Sei sulla pagina 1di 15

Am J Physiol Lung Cell Mol Physiol 305: L93L107, 2013.

First published May 24, 2013; doi:10.1152/ajplung.00072.2013.

Physiology in Medicine

Autophagy: a potential therapeutic target in lung diseases


Kiichi Nakahira and Augustine M. K. Choi
Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Womens Hospital, Harvard
Medical School, Boston, Massachusetts
Submitted 18 March 2013; accepted in final form 17 May 2013

Nakahira K, Choi AM. Autophagy: a potential therapeutic target in lung diseases.


Am J Physiol Lung Cell Mol Physiol 305: L93L107, 2013. First published May 24,
2013; doi:10.1152/ajplung.00072.2013.Macroautophagy (hereafter referred to as
autophagy) is an evolutionally conserved intracellular process to maintain cellular
homeostasis by facilitating the turnover of protein aggregates, cellular debris, and
damaged organelles. During autophagy, cytosolic constituents are engulfed into
double-membrane-bound vesicles called autophagosomes, which are subsequently delivered to the lysosome for degradation. Accumulated evidence suggests
that autophagy is critically involved not only in the basal physiological states but
also in the pathogenesis of various human diseases. Interestingly, a diverse variety
of clinically approved drugs modulate autophagy to varying extents, although they
are not currently utilized for the therapeutic purpose of manipulating autophagy. In
this review, we highlight the functional roles of autophagy in lung diseases with
focus on the recent progress of the potential therapeutic use of autophagymodifying drugs in clinical medicine. The purpose of this review is to discuss the
merits, and the pitfalls, of modulating autophagy as a therapeutic strategy in lung
diseases.
autophagy

scarcity, cells survive by eating


and recycling part of themselves (73, 94). These cellular
processes are collectively named autophagy (in Greek, eating oneself). Autophagy is a cellular self-degradation process
in which cytosolic materials and organelles are sequestered and
delivered to lysosome for degradation and recycling (73, 94).
In addition to nutrient starvation, autophagy is induced by
various physiological and pathological conditions. Importantly, autophagy is regulated in various human diseases such
as cancer (120), metabolic diseases (e.g., obesity and Type 2
diabetes) (91), neurodegenerative diseases (e.g., Alzheimers
disease and Parkinson disease) (122), and infectious diseases
[e.g., human immunodeficiency virus (HIV) and tuberculosis]
(54, 60). These reports suggest that autophagy can serve as a
potential therapeutic target for human diseases. Here, we summarize the pathophysiological roles of autophagy in lung
disease, and we explore the possibility that modulating autophagy activity using a pharmacological approach can be a
useful therapeutic intervention.

IN TIMES OF NUTRIENT OR OXYGEN

MOLECULAR MECHANISM OF AUTOPHAGY

Autophagy encloses cytosolic materials by isolation membranes (phagophores) to form double membrane-bound vesicles
The aim of this series is to provide practicing MDs and researchers an
up-to-date physiological understanding of disease. Articles published in this
series are intended to provide a thoughtful and lucid linkage of science to the
patient. Go here for more information on this series and to browse the content
(http://www.the-aps.org/mm/Publications/Journals/PIM).
Address for reprint requests and other correspondence: A. M. K. Choi,
Division of Pulmonary and Critical Care Medicine, Brigham and Womens
Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115 (e-mail:
amchoi@rics.bwh.harvard.edu).
http://www.ajplung.org

called autophagosomes (Fig. 1). The isolation membranes are


acquired from multiple sources including endoplasmic reticulum,
Golgi apparatus, mitochondrial outer membrane, and plasma
membrane (35, 76, 93, 116). Subsequently, the autophagosome
containing the cytosolic components and organelles fuses with the
lysosome to become autolysosome, with subsequent degradation
of cytosolic components (Fig. 1). More than 30 autophagy-related
gene (Atg) proteins have been identified to be involved in autophagosome formation (73, 94). The activation of autophagy is
mainly regulated by two signaling pathways including mammalian target of rapamycin (mTOR)-dependent and mTOR-independent pathways (94). In normal physiological condition, autophagy
activity is regulated at basal level by activation of mTOR, a
serine/threonine kinase that has diverse cellular functions. When
cells encounter nutritional starvation, mTOR is inactivated and
promotes the autophagosome formation (Fig. 2, left). In mTORindependent pathways, Ca2 calpainGs and cyclic AMP
(cAMP)phospholipase C (PLC)inositol-(1,4,5)-trisphosphate
(IP3) pathways are involved in autophagy activation (Fig. 2,
right).
FUNCTION OF AUTOPHAGY

Whereas autophagy nonselectively engulfs and degrades


intracellular proteins for recycling under nutritional starvation,
autophagy can selectively target and remove specific subcellular components (selective autophagy) (18). This selective
degradation pathway includes eliminating invading pathogens
(xenophagy) (60), dysfunctional cellular organelles such as
mitochondria (mitophagy) (126), and polyubiquitinated protein
aggregates (aggrephagy) (56). Autophagy is also involved in
lipid metabolism (lipophagy) (108). Selective autophagy

1040-0605/13 Copyright 2013 the American Physiological Society

L93

Physiology in Medicine
L94

AUTOPHAGY MODULATION IN LUNG DISEASES

Lysosome
ULK1 complex

Phagophore
3-MA

Autolysosome

Autophagosome

Class III
PI3K
Beclin 1

BCL-2

Initiation,
isolation membrane
BH3 mimetics

LC3II

Elongation

Completion

Autophagosome and
Lysosome fusion

Degradation

Spautin 1

Atg5-Atg12-Atg16L

Acid hydrolase

Class III PI3K complex

Cargos

Chloroquine

Cystatin B

Bafilomycin A

Pepstatin A

Azithromycin

Autophagy activator
Autophagy inhibitor

Fig. 1. Autophagy process and potential targets for modulating autophagy. Activation of UNC51-like kinase (ULK) complex in response to certain signals
initiates isolation membrane and the formation of phagophore. (Upstream pathway of ULK1 complex is depicted in Fig. 2.) Class III phosphoinositide 3-kinase
(PI3K) complex composed of Beclin 1, class III PI3K, PIK3R4, and activating molecule in BECN1-regulated autophagy protein 1 (AMBRA1) is also required
for the phagophore formation. The Atg-Atg12-Atg16L complex and LC3-II phosphatidylethanolamine (PE) conjugate promote the elongation and enwrap the
cytosolic cargos including mitochondria, leading to the formation of autophagosome. Subsequently, lysosome fuses with the autophagosome (the formation of
autolysosome) and releases acid hydrases into the interiors to degrade the cytosolic cargos. Autophagy can be activated by drugs such as BCL-2 homology 3
(BH3) mimetics that prevent the formation of autophagy inhibitory complex of Beclin 1 and BCL-2. In contrast, ubiquitin-mediated decrease of Beclin 1 protein
by Spautin-1 and inhibition of class III PI3K by 3-methyladenine (3-MA) can inhibit autophagy. In addition, there are drugs to inhibit the late phase of autophagy
process. Lysosomotropic agents that enhance lysosomal pH such as chloroquine inhibit lysosomal enzymes and also prevent the fusion of autophagosome and
lysosome, resulting in inhibition of autophagy. Bafilomycin A1 inhibits the fusion of autophagosome and lysosome by inhibiting vacuolar ATPase (V-ATPase)
located in the lysosomal membrane. Compounds such as pepstatin A and cystatin B are inhibitors for lysosomal protease. (Please also see Table 4.)

plays important roles in maintaining cellular homeostasis in


basal physiological states and in response to various cellular
stresses.
Cytoprotective roles of autophagy under stress conditions
such as starvation have been well documented (73). However,
when cells receive lethal signals, the cellular stresses cause
autophagy and also cell death including apoptosis. Although
interactions of autophagy- and apoptosis-related molecules
such as Beclin 1 and B cell lymphoma 2 (BCL-2), or LC3B and
Fas have been reported in various models (16, 87), it is still
unclear whether autophagy induced by lethal signals promotes
cell death or is an independent process from cell death (94).
The functional role of autophagy on cell death is likely to be
dependent on stress models.
Against invading microbes, autophagy actively participates
in innate immune responses (54, 60). For example, autophagy
exerts its host defense role by degrading various pathogens by
lysosomal system (xenophagy) (60). The target pathogens
include bacteria, such as group A Streptococcus pyrogen (60),
Mycobacterium tuberculosis (Mtb) (23), Salmonella enterica
(54), and Pseudomonas aeruginosa (127); viruses such as
herpes simplex virus 1 (54, 60); and parasites such as Toxoplasma gondii (54). In contrast, recent studies also suggest that

autophagy machinery contributes to the replication and survival of microbes in the host cells. The list of pathogens
exploiting autophagy machinery includes clinically important
microbes, e.g., bacterial agents such as Brucella abortus (99)
and Coxiella burnetii (99) and viruses such as HIV (99),
hepatitis B virus (HBV) (54, 99), and avian influenza A H5N1
(H5N1) (109). Of note, autophagy exerts both killing and
prosurvival properties for HIV (60, 99). Autophagy has diverse
effects on both innate and adaptive immune systems (54, 60).
Atg5 is involved with the production of type I interferons in
response to single-stranded RNA viruses (54, 60). Autophagy
proteins are also involved in the regulation of inflammasome,
cytosolic multiprotein complexes responsible for activation of
caspase-1 and its downstream signaling including secretion of
IL-1 and IL-18 (20, 79). Beclin 1 and LC3B negatively
regulate inflammasome activation by preserving mitochondrial
homeostasis (79). The roles of autophagy in adaptive immunity
include antigen presentation and antibody production. Autophagy is required for presenting antigen on both major
histocompatibility complex class I and II molecules and activating CD8 and CD4 T lymphocytes, respectively (60).
Autophagy is also required for B cell differentiation (19) and
for sustainable antibody production in plasma cells (88). Thus

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L95

AUTOPHAGY MODULATION IN LUNG DISEASES

Cell membrane

Cytoplasm

L type Ca2+ channel blockers


(verapamil, amidarone)

Clonidine

EGFR antagonists (ertinib)

Ca2+ channel

I1R

Receptor
tyrosine kinase

AC

Class I
PI3K

Gs

Insulin and IGF


receptor
Ca2+

25ddA
cAMP

ATP

Akt

Epac

Metformin

Calpastatin,
calpeptin

Calpain

PI103
Rap2B

TSC1/2

AMPK

Xestospongin B

PLC-

IP3 receptor

PIP2

IP3
Rheb

ER
IP2
Ca2+

Rapamycin and
analogues, Torin 1

Ins
IMPase

mTOR

Lithium, L-960,330

Autophagy activator
Autophagy activator

IP1

Valproic acid,
Carbamazepine

ULK1 complex
via cleavage of Atg5

Autophagy inhibitor

Autophagy

Fig. 2. Overview of the regulation of autophagy pathways and targets for modulating autophagy by drugs/compounds. Two major signaling pathways to regulate
autophagy are depicted in this figure: mammalian target of rapamycin (mTOR) signaling pathways and mTOR-independent signaling pathways. Autophagy is
modulated by mTOR in response to certain nutritional stimulations. Insulin or growth factors activate class I PI3K pathway, leading to activation of mTOR and
inhibition of autophagy. Glucose starvation activates AMPK and inhibits mTOR, followed by activating of ULK1 complex and activating autophagy. mTOR can
be pharmacologically inhibited by activating AMPK (e.g., metformin) or inhibiting class I PI3K (e.g., EGFR antagonist). Recent new drugs such as PI-103 can
activate autophagy by inhibiting both class I PI3K and mTOR. In the mTOR-independent pathway, increase of intracellular cAMP level and Ca2 inhibits
autophagy. Intracellular level of cAMP is upregulated by adenylyl cyclase (AC) and the calpain-Gs pathway, which is activated by intracellular Ca2 level.
The inhibitory effect of cAMP on autophagy is mediated by increase of synthesis of inositol-(1,4,5)-trisphosphate (IP3) and inositol. Increased level of IP3
activates IP3 receptor in endoplasmic reticulum (ER) and release Ca2 into cytosol, leading to inhibition of autophagy. Activation of L-type Ca2 channel triggers
Ca2 influx and elevates intracellular Ca2 level, which activates cysteine protease called calpain. In addition to elevating cAMP by the calpain-Gs pathway,
calpain inhibits autophagy by cleavage of Atg5. However, the concise mechanism by which cytosolic cAMP, Ca2, inositol, and IP3 regulate autophagy is still
unclear. In this cyclical mTOR-independent pathway, autophagy can be modulated by mainly targeting intracellular levels of cAMP or Ca2. L-type Ca2
receptor blockers such as verapamil inhibit Ca2 influx, which leads inhibition of calpain activity and cAMP synthesis. Activation of autophagy by drugs such
as lithium or carbamazepine is mediated by at least reduction of Ca2 release by IP3 receptor (IP3R). EGFR, epidermal growth factor receptor; IGF, insulin-like
growth; TSC, tuberous sclerosis 2; AMPK, AMP-activated protein kinase; V-ATPase, vacuolar ATPase; I1R, imidazoline-1 receptor; PLC, phospholipase C
epsilon; 2=5=ddA, 2=,5=-dideoxyadenosine; PIP2, phosphatidylinositol-4,5-bisphosphate; cAMP, cyclic AMP; Ins, inositol; IP2, inositol-(1,4)-bisphosphate; IP3,
Inositol-(1,4,5)-trisphosphate; IMPase, inositol monophosphatase.

autophagy plays crucial diverse roles in immune systems


including pathogen degradation and immune signaling, suggesting that the involvement of autophagy in infectious and
inflammatory diseases.
Autophagy is critically involved in various metabolic pathways for maintaining cellular homeostasis (91). For example,
autophagy is an important energy generator in the liver. Mice
with specific deletion of atg7 gene in liver display decreased
level of blood glucose and amino acids after starvation (25).
Autophagy is also involved in lipid metabolism by breaking

down lipid droplets to generate energy (lipophagy) (108).


Metabolome analysis also suggests autophagy is required for
maintenance of tricarboxylic acid cycle-related metabolites
(33), suggesting that autophagy is involved in the quality
control of mitochondria. Damaged mitochondria are selectively targeted and removed by autophagy to maintain normal
mitochondrial function (mitophagy) (126). Although the concise molecular mechanism of mitophagy is still unclear, the
importance of preserving mitochondrial homeostasis by autophagy is now further linked to the pathogenesis of human

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L96

AUTOPHAGY MODULATION IN LUNG DISEASES

diseases such as Parkinson disease (68, 80, 81, 118). Thus the
autophagy process is critically involved in not only physiological but also pathological metabolic responses.

AUTOPHAGY PATHWAY: POTENTIAL THERAPEUTIC TARGET


FOR LUNG DISEASES

MEASUREMENT OF AUTOPHAGY

The functional roles of autophagy on various lung diseases


have been studied both in vitro and in vivo. Tables 2 and 3
represent the functions of autophagy or autophagic proteins in
lung diseases on the basis of studies using genetic or biochemical perturbation of autophagy. Although autophagy has been
initially thought as a cytoprotective response in pathophysiological states, accumulating data reveal diverse functions of
autophagy in lung diseases (Tables 2 and 3). For example,
chronic obstructive pulmonary disease (COPD) is one lung
disease that has been shown to be associated with autophagy
(15). Increased number of autophagosomes is observed by
electron microscopy analysis, and expression of autophagic
protein LC3B-II is increased in lung tissues from patients with
COPD (15). Several molecules involved in autophagy-mediated COPD include FAS (16), Toll-like receptor 4 (5), and
cystic fibrosis transmembrane conductance regulator (CFTR)
(11). In addition, alveolar macrophages isolated from smokers
show increased of LC3 expression with defect of autophagy
flux (77). In vivo, genetic deletion of LC3B displays resistance
to cigarette smoke-induced airway space enlargement compared with the control mice (16). Similar adverse effects of

As the research of autophagy continues to evolve, methods


for monitoring autophagy have been evaluated and discussed in
detail. Klionsky et al. (51) have recently reported the guideline
for monitoring autophagy in higher eukaryotes and described
useful methods and how to interpret the data. A key tenet to
emphasize is that investigators need to recognize whether they
are evaluating the steady-state levels of autophagosomes or
dynamic state of autophagy generated by their models (51,
101). If they are seeking to assess the change of autophagy flux
or activity (e.g., the rate of delivery of autophagosomes substrate to lysosomes) in certain points, it is likely that monitoring steady state of autophagosome (e.g., measuring LC3-II
expression without examining turnover by Western blot, counting autophagosomes by using electron microscopy, or counting
puncta formation of LC3 by immunofluorescence microscopy)
is not sufficient. Table 1 demonstrates the various methods to
monitor autophagy both in vitro and in vivo. Generally, it is
recommended to use multiple different assays (ideally for both
steady and dynamic states), rather than relying on the results
from a single method (51, 101).

Autophagy in Models of Human Lung Diseases

Table 1. Methods for monitoring autophagy in vitro and in vivo


Monitoring static state autophagy
Quantifying autophagosomes

Methods

Key Points

Electron microscopy

Positive correlation of elevated number of


autophagosomes and increased autophagy flux has not
been proved reliably in all models.
Increased level of LC3-II (ratio of LC3-II/LC3-I) does not
always correlate with autophagy flux.
This method can be applied to in vivo using GFP-LC3
transgenic mice.
Increased puncta formation of LC3 does not reliably
correlate with autophagic flux.

LC3-II level (ratio of LC3-II/LC3-I)

Western blot

Puncta formation of LC3

Immunofluorescence
microscopy

Monitoring dynamic state autophagy


Turnover of LC3-II

Western blot

Turnover of p62/SQSTMI

Western blot

Cytosolic protein sequestration assays

Western blot

LC3-II on the autophagosome membrane is normally


continuously degraded during autophagy process. LC3-II
can be accumulated by adding lysosomal proteolysis
inhibitors such as leupeptin, chloroquine or bafilomycin.
Cells or animals can be treated with lysosomal protease
inhibitors.
Autophagic flux can be expressed as the difference in
LC3B-II signal on Western blot obtained in the presence
or absence of lysosome protein inhibitors.
P62/SQSTMI is a cytosolic protein that has an LC3
binding domain (REF). It binds to ubiquitinated protein
and carry them to autophagosome. Subsequently, both
p62/SQSTMI and the cargo proteins are degraded by
autophagy.
Similar with LC3-II, p62/SQSTMI turnover can be
measured by adding lysosome protease inhibitors in vitro
or in vivo.
Cells are incubated (or animals are intraperitoneally
administered) with leupeptin to inhibit lysosomal
proteolysis. Autophagosomes are purified via mechanical
disruption (34) followed by density centrifugation.
Autophagy flux is expressed by the amount of cytosolic
protein, such as LDH recovered in the autophagosome
fraction (Western blot).

LDH, lactate dehydrogenase.


AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

References

110
75
72, 74

38, 41

10, 38

38, 52, 106

Physiology in Medicine
L97

AUTOPHAGY MODULATION IN LUNG DISEASES

Table 2. The functional role of autophagic proteins in experimental models of lung diseases
Lung Diseases

COPD

IPF

PH (PAH)

CF

Lung cancer

LAM

HALI
Sepsis

Nanoparticle-induced ALI

Function of Autophagic Proteins

References

LC3B and Beclin 1 deficiency inhibits CSE-induced cell death in pulmonary epithelial cells.
LC3B-deficient mice display inhibition of airspace enlargement and apoptosis in lungs after cigarette smoke
exposure.
LC3 deficiency promotes CSE-induced IL-8 secretion in HBEC.
Autophagy inhibition by bafilomycin A enhances CSE-induced IL-8 secretion in HBEC.
Autophagy activation by rapamycin inhibits IL-17A-induced collagen production in lung epithelial cells.
Autophagy inhibition by 3-MA suppresses degradation of collagen in epithelial cells.
3-MA reverses the therapeutic effect of IL-17 antagonism on bleomycin-induced lung fibrosis and mortality
of mice.
LC3B and Beclin 1 deficiency promotes TGF--induced activation of lung fibroblast in vitro.
Autophagy activation by rapamycin inhibits TGF--induced fibronectin and -SMA expression in lung
fibroblast.
Rapamycin inhibits bleomycin-induced lung fibrosis in mice.
Rapamycin inhibits anti-TLR4 antibody-induced lung fibrosis in mice.
Atg5 and LC3 deficiency promotes TGF--induced fibroblast activation in vitro.
Inhibition of mTOR by Torin1 inhibits tunicamycin (ER stress inducer)-induced senescence in HBEC.
Atg5 and LC3 deficiency promotes tunicamycin-induced senescence in HBEC.
LC3B deficiency promotes hypoxia-induced pulmonary hypertension in mice.
LC3B deficiency promotes cell proliferation in pulmonary artery endothelial cells and vascular smooth
muscle cells.
Autophagy inhibition by 3-MA and chloroquine increase angiogenesis in PAEC from fetal lambs with
persistent pulmonary hypertension (PPHN-PAEC).
Beclin-1 knockdown promotes angiogenesis in PPHN-PAEC.
Beclin 1 deficiency promotes aggresome accumulation in CF epithelia.
Beclin 1 deficiency promotes macrophage infiltration and MPO activity in nasal mucosa of CF.
Autophagy inhibition by 3-MA abrogates CF phenotype in nasal mucosa.
LC3 deficiency promotes growth of B. cepacia and IL-1 secretion in macrophages.
Autophagy activation by rapamycin inhibits growth of B. cepacia and IL-1 secretion in macrophages.
Autophagy activation reduces bacterial burden of lung and inflammation in lung of CF mice after B.
cepacia infection.
Overexpression of SQSTM1/p62 inhibits intracellular survival of B. cepacia in macrophages.
Deficiency of SQSTM1/p62 promotes intracellular survival of B. cepacia in macrophages.
Blocking chaperone-mediated autophagy by LAMP-2A depletion suppresses cell proliferation and
increases cell death in lung cancer cells.
Injection of lung cancer cells with LAMP-2A deficiency reduces tumor formation in xenograft mouse
model.
Autophagy inhibition by chloroquine enhances cytotoxicity of gefinitib and erlotinib in lung cancer cells.
Atg5 and Atg7 deficiency enhances cytotoxicity of gefinitib and erlotinib in lung cancer cells.
Atg7 deficiency reduces cancer cell proliferation in NSCLC cell lines.
Atg7 deficiency sensitizes the cancer cells to cisplatin-induced apoptosis in NSCLC cell lines.
Atg3 deficiency reverses erlotinib resistance in erlotinib-resistant lung cancer cells.
TSC2 knockdown increases autophagy-dependent cell survival in mouse embryonic fibroblasts.
Atg5 deficiency inhibits TSC2-null xenograft tumor cell survival.
Autophagy inhibition by chloroquine inhibits xenograft tumor size in mice of TSC2-null xenograft tumor.
LC3B knockdown promotes hyperoxia-induced cell death in lung epithelial cells.
Depletion of LC3B and Beclin 1 leads mitochondrial dysfunction and enhances NLRP3 inflammasomemediated IL-1 and IL-18 secretion in macrophages.
LC3B knockdown increases serum level of IL-1 and IL-18 production in CLP-induced polymicrobial
sepsis and sensitizes mice to endotoxic shock.
Autophagy activation by rapamycin restores CLP-induced myocardial dysfunction in mice.
LC3 overexpression ameliorates acute lung injury and survival in CLP-induced polymicrobial model of
sepsis.
Knockdown of VPS34 increases cell death and liver injury in CLP-induced polymicrobial models of
sepsis.
Autophagy inhibition suppresses releases of neutrophil extracellular trap (NET) induced by E. coli in
human neutrophils.
Knockdown of Atg6 improves nanoparticle-induced cell death in lung epithelial cells.
Autophagy inhibition by 3-MA ameliorates nanoparticle-induced ALI in mice.

15, 16, 47

28
71

85

124
7

59

114

65

2
53

36
46
57
84

112
79

40
64
14
45
61, 62

COPD, chronic obstructive pulmonary disease; CSE, cigarette smoke extract; HBEC, human bronchial epithelial cells; IPF, idiopathic pulmonary fibrosis;
TGF-, transforming growth factor-; -SMA, -smooth muscle actin; 3-MA, 3-methyladenine; TLR4, Toll-like receptor 4; ER, endoplasmic reticulum; P(A)H,
pulmonary (artery) hypertension; PAEC, pulmonary artery endothelial cells; CF, cystic fibrosis; MPO, myeloperoxidase; B. cepacia, Burkholderia cenocepacia;
NSCLC, nonsmall cell lung cancer; LAMP2, lysosome-associated membrane protein 2; LAM, Lymphangioleiomyomatosis; TSC, tuberous sclerosis; HALI,
hyperoxia-induced acute lung injury; NLRP3, NOD-like receptor family, pyrin domain containing 3; CLP, cecal ligation and puncture; E. coli, Escherichia coli;
ALI, acute lung injury.
AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L98

AUTOPHAGY MODULATION IN LUNG DISEASES

Table 3. The functional role of autophagic proteins on pathogen(s) causing respiratory tract infection
Pathogens

Role of Autophagy or Autophagic Proteins

References

Tuberculosis (M. tuberculosis)

Autophagy induction by rapamycin or starvation inhibits M. tuberculosis survival in


infected macrophages.
IL-4 and IL-13 inhibit autophagy activation and autophagy-mediated killing of
intracellular M. tuberculosis in macrophages.
Autophagy activation by rapamycin enhances presentation of mycobacterial antigen
in macrophages.
Subcutaneous injection of mycobacteria-infected dendritic cells pretreated with
rapamycin enhances Th1 response and increases vaccine efficacy in mice.
Autophagy induction by Vitamin D3 promotes killing of M. tuberculosis.
Isoniazid-induced autophagy is associated with antimicrobial activity in M.
tuberculosis-infected macrophages.
Knockdown of Atg5 and TSC2 inhibits H5N1-induced cell death in lung epithelial
cells.
Autophagy inhibition by 3-MA ameliorates acute lung injury and mice survival rate in
mice infected with H5N1
Knockdown of Atg5 ameliorates acute lung injury and mice survival rate in mice
infected with H5N1.
Autophagy induction by rapamycin promotes P. aeruginosa clearance and
autophagy inhibition by 3-MA inhibits the bacterial clearances in macrophages.
Knockdown of Beclin 1 inhibits P. aeruginosa clearance in macrophages.
Autophagy inhibition by Azithromycin inhibits intracellular killing of M. abscessus
in macrophages.
Autophagy inhibition by Azithromycin promotes M. abscessus infection in mice.
Autophagy activation by rapamycin increases replication of human rhinovirus 2
(HRV-2).
Autophagy inhibition by 3-MA suppresses replication of HRV-2.
Knockdown of Atg5 inhibits human adenovirus-mediated cell lysis.
Knockdown of Atg5 inhibits replication of S. aureus and S. aureus-mediated cell
death.
Knockdown of Beclin 1 or LC3 attenuates respiratory syncytial virus-induced
proinflammatory cytokines production.

34

Avian influenza A H5N1

P. aeruginosa

M. abscessus

Human rhinovirus 2

Human adenovirus type 5


S. aureus
Respiratory syncytial virus

37
42

128
49
109

127

96

50

44
105
78

M. tuberculosis, Mycobacterium tuberculosis; P. aeruginosa, Pseudomonas aeruginosa; M. abscessus, Mycobacterium abscessus; S. aureus, Staphylococcus
aureus.

autophagy are observed in other lung diseases including lung


cancer (36, 46, 53) and acute lung injury induced by H5N1
infection (109). In contrast, the beneficial roles of autophagy
are also observed in the diseases such as cystic fibrosis (CF) (1,
2, 65), tuberculosis (23, 34, 42), and sepsis (14, 40, 45, 64, 79).
Thus autophagy is an important cellular process to regulate or
contribute to the pathogenesis of lung diseases. Importantly,
Fig. 3 demonstrates specificity of the pathophysiological functions of autophagy in each lung disease, resulting in either
favorable or deleterious phenotype depending in the disease
process.
Autophagy in Clinical Trials
Although drugs modulating autophagy such as rapamycin or
chloroquine have been clinically used for years without the
intent to modulate autophagy activity, these drugs are now
studied as autophagy modulators in clinical trials for lung
diseases (17, 99, 120, 125). For example, chloroquine, which
inhibits autophagy-mediated cell survival in tumor cells, is
used as an intervention for patients with small cell lung cancer
in a clinical trial (phase 1). In addition, the interventions using
hydrochloroquine and other anticancer drugs such as erlotinib
are used for non-small cell lung cancer in two clinical trials
(phase 1/2 and 2) (125). Although autophagy has been initially
shown to be associated with anti-tumorigenesis in rodent ani-

mal studies (90, 111), the current strategy for cancer therapy is
more likely to be based on inhibition of autophagy (17, 120,
125).
This paradox may be due to the differential roles of autophagy in different stages of tumorigenesis (99, 120). Although initiation of tumorigenesis in normal cells is associated
with defect of autophagy, autophagy subsequently may exert
prosurvival effect in tumor cells (99, 120). Lymphangioleiomyomatosis (LAM), a progressive lung disease caused by
mutation in the tuberous sclerosis genes (tsc), is associated
with inappropriate activation of mTOR signaling, which regulates cellular growth and lymphangiogenesis (39). The pathogenesis of LAM is in part similar with those of tumorigenesis
including inappropriate cell growth and survival (39). Chloroquine is also used with rapamycin for patients with LAM
(phase 1). This clinical trial is based on the rationale that
rapamycin blocks mTOR of downstream kinases and restores
homeostasis in cells with defective tsc function (39, 69, 84).
The use of chloroquine aims to inhibit autophagy-mediated
survival of LAM cells induced by rapamycin (39, 84). A recent
clinical study concludes chloroquine does not prevent infection
with influenza including H1N1 strain (86). Interestingly, autophagy is involved in infection with H5N1, rather than H1N1
(109). Therefore, it is possible that chloroquine has a differential effect on infection with H5N1 vs. H1N1, with genetic and

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L99

AUTOPHAGY MODULATION IN LUNG DISEASES

Cystic fibrosis
Clearance of protein aggregate
and regulating oxidative stress

LAM
Promoting survival and
proliferation of LAM cells

Lung infection
Promoting or inhibiting
replication of microbes in host
cells

Lung cancer

Xenophagy

Aggrephagy

Promoting survival and


proliferation of cancer cells

Sepsis

Tumor suppression

Mitophagy

Regulating inflammatory
response (i.e. inflammasome)

Lipophagy

COPD
Promoting apoptosis

PAH
IPF

Fig. 3. Diverse effects of autophagy in lung


diseases. The functional modes of autophagy
at cellular levels include eliminating intracellular microbes (xenophagy), dysfunctional
mitochondria (mitophagy), and protein aggregate (aggrephagy). Autophagy also regulates lipid metabolism (lipophagy). Autophagy is involved in pathogenesis of various lung diseases. The roles of autophagy in
each disease are shown. In lung cancer, autophagy may prevent tumorigenesis, whereas
autophagy can promote survival and proliferation of tumor cells. In infectious diseases,
the roles of autophagy are dependent on microbes. Autophagy may promote bacterial
killing and inhibit intracellular survival of
microbes such as Mycobacterium tuberculosis. On the other hand, autophagy may promote replication and survival of microbes
such as H5N1.

Regulating vascular cell


proliferation

Regulating fibroblast
activation and proliferation

biochemical inhibition of autophagy rescue in mice infected


with H5N1 (109). Since chloroquine also has autophagyindependent pharmacological effects such as anti-inflammatory property, the effect of chloroquine on diseases including
tumor may not be entirely explained by modulating autophagy
(120). Although further studies are needed to evaluate the
off-target effects of chloroquine on diseases, these reports
suggest that autophagy modulation by clinically used drugs
may be more accessible to study in clinical trials because of
their favorable safety profiles. The details of ongoing clinical
trial targeting modulating autophagy are listed (http://www.
clinicaltrials.gov/ct2/results?termautophagy&SearchSearch).
Autophagy Modulators
Given the important association between autophagy and
human lung diseases, it is worthwhile to review whether
modulating autophagy can serve as potent therapeutic targets
for various lung diseases. Table 4 demonstrates a list of
autophagy modulating drugs that are clinically used or being
studied in clinical trials. Table 5 shows conventional and newly
developed compounds or molecules modulating autophagy
activity. There are several target signaling pathways wherein
autophagy activity is modulated by drugs.
mTOR signaling pathways. The mTOR is one of the main
targets for modulating autophagy by drugs and forms two
distinct complexes to regulate autophagy: mTOR complex 1
(mTORC1) and mTOR complex 2 (mTORC2) (94) (Fig. 2,
left). Rapamycin and its analogs inhibit mTORC1, a main
complex responsible for autophagy regulation, and promote
autophagy induction (92, 95, 121). A recently identified com-

pound Torin1 directly inhibits both mTORC1 and mTORC2


and induces greater autophagy than does rapamycin (115). Of
note, another new class of drugs has dual targets in autophagy
pathways. PI-103 hydrochloride inhibits both mTOR and class
I phosphoinositide 3-kinase (class I PI3K), suggesting an
effective autophagy inducer (22).
mTOR-independent pathways. Figure 2, right, shows mTORindependent pathways.
PHOSPHOINOSITOL SIGNALING PATHWAY. Intracellular inositol
and IP3 levels negatively regulate autophagy (94). Drugs such
as lithium (103) or carbamazepine (103, 121) increase autophagy activity by decreasing intracellular level of inositol.
CA2-CAMP-PLC-IP3 PATHWAY. Intracellular Ca2 and cAMP
negatively regulate autophagy by increasing intracellular inositol level (94). In addition, calpain activated by elevated level
of intracellular Ca2 inhibits autophagy by cleavage of Atg5
(94, 99). Antihypertensive drugs including Ca2 receptor
blockers (9, 121, 129) or imidazolin receptor agonists (98, 121)
induce autophagy by decreasing cAMP.
Degradation steps of autophagy. Inhibiting formation of
autolysosome and lysosomal protease are also important targets to inhibit autophagy (Fig. 1). Chloroquine is a lysosomotropic and inhibits the fusion of autophagosome and lysosome
(4, 12). Cystatin B is a potent inhibitor of cystatin protease
(cathepsin B) in lysosome (99).
Other pathways. BCL-2 homology 3 (BH3) mimics inhibit
the interaction of BCL-2 and BCL-x with Beclin 1, an inhibitory complex for autophagy (66) (Fig. 1). Resveratrol induces
autophagy by activating sirtuin 1 and inhibits P70 S6 kinase (8,
43, 82). There are several compounds or drugs such as L-

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L100

AUTOPHAGY MODULATION IN LUNG DISEASES

Table 4. Drugs that modulate autophagy activity


Drugs

Autophagy inducers
Rapamycin and analogs

Clinical Application or Pharmacological Class

Amidarone

Immunosuppressant for preventing rejection


in organ transplant and coronary stent
coating for anti-proliferation
Class III antiarrhytmic

Nicosamide

Antiparasitic

EGFR antagonists (erltinib


hydrochlorine)
Resveratrol (Stilbenoids)

Anticancer

Suberoylanilide hydroxamic acid


Dexamethasone (glucocorticoide)

Supplementary diet. Secondary products of


heartwood formation in trees that act as
phytoalexins
Anticutaneous T cell lymphoma
Anti-inflammatory or immunosuppressant

Metformin

Antidiabetic

Verapamil, nicardipine, nimodipine


(L-type Ca2 channel blockers)
Sodium valproate

Vasodilator

Clonidine
Rilmenidine

Antihypertensive, treatment for ADHD and


anxiety/panic disorder
Antihypertensive

Lithium, L-690 330

Mood-stabilizing drug

Carbamazepine
Tamoxifen (estrogen receptor
antagonist)
Statin (HMG-CoA reductase
inhibitor)
Vitamin D3

Antiepileptics
Anti-breast cancer and anti-bipolar disorder

BH3 mimetics (ABT-737)


Carbon monoxide (at 250 ppm)

Minoxidil (potassium channel


opener)
Salbutamol (2 adrenergic receptor
agonist)
Isoniazid, pyrazinamide
Autophagy inhibitors
Azithromycin
Chloroquine/hydroxychloroquine
Vinblastine
IL-4
IL-13

Anticonvulsant

Lower cholesterol
Supplementary diet to reduce the risk of
fractures and falls
Undergoing clinical trial for ovarian cancer
Vasodilator, anti-inflammation and
antiproliferation Undergoing clinical
trials for idiopathic pulmonary fibrosis
and sepsis
Vasodilator

Mechanism of Action in Autophagy Pathway

References

Inhibit mTORC1

92, 95, 121

Inhibits mTORC1 or upstream in


mTOR pathway
Inhibits mTORC1 or upstream in
mTOR pathway
Inhibit PI3K-Akt-mTOR pathway

Activates sirtuin 1 (histone deacetylase)


and inhibits S6 kinase

8, 43, 82

Inhibits mTOR
Upregulates PML and Akt
dephosphorylation
Upregulates AMPK (and ULK1
phosphorylation)
Reduce intracellular Ca2 levels

13
31, 55

Inhibits histone deacetylase and reduces


intracellular Ca2 levels
Reduces cAMP levels (Imidazoline-1
receptor agonist)
Reduces cAMP levels (imidazoline-1
receptor agonist)
Inhibit IMPase and reduce inositol and
IP3 levels
Reduces inositol and IP3 levels
Accumulation of sterol, Increases
Beclin 1
Depletes geranylgeranyl diphosphate

9
30, 36

48, 70
9, 121, 129
103, 121
121
98, 121
103
103, 121
21, 99
83

Increases Beclin 1 expression by


upregulating cathelicidin
Disrupts interaction between the BH3
domain of Beclin 1 and the
antiapoptotic proteins BCL-2
Increases mitochondrial reactive oxygen
species

128

Unknown

121

66
58

Treatment for asthma by smooth muscle


relaxant
Antibiotics for tuberculosis

Unknown
AMPK and intracellular Ca
dependent

49

Macrolide antibiotic

Inhibits fusion of autophagosome and


lysosome
Inhibits fusion of autophagosome and
lysosome

96

Inhibits microtubule formation


Activates Akt and STAT6

89
37

Activates Akt and STAT6

37

Treatment for RA, SLE, and malaria


Undergoing clinical trials for various
cancers
Anticancer
Undergoing clinical trial for tuberculosis by
blocking IL-4
Undergoing clinical trial for advanced
cancers

6
2

4, 12

mTOR, mammalian target of rapamycin; mTORC1, mTOR complex 1; PI3K, phosphoinositide 3-kinase; NF-B, nuclear factor-B, AMPK, AMP-activated
protein kinase: ULK1, UNC51-like kinase; cAMP, cyclic AMP; IP3, inositol-(1,4,5)-trisphosphate; IMPase, inositol monophosphatase; BH3, BCL-2 homology
3; BCL-2, B cell lymphoma 2; EGFR, epidermal growth factor receptor; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; STAT6, signal transduction and
transcription 6; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus; PML, promyelocytic leukemia protein; ADHD, attention deficit hyperactivity
disorder; IL-4, interleukin 4; IL-13, interleukin 13. For details of clinical trials, please see the website http://www.clinicaltrials.gov/.

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
AUTOPHAGY MODULATION IN LUNG DISEASES

L101

Table 5. Molecules or compounds that modulate autophagy activity


Molecules or Compounds

Autophagy activators
Torin1
PP242
PI103 hydrochloride
2=,5=- Dideoxyadenosine
Xestospongin B
Spermidine
Tat-beclin1
Calpastatin, calpeptin
Trehalose (an -linked disaccharide
L-NAME (inhibition of NOS and decrease of NO
production)
GGTi-298 (inhibition of geranylgeranyl transferase 1)
Autophagy inhibitors
Spautin-1
3-MA
Pepstatin A
Cystatin B
Leupeptin
Bafilomycin A1
Nocodazole

Mechanism of Action in Autophagy Pathway

References

Directly inhibits both mTORC1 and mTORC2


Inhibits mTORC1
Highly selective class I PI3K inhibitor and ATP-competitive mTOR inhibitor
Reduces cAMP levels (Adenynyl cyclase inhibitor)
IP3R antagonist
Postulated to affect expression of Atg genes
A cell permeable peptide derived from a region (267284) of Beclin 1
Inhibit calpain
mTOR independent
Unknown

115
26
22
121
117
24
107
121
102
104

P53 dependent

29

Lowers Beclin 1 levels by promoting its ubiquitination


Inhibits class III PI3K
Inhibits aspartyl protease (cathepsin D)
Inhibits cystatine protease (cathepsin B)
Inhibits serine and cysteine proteases
Inhibits V-ATPase; Inhibits fusion of autophagosome and lysosome
Inhibits microtubule formation

63
100
113
99
38
92, 123
97, 119

IP3R, inositol-(1,4,5)-trisphosphate receptor; L-NAME, N-L-arginine methyl ester; 3-MA, 3-methyladenine; NO, nitric oxide; NOS, nitric oxide synthase; Atg,
autophagy-related; V-ATPase, vacuolar ATPase.

NAME (104), trehalose (102), or 2-adrenergic receptor agonist (6) whose mechanisms to modulate autophagy are still
unclear (Table 4). Further studies will be needed to identify
concise pathways by which those drugs or compounds modulate autophagy.
Since there are several pathways regulating autophagy, it
is possible that some drugs can stimulate both proautophagy
and antiautophagy pathways. For example, although lithium
has been shown to activate autophagy via an mTOR-independent pathway, lithium also inhibits GSK-3, which can
activate mTOR and lead to inhibition of autophagy (27)
(Fig. 2). In addition, some drugs may inhibit Ca2 channel
(activating autophagy by mTOR-independent pathways) and
also inhibit EGFR receptor (inhibiting autophagy by activating mTOR), which offset both the cellular signaling
pathways against each other and result in no autophagy
activation being observed. Thus it is also important to
analyze the effect of drugs on the individual pathways to
regulate autophagy. These profiles may lead to developing
the combined use of different types of autophagy modulators such as lithium and rapamycin, which can gain the
greater activation of autophagy (27) (Fig. 2).
Autophagy Modulation as a Potential Therapeutic
Intervention for Lung Diseases
Although various classes of clinically used drug modulate
autophagy pathways (Table 4), it is still unclear in many cases
whether the therapeutic effects of those drugs arise from
autophagy or autophagy-independent pathways. Even if autophagy is involved, there is still an important question remaining whether modulating autophagy by drugs contributes to
their beneficial pharmacological effects. For example, metformin, one of the most commonly used drugs for treatment of
Type 2 diabetes, induces autophagy (48, 70). The role of
autophagy on the pathogenesis of diabetes is still controversial
(94). Antidiabetic effects of metformin may be mediated by

metformin-induced autophagy activation or conversely may be


compromised by the autophagy activation. Recent clinical
studies show new possible interventions using drugs for lung
diseases including COPD (3) and tuberculosis (67). Azithromycin, a macrolide antibiotic, inhibits the frequency of exacerbation of COPD (3). High-dose supplement of vitamin D3
(VitD3) reduces time of sputum culture conversion of Mycobacterium tuberculosis (Mbt) in patients with polymorphism of
VitD3 receptor (67). Although the concise mechanisms by
which these drugs exert the beneficial effects are still unclear,
they commonly modulate autophagy activity. The pharmacological effect of azithromycin is similar with bafilomycin A1,
a family of macrolide antibiotic and also a well-known autophagy inhibitor (96) (Fig. 1). The beneficial roles of azithromycin on COPD may be associated with the inhibitory effect of
azithromycin on autophagy, since autophagy gene deficiency
displays protective effects in a mouse model of COPD. In
contrast, VitD3 inhibits replication of Mbt by increasing autophagy activity (128). Interestingly, drugs can also increase
autophagy activity in specific pathological condition. Isoniazid, an antibiotic for Mbt, induces autophagy in macrophages
infected with Mbt, rather than in uninfected cells (49). Although further studies are needed to clarify how autophagy is
involved in the beneficial effects of those drugs, these reports
suggest that pharmacological manipulation on autophagy activity can serve as a potential therapeutic strategy in lung
diseases. In addition, investigating the effect of drugs on
autophagy activity can reveal the new mechanism of disease
pathogenesis and previously unknown pharmacological actions
of the drugs.
It is also important to explore the possibility to use autophagy modulators as an intervention for treatment of lung
diseases. For example, an intervention to enhance autophagy
activity may have beneficial effects in lung diseases such as
tuberculosis or CF, since the effective drugs reported for those
diseases activate autophagy (32, 67). However, the caution

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L102

AUTOPHAGY MODULATION IN LUNG DISEASES

should be taken for this strategy. Autophagy has diverse effects


depending on diseases or pathological conditions. Although
activation of autophagy is beneficial for Mbt infection, the
activated autophagy may also exert protective roles for microbes such as HIV or HBV (99). If patients with tuberculosis
have latent infections with HIV or HBV, it is possible that
autophagy activation promotes replication and survival of
those microbes in the patients (99). On the other hand, autophagy inducers may have the additive beneficial effects in
patients enrolled in clinical trials for treatment of tuberculosis,
such as patients with Parkinson disease, where autophagy plays
protective roles (99). Finally, therapeutic interventions by
modulating autophagy can add to the current therapeutic strategies, rather than be used solely. For example, a recent clinical
trial for lung cancer utilizes the combined interventions of
erlotinib, a tyrosine kinase inhibitor, and chloroquine (125).
The anticancer mechanism of erlotinib is likely to be autophagy independent (36, 57). Inhibiting autophagy by chloroquine enhances anticancer effect of erlotinib by minimizing
prosurvival effect of erlotinib-mediated autophagy in tumor
cells (36, 57, 120, 125). Modulating autophagy activity may
enhance the beneficial roles of autophagy or minimize the
adverse effect of autophagy on each disease. Furthermore,
adding autophagy modulators may improve the current pharmacological effect of drugs used for the treatment.
Although clinical trials for investigating the therapeutic
effects of autophagy modulators are most desirable to observe the usefulness of drugs, cohort studies to examine the
effect of autophagy modulator on patients prognosis would
be more accessible. For example, there are a number of
patients with COPD who use autophagy modulators such as
Ca2 blockers, statin, or metformin for their medication;
these drugs may influence patients quality of life, lung
function, and frequency of acute exacerbation. In addition,
these studies may help to identify drugs which are suitable
for the clinical trials.
CONCLUSION

Until now more than 20 different classes of drug used for


treatment have been identified as autophagy modulators (9,
129) (Table 4). Moreover, the new-class autophagy modulators
with improved specificity and effectiveness have been developed for human disease indications as potential therapeutics
(Table 5). Given the important roles of autophagy in various
lung diseases (Fig. 3 and Tables 2 and 3), utilizing autophagy
modulators may improve the therapeutic effects of the current
interventions in various lung diseases. However, there are also
several important points to keep in mind. Although similar
situations are observed in other medical interventions, effective
and less-invasive methods to monitor autophagy activity for
patients (e.g., biomarkers) are not currently available. Secondly, since autophagy is such a fundamental cellular process
that affects various pathophysiological conditions, it is plausible that modulating autophagy causes other untoward effects
that may be clinically deleterious. Finally, the autophagy modulators listed in Tables 4 and 5 may have off-target effects in
addition to modulating autophagy. Nonetheless, recent clinical
studies using autophagy modulators (3, 32, 67) and ongoing
clinical trials (more than 15 active trials targeting autophagy)
still suggest that autophagy modulators unravel new avenues in

therapeutic interventions of various human diseases including


lung diseases. Although further development of assays for
autophagy activity and drugs with improved selectivity are
needed, the day when we use autophagy-modifying drugs to
treat lung disease may come sooner than later.
ACKNOWLEDGMENTS
The authors thank Jeffrey Adam Haspel for critical review of this manuscript.
GRANTS
This study is supported by NIH T32HL007633-27 (K. Nakahira) and NIH
PO1 HL105339 (A. M. K. Choi) and R01 HL05530 (A. M. K. Choi).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the author(s).
AUTHOR CONTRIBUTIONS
K.N. and A.M.K.C. conception and design of research; K.N. drafted
manuscript; K.N. and A.M.K.C. edited and revised manuscript; K.N. and
A.M.K.C. approved final version of manuscript.
REFERENCES
1. Abdulrahman BA, Khweek AA, Akhter A, Caution K, Kotrange S,
Abdelaziz DH, Newland C, Rosales-Reyes R, Kopp B, McCoy K,
Montione R, Schlesinger LS, Gavrilin MA, Wewers MD, Valvano
MA, Amer AO. Autophagy stimulation by rapamycin suppresses lung
inflammation and infection by Burkholderia cenocepacia in a model of
cystic fibrosis. Autophagy 7: 1359 1370, 2011.
2. Abdulrahman BA, Khweek AA, Akhter A, Caution K, Tazi M,
Hassan H, Zhang Y, Rowland PD, Malhotra S, Aeffner F, Davis IC,
Valvano MA, Amer AO. Depletion of the ubiquitin-binding adaptor
molecule SQSTM1/p62 from macrophages harboring cftr DeltaF508
mutation improves the delivery of Burkholderia cenocepacia to the
autophagic machinery. J Biol Chem 288: 2049 2058, 2013.
3. Albert RK, Connett J, Bailey WC, Casaburi R, Cooper JA Jr, Criner
GJ, Curtis JL, Dransfield MT, Han MK, Lazarus SC, Make B,
Marchetti N, Martinez FJ, Madinger NE, McEvoy C, Niewoehner
DE, Porsasz J, Price CS, Reilly J, Scanlon PD, Sciurba FC, Scharf
SM, Washko GR, Woodruff PG, Anthonisen NR. Azithromycin for
prevention of exacerbations of COPD. N Engl J Med 365: 689 698,
2011.
4. Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI,
Thomas-Tikhonenko A, Thompson CB. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma.
J Clin Invest 117: 326 336, 2007.
5. An CH, Wang XM, Lam HC, Ifedigbo E, Washko GR, Ryter SW,
Choi AM. TLR4 deficiency promotes autophagy during cigarette smokeinduced pulmonary emphysema. Am J Physiol Lung Cell Mol Physiol
303: L748 L757, 2012.
6. Aranguiz-Urroz P, Canales J, Copaja M, Troncoso R, Vicencio JM,
Carrillo C, Lara H, Lavandero S, Diaz-Araya G. Beta(2)-adrenergic
receptor regulates cardiac fibroblast autophagy and collagen degradation.
Biochim Biophys Acta 1812: 2331, 2011.
7. Araya J, Kojima J, Takasaka N, Ito S, Fujii S, Hara H, Yanagisawa
H, Kobayashi K, Tsurushige C, Kawaishi M, Kamiya N, Hirano J,
Odaka M, Morikawa T, Nishimura SL, Kawabata Y, Hano H,
Nakayama K, Kuwano K. Insufficient autophagy in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 304: L56 L69, 2013.
8. Armour SM, Baur JA, Hsieh SN, Land-Bracha A, Thomas SM,
Sinclair DA. Inhibition of mammalian S6 kinase by resveratrol suppresses autophagy. Aging (Albany NY) 1: 515528, 2009.
9. Balgi AD, Fonseca BD, Donohue E, Tsang TC, Lajoie P, Proud CG,
Nabi IR, Roberge M. Screen for chemical modulators of autophagy
reveals novel therapeutic inhibitors of mTORC1 signaling. PLoS One 4:
e7124, 2009.
10. Bjorkoy G, Lamark T, Pankiv S, Overvatn A, Brech A, Johansen T.
Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol
452: 181197, 2009.

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
AUTOPHAGY MODULATION IN LUNG DISEASES
11. Bodas M, Min T, Vij N. Critical role of CFTR-dependent lipid rafts in
cigarette smoke-induced lung epithelial injury. Am J Physiol Lung Cell
Mol Physiol 300: L811L820, 2011.
12. Boya P, Gonzalez-Polo RA, Casares N, Perfettini JL, Dessen P,
Larochette N, Metivier D, Meley D, Souquere S, Yoshimori T,
Pierron G, Codogno P, Kroemer G. Inhibition of macroautophagy
triggers apoptosis. Mol Cell Biol 25: 10251040, 2005.
13. Cao Q, Yu C, Xue R, Hsueh W, Pan P, Chen Z, Wang S, McNutt M,
Gu J. Autophagy induced by suberoylanilide hydroxamic acid in Hela S3
cells involves inhibition of protein kinase B and up-regulation of Beclin
1. Int J Biochem Cell Biol 40: 272283, 2008.
14. Carchman EH, Rao J, Loughran PA, Rosengart MR, Zuckerbraun
BS. Heme oxygenase-1-mediated autophagy protects against hepatocyte
cell death and hepatic injury from infection/sepsis in mice. Hepatology
53: 20532062, 2011.
15. Chen ZH, Kim HP, Sciurba FC, Lee SJ, Feghali-Bostwick C, Stolz
DB, Dhir R, Landreneau RJ, Schuchert MJ, Yousem SA, Nakahira
K, Pilewski JM, Lee JS, Zhang Y, Ryter SW, Choi AM. Egr-1
regulates autophagy in cigarette smoke-induced chronic obstructive pulmonary disease. PLoS One 3: e3316, 2008.
16. Chen ZH, Lam HC, Jin Y, Kim HP, Cao J, Lee SJ, Ifedigbo E,
Parameswaran H, Ryter SW, Choi AM. Autophagy protein microtubule-associated protein 1 light chain-3B (LC3B) activates extrinsic apoptosis during cigarette smoke-induced emphysema. Proc Natl Acad Sci
USA 107: 18880 18885, 2010.
17. Cheong H, Lu C, Lindsten T, Thompson CB. Therapeutic targets in
cancer cell metabolism and autophagy. Nat Biotechnol 30: 671678,
2012.
18. Choi AM, Ryter SW, Levine B. Autophagy in human health and
disease. N Engl J Med 368: 18451846, 2013.
19. Conway KL, Kuballa P, Khor B, Zhang M, Shi HN, Virgin HW,
Xavier RJ. ATG5 regulates plasma cell differentiation. Autophagy 9:
2013.
20. Davis BK, Wen H, Ting JP. The inflammasome NLRs in immunity,
inflammation, and associated diseases. Annu Rev Immunol 29: 707735,
2011.
21. de Medina P, Silvente-Poirot S, Poirot M. Tamoxifen and AEBS
ligands induced apoptosis and autophagy in breast cancer cells through
the stimulation of sterol accumulation. Autophagy 5: 1066 1067, 2009.
22. Degtyarev M, De Maziere A, Orr C, Lin J, Lee BB, Tien JY, Prior
WW, van Dijk S, Wu H, Gray DC, Davis DP, Stern HM, Murray LJ,
Hoeflich KP, Klumperman J, Friedman LS, Lin K. Akt inhibition
promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic
agents. J Cell Biol 183: 101116, 2008.
23. Deretic V, Delgado M, Vergne I, Master S, De Haro S, Ponpuak M,
Singh S. Autophagy in immunity against mycobacterium tuberculosis: a
model system to dissect immunological roles of autophagy. Curr Top
Microbiol Immunol 335: 169 188, 2009.
24. Eisenberg T, Knauer H, Schauer A, Buttner S, Ruckenstuhl C,
Carmona-Gutierrez D, Ring J, Schroeder S, Magnes C, Antonacci L,
Fussi H, Deszcz L, Hartl R, Schraml E, Criollo A, Megalou E,
Weiskopf D, Laun P, Heeren G, Breitenbach M, Grubeck-Loebenstein B, Herker E, Fahrenkrog B, Frohlich KU, Sinner F, Tavernarakis N, Minois N, Kroemer G, Madeo F. Induction of autophagy by
spermidine promotes longevity. Nat Cell Biol 11: 13051314, 2009.
25. Ezaki J, Matsumoto N, Takeda-Ezaki M, Komatsu M, Takahashi K,
Hiraoka Y, Taka H, Fujimura T, Takehana K, Yoshida M, Iwata J,
Tanida I, Furuya N, Zheng DM, Tada N, Tanaka K, Kominami E,
Ueno T. Liver autophagy contributes to the maintenance of blood
glucose and amino acid levels. Autophagy 7: 727736, 2011.
26. Feldman ME, Apsel B, Uotila A, Loewith R, Knight ZA, Ruggero D,
Shokat KM. Active-site inhibitors of mTOR target rapamycin-resistant
outputs of mTORC1 and mTORC2. PLoS Biol 7: e38, 2009.
27. Fleming A, Noda T, Yoshimori T, Rubinsztein DC. Chemical modulators of autophagy as biological probes and potential therapeutics. Nat
Chem Biol 7: 9 17, 2011.
28. Fujii S, Hara H, Araya J, Takasaka N, Kojima J, Ito S, Minagawa S,
Yumino Y, Ishikawa T, Numata T, Kawaishi M, Hirano J, Odaka M,
Morikawa T, Nishimura S, Nakayama K, Kuwano K. Insufficient
autophagy promotes bronchial epithelial cell senescence in chronic obstructive pulmonary disease. Oncoimmunology 1: 630 641, 2012.
29. Ghavami S, Mutawe MM, Schaafsma D, Yeganeh B, Unruh H,
Klonisch T, Halayko AJ. Geranylgeranyl transferase 1 modulates au-

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.
40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

L103

tophagy and apoptosis in human airway smooth muscle. Am J Physiol


Lung Cell Mol Physiol 302: L420 L428, 2012.
Gorzalczany Y, Gilad Y, Amihai D, Hammel I, Sagi-Eisenberg R,
Merimsky O. Combining an EGFR directed tyrosine kinase inhibitor
with autophagy-inducing drugs: a beneficial strategy to combat non-small
cell lung cancer. Cancer Lett 310: 207215, 2011.
Grander D, Kharaziha P, Laane E, Pokrovskaja K, Panaretakis T.
Autophagy as the main means of cytotoxicity by glucocorticoids in
hematological malignancies. Autophagy 5: 1198 1200, 2009.
Grossmann RE, Zughaier SM, Kumari M, Seydafkan S, Lyles RH,
Liu S, Sueblinvong V, Schechter MS, Stecenko AA, Ziegler TR,
Tangpricha V. Pilot study of vitamin D supplementation in adults with
cystic fibrosis pulmonary exacerbation: a randomized, controlled trial.
Dermatoendocrinology 4: 191197, 2012.
Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, KarsliUzunbas G, Kamphorst JJ, Chen G, Lemons JM, Karantza V, Coller
HA, Dipaola RS, Gelinas C, Rabinowitz JD, White E. Activated Ras
requires autophagy to maintain oxidative metabolism and tumorigenesis.
Genes Dev 25: 460 470, 2011.
Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI,
Deretic V. Autophagy is a defense mechanism inhibiting BCG and
Mycobacterium tuberculosis survival in infected macrophages. Cell 119:
753766, 2004.
Hailey DW, Rambold AS, Satpute-Krishnan P, Mitra K, Sougrat R,
Kim PK, Lippincott-Schwartz J. Mitochondria supply membranes for
autophagosome biogenesis during starvation. Cell 141: 656 667, 2010.
Han W, Pan H, Chen Y, Sun J, Wang Y, Li J, Ge W, Feng L, Lin X,
Wang X, Jin H. EGFR tyrosine kinase inhibitors activate autophagy as
a cytoprotective response in human lung cancer cells. PLoS One 6:
e18691, 2011.
Harris J, De Haro SA, Master SS, Keane J, Roberts EA, Delgado M,
Deretic V. T helper 2 cytokines inhibit autophagic control of intracellular
Mycobacterium tuberculosis. Immunity 27: 505517, 2007.
Haspel J, Shaik RS, Ifedigbo E, Nakahira K, Dolinay T, Englert JA,
Choi AM. Characterization of macroautophagic flux in vivo using a
leupeptin-based assay. Autophagy 7: 629 642, 2011.
Henske EP, McCormack FX. Lymphangioleiomyomatosis a wolf in
sheeps clothing. J Clin Invest 122: 38073816, 2012.
Hsieh CH, Pai PY, Hsueh HW, Yuan SS, Hsieh YC. Complete
induction of autophagy is essential for cardioprotection in sepsis. Ann
Surg 253: 1190 1200, 2011.
Iwai-Kanai E, Yuan H, Huang C, Sayen MR, Perry-Garza CN, Kim
L, Gottlieb RA. A method to measure cardiac autophagic flux in vivo.
Autophagy 4: 322329, 2008.
Jagannath C, Lindsey DR, Dhandayuthapani S, Xu Y, Hunter RL Jr,
Eissa NT. Autophagy enhances the efficacy of BCG vaccine by increasing peptide presentation in mouse dendritic cells. Nat Med 15: 267276,
2009.
Jeong JK, Moon MH, Bae BC, Lee YJ, Seol JW, Kang HS, Kim JS,
Kang SJ, Park SY. Autophagy induced by resveratrol prevents human
prion protein-mediated neurotoxicity. Neurosci Res 73: 99 105, 2012.
Jiang H, White EJ, Rios-Vicil CI, Xu J, Gomez-Manzano C, Fueyo J.
Human adenovirus type 5 induces cell lysis through autophagy and
autophagy-triggered caspase activity. J Virol 85: 4720 4729, 2011.
Kambas K, Mitroulis I, Apostolidou E, Girod A, Chrysanthopoulou
A, Pneumatikos I, Skendros P, Kourtzelis I, Koffa M, Kotsianidis I,
Ritis K. Autophagy mediates the delivery of thrombogenic tissue factor
to neutrophil extracellular traps in human sepsis. PLoS One 7: e45427,
2012.
Kaminskyy VO, Piskunova T, Zborovskaya IB, Tchevkina EM,
Zhivotovsky B. Suppression of basal autophagy reduces lung cancer cell
proliferation and enhances caspase-dependent and -independent apoptosis by stimulating ROS formation. Autophagy 8: 2012.
Kim HP, Wang X, Chen ZH, Lee SJ, Huang MH, Wang Y, Ryter
SW, Choi AM. Autophagic proteins regulate cigarette smoke-induced
apoptosis: protective role of heme oxygenase-1. Autophagy 4: 887895,
2008.
Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate
autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13:
132141, 2011.
Kim JJ, Lee HM, Shin DM, Kim W, Yuk JM, Jin HS, Lee SH, Cha
GH, Kim JM, Lee ZW, Shin SJ, Yoo H, Park YK, Park JB, Chung
J, Yoshimori T, Jo EK. Host cell autophagy activated by antibiotics is

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L104

AUTOPHAGY MODULATION IN LUNG DISEASES

required for their effective antimycobacterial drug action. Cell Host


Microbe 11: 457468, 2012.
50. Klein KA, Jackson WT. Human rhinovirus 2 induces the autophagic
pathway and replicates more efficiently in autophagic cells. J Virol 85:
96519654, 2011.
51. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, AcevedoArozena A, Adeli K, Agholme L, Agnello M, Agostinis P, AguirreGhiso JA, Ahn HJ, Ait-Mohamed O, Ait-Si-Ali S, Akematsu T, Akira
S, Al-Younes HM, Al-Zeer MA, Albert ML, Albin RL, AlegreAbarrategui J, Aleo MF, Alirezaei M, Almasan A, Almonte-Becerril
M, Amano A, Amaravadi R, Amarnath S, Amer AO, AndrieuAbadie N, Anantharam V, Ann DK, Anoopkumar-Dukie S, Aoki H,
Apostolova N, Arancia G, Aris JP, Asanuma K, Asare NY, Ashida H,
Askanas V, Askew DS, Auberger P, Baba M, Backues SK, Baehrecke
EH, Bahr BA, Bai XY, Bailly Y, Baiocchi R, Baldini G, Balduini W,
Ballabio A, Bamber BA, Bampton ET, Banhegyi G, Bartholomew
CR, Bassham DC, Bast RC Jr, Batoko H, Bay BH, Beau I, Bechet
DM, Begley TJ, Behl C, Behrends C, Bekri S, Bellaire B, Bendall LJ,
Benetti L, Berliocchi L, Bernardi H, Bernassola F, Besteiro S,
Bhatia-Kissova I, Bi X, Biard-Piechaczyk M, Blum JS, Boise LH,
Bonaldo P, Boone DL, Bornhauser BC, Bortoluci KR, Bossis I, Bost
F, Bourquin JP, Boya P, Boyer-Guittaut M, Bozhkov PV, Brady NR,
Brancolini C, Brech A, Brenman JE, Brennand A, Bresnick EH,
Brest P, Bridges D, Bristol ML, Brookes PS, Brown EJ, Brumell JH,
Brunetti-Pierri N, Brunk UT, Bulman DE, Bultman SJ, Bultynck G,
Burbulla LF, Bursch W, Butchar JP, Buzgariu W, Bydlowski SP,
Cadwell K, Cahova M, Cai D, Cai J, Cai Q, Calabretta B, CalvoGarrido J, Camougrand N, Campanella M, Campos-Salinas J,
Candi E, Cao L, Caplan AB, Carding SR, Cardoso SM, Carew JS,
Carlin CR, Carmignac V, Carneiro LA, Carra S, Caruso RA, Casari
G, Casas C, Castino R, Cebollero E, Cecconi F, Celli J, Chaachouay
H, Chae HJ, Chai CY, Chan DC, Chan EY, Chang RC, Che CM,
Chen CC, Chen GC, Chen GQ, Chen M, Chen Q, Chen SS, Chen W,
Chen X, Chen YG, Chen Y, Chen YJ, Chen Z, Cheng A, Cheng CH,
Cheng Y, Cheong H, Cheong JH, Cherry S, Chess-Williams R,
Cheung ZH, Chevet E, Chiang HL, Chiarelli R, Chiba T, Chin LS,
Chiou SH, Chisari FV, Cho CH, Cho DH, Choi AM, Choi D, Choi
KS, Choi ME, Chouaib S, Choubey D, Choubey V, Chu CT, Chuang
TH, Chueh SH, Chun T, Chwae YJ, Chye ML, Ciarcia R, Ciriolo
MR, Clague MJ, Clark RS, Clarke PG, Clarke R, Codogno P, Coller
HA, Colombo MI, Comincini S, Condello M, Condorelli F, Cookson
MR, Coombs GH, Coppens I, Corbalan R, Cossart P, Costelli P,
Costes S, Coto-Montes A, Couve E, CoxonFP, Cregg JM, Crespo JL,
Cronje MJ, Cuervo AM, Cullen JJ, Czaja MJ, DAmelio M, Darfeuille-Michaud A, Davids LM, Davies FE, De Felici M, de Groot JF,
de Haan CA, De Martino L, De Milito A, De Tata V, Debnath J,
Degterev A, Dehay B, Delbridge LM, Demarchi F, Deng YZ, Dengjel
J, Dent P, Denton D, Deretic V, Desai SD, Devenish RJ, Di Gioacchino M, Di Paolo G, Di Pietro C, Diaz-Araya G, Diaz-Laviada I,
Diaz-Meco MT, Diaz-Nido J, Dikic I, Dinesh-Kumar SP, Ding WX,
Distelhorst CW, Diwan A, Djavaheri-Mergny M, Dokudovskaya S,
Dong Z, Dorsey FC, Dosenko V, Dowling JJ, Doxsey S, Dreux M,
Drew ME, Duan Q, Duchosal MA, Duff K, Dugail I, Durbeej M,
Duszenko M, Edelstein CL, Edinger AL, Egea G, Eichinger L, Eissa
NT, Ekmekcioglu S, El-Deiry WS, Elazar Z, Elgendy M, Ellerby LM,
Eng KE, Engelbrecht AM, Engelender S, Erenpreisa J, Escalante R,
Esclatine A, Eskelinen EL, Espert L, Espina V, Fan H, Fan J, Fan
QW, Fan Z, Fang S, Fang Y, Fanto M, Fanzani A, Farkas T, Farre
JC, Faure M, Fechheimer M, Feng CG, Feng J, Feng Q, Feng Y,
Fesus L, Feuer R, Figueiredo-Pereira ME, Fimia GM, Fingar DC,
Finkbeiner S, Finkel T, Finley KD, Fiorito F, Fisher EA, Fisher PB,
Flajolet M, Florez-McClure ML, Florio S, Fon EA, Fornai F, Fortunato F, Fotedar R, Fowler DH, Fox HS, Franco R, Frankel LB,
Fransen M, Fuentes JM, Fueyo J, Fujii J, Fujisaki K, Fujita E,
Fukuda M, Furukawa RH, Gaestel M, Gailly P, Gajewska M, Galliot
B, Galy V, Ganesh S, Ganetzky B, Ganley IG, Gao FB, Gao GF, Gao
J, Garcia L, Garcia-Manero G, Garcia-Marcos M, Garmyn M,
Gartel AL, Gatti E, Gautel M, Gawriluk TR, Gegg ME, Geng J,
Germain M, Gestwicki JE, Gewirtz DA, Ghavami S, Ghosh P,
Giammarioli AM, Giatromanolaki AN, Gibson SB, Gilkerson RW,
Ginger ML, Ginsberg HN, Golab J, Goligorsky MS, Golstein P,
Gomez-Manzano C, Goncu E, Gongora C, Gonzalez CD, Gonzalez
R, Gonzalez-Estevez C, Gonzalez-Polo RA, Gonzalez-Rey E, Gorbunov NV, Gorski S, Goruppi S, Gottlieb RA, Gozuacik D, Granato

GE, Grant GD, Green KN, Gregorc A, Gros F, Grose C, Grunt TW,
Gual P, Guan JL, Guan KL, Guichard SM, Gukovskaya AS, Gukovsky I, Gunst J, Gustafsson AB, Halayko AJ, Hale AN, Halonen SK,
Hamasaki M, Han F, Han T, Hancock MK, Hansen M, Harada H,
Harada M, Hardt SE, Harper JW, Harris AL, Harris J, Harris SD,
Hashimoto M, Haspel JA, Hayashi S, Hazelhurst LA, He C, He YW,
Hebert MJ, Heidenreich KA, Helfrich MH, Helgason GV, Henske
EP, Herman B, Herman PK, Hetz C, Hilfiker S, Hill JA, Hocking LJ,
Hofman P, Hofmann TG, Hohfeld J, Holyoake TL, Hong MH, Hood
DA, Hotamisligil GS, Houwerzijl EJ, Hoyer-Hansen M, Hu B, Hu
CA, Hu HM, Hua Y, Huang C, Huang J, Huang S, Huang WP,
Huber TB, Huh WK, Hung TH, Hupp TR, Hur GM, Hurley JB,
Hussain SN, Hussey PJ, Hwang JJ, Hwang S, Ichihara A, Ilkhanizadeh S, Inoki K, Into T, Iovane V, Iovanna JL, Ip NY, Isaka Y,
Ishida H, Isidoro C, Isobe K, Iwasaki A, Izquierdo M, Izumi Y,
Jaakkola PM, Jaattela M, Jackson GR, Jackson WT, Janji B,
Jendrach M, Jeon JH, Jeung EB, Jiang H, Jiang JX, Jiang M, Jiang
Q, Jiang X, Jimenez A, Jin M, Jin S, Joe CO, Johansen T, Johnson
DE, Johnson GV, Jones NL, Joseph B, Joseph SK, Joubert AM,
Juhasz G, Juillerat-Jeanneret L, Jung CH, Jung YK, Kaarniranta K,
Kaasik A, Kabuta T, Kadowaki M, Kagedal K, Kamada Y, Kaminskyy VO, Kampinga HH, Kanamori H, Kang C, Kang KB, Kang KI,
Kang R, Kang YA, Kanki T, Kanneganti TD, Kanno H, Kanthasamy
AG, Kanthasamy A, Karantza V, Kaushal GP, Kaushik S, Kawazoe
Y, Ke PY, Kehrl JH, Kelekar A, Kerkhoff C, Kessel DH, Khalil H,
Kiel JA, Kiger AA, Kihara A, Kim DR, Kim DH, Kim EK, Kim HR,
Kim JS, Kim JH, Kim JC, Kim JK, Kim PK, Kim SW, Kim YS, Kim
Y, Kimchi A, Kimmelman AC, King JS, Kinsella TJ, Kirkin V,
Kirshenbaum LA, Kitamoto K, Kitazato K, Klein L, Klimecki WT,
Klucken J, Knecht E, Ko BC, Koch JC, Koga H, Koh JY, Koh YH,
Koike M, Komatsu M, Kominami E, Kong HJ, Kong WJ, Korolchuk
VI, Kotake Y, Koukourakis MI, Kouri Flores JB, Kovacs AL, Kraft
C, Krainc D, Kramer H, Kretz-Remy C, Krichevsky AM, Kroemer
G, Kruger R, Krut O, Ktistakis NT, Kuan CY, Kucharczyk R,
Kumar A, Kumar R, Kumar S, Kundu M, Kung HJ, Kurz T, Kwon
HJ, La Spada AR, Lafont F, Lamark T, Landry J, Lane JD,
Lapaquette P, Laporte JF, Laszlo L, Lavandero S, Lavoie JN,
Layfield R, Lazo PA, Le W, Le Cam L, Ledbetter DJ, Lee AJ, Lee
BW, Lee GM, Lee J, Lee JH, Lee M, Lee MS, Lee SH, Leeuwenburgh C, Legembre P, Legouis R, Lehmann M, Lei HY, Lei QY, Leib
DA, Leiro J, Lemasters JJ, Lemoine A, Lesniak MS, Lev D, Levenson VV, Levine B, Levy E, Li F, Li JL, Li L, Li S, Li W, Li XJ, Li
YB, Li YP, Liang C, Liang Q, Liao YF, Liberski PP, Lieberman A,
Lim HJ, Lim KL, Lim K, Lin CF, Lin FC, Lin J, Lin JD, Lin K, Lin
WW, Lin WC, Lin YL, Linden R, Lingor P, Lippincott-Schwartz J,
Lisanti MP, Liton PB, Liu B, Liu CF, Liu K, Liu L, Liu QA, Liu W,
Liu YC, Liu Y, Lockshin RA, Lok CN, Lonial S, Loos B, LopezBerestein G, Lopez-Otin C, Lossi L, Lotze MT, Low P, Lu B, Lu Z,
Luciano F, Lukacs NW, Lund AH, Lynch-Day MA, Ma Y, Macian F,
MacKeigan JP, Macleod KF, Madeo F, Maiuri L, Maiuri MC,
Malagoli D, Malicdan MC, Malorni W, Man N, Mandelkow EM,
Manon S, Manov I, Mao K, Mao X, Mao Z, Marambaud P, Marazziti D, Marcel YL, Marchbank K, Marchetti P, Marciniak SJ,
Marcondes M, Mardi M, Marfe G, Marino G, Markaki M, Marten
MR, Martin SJ, Martinand-Mari C, Martinet W, Martinez-Vicente
M, Masini M, Matarrese P, Matsuo S, Matteoni R, Mayer A, Mazure
NM, McConkey DJ, McConnell MJ, McDermott C, McDonald C,
McInerney GM, McKenna SL, McLaughlin B, McLean PJ, McMaster CR, McQuibban GA, Meijer AJ, Meisler MH, Melendez A, Melia
TJ, Melino G, Mena MA, Menendez JA, Menna-Barreto RF, Menon
MB, Menzies FM, Mercer CA, Merighi A, Merry DE, Meschini S,
Meyer CG, Meyer TF, Miao CY, Miao JY, Michels PA, Michiels C,
Mijaljica D, Milojkovic A, Minucci S, Miracco C, Miranti CK,
Mitroulis I, Miyazawa K, Mizushima N, Mograbi B, Mohseni S,
Molero X, Mollereau B, Mollinedo F, Momoi T, Monastyrska I,
Monick MM, Monteiro MJ, Moore MN, Mora R, Moreau K,
Moreira PI, Moriyasu Y, Moscat J, Mostowy S, Mottram JC, Motyl
T, Moussa CE, Muller S, Munger K, Munz C, Murphy LO, Murphy
ME, Musaro A, Mysorekar I, Nagata E, Nagata K, Nahimana A,
Nair U, Nakagawa T, Nakahira K, Nakano H, Nakatogawa H,
Nanjundan M, Naqvi NI, Narendra DP, Narita M, Navarro M,
Nawrocki ST, Nazarko TY, Nemchenko A, Netea MG, Neufeld TP,
Ney PA, Nezis IP, Nguyen HP, Nie D, Nishino I, Nislow C, Nixon RA,
Noda T, Noegel AA, Nogalska A, Noguchi S, Notterpek L, Novak I,

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
AUTOPHAGY MODULATION IN LUNG DISEASES
Nozaki T, Nukina N, Nurnberger T, Nyfeler B, Obara K, Oberley
TD, Oddo S, Ogawa M, Ohashi T, Okamoto K, Oleinick NL, Oliver
FJ, Olsen LJ, Olsson S, Opota O, Osborne TF, Ostrander GK, Otsu
K, Ou JH, Ouimet M, Overholtzer M, Ozpolat B, Paganetti P,
Pagnini U, Pallet N, Palmer GE, Palumbo C, Pan T, Panaretakis T,
Pandey UB, Papackova Z, Papassideri I, Paris I, Park J, Park OK,
Parys JB, Parzych KR, Patschan S, Patterson C, Pattingre S,
Pawelek JM, Peng J, Perlmutter DH, Perrotta I, Perry G, Pervaiz S,
Peter M, Peters GJ, Petersen M, Petrovski G, Phang JM, Piacentini
M, Pierre P, Pierrefite-Carle V, Pierron G, Pinkas-Kramarski R,
Piras A, Piri N, Platanias LC, Poggeler S, Poirot M, Poletti A, Pous
C, Pozuelo-Rubio M, Praetorius-Ibba M, Prasad A, Prescott M,
Priault M, Produit-Zengaffinen N, Progulske-Fox A, Proikas-Cezanne T, Przedborski S, Przyklenk K, Puertollano R, Puyal J, Qian
SB, Qin L, Qin ZH, Quaggin SE, Raben N, Rabinowich H, Rabkin
SW, Rahman I, Rami A, Ramm G, Randall G, Randow F, Rao VA,
Rathmell JC, Ravikumar B, Ray SK, Reed BH, Reed JC, Reggiori F,
Regnier-Vigouroux A, Reichert AS, Reiners JJ Jr, Reiter RJ, Ren J,
Revuelta JL, Rhodes CJ, Ritis K, Rizzo E, Robbins J, Roberge M,
Roca H, Roccheri MC, Rocchi S, Rodemann HP, Rodriguez de
Cordoba S, Rohrer B, Roninson IB, Rosen K, Rost-Roszkowska
MM, Rouis M, Rouschop KM, Rovetta F, Rubin BP, Rubinsztein
DC, Ruckdeschel K, Rucker EB 3rd, Rudich A, Rudolf E, RuizOpazo N, Russo R, Rusten TE, Ryan KM, Ryter SW, Sabatini DM,
Sadoshima J, Saha T, Saitoh TSakagami H, Sakai Y, Salekdeh GH,
Salomoni P, Salvaterra PM, Salvesen G, Salvioli R, Sanchez AM,
Sanchez-Alcazar JA, Sanchez-Prieto R, Sandri M, Sankar U,
Sansanwal P, Santambrogio L, Saran S, Sarkar S, Sarwal M, Sasakawa C, Sasnauskiene A, Sass M, Sato K, Sato M, Schapira AH,
Scharl M, Schatzl HM, Scheper W, Schiaffino S, Schneider C,
Schneider ME, Schneider-Stock R, Schoenlein PV, Schorderet DF,
Schuller C, Schwartz GK, Scorrano L, Sealy L, Seglen PO, SeguraAguilar J, Seiliez I, Seleverstov O, Sell C, Seo JB, Separovic D,
Setaluri V, Setoguchi T, Settembre C, Shacka JJ, Shanmugam M,
Shapiro IM, Shaulian E, Shaw RJ, Shelhamer JH, Shen HM, Shen
WC, Sheng ZH, Shi Y, Shibuya K, Shidoji Y, Shieh JJ, Shih CM,
Shimada Y, Shimizu S, Shintani T, Shirihai OS, Shore GC, Sibirny
AA, Sidhu SB, Sikorska B, Silva-Zacarin EC, Simmons A, Simon
AK, Simon HU, Simone C, Simonsen A, Sinclair DA, Singh R, Sinha
D, Sinicrope FA, Sirko A, Siu PM, Sivridis E, Skop V, Skulachev VP,
Slack RS, Smaili SS, Smith DR, Soengas MS, Soldati T, Song X, Sood
AK, Soong TW, Sotgia F, Spector SA, Spies CD, Springer W,
Srinivasula SM, Stefanis L, Steffan JS, Stendel R, Stenmark H,
Stephanou A, Stern ST, Sternberg C, Stork B, Stralfors P, Subauste
CS, Sui X, Sulzer D, Sun J, Sun SY, Sun ZJ, Sung JJ, Suzuki K,
Suzuki T, Swanson MS, Swanton C, Sweeney ST, Sy LK, Szabadkai
G, Tabas I, Taegtmeyer H, Tafani M, Takacs-Vellai K, Takano Y,
Takegawa K, Takemura G, Takeshita F, Talbot NJ, Tan KS, Tanaka
K, Tang D, Tanida I, Tannous BA, Tavernarakis N, Taylor GS,
Taylor GA, Taylor JP, Terada LS, Terman A, Tettamanti G, Thevissen K, Thompson CB, Thorburn A, Thumm M, Tian F, Tian Y,
Tocchini-Valentini G, Tolkovsky AM, Tomino Y, Tonges L, Tooze
SA, Tournier C, Tower J, Towns R, Trajkovic V, Travassos LH, Tsai
TF, Tschan MP, Tsubata T, Tsung A, Turk B, Turner LS, Tyagi SC,
Uchiyama Y, Ueno T, Umekawa M, Umemiya-Shirafuji R, Unni VK,
Vaccaro MI, Valente EM, Van den Berghe G, van der Klei IJ, van
Doorn W, van Dyk LF, van Egmond M, van Grunsven LA, Vandenabeele P, Vandenberghe WP, Vanhorebeek I, Vaquero EC,
Velasco G, Vellai T, Vicencio JM, Vierstra RD, Vila M, Vindis C,
Viola G, Viscomi MT, Voitsekhovskaja OV, von Haefen C, Votruba
M, Wada K, Wade-Martins R, Walker CL, Walsh CM, Walter J,
Wan XB, Wang A, Wang C, Wang D, Wang F, Wang G, Wang H,
Wang HG, Wang HD, Wang J, Wang K, Wang M, Wang RC, Wang
X, Wang YJ, Wang Y, Wang Z, Wang ZC, Wansink DG, Ward DM,
Watada H, Waters SL, Webster P, Wei L, Weihl CC, Weiss WA,
Welford SM, Wen LP, Whitehouse CA, Whitton JL, Whitworth AJ,
Wileman T, Wiley JW, Wilkinson S, Willbold D, Williams RL,
Williamson PR, Wouters BG, Wu C, Wu DC, Wu WK, Wyttenbach
A, Xavier RJ, Xi Z, Xia P, Xiao G, Xie Z, Xu DZ, Xu J, Xu L, Xu X,
Yamamoto A, Yamashina S, Yamashita M, Yan X, Yanagida M,
Yang DS, Yang E, Yang JM, Yang SY, Yang W, Yang WY, Yang Z,
Yao MC, Yao TP, Yeganeh B, Yen WL, Yin JJ, Yin XM, Yoo OJ,
Yoon G, Yoon SY, Yorimitsu T, Yoshikawa Y, Yoshimori T, Yoshimoto K, You HJ, Youle RJ, Younes A, Yu L, Yu SW, Yu WH,

52.

53.

54.
55.

56.
57.
58.

59.

60.
61.

62.

63.

64.

65.

66.

67.

L105

Yuan ZM, Yue Z, Yun CH, Yuzaki M, Zabirnyk O, Silva-Zacarin E,


Zacks D, Zacksenhaus E, Zaffaroni N, Zakeri Z, Zeh HJ 3rd, Zeitlin
SO, Zhang H, Zhang HL, Zhang J, Zhang JP, Zhang L, Zhang MY,
Zhang XD, Zhao M, Zhao YF, Zhao Y, Zhao ZJ, Zheng X, Zhivotovsky B, Zhong Q, Zhou CZ, Zhu C, Zhu WG, Zhu XF, Zhu X, Zhu
Y, Zoladek T, Zong WX, Zorzano A, Zschocke J, and Zuckerbraun
B. Guidelines for the use and interpretation of assays for monitoring
autophagy. Autophagy 8: 445544, 2012.
Kominami E, Hashida S, Khairallah EA, Katunuma N. Sequestration
of cytoplasmic enzymes in an autophagic vacuole-lysosomal system
induced by injection of leupeptin. J Biol Chem 258: 60936100, 1983.
Kon M, Kiffin R, Koga H, Chapochnick J, Macian F, Varticovski L,
Cuervo AM. Chaperone-mediated autophagy is required for tumor
growth. Sci Transl Med 3: 109ra117, 2011.
Kuballa P, Nolte WM, Castoreno AB, Xavier RJ. Autophagy and the
immune system. Annu Rev Immunol 30: 611646, 2012.
Laane E, Tamm KP, Buentke E, Ito K, Kharaziha P, Oscarsson J,
Corcoran M, Bjorklund AC, Hultenby K, Lundin J, Heyman M,
Soderhall S, Mazur J, Porwit A, Pandolfi PP, Zhivotovsky B, Panaretakis T, Grander D. Cell death induced by dexamethasone in lymphoid leukemia is mediated through initiation of autophagy. Cell Death
Differ 16: 1018 1029, 2009.
Lamark T, Johansen T. Aggrephagy: selective disposal of protein
aggregates by macroautophagy. Int J Cell Biol 2012: 736905, 2012.
Lee JG, Wu R. Combination erlotinib-cisplatin and Atg3-mediated
autophagy in erlotinib resistant lung cancer. PLoS One 7: e48532, 2012.
Lee SJ, Ryter SW, Xu JF, Nakahira K, Kim HP, Choi AM, Kim YS.
Carbon monoxide activates autophagy via mitochondrial reactive oxygen
species formation. Am J Respir Cell Mol Biol 45: 867873, 2011.
Lee SJ, Smith A, Guo L, Alastalo TP, Li M, Sawada H, Liu X, Chen
ZH, Ifedigbo E, Jin Y, Feghali-Bostwick C, Ryter SW, Kim HP,
Rabinovitch M, Choi AM. Autophagic protein LC3B confers resistance
against hypoxia-induced pulmonary hypertension. Am J Respir Crit Care
Med 183: 649 658, 2011.
Levine B, Mizushima N, Virgin HW. Autophagy in immunity and
inflammation. Nature 469: 323335, 2011.
Li C, Liu H, Sun Y, Wang H, Guo F, Rao S, Deng J, Zhang Y, Miao
Y, Guo C, Meng J, Chen X, Li L, Li D, Xu H, Li B, Jiang C. PAMAM
nanoparticles promote acute lung injury by inducing autophagic cell
death through the Akt-TSC2-mTOR signaling pathway. J Mol Cell Biol
1: 3745, 2009.
Liu HL, Zhang YL, Yang N, Zhang YX, Liu XQ, Li CG, Zhao Y,
Wang YG, Zhang GG, Yang P, Guo F, Sun Y, Jiang CY. A
functionalized single-walled carbon nanotube-induced autophagic cell
death in human lung cells through Akt-TSC2-mTOR signaling. Cell
Death Dis 2: e159, 2011.
Liu J, Xia H, Kim M, Xu L, Li Y, Zhang L, Cai Y, Norberg HV,
Zhang T, Furuya T, Jin M, Zhu Z, Wang H, Yu J, Hao Y, Choi A,
Ke H, Ma D, Yuan J. Beclin1 controls the levels of p53 by regulating
the deubiquitination activity of USP10 and USP13. Cell 147: 223234,
2011.
Lo S, Yuan SS, Hsu C, Cheng YJ, Chang YF, Hsueh HW, Lee PH,
Hsieh YC. Lc3 Over-expression improves survival and attenuates lung
injury through increasing autophagosomal clearance in septic mice. Ann
Surg 257: 352363, 2013.
Luciani A, Villella VR, Esposito S, Brunetti-Pierri N, Medina D,
Settembre C, Gavina M, Pulze L, Giardino I, Pettoello-Mantovani
M, DApolito M, Guido S, Masliah E, Spencer B, Quaratino S, Raia
V, Ballabio A, Maiuri L. Defective CFTR induces aggresome formation
and lung inflammation in cystic fibrosis through ROS-mediated autophagy inhibition. Nat Cell Biol 12: 863875, 2010.
Malik SA, Orhon I, Morselli E, Criollo A, Shen S, Marino G,
BenYounes A, Benit P, Rustin P, Maiuri MC, Kroemer G. BH3
mimetics activate multiple pro-autophagic pathways. Oncogene 30:
3918 3929, 2011.
Martineau AR, Timms PM, Bothamley GH, Hanifa Y, Islam K,
Claxton AP, Packe GE, Moore-Gillon JC, Darmalingam M, Davidson RN, Milburn HJ, Baker LV, Barker RD, Woodward NJ, Venton
TR, Barnes KE, Mullett CJ, Coussens AK, Rutterford CM, Mein
CA, Davies GR, Wilkinson RJ, Nikolayevskyy V, Drobniewski FA,
Eldridge SM, Griffiths CJ. High-dose vitamin D(3) during intensivephase antimicrobial treatment of pulmonary tuberculosis: a double-blind
randomised controlled trial. Lancet 377: 242250, 2011.

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
L106

AUTOPHAGY MODULATION IN LUNG DISEASES

68. Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, Sou


YS, Saiki S, Kawajiri S, Sato F, Kimura M, Komatsu M, Hattori N,
Tanaka K. PINK1 stabilized by mitochondrial depolarization recruits
Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol 189: 211221, 2010.
69. McCormack FX, Inoue Y, Moss J, Singer LG, Strange C, Nakata K,
Barker AF, Chapman JT, Brantly ML, Stocks JM, Brown KK,
Lynch JP 3rd, Goldberg HJ, Young LR, Kinder BW, Downey GP,
Sullivan EJ, Colby TV, McKay RT, Cohen MM, Korbee L, TaveiraDaSilva AM, Lee HS, Krischer JP, Trapnell BC. Efficacy and safety
of sirolimus in lymphangioleiomyomatosis. N Engl J Med 364: 1595
1606, 2011.
70. Meley D, Bauvy C, Houben-Weerts JH, Dubbelhuis PF, Helmond
MT, Codogno P, Meijer AJ. AMP-activated protein kinase and the
regulation of autophagic proteolysis. J Biol Chem 281: 34870 34879,
2006.
71. Mi S, Li Z, Yang HZ, Liu H, Wang JP, Ma YG, Wang XX, Liu HZ,
Sun W, Hu ZW. Blocking IL-17A promotes the resolution of pulmonary
inflammation and fibrosis via TGF-beta1-dependent and -independent
mechanisms. J Immunol 187: 30033014, 2011.
72. Mizushima N. Methods for monitoring autophagy using GFP-LC3
transgenic mice. Methods Enzymol 452: 1323, 2009.
73. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues.
Cell 147: 728 741, 2011.
74. Mizushima N, Kuma A. Autophagosomes in GFP-LC3 transgenic mice.
Methods Mol Biol 445: 119 124, 2008.
75. Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting.
Autophagy 3: 542545, 2007.
76. Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in
autophagosome formation. Annu Rev Cell Dev Biol 27: 107132, 2011.
77. Monick MM, Powers LS, Walters K, Lovan N, Zhang M, Gerke A,
Hansdottir S, Hunninghake GW. Identification of an autophagy defect
in smokers alveolar macrophages. J Immunol 185: 54255435, 2010.
78. Morris S, Swanson MS, Lieberman A, Reed M, Yue Z, Lindell DM,
Lukacs NW. Autophagy-mediated dendritic cell activation is essential
for innate cytokine production and APC function with respiratory syncytial virus responses. J Immunol 187: 39533961, 2011.
79. Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC,
Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA,
Ryter SW, Choi AM. Autophagy proteins regulate innate immune
responses by inhibiting the release of mitochondrial DNA mediated by
the NALP3 inflammasome. Nat Immunol 12: 222230, 2011.
80. Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited
selectively to impaired mitochondria and promotes their autophagy. J
Cell Biol 183: 795803, 2008.
81. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J,
Cookson MR, Youle RJ. PINK1 is selectively stabilized on impaired
mitochondria to activate Parkin. PLoS Biol 8: e1000298, 2010.
82. Opipari AW Jr, Tan L, Boitano AE, Sorenson DR, Aurora A, Liu
JR. Resveratrol-induced autophagocytosis in ovarian cancer cells. Cancer Res 64: 696 703, 2004.
83. Parikh A, Childress C, Deitrick K, Lin Q, Rukstalis D, Yang W.
Statin-induced autophagy by inhibition of geranylgeranyl biosynthesis in
prostate cancer PC3 cells. Prostate 70: 971981, 2010.
84. Parkhitko A, Myachina F, Morrison TA, Hindi KM, Auricchio N,
Karbowniczek M, Wu JJ, Finkel T, Kwiatkowski DJ, Yu JJ, Henske
EP. Tumorigenesis in tuberous sclerosis complex is autophagy and
p62/sequestosome 1 (SQSTM1)-dependent. Proc Natl Acad Sci USA
108: 1245512460, 2011.
85. Patel AS, Lin L, Geyer A, Haspel JA, An CH, Cao J, Rosas IO,
Morse D. Autophagy in idiopathic pulmonary fibrosis. PLoS One 7:
e41394, 2012.
86. Paton NI, Lee L, Xu Y, Ooi EE, Cheung YB, Archuleta S, Wong G,
Wilder-Smith A. Chloroquine for influenza prevention: a randomised,
double-blind, placebo controlled trial. Lancet Infect Dis 11: 677683,
2011.
87. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N,
Packer M, Schneider MD, Levine B. Bcl-2 antiapoptotic proteins
inhibit Beclin 1-dependent autophagy. Cell 122: 927939, 2005.
88. Pengo N, Scolari M, Oliva L, Milan E, Mainoldi F, Raimondi A,
Fagioli C, Merlini A, Mariani E, Pasqualetto E, Orfanelli U, Ponzoni
M, Sitia R, Casola S, Cenci S. Plasma cells require autophagy for
sustainable immunoglobulin production. Nat Immunol 14: 298 305,
2013.

89. Punnonen EL, Reunanen H. Effects of vinblastine, leucine, and histidine, and 3-methyladenine on autophagy in Ehrlich ascites cells. Exp Mol
Pathol 52: 8797, 1990.
90. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J,
Eskelinen EL, Mizushima N, Ohsumi Y, Cattoretti G, Levine B.
Promotion of tumorigenesis by heterozygous disruption of the beclin 1
autophagy gene. J Clin Invest 112: 1809 1820, 2003.
91. Rabinowitz JD, White E. Autophagy and metabolism. Science 330:
1344 1348, 2010.
92. Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone proteins
with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 11: 11071117, 2002.
93. Ravikumar B, Moreau K, Jahreiss L, Puri C, Rubinsztein DC.
Plasma membrane contributes to the formation of pre-autophagosomal
structures. Nat Cell Biol 12: 747757, 2010.
94. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M,
Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S, Massey DC, Menzies FM, Moreau K, Narayanan U,
Renna M, Siddiqi FH, Underwood BR, Winslow AR, Rubinsztein
DC. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev 90: 13831435, 2010.
95. Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, Oroz LG,
Scaravilli F, Easton DF, Duden R, OKane CJ, Rubinsztein DC.
Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat
Genet 36: 585595, 2004.
96. Renna M, Schaffner C, Brown K, Shang S, Tamayo MH, Hegyi K,
Grimsey NJ, Cusens D, Coulter S, Cooper J, Bowden AR, Newton
SM, Kampmann B, Helm J, Jones A, Haworth CS, Basaraba RJ,
DeGroote MA, Ordway DJ, Rubinsztein DC, Floto RA. Azithromycin
blocks autophagy and may predispose cystic fibrosis patients to mycobacterial infection. J Clin Invest 121: 3554 3563, 2011.
97. Reunanen H, Marttinen M, Hirsimaki P. Effects of griseofulvin and
nocodazole on the accumulation of autophagic vacuoles in Ehrlich ascites
tumor cells. Exp Mol Pathol 48: 97102, 1988.
98. Rose C, Menzies FM, Renna M, Acevedo-Arozena A, Corrochano S,
Sadiq O, Brown SD, Rubinsztein DC. Rilmenidine attenuates toxicity
of polyglutamine expansions in a mouse model of Huntingtons disease.
Hum Mol Genet 19: 2144 2153, 2010.
99. Rubinsztein DC, Codogno P, Levine B. Autophagy modulation as a
potential therapeutic target for diverse diseases. Nat Rev Drug Discov 11:
709 730, 2012.
100. Rubinsztein DC, Gestwicki JE, Murphy LO, Klionsky DJ. Potential
therapeutic applications of autophagy. Nat Rev Drug Discov 6: 304 312,
2007.
101. Ryter SW, Nakahira K, Haspel JA, Choi AM. Autophagy in pulmonary diseases. Annu Rev Physiol 74: 377401, 2012.
102. Sarkar S, Davies JE, Huang Z, Tunnacliffe A, Rubinsztein DC.
Trehalose, a novel mTOR-independent autophagy enhancer, accelerates
the clearance of mutant huntingtin and alpha-synuclein. J Biol Chem 282:
56415652, 2007.
103. Sarkar S, Floto RA, Berger Z, Imarisio S, Cordenier A, Pasco M,
Cook LJ, Rubinsztein DC. Lithium induces autophagy by inhibiting
inositol monophosphatase. J Cell Biol 170: 11011111, 2005.
104. Sarkar S, Korolchuk VI, Renna M, Imarisio S, Fleming A, Williams
A, Garcia-Arencibia M, Rose C, Luo S, Underwood BR, Kroemer G,
OKane CJ, Rubinsztein DC. Complex inhibitory effects of nitric oxide
on autophagy. Mol Cell 43: 19 32, 2011.
105. Schnaith A, Kashkar H, Leggio SA, Addicks K, Kronke M, Krut O.
Staphylococcus aureus subvert autophagy for induction of caspaseindependent host cell death. J Biol Chem 282: 26952706, 2007.
106. Seglen PO, Overbye A, Saetre F. Sequestration assays for mammalian
autophagy. Methods Enzymol 452: 6383, 2009.
107. Shoji-Kawata S, Sumpter R, Leveno M, Campbell GR, Zou Z, Kinch
L, Wilkins AD, Sun Q, Pallauf K, Macduff D, Huerta C, Virgin HW,
Helms JB, Eerland R, Tooze SA, Xavier R, Lenschow DJ, Yamamoto
A, King D, Lichtarge O, Grishin NV, Spector SA, Kaloyanova DV,
Levine B. Identification of a candidate therapeutic autophagy-inducing
peptide. Nature 494: 201206, 2013.
108. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M,
Tanaka K, Cuervo AM, Czaja MJ. Autophagy regulates lipid metabolism. Nature 458: 11311135, 2009.
109. Sun Y, Li C, Shu Y, Ju X, Zou Z, Wang H, Rao S, Guo F, Liu H, Nan
W, Zhao Y, Yan Y, Tang J, Zhao C, Yang P, Liu K, Wang S, Lu H,

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Physiology in Medicine
AUTOPHAGY MODULATION IN LUNG DISEASES

110.
111.
112.

113.
114.

115.

116.
117.

118.

Li X, Tan L, Gao R, Song J, Gao X, Tian X, Qin Y, Xu KF, Li D, Jin


N, Jiang C. Inhibition of autophagy ameliorates acute lung injury caused
by avian influenza A H5N1 infection. Sci Signal 5: ra16, 2012.
Swanlund JM, Kregel KC, Oberley TD. Investigating autophagy:
quantitative morphometric analysis using electron microscopy. Autophagy 6: 270 277, 2010.
Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C, Waguri S,
Eishi Y, Hino O, Tanaka K, Mizushima N. Autophagy-deficient mice
develop multiple liver tumors. Genes Dev 25: 795800, 2011.
Tanaka A, Jin Y, Lee SJ, Zhang M, Kim HP, Stolz DB, Ryter SW,
Choi AM. Hyperoxia-induced LC3B interacts with the Fas apoptotic
pathway in epithelial cell death. Am J Respir Cell Mol Biol 46: 507514,
2012.
Tanida I, Minematsu-Ikeguchi N, Ueno T, Kominami E. Lysosomal
turnover, but not a cellular level, of endogenous LC3 is a marker for
autophagy. Autophagy 1: 84 91, 2005.
Teng RJ, Du J, Welak S, Guan T, Eis A, Shi Y, Konduri GG. Cross
talk between NADPH oxidase and autophagy in pulmonary artery endothelial cells with intrauterine persistent pulmonary hypertension. Am J
Physiol Lung Cell Mol Physiol 302: L651L663, 2012.
Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y, Reichling
LJ, Sim T, Sabatini DM, Gray NS. An ATP-competitive mammalian
target of rapamycin inhibitor reveals rapamycin-resistant functions of
mTORC1. J Biol Chem 284: 80238032, 2009.
Tooze SA, Yoshimori T. The origin of the autophagosomal membrane.
Nat Cell Biol 12: 831835, 2010.
Vicencio JM, Ortiz C, Criollo A, Jones AW, Kepp O, Galluzzi L,
Joza N, Vitale I, Morselli E, Tailler M, Castedo M, Maiuri MC,
Molgo J, Szabadkai G, Lavandero S, Kroemer G. The inositol
1,4,5-trisphosphate receptor regulates autophagy through its interaction
with Beclin 1. Cell Death Differ 16: 1006 1017, 2009.
Vives-Bauza C, Zhou C, Huang Y, Cui M, de Vries RL, Kim J, May
J, Tocilescu MA, Liu W, Ko HS, Magrane J, Moore DJ, Dawson VL,
Grailhe R, Dawson TM, Li C, Tieu K, Przedborski S. PINK1dependent recruitment of Parkin to mitochondria in mitophagy. Proc Natl
Acad Sci USA 107: 378 383, 2010.

L107

119. Webb JL, Ravikumar B, Rubinsztein DC. Microtubule disruption


inhibits autophagosome-lysosome fusion: implications for studying the
roles of aggresomes in polyglutamine diseases. Int J Biochem Cell Biol
36: 25412550, 2004.
120. White E. Deconvoluting the context-dependent role for autophagy in
cancer. Nat Rev Cancer 12: 401410, 2012.
121. Williams A, Sarkar S, Cuddon P, Ttofi EK, Saiki S, Siddiqi FH,
Jahreiss L, Fleming A, Pask D, Goldsmith P, OKane CJ, Floto RA,
Rubinsztein DC. Novel targets for Huntingtons disease in an mTORindependent autophagy pathway. Nat Chem Biol 4: 295305, 2008.
122. Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative
diseases. Nat Neurosci 13: 805811, 2010.
123. Yamamoto A, Tagawa Y, Yoshimori T, Moriyama Y, Masaki R,
Tashiro Y. Bafilomycin A1 prevents maturation of autophagic vacuoles
by inhibiting fusion between autophagosomes and lysosomes in rat
hepatoma cell line, H-4-II-E cells. Cell Struct Funct 23: 3342, 1998.
124. Yang HZ, Wang JP, Mi S, Liu HZ, Cui B, Yan HM, Yan J, Li Z, Liu
H, Hua F, Lu W, Hu ZW. TLR4 activity is required in the resolution of
pulmonary inflammation and fibrosis after acute and chronic lung injury.
Am J Pathol 180: 275292, 2012.
125. Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in
cancer: therapeutic implications. Mol Cancer Ther 10: 15331541, 2011.
126. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell
Biol 12: 9 14, 2011.
127. Yuan K, Huang C, Fox J, Laturnus D, Carlson E, Zhang B, Yin Q,
Gao H, Wu M. Autophagy plays an essential role in the clearance of
Pseudomonas aeruginosa by alveolar macrophages. J Cell Sci 125:
507515, 2012.
128. Yuk JM, Shin DM, Lee HM, Yang CS, Jin HS, Kim KK, Lee ZW,
Lee SH, Kim JM, Jo EK. Vitamin D3 induces autophagy in human
monocytes/macrophages via cathelicidin. Cell Host Microbe 6: 231243,
2009.
129. Zhang L, Yu J, Pan H, Hu P, Hao Y, Cai W, Zhu H, Yu AD, Xie X,
Ma D, Yuan J. Small molecule regulators of autophagy identified by an
image-based high-throughput screen. Proc Natl Acad Sci USA 104:
1902319028, 2007.

AJP-Lung Cell Mol Physiol doi:10.1152/ajplung.00072.2013 www.ajplung.org

Potrebbero piacerti anche