You are on page 1of 16

Molecular Endocrinology. First published ahead of print December 18, 2012 as doi:10.1210/me.

2012-1260

ORIGINAL RESEARCH

Androgens Promote Prostate Cancer Cell Growth


through Induction of Autophagy

Yan Shi, Jenny J. Han, Jayantha B. Tennakoon, Fabiola F. Mehta,


Fatima A. Merchant, Alan R. Burns, Matthew K. Howe, Donald P. McDonnell,
and Daniel E. Frigo
Center for Nuclear Receptors and Cell Signaling (Y.S., J.J.H., J.B.T., F.F.M., D.E.F.), Departments of
Biology and Biochemistry (Y.S., J.J.H., J.B.T., F.F.M., D.E.F.) and Engineering Technology (F.A.M.), and
College of Optometry (A.R.B.), University of Houston, Houston, Texas 77204; and Department of
Pharmacology and Cancer Biology (M.K.H., D.P.M.), Duke University Medical Center, Durham, North
Carolina 27710

Androgens regulate both the physiological development of the prostate and the pathology of
prostatic diseases. However, the mechanisms by which androgens exert their regulatory activities
on these processes are poorly understood. In this study, we have determined that androgens
regulate overall cell metabolism and cell growth, in part, by increasing autophagy in prostate
cancer cells. Importantly, inhibition of autophagy using either pharmacological or molecular
inhibitors significantly abrogated androgen-induced prostate cancer cell growth. Mechanistically,
androgen-mediated autophagy appears to promote cell growth by augmenting intracellular lipid
accumulation, an effect previously demonstrated to be necessary for prostate cancer cell growth.
Further, autophagy and subsequent cell growth is potentiated, in part, by androgen-mediated
increases in reactive oxygen species. These findings demonstrate a role for increased fat metab-
olism and autophagy in prostatic neoplasias and highlight the potential of targeting underex-
plored metabolic pathways for the development of novel therapeutics. (Molecular Endocrinology
27: 0000 – 0000, 2013)

oth the basic physiology of the prostate and the de- tions, neurodegeneration, cancer, and aging (3, 4). Au-
B velopment of diseases such as benign prostatic hyper-
plasia and prostate cancer are influenced by androgens
tophagy is a process that involves the de novo formation
of a limiting membrane, called an autophagosome,
(1). Androgens are steroid hormones that function by around cytosolic components destined for degradation.
binding to and activating their target protein, the andro- Degradation occurs when the autophagosome fuses
gen receptor (AR). Ligand-mediated activation of AR with a lysosome to create an autophagolysosome or
controls the expression of a large number of genes, which autolysosome. Subsequently, the contents of the au-
together enable the same AR-ligand complex to exert reg- tolysosome are degraded by numerous acidic lysosomal
ulatory activities over a wide range of cellular processes hydrolases. In addition to removing damaged organ-
(2). Although AR and androgens are involved in the de- elles and proteins, this process can also facilitate the
cellular remodeling that is necessary for cells to carry
velopment and functioning of the normal prostate and are
out specialized functions (5, 6).
causally involved in prostate cancer progression, the
pathways responsible for these activities remain to be Abbreviations: ACC, Acetyl-CoA carboxylase; ACL, ATP citrate lyase; AMPK, AMP-acti-
determined. vated protein kinase; AR, androgen receptor; ATG7, autophagy-related protein 7; AV,
autophagic vesicle; CM-H2DCFDA, chloromethyl-2⬘,7⬘-dichlorodihydrofluorescein diace-
Macroautophagy, herein referred to as autophagy, is a tate; CoA, coenzyme A; CS-FBS, charcoal-stripped FBS; DAPI, 4⬘,6-diamidino-2-phenylin-
cellular process, which occurs in all eukaryotic cells and dole; DHT, dihydrotestosterone; Dox, doxycycline; E-64-d, (2S,3S)-trans-epoxysuccinyl-L-
leucylamido-3-methylbutane ethyl ester; eGFP, enhanced green fluorescent protein;
has been linked to numerous pathologies, including infec- FACS, fluorescence-activated cell sorting; FAS, fatty acid synthase; FBS, fetal bovine se-
rum; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; GFP, green fluorescent pro-
ISSN Print 0888-8809 ISSN Online 1944-9917 tein; LC3BI/II, microtubule-associated light chain 3 ␤ I/II; 3-MA, 3-methyladenine; qPCR,
Printed in U.S.A. quantitative RT-PCR; R1881, methyltrienolone; ROS, reactive oxygen species; shRNA,
Copyright © 2013 by The Endocrine Society short hairpin RNA; siRNA, small interfering RNA; TEM, transmission electron microscopy;
doi: 10.1210/me.2012-1260 Received August 7, 2012. Accepted November 2, 2012. TEMPOL, 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl; TG, triglyceride.

Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 1

Copyright (C) 2012 by The Endocrine Society


2 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

While autophagy is commonly thought of as a cata- zymes (36, 47, 48), it is not known whether androgens
bolic process that is activated either during times of star- may promote the formation of these lipid reservoirs by
vation or upon elevation of misfolded proteins, it is clear additional mechanisms that may therefore also be critical
that autophagy is also required for both normal and, for AR-mediated tumorigenesis. The goal of this study
more recently, tumorigenic processes that require ana- was to determine whether autophagy has a role in AR-
bolic activities (5–9). Initially, autophagy was thought to mediated prostate cancer cell growth and, if so, mecha-
function solely as a tumor suppressor largely because of nistically how this occurs. By identifying novel processes
data indicating that the genetic deletion of Beclin1, a com- that modulate prostate cancer cell growth, we hoped to
ponent of the autophagic machinery, increases the inci- discover new targets that could be exploited with future
dence of lung cancers, hepatocellular carcinomas, and therapeutics.
lymphomas (10, 11). Further, the commonly mutated tu-
mor suppressors p53 and PTEN have both been demon-
strated to activate autophagy (12, 13). Conversely, it has Materials and Methods
recently been demonstrated using both pharmacological
and molecular inhibition that autophagy is required for Reagents
the growth and viability of multiple types of cancers, in- Fetal bovine serum (FBS), L-glutamine, RPMI 1640 medium,
cluding breast, pancreatic, colon, ovarian, cervical, brain, DMEM, and chloromethyl-2⬘,7⬘-dichlorodihydrofluorescein di-
lung, skin, and lymphomas (14 –27). For example, chlo- acetate (CM-H2DCFDA) were obtained from Life Technologies
(Carlsbad, CA). Charcoal-stripped FBS (CS-FBS) was purchased
roquine, an inhibitor of autophagic flux, decreased tumor
from HyClone (Logan, UT). Polyclonal antibodies recognizing p27
formation in both xenograft and transgenic mouse mod- Kip1, p15 INK4B, long-chain acyl-CoA synthetase, ACL, lipin1,
els of pancreatic cancer (26). Additionally, genetic dele- microtubule-associated light chain 3 ␤ I/II (LC3BI/II), FAS, and
tion of either atg5 or atg7, core components of the au- monoclonal antibodies recognizing cyclin D1, cyclin D3, p21
tophagic machinery, suppressed the growth of Ras-driven Waf1/Cip1, CDK4, p18 INK4C, acetyl-CoA synthetase, and ACC
tumors (17). Thus, depending on the circumstances, au- were from Cell Signaling (Danvers, MA). Antiglyceraldehyde-3-
tophagy may function either as a tumor suppressor or a phosphate dehydrogenase (GAPDH) antibody, N-acetyl cysteine,
4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (TEMPOL),
tumor promoter. As a result, our understanding of
doxycycline (Dox), chloroquine, 3-methyladenine (3-MA), bafilo-
whether to target autophagy for therapeutic purposes, mycin A1, pepstatin A, and (2S,3S)-trans-epoxysuccinyl-L-leucyl-
and if so how, is limited. amido-3-methylbutane ethyl ester (E-64-d) were from Sigma (St.
It is currently unclear how exactly autophagy could Louis, MO). Dihydrotestosterone (DHT) and methyltrienolone
promote cancer. The prevailing thought is that autophagy (R1881) were purchased from Steraloids (Newport, RI) and
induces a dormant state similar to that of a stem cell (7, 8, PerkinElmer (Waltham, MA), respectively.
28). This state enables a level of independence that makes
Cell culture and plasmid transfection
the cell resistant to outside insults, such as hypoxia or
Androgen-sensitive LNCaP, VCaP, CWR22, and 22Rv1 hu-
even chemotherapeutic drugs. Alternatively, autophagy
man prostate cancer cell lines were obtained from American
may take on a more active role in cell growth. Since its Type Culture Collection (Manassas, VA) and maintained as
initial discovery in the 1960s, the major focus of studies recommended. Androgen-sensitive LAPC4 cells were a gift from
on autophagy has been on its role in protein and organelle Charles L. Sawyers (Memorial Sloan-Kettering Cancer Center,
turnover. However, several groups have recently demon- New York, NY) and maintained as previously described (49).
strated a role for autophagy in differentiation and corre- All experiments were carried out with cells of passage less than
25. All cells were authenticated by morphological inspection
sponding anabolic processes, such as skeletal muscle
and mycoplasma testing. Their response to androgens was au-
growth and lipogenesis (29 –35). Interestingly, altered thenticated using growth and reporter gene assays. Unless oth-
lipid metabolism has been demonstrated by multiple erwise noted, for all experiments, cells were steroid starved for
groups to play an important role in prostate cancer (36 – 72 h in steroid-reduced medium, that is, phenol red-free medium
40). Fatty acid synthase (FAS), a rate-limiting enzyme in containing 8% CS-FBS (15% CS-FBS for LAPC4 cells). For
de novo lipogenesis, is frequently overexpressed in pros- transfections, cells were transfected with the mCherry-green flu-
tate cancers (41– 44). Correspondingly, pharmacological orescent protein (GFP)-LC3 construct (50) using Lipofectamine
2000 (Life Technologies) according to the manufacturer’s
or molecular inhibition of either FAS or other lipogenic
instructions.
enzymes like acetyl-coenzyme A (CoA) carboxylase
(ACC) and ATP citrate lyase (ACL) suppresses both in Cell proliferation assay
vitro and in vivo tumor growth (45, 46). Although andro- Cell proliferation assays were carried out as previously de-
gens promote prostate cancer cell growth in part by in- scribed (49) by measuring the cellular DNA content using a
creasing the expression of several of these lipogenic en- FluoReporter Blue fluorometric double-stranded DNA Quanti-
Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 3

