUFM), which was previously shown to be a direct binding partner of Kaem. Collectively, these results demonstrate that Kaem could be a potential therapeutic for tauopathy and reveal that TUFM can be a drug target for autophagy-driven disorders.
Article
Full-text available
Aim: To investigate the regulation of vascular endothelial growth factors (VEGF) and pigment epithelium-derived factor (PEDF) expression by autophagy in retinal pigment epithelium (RPE) cells on exposure to hypoxia. Methods: ARPE-19, an RPE cell line, was treated as following: the control group was kept in a normoxic incubator; the hypoxia group was incubated in a hypoxic incubator with 1% O2/5% CO2/94% N2 for 24h; the hypoxia + 3-methyladenine (3-MA) group was pretreated with 10 mmol/L 3-MA for 1h and then in the hypoxic incubator for 24h; and the hypoxia + chloroquine (CQ) group was pretreated with 50 µmol/L CQ for 1h and then in the hypoxic incubator for 24h. The morphology and ultrastructure of the cells was observed by an inverted microscope or a transmission electronic microscope (TEM). Western blot was performed to assay the expression of autophagy-associated markers, including microtubule associated protein 1 light chain 3 B (LC3B), Beclin-1, Atg5 and p62. The concentration of VEGF and PEDF in the culture supernatant was determined by ELISA, and the ratio of VEGF/PEDF was calculated. Results: There were no obvious differences in cell morphology among different groups and autolysosomes could be observed in the cytoplasm in all groups. Compared to the control cells, the LC3B-II/I ratio and levels of Beclin-1 and Atg5 were significantly increased and p62 level was significantly decreased in the hypoxia group. With the increase of VEGF and decrease of PEDF concentration, the VEGF/PEDF ratio was significantly increased in the hypoxia group compared to the control cells. The LC3B-II/I ratio was significantly reduced by 3-MA treatment and increased by CQ treatment. The expressions of Beclin-1 and Atg5 were significantly reduced by 3-MA or CQ treatment, while expression of p62 was increased in the 3-MA or CQ treated cells. The concentration of VEGF was significantly decreased and PEDF increased, thereby the VEGF/PEDF ratio was decreased in the hypoxia + 3-MA group and hypoxia + CQ group compared with that in the hypoxia group. Conclusion: Hypoxia leads to elevated autophagy in RPE cells, and expression of VEGF and PEDF might be regulated by autophagy on exposure to hypoxia to further participate in regulating the formation of retinal neovascularization.
Article
Full-text available
Manipulating autophagy is a promising strategy for treating cancer as several autophagy inhibitors are shown to induce autophagic cell death. One of these, autophagonizer (APZ), induces apoptosis-independent cell death by binding an unknown target via an unknown mechanism. To identify APZ targets, we used a label-free drug affinity responsive target stability (DARTS) approach with a liquid chromatography/tandem mass spectrometry (LC–MS/MS) readout. Of 35 protein interactors, we identified Hsp70 as a key target protein of unmodified APZ in autophagy. Either APZ treatment or Hsp70 inhibition attenuates integrity of lysosomes, which leads to autophagic cell death exhibiting an excellent synergism with a clinical drug, temozolomide, in vitro, in vivo, and orthotropic glioma xenograft model. These findings demonstrate the potential of APZ to induce autophagic cell death and its development to combinational chemotherapeutic agent for glioma treatment. Collectively, our study demonstrated that APZ, a new autophagy inhibitor, can be used as a potent antitumor drug candidate to get over unassailable glioma and revealed a novel function of Hsp70 in lysosomal integrity regulation of autophagy.
Article
Full-text available
Autophagy is an essential cellular process affecting virus infections and other diseases and Beclin1 (BECN1) is one of its key regulators. Here, we identified S-phase kinase-associated protein 2 (SKP2) as E3 ligase that executes lysine-48-linked poly-ubiquitination of BECN1, thus promoting its proteasomal degradation. SKP2 activity is regulated by phosphorylation in a hetero-complex involving FKBP51, PHLPP, AKT1, and BECN1. Genetic or pharmacological inhibition of SKP2 decreases BECN1 ubiquitination, decreases BECN1 degradation and enhances autophagic flux. Middle East respiratory syndrome coronavirus (MERS-CoV) multiplication results in reduced BECN1 levels and blocks the fusion of autophagosomes and lysosomes. Inhibitors of SKP2 not only enhance autophagy but also reduce the replication of MERS-CoV up to 28,000-fold. The SKP2-BECN1 link constitutes a promising target for host-directed antiviral drugs and possibly other autophagy-sensitive conditions.
Article
Full-text available
Background Cannabis benefits patients with inflammatory bowel disease (IBD). Cannabinoid receptors are expressed in gut immune cells and in epithelial cells of inflamed guts. Mucosal healing (MH) requires epithelial layer restoration. Objective To analyze the effects of CB2 agonist on parameters implicated in gut inflammation and MH. Methods Mucosal samples from areas of inflamed/uninflamed colon from 16 patients with IBD were cultured without/with cannabinoid receptor 2 (CB2) agonist (JWH-133, 10 µM, 6 hours (hr)), and analyzed for epithelial/stromal cell proliferation, apoptosis (secretome matrix metalloproteinase 9 (MMP9) activity, which impairs epithelial permeability) and interleukin-8 (IL-8) levels ( n = 5–9). In addition, Caco-2 (colon carcinoma epithelial cells) were cultured with biopsy secretomes (48 hr), and analyzed for phenotype and protein markers of proliferation (proliferating cell nuclear antigen), autophagy (LC3IIB) and permeability (Zonula occludens-1) ( n = 4–6). Results Uninflamed tissue had higher epithelial proliferation (Ki67: 50%↑, p < 0.05), and reduced secretome MMP9 activity and IL-8 levels (>50%↓, p < 0.05) compared to inflamed tissue. Treatment with CB2 agonist had no effect on epithelial apoptosis, but increased epithelial Ki67 expression (25%), and reduced secretome MMP9 and IL-8 levels in inflamed biopsies. Secretomes of CB2-treated biopsies increased Caco-2 number, migration, proliferating cell nuclear antigen and LC3IIB expression (all, p < 0.05), but had no effect on ZO-1. Conclusion Using ex vivo and in vitro human models, we demonstrated that manipulating the cannabinoid system affects colon cells and secretome characteristics that facilitate MH in IBD.
Article
Full-text available
Prion diseases are fatal transmissible neurodegenerative disorders that affect animals and humans. Prions are proteinaceous infectious particles consisting of a misfolded isoform of the cellular prion protein PrPC, termed PrPSc. PrPSc accumulates in infected neurons due to partial resistance to proteolytic digestion. Using compounds that interfere with the production of PrPSc or enhance its degradation cure prion infection in vitro, but most drugs failed when used to treat prion-infected rodents. In order to synergize the effect of anti-prion drugs, we combined drugs interfering with the generation of PrPSc with compounds inducing PrPSc degradation. Here, we tested autophagy stimulators (rapamycin or AR12) and cellulose ether compounds (TC-5RW or 60SH-50) either as single or combination treatment of mice infected with RML prions. Single drug treatments significantly extended the survival compared to the untreated group. As anticipated, also all the combination therapy groups showed extended survival compared to the untreated group, but no combination treatment showed superior effects to 60SH-50 or TC-5RW treatment alone. Unexpectedly, we later found that combining autophagy stimulator and cellulose ether treatment in cultured neuronal cells mitigated the pro-autophagic activity of AR12 and rapamycin, which can in part explain the in vivo results. Overall, we show that it is critical to exclude antagonizing drug effects when attempting combination therapy. In addition, we identified AR-12 as a pro-autophagic drug that significantly extends survival of prion-infected mice, has no adverse side effects on the animals used in this study, and can be useful in future studies.
Article
Full-text available
Objective:Browning, the conversion of white adipose tissue (WAT) to a beige phenotype, has gained interest as a strategy to induce weight lossand improve insulin resistance in metabolic disorders. However, for hypermetabolic conditions stemming from burn trauma or cancer cachexia,browning is thought to contribute to energy wasting and supraphysiological nutritional requirements. Metformin’s impact on this phenomenon andunderlying mechanisms have not been explored. Methods:We used both a murine burn model and humanex vivoadipose explants to assess metformin and 5-aminoimidazole-4-carboxamideribonucleotide (AICAR)’s effects on the development of subcutaneous beige adipose. Enzymes involved in fat homeostasis and browning, as wellas mitochondrial dynamics, were assessed to determine metformin’s effects. Results:Treatment with the biguanide metformin lowers lipolysis in beige fat by inducing protein phosphatase 2A (PP2A) independently ofadenosine monophosphate kinase (AMPK) activation. Increased PP2A activity catalyzes the dephosphorylation of acetyl-CoA carboxylase (Ser 79)and hormone sensitive lipase (Ser 660), thus promoting fat storage and the“whitening”of otherwise lipolytic beige adipocytes. Moreover, co-incubation of metformin with the PP2A inhibitor okadaic acid countered the anti-lipolytic effects of this biguanide in human adipose. Additionally,we show that metformin does not activate this pathway in the WAT of control mice and that AICAR sustains the browning of white adipose,offering further evidence that metformin acts independently of this cellular energy sensor. Conclusions:This work provides novel insights into the mechanistic underpinnings of metformin’s therapeutic benefits and potential as an agent to reduce the lipotoxicity associated with hypermetabolism and adipose browning.
Article
Full-text available
One of the most significant challenges of inflammatory bowel disease (IBD) research is to understand how alterations in the symbiotic relationship between the genetic composition of the host and the intestinal microbiota, under impact of specific environmental factors, lead to chronic intestinal inflammation. Genome-wide association studies, followed by functional studies, have identified a role for numerous autophagy genes in IBD, especially in Crohn disease. Studies using in vitro and in vivo models, in addition to human clinical studies have revealed that autophagy is pivotal for intestinal homeostasis maintenance, gut ecology regulation, appropriate intestinal immune responses and anti-microbial protection. This review describes the latest researches on the mechanisms by which dysfunctional autophagy leads to disrupted intestinal epithelial function, gut dysbiosis, defect in anti-microbial peptide secretion by Paneth cells, endoplasmic reticulum stress response and aberrant immune responses to pathogenic bacteria. A better understanding of the role of autophagy in IBD pathogenesis may provide better sub-classification of IBD phenotypes and novel approaches for disease management. Abbreviations: AIEC: adherent-invasive Escherichia coli; AMPK: AMP-activated protein kinase; ATF6: activating transcription factor 6; ATG: autophagy related; Atg16l1[ΔIEC] mice: mice with Atg16l1 depletion specifically in intestinal epithelial cells; Atg16l1[HM] mice: mice hypomorphic for Atg16l1 expression; BCL2: B cell leukemia/lymphoma 2; BECN1: beclin 1, autophagy related; CALCOCO2: calcium binding and coiled-coil domain 2; CASP: caspase; CD: Crohn disease; CGAS: cyclic GMP-AMP synthase; CHUK/IKKA: conserved helix-loop-helix ubiquitous kinase; CLDN2: claudin 2; DAPK1: death associated protein kinase 1; DCs: dendritic cells; DSS: dextran sulfate sodium; EIF2A: eukaryotic translation initiation factor 2A; EIF2AK: eukaryotic translation initiation factor 2 alpha kinase; ER: endoplasmic reticulum; ERBIN: Erbb2 interacting protein; ERN1/IRE1A: ER to nucleus signaling 1; FNBP1L: formin binding protein 1-like; FOXP3: forkhead box P3; GPR65: G-protein coupled receptor 65; GSK3B: glycogen synthase kinase 3 beta; IBD: inflammatory bowel disease; IECs: intestinal epithelial cells; IFN: interferon; IL: interleukin; IL10R: interleukin 10 receptor; IRGM: immunity related GTPase M; ISC: intestinal stem cell; LAMP1: lysosomal-associated membrane protein 1; LAP: LC3-associated phagocytosis; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; LPS: lipopolysaccharide; LRRK2: leucine-rich repeat kinase 2; MAPK: mitogen-activated protein kinase; MHC: major histocompatibility complex; MIF: macrophage migration inhibitory factor; MIR/miRNA: microRNA; MTMR3: myotubularin related protein 3; MTOR: mechanistic target of rapamycin kinase; MYD88: myeloid differentiation primary response gene 88; NLRP3: NLR family, pyrin domain containing 3; NOD2: nucleotide-binding oligomerization domain containing 2; NPC: Niemann-Pick disease type C; NPC1: NPC intracellular cholesterol transporter 1; OMVs: outer membrane vesicles; OPTN: optineurin; PI3K: phosphoinositide 3-kinase; PRR: pattern-recognition receptor; PTPN2: protein tyrosine phosphatase, non-receptor type 2; PTPN22: protein tyrosine phosphatase, non-receptor type 22 (lymphoid); PYCARD/ASC: PYD and CARD domain containing; RAB2A: RAB2A, member RAS oncogene family; RELA: v-rel reticuloendotheliosis viral oncogene homolog A (avian); RIPK2: receptor (TNFRSF)-interacting serine-threonine kinase 2; ROS: reactive oxygen species; SNPs: single nucleotide polymorphisms; SQSTM1: sequestosome 1; TAX1BP1: Tax1 binding protein 1; Th: T helper 1; TIRAP/TRIF: toll-interleukin 1 receptor (TIR) domain-containing adaptor protein; TLR: toll-like receptor; TMEM173/STING: transmembrane protein 173; TMEM59: transmembrane protein 59; TNF/TNFA: tumor necrosis factor; Treg: regulatory T; TREM1: triggering receptor expressed on myeloid cells 1; UC: ulcerative colitis; ULK1: unc-51 like autophagy activating kinase 1; WT: wild-type; XBP1: X-box binding protein 1; XIAP: X-linked inhibitor of apoptosis.
Article
Wound healing is a sequester program that involves diverse cell signalling cascades. Notwithstanding, complete signal transduction pathways underpinning acidic milieu derived from cancer cells is not clear, yet. MTT (3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide) assay, fluorescein diacetate/propidium iodide staining, and cell cycle flow cytometry revealed that acidic media decreased cell viability and cell number along with enhanced dead cells and S‐phase arrest in normal fibroblasts. Notably, the trends of intracellular reactive oxygen species production and lactate dehydrogenase release significantly increased with time. It seems the downregulation of Klf4 is in part due to acidosis‐induced DNA damage. It promoted cells towards S‐phase arrest and diminished cell proliferation. Klf4 downregulation had a direct correlation with the P53 level while acidic microenvironment promotes cells towards cell death mechanisms including apoptosis and autophagy. Noteworthily, the unchanged levels of Rb and Mlh1 indicated in those genes had no dominant role in the repairing of DNA damage in fibroblasts treated with the acidic microenvironment. Therefore, cells owing to not entering to mitosis and accumulation of DNA damage were undergone cell death to preserve cell homeostasis. Since acidic media decreased the level of tumour suppressor and DNA repair genes and altered the normal survival pathways in fibroblasts, caution should be exercised to not lead to cancer rather than wound healing.
Article
The injury of intestinal epithelial barrier is considered as the key pathophysiological process in response to gastrointestinal infection and inflammation, and plays an important role in the initiation and development of colitis. Alpinetin has been shown to improve intestinal barrier homeostasis under colitis condition, but the mechanism is still unclear. Here, we showed that alpinetin significantly improved transepithelial electrical resistance (TEER) in TNF-α-stimulated Caco-2 cells, which was mainly mediated by inhibiting the apoptosis. Mechanistic studies demonstrated that alpinetin markedly increased the production of autophagosomes, along with obvious regulation of LC3B-II, beclin-1, p62, Atg7 and Atg5 expressions. In addition, it also markedly repressed the activation of mTORC1 signaling pathway, which was ascribed to TSC2 rather than p-AKT, p-ERK, p-AMPKα or PTEN expressions in Caco-2 and NCM460 cells. Furthermore, the enrichment of H3K9me3 at TSC2 promoter region was decreased and ubiquitin proteasome degradation of suv39h1 was increased. Additionally, alpinetin activated aryl hydrocarbon receptor (AhR) and promoted co-localization of AhR with suv39h1 in the cytoplasm. The relationship between alpinetin-regulated AhR/suv39h1/TSC2/mTORC1 signals, autophagy and apoptosis of Caco-2 and NCM460 cells was confirmed by using CH223191, siAhR, siTSC2 and chloroquine. Finally, CH223191 and leucine abolished alpinetin-mediated inhibition of intestinal epithelial cells apoptosis, improvement of intestinal epithelial barrier and amelioration of colitis.
Article
The microbiome mediates constant bidirectional communication with the host immune system in a delicate balance of inducing pathogenic infection or residing in the human body in a commensal state. Dysbiosis is a hallmark of several autoimmune and immune-mediated inflammatory diseases. Various pathways involved in microbiome-induced autoimmunity are activated when a genetic defect or microbial trigger is found in the inflammasome, neutrophil extracellular trap, or receptors for microbial sensitization in the innate immune system or when similar defects are present in T or B lymphocytes of the adaptive immune system. Regulating autoimmunity through microbiome modulation may be highly effective as a therapeutic intervention.