tation kit (Life Technologies) following the manufacturer’s time for 10 min in 2% gluteraldehyde, washed, and stained with
protocol. 1% uranyl acetate for 15 min. Samples were then embedded and
sectioned by the MD Anderson Cancer Center High Resolution
Cell viability assay Electron Microscopy Facility (Houston, TX). All grids were
Cell viability assays were carried out as previously described viewed on a JEOL 100CX II TEM at 80 kV.
(51) using a CellTiter-Blue Cell Viability Assay (Promega, Mad-
ison, WI) following the manufacturer’s protocol. Creation of stable cell lines using retrovirus
LNCaP and VCaP enhanced GFP (eGFP)-LC3B stable cells
Small interfering RNA (siRNA) transfection of were created using a previously described GFP-LC3 retroviral
human prostate cells vector (56) and previously described approach (53).
Stealth siRNA (Life Technologies) transfections were carried
out as previously described (52). The sequences of all siRNAs Fluorescence microscopy
used in this study are listed in Supplemental Table 1, published Cells were seeded on coverslips coated with poly-L-lysine,
on The Endocrine Society’s Journals Online web site at cultured and treated with vehicle (ethanol), DHT (10 nM), or
http://mend.endojournals.org. R1881 (10 nM) for 72 h. To visualize punctae in mCherry-GFP-
LC3 or eGFP-LC3-expressing cells, cultures were fixed in 4%
Western blot analysis paraformaldehyde, washed with PBS, and then stained with 50
Western blottings were conducted as previously described nM LysoTracker or 200 nM 4⬘,6-diamidino-2-phenylindole
(53). Densitometry was performed in Figure 2 using ImageJ (DAPI) as a counter stain and mounted in ProLong antifade
software (National Institutes of Health, Bethesda, MD). Here, (Life Technologies). Lipid droplets were stained using HCS Lip-
LC3BII levels (functional readout of levels of autophagy) were idTOX 1:1000 dilution from stock (Life Technologies). Of cau-
normalized to GAPDH (loading control) and not LC3BI, be- tionary note, BODIPY 493/503 stain was initially used to detect
cause the low immunoreactivity of LC3BI can lead to erroneous intracellular lipids but yielded false positive staining (data not
results. Hence, it is highly recommended that LC3BII levels be shown). This commonly used fluorescent marker of neutral lip-
normalized only to loading control to compare samples (54, 55). ids is highly susceptible to bleed through into the other fluores-
cent channels, unlike the LipidTOX stain that has a narrow
RNA isolation, cDNA preparation, and quantitative emission spectrum. Therefore, BODIPY 493/503 should be used
RT-PCR (qPCR) with caution when performing costains (especially in the green
RNA isolation, cDNA preparation, and qPCR were carried and red spectra). Stable cells were visualized using an Olympus
out as previously described using 36B4 as a control (49). The FV-1000 confocal microscope (Olympus, New York, NY) un-
sequences of the primers were as follows: autophagy-related der ⫻63 objective or a DeltaVision core deconvolution micro-
protein 7 (ATG7) forward, 5⬘-GATCCGGGATTTCTT- scope (Olympus IX-71) with a ⫻60 oil immersion objective lens
TCACG-3⬘ and reverse, 5⬘-CAGCAATGTAAGACCAGT- and analyzed using ImageJ software (National Institutes of
CAAGT-3⬘; and FAS forward, 5⬘-CGCTCTGGTTCATCT- Health). The Pearson correlation coefficient for lysosome and
GCTCTG-3⬘ and reverse, 5⬘-TCATCAAAGGTGCTCTCG- autophagosome colocalization was determined using the Soft-
TCTG-3⬘. The sequences of all other qPCR primers used in this Worx software (Applied Precision, Issaquah, WA).
study have been previously described (49). For rapid characterization of the tet-inducible ATG7 short
hairpin RNA (shRNA) stable cell line or reactive oxygen species
Transmission electron microscopy (TEM) and (ROS) imaging described below (ROS measurement), fluores-
cence microscopy was performed on plated and treated cells
immunogold TEM
using a Zeiss Axiovert 40 CFL fluorescence microscope with a
For basic TEM, cells were plated in six-well plates at 500,000
⫻10 objective (Zeiss, Oberkochen, Germany).
cells/well and treated as described above. Cells were then
washed twice in serum-free media and fixed for 3–5 min at room
temperature with 4% glutaraldehyde in 0.1 M cacodylate buffer Lipogenesis assay
(pH 7.0). Cells were gently scraped and centrifuged at 5000 ⫻ g For lipogenesis assays, cells were plated in CS-FBS-contain-
for 3 min in a swinging bucket rotor followed by additional ing media at 20,000 cells/well in poly-L-lysine-coated clear bot-
fixing for 1 h. Pellets were then embedded and sectioned by the tom/black walled 96-well plates. Cells were then either trans-
Duke University Department of Pathology’s Electron Micros- fected with or without indicated siRNAs as described above and
copy Core. Grids were viewed using a Philips/FEI CM 12 Trans- then treated for 3 d with vehicle, R1881, or chloroquine. Intra-
mission EM with AMT 2k x 2k digital camera. cellular triglyceride (TG) levels were then determined using an
For immunogold TEM, cells were plated in 24-well plates at AdipoRed Nile Red-based fluorometric assay according to the
100,000 cells/well and treated as described above. Preembed- manufacturer’s instructions (Lonza, Allendale, NJ). The relative
ding immunogold labeling was performed on samples fixed in fluorescence intensity was corrected for the background and
4% paraformaldehyde/0.1% glutaraldehyde after free alde- normalized to cell number using duplicate plates and the fluo-
hydes were quenched in 0.1 M sodium borohydride in PBS for 15 rescent DNA-binding dye assay described above. For stable LN-
min, blocked with 3% BSA, incubated with LC3 antibody (1: CaP cells with inducible ATG7-targeting shRNA, cells were
100) overnight at 4 C, washed extensively, and incubated with plated (10,000 cells/well) and then treated ⫾800 ng/ml Dox and
the gold-conjugated secondary antibody (1:100) overnight at 4 ⫾R1881 in 96-well plates containing CS-FBS. Neutral lipid con-
C. After further extensive washing, samples were fixed a second tent was then quantitated as described above.
4 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

Creation of inducible stable cell lines using VCaP human prostate cancer cells, which express endog-
lentivirus enous AR, we determined the effects of three different
Commercially available shRNAs (sequences are listed in Sup- inhibitors of autophagy (3-MA blocks autophagosome
plemental Table 1) in the pGIPZ backbone were obtained from formation; chloroquine and bafilomycin A1 block au-
Thermo Scientific (Lafayette, CO). An shRNA that we deter- tophagic flux) on androgen-mediated cell growth using
mined in preliminary experiments (data not shown) to give the
inhibitor concentrations that are both recommended and
best knockdown of ATG7 was used to produce lentivirus. For
inducible RNA interference experiments, shRNAs were sub- widely used in the field of autophagy (14 –19, 26, 34, 56,
cloned into the pINDUCER11 Dox-inducible lentiviral expres- 62– 66). In both cell models, all three inhibitors sup-
sion system by XhoI and MluI double digestion and ligation pressed androgen-mediated cell growth in a dose-depen-
(57). Lentiviruses were generated by cotransfecting the appro- dent manner (Fig. 1A). Although there were significant
priate shRNA constructs along with the packaging plasmids effects of the higher concentrations of chloroquine and
pMDL, pRSV-Rev, and pCMV-VSV-G into 293T cells. Viral
bafilomycin A1 on basal cell growth, these compounds
supernatants were harvested 48 h after transfection and com-
bined with 8 ␮g/ml polybrene to increase infection efficiency. had a more dramatic effect on androgen-mediated
Virus was then used to infect target cells, and 96 h later, target growth, in most instances suppressing growth to vehicle-
cells were GFP sorted using flow cytometry. Induction of alone-treated levels. Additional assays using these phar-
shRNA expression was done using 800 ng/ml Dox. macological inhibitors suggest that some of the effects on
cell growth, particularly at the higher concentrations, are
ROS measurement due to changes in cell viability (Supplemental Fig. 1A).
Intracellular ROS levels were determined using CM- Like most pharmacological modulators, potential off-
H2DCFDA and subsequent fluorescence-activated cell sorting
target effects of the autophagy inhibitors are a concern.
(FACS) analysis (58) or fluorescence microscopy described
above. Briefly, for FACS analysis, treated cells were rinsed in Therefore, to complement our pharmacological studies,
PBS buffer and exposed to 5 ␮M CM-H2DCFDA in the dark for we also blocked autophagy using molecular techniques.
30 min at 37 C. Subsequently, the cells were trypsinized and To determine the effects of molecular inhibition on an-
resuspended in PBS buffer. Intracellular fluorescence was then drogen-mediated prostate cell growth, we used siRNAs
quantified using a BD Aria II flow cytometer (Becton Dickinson, targeting three separate regions of ATG7, a core compo-
Franklin Lakes, NJ).
nent of the autophagic machinery that is required for LC3
To visualize intracellular ROS levels, cells were plated and
treated as described above for immunofluorescence microscopy. lipidation, a hallmark of functional autophagy (67). Dur-
Cells were then incubated with 2.5 ␮M CM2-H2DCFDA for 30 ing autophagy, the cytoplasmically diffuse form of LC3B,
min at 37 C and imaged according to previously described meth- LC3BI, is conjugated to a phospholipid to create a mod-
ods (59, 60). ified form, LC3BII, which localizes to autophagosomes
and autolysosomes. LC3BII, despite having a phospho-
Statistical analysis lipid conjugate, migrates faster on an SDS-PAGE gel and
Two-sample comparisons were performed using Student’s t hence can be distinguished from LC3BI using Western
tests. Multiple comparisons were performed using a one-way
blot analysis. Depletion of ATG7 levels (Supplemental
ANOVA and post hoc Dunnett’s test with GraphPad Prism,
Version 4 (GraphPad Software, Inc., San Diego, CA). Unless Fig. 1B) not only blocked the conversion of LC3BI to
otherwise noted, statistically significant changes were deter- LC3BII (Fig. 1B, right) but also inhibited androgen-me-
mined at the P ⬍ 0.05 level. diated cell growth approximately 50% (Fig. 1B, left). Im-
portantly, molecular inhibition of autophagy had mini-
mal effect on cell viability (Supplemental Fig. 1C).
Results Collectively, these findings indicate that autophagy is
needed for maximal prostate cell growth and, more spe-
Functional autophagy is required for maximal cifically, for androgen-mediated prostate cancer cell
androgen-mediated prostate cancer cell growth growth.
There has been a recent shift in our understanding of
the role of autophagy in cancer that suggests that au- Androgens increase autophagy
tophagy plays a major role in the progression of many Because both molecular and pharmacological inhibi-
types of cancers (14 –27). Given the importance of andro- tors suppressed androgen-mediated prostate cancer cell
gens and AR signaling in both the development of the growth, we wanted to know whether androgens could
prostate and the progression of prostate cancer (61), we induce autophagy. To assess cellular autophagy, three
sought to determine whether autophagy was important classical approaches are primarily used: 1) TEM (still of-
for androgen-mediated cell growth in cellular models of ten considered the gold standard), 2) LC3B immunofluo-
androgen-sensitive prostate cancer. Using LNCaP or rescence microscopy to assess LC3B relocalization, and 3)
Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 5

FIG. 1. Autophagy is required for androgen-mediated prostate cell growth. A, LNCaP or VCaP cells were treated for 7 d with vehicle or the
synthetic androgen R1881 in combination with vehicle or increasing concentrations of various inhibitors of autophagy (chloroquine: 1, 10, and 40
␮M; 3-MA: 1 and 10 mM; bafilomycin A1: 1, 10, and 100 nM). Cells were then lysed, and the relative number of cells was quantified using a
fluorescent DNA-binding dye. Each sample was performed in triplicate, and results from a representative experiment are shown. Results are
expressed as mean relative cell number ⫹ SE (n ⫽ 3). *, Significant changes from vehicle (no R1881)-treated cells; #, significant changes from
vehicle (no autophagy inhibitor)-treated cells. B, LNCaP cells were transfected with mock, a control siRNA or siRNAs targeting three separate
regions of the core autophagy molecule ATG7 and then treated for 7 d ⫾ R1881. Relative cell numbers were then quantitated as in A. Each
sample was performed in triplicate, and results from a representative experiment are shown. Results are expressed as mean relative cell number ⫹
SE (n ⫽ 3). *, Significant changes from vehicle (no R1881)-treated cells; #, significant changes from mock-transfected cells. D, Western blot analysis
of transfected LNCaP cell lysates to demonstrate ATG7 knockdown blocks autophagy as assessed by the decrease in the levels of LC3BII.

LC3B Western blot analysis to assess LC3BII levels. cantly increased GFP punctae formation, indicating
Therefore, we treated LNCaP cells for 3 d with vehicle or LC3B relocalization and hence autophagosome forma-
R1881 and assessed the formation of autophagic vesicles tion. Interestingly, in our hands, it took at least 2 d to
(AVs) (autophagosomes ⫹ autolysosomes) using TEM detect androgen-mediated autophagy, indicating an indi-
followed by blinded scoring (Fig. 2A). Using this rela- rect induction of autophagy by androgens (data not
tively low-throughput approach, we determined there to shown). Next, we wanted to confirm that androgens ex-
be a roughly 4-fold increase in AV levels after androgen erted a similar effect on autophagy in other androgen-
treatment. To further corroborate and expand our find- sensitive prostate cancer cell models. Using Western blot
ings, we created, using a retroviral approach, both LN- analysis to assess the formation of the lipid-modified
CaP and VCaP prostate cancer cells that stably expressed LC3BII form, it was determined that androgens increased
an eGFP-LC3B construct. We then treated these cells for autophagy in LNCaP, VCaP, LAPC4, CWR22, and
3 d with vehicle, R1881, or the physiological androgen 22Rv1 cells after R1881 treatment (Fig. 2C). To demon-
DHT and quantified GFP-LC3 punctae formation using strate the role of AR, LNCaP cells were treated with in-
immunofluorescence microscopy (Fig. 2B). In either LN- creasing concentrations of R1881 or DHT, and it was
CaP- or VCaP-GFP-LC3 cells, both the synthetic andro- observed that either androgen increased LC3BII levels in
gen R1881 or the physiological androgen DHT signifi- a dose-dependent manner (Fig. 2D). This androgen-me-
6 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

FIG. 2. Androgens increase autophagy in an AR-dependent manner in prostate cancer cells. A, LNCaP cells were treated for 3 d with vehicle
(ethanol) or 10 nM R1881. Cells were then fixed and examined by TEM. Left, AVs, autophagosomes and autolysosomes. Right, Quantification of
the number of AVs in vehicle- and R1881-treated cells. Data are presented as the mean number of AVs/cell (n ⫽ 19 cells each). *, Significant
changes from vehicle-treated cells. B, LNCaP or VCaP cells stably expressing an eGFP-LC3B fusion were treated for 3 d with vehicle, R1881 (10 nM),
or the endogenous androgen DHT (10 nM). Redistribution of eGFP-LC3B (green) was covisualized with a DAPI nuclear stain (blue) using
immunofluorescence confocal microscopy. Left, Representative pictures are shown. Scale bar, 20 ␮m. Right, Average number of punctae/cell (n ⫽
30 cells) was analyzed and plotted. *, Significant changes from vehicle-treated cells. C, Indicated prostate cancer cells were treated for 3 d with
vehicle or 10 nM R1881 and then subjected to Western blot analysis to determine LC3BI/II and GAPDH (loading control) levels. Below the images
are the average LC3BII densitometry values normalized to GAPDH ⫾ SE (n ⱖ 3). D, LNCaP cells were treated for 3 d with vehicle (⫺), 40 ␮M
chloroquine (CQ) (blocks autophagic flux leading to build up of LC3BII), or increasing doses (0.1, 1, and 10 nM) of R1881 or DHT. Cells were then
subjected to Western blot and densitometry analysis as in C. E, LNCaP cells were treated for 3 d ⫾ 10 ␮M Casodex and ⫾10 nM R1881. Cells were
then subjected to Western blot and densitometry analysis as in C.
Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 7

diated effect could be blocked by cotreatment with the Autophagy enhances androgen-mediated
antiandrogen Casodex, indicating that LC3B conversion intracellular lipid accumulation
is AR dependent (Fig. 2E). Although our data here, as well as studies performed
by other groups, indicate that autophagy can contribute
Androgens increase autophagic flux in prostate to cancer cell growth, it is still not fully understood mech-
cancer cells anistically how autophagy could augment prostate cancer
Although LC3 lipidation and relocalization is a marker cell growth. We initially tested whether blocking au-
for autophagosome formation, it does not report on the tophagy could influence the levels of various cell cycle
rate of autophagic flux. This is because LC3BII is de- regulatory proteins (Supplemental Fig. 3). In our hands,
graded over time by lysosomal enzymes and hence the suppression of autophagy did not consistently alter the
accumulation of LC3BII can result from either increased levels of cyclin D1, cyclin D3, p21 Waf1/Cip1, p27 Kip1,
autophagic entry or decreased autophagic flux (i.e. failure CDK4, p15 INK4B, or p18 INK4C. Although this is cer-
tainly not a comprehensive list of all potential growth
of autophagosomes to fuse with lysosomes) (68). There-
regulatory proteins, it suggested that perhaps autophagy
fore, we took three different approaches to assess the
was not directly influencing the cell cycle machinery.
impact of androgens on autophagic flux in prostate can-
Recently, several groups have demonstrated that pros-
cer cells. First, if functional autophagy is occurring, au-
tate cancers have elevated levels of intracellular lipids and
tophagosomes need to fuse with existing lysosomes to
that blocking the accumulation of these lipids inhibits
degrade their contents. To test this, we used our LNCaP tumorigenesis (36 – 40). Interestingly, although au-
and VCaP stably expressing eGFP-LC3B cell lines to dem- tophagy is classically thought of as a catabolic process, it
onstrate that androgens increased LC3B and lysosome has also become apparent that autophagy plays a role in
colocalization (Fig. 3A and Supplemental Fig. 2A). A sec- certain anabolic processes, such as lipogenesis (31–35).
ond, newer assay uses a tandem mCherry-GFP fusion pro- Therefore, we tested the hypothesis that androgen-medi-
tein linked to LC3 (69). This GFP signal is sensitive to the ated autophagy contributes to prostate cancer cell growth
acidic and/or proteolytic environment within the lyso- through altering lipid metabolism. Androgen treatment
some, unlike mCherry, which is more stable. Thus, punc- of either LNCaP or VCaP cells leads to a significant AR-
tate colocalization (yellow) of both GFP and mCherry dependent increase in intracellular levels of lipids as de-
indicates the formation of an early autophagosome that termined using either TEM or a Nile Red-based neutral
has not yet fused with a lysosome, whereas mCherry (red) lipid assay (Fig. 4A and Supplemental Fig. 4). Further,
punctate staining alone suggests LC3 has been delivered using immunofluorescence confocal microscopy with a
to the lysosomes, marking late autophagy. After transient HCS LipidTOX stain highly specific for neutral lipids, we
transfection of the mCherry-GFP-LC3 construct and an- determined that, although there was not a high level of
drogen treatment in either LNCaP or VCaP cells, we im- lipid and LC3 punctae colocalization, the androgen-me-
aged cells and quantitated the different LC3 punctate diated rise in intracellular lipid levels correlated with the
populations (Fig. 3B and Supplemental Fig. 2B). In both appearance of autophagy (Fig. 4B and Supplemental Fig.
cell lines, R1881 or DHT treatment lead to a significant 5). This is consistent with what has been observed by
other laboratories studying the link between autophagy
increase in GFP⫺mCherry⫹ (red) punctate formation, in-
and fat metabolism (35, 65, 70, 71). To get a better idea
dicating that androgens promote autophagic flux. Fi-
of the subcellular targets of autophagy, we performed
nally, to complement our immunofluorescence studies,
LC3 immunogold TEM with androgen-treated cells (Fig.
we assessed autophagic flux using a third approach that is
4C). After androgen treatment, LC3 immunogold stain-
based on Western blot analysis of LC3BII levels before
ing could be detected in discrete regions adjacent to lipid
and after lysosomal inhibition (Fig. 3C and Supplemental
droplets (left two pictures), consistent with findings from
Fig. 2C). Here, cells were pretreated with a cocktail of other laboratories (65), or localized to AVs (far right pic-
E-64-d and pepstatin A (Fig. 3C), specific inhibitors of tures). In vehicle-treated cells, LC3 staining was difficult
lysosomal proteases, or chloroquine (Supplemental Fig. to find and did not localize to lipid droplets since vehicle-
2C) before androgen treatment. After inhibitor treat- treated prostate cancer cells do not accumulate lipid drop-
ment, LC3BII levels accumulated due to lysosomal turn- lets (data not shown). Unexpectedly, we did not observe a
over being blocked. Importantly, androgen treatment fur- robust localization of LC3 density in the proximity of fat
ther increased LC3BII levels despite impaired lysosomal droplets, a consequence we believe of the ultrathin sec-
function, implying that AR-signaling promotes entry into tions required for TEM or the fixing procedure. There-
autophagy rather than blocking the autophagic flux. fore, to complement these qualitative studies, we used the
8 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

FIG. 3. Androgens increase autophagic flux in prostate cancer cells. A, LNCaP (representative pictures shown on left) or VCaP (representative
pictures shown in Supplemental Fig. 2A) cells stably expressing an eGFP-LC3B fusion were treated for 3 d with vehicle or 10 nM R1881. Cells were
then fixed and stained with LysoTracker to identify acidic organelles. eGFP-LC3B and LysoTracker colocalization was determined using
immunofluorescence confocal microscopy. Scale bars, 10 ␮m. Quantification of colocalization (right) was done using ImageJ software. B, LNCaPs
(representative pictures shown in Supplemental Fig. 2B) or VCaPs (representative pictures shown on left) were transfected with an mCherry-GFP-
LC3B construct and treated for 3 d with vehicle, DHT (10 nM), or R1881 (10 nM) to assess autophagic flux. Cells were then fixed and visualized
using a confocal immunofluorescence microscope. Scale bars, 10 ␮m. Right, Quantification of average total, GFP⫹mCherry⫹ (yellow, early
autophagy/autophagosome) and GFP⫺mCherry⫹ (red, late autophagy/autolysosomes) punctae/cell ⫾ SE for both LNCaP and VCaP cells (n ⬎ 10
cells each condition). *, Significant changes (P ⬍ 0.05) from vehicle-treated cells. **, Significant changes (P ⬍ 0.01) from vehicle-treated cells. C,
LNCaP cells were treated for 3 d ⫾ 10 ␮g/ml each of E-64-d and pepstatin A (specific lysosomal protease inhibitors) and ⫾ 10 nM R1881. Cells
were then subjected to Western blot analysis as in Fig. 2.

Nile Red-based assay to quantitate total neutral lipid levels quine (Fig. 4E and Supplemental Fig. 6B) significantly
after androgen treatment with or without molecular (ATG7 decreased neutral lipid levels after androgen treatment.
siRNA) or pharmacological (chloroquine) inhibition of au- To avoid the potential side effects that can occur with
tophagy. Both siRNAs targeting ATG7 (Fig. 4D and Sup- the use of chemical siRNAs, we created an LNCaP stable
plemental Fig. 6A) or increasing concentrations of chloro- cell line using the pINDUCER system (57). This platform
Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 9

Dox, promotes the pol II-driven ex-


pression of an shRNA transcript (in
our case ATG7-targeting) expressed as
a microRNA mimic (Fig. 5A). These
microRNA mimics have the advantage
of minimizing off-target RNA interfer-
ence effects compared with their classi-
cal shRNA predecessors (72). After the
creation and validation of this cell
model (Fig. 5, B and C), we confirmed
that the inducible knockdown of
ATG7 decreased both androgen-medi-
ated lipogenesis (Fig. 5D) and cell
growth (Fig. 5E). Collectively, these
data indicate that autophagy facili-
tated the formation of intracellular fat
depots. Importantly, this was not the
result of shutting down basic cellular
processes like transcription, because
knockdown of ATG7 had no effect on
the expression of classic AR-regulated
genes (Supplemental Fig. 7). Further, in-
hibition of autophagy using either induc-
ible ATG7 shRNA or E-64-d and pepsta-
tin A cotreatment did not prevent the
expression of FAS, a bona fide AR-target
and the rate-limiting step of de novo li-
pogenesis (Supplemental Fig. 8, A and B)
(42, 43, 48, 73, 74). Additionally,
knockdown of ATG7 did not affect the
levels of a sampling of other proteins
classically involved in lipid metabolism,
including ACC, ACL, acetyl-CoA syn-
FIG. 4. Autophagy promotes androgen-mediated lipid accumulation. A, LNCaP cells were thetase, acyl-CoA synthetase, and lipin1
treated for 3 d with vehicle (ethanol) or 10 nM R1881. Cells were then fixed and examined by (Supplemental Fig. 8C), suggesting that
TEM as in Fig. 2. B, LNCaP cells stably expressing an eGFP-LC3B fusion were treated for 3 d
with vehicle, DHT (10 nM), or R1881 (10 nM). Cells were then fixed and costained with autophagy may not regulate the levels of
LipidTOX (neutral lipid depots) and DAPI (nucleus). eGFP-LC3B, LipidHTX, and DAPI lipogenic enzymes.
colocalization was visualized using immunofluorescence confocal microscopy. Scale bars, 20
␮m; 5 ␮m for higher resolution images on far right. C, LNCaP or VCaP cells were treated as in
A and subjected to immunogold TEM to determine LC3B subcellular localization (yellow
Autophagy promotes androgen-
arrows). D, LNCaP cells were transfected with siRNAs as in Fig. 1B and treated for 3 d with
vehicle or 10 nM R1881. Intracellular TG levels were then determined using a fluorescent Nile mediated lipid accumulation
Red-based stain (AdipoRed) and normalized to cell numbers that were determined using independent of FAS expression
duplicate plates that were instead subjected to the fluorescent DNA-binding dye described in Because inhibition of autophagy did
Fig. 1. Each sample was performed in triplicate, and results from a representative experiment
are shown. Results are expressed as TG levels normalized to cell numbers ⫹ SE (n ⫽ 4). *, not alter the levels of FAS or other li-
Significant changes from mock-transfected cells. E, LNCaP cells were treated with vehicle or pogenic enzymes (Supplemental Fig.
increasing concentrations of chloroquine (CQ) ⫾ 10 nM R1881 for 3 d. Intracellular TG levels 8), we asked the reciprocal question of
were then determined as described in D. Each sample was performed in triplicate and results
from a representative experiment are shown. Results are expressed as TG levels normalized to whether de novo lipogenesis could reg-
cell numbers ⫹ SE (n ⫽ 4). *, Significant changes from vehicle (no CQ)-treated cells. ulate autophagy. In other words, we
wanted to know whether the accumu-
is advantageous, because it uses a single vector to consti- lation of intracellular fat was the cue that stimulated au-
tutively express a third-generation reverse Tet transacti- tophagy. To test this, we knocked down FAS levels using
vator that, upon addition of tetracycline or its derivative three separate siRNAs and then treated cells with or with-
10 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

shown, CM2-H2DCF-DA and subse-


quent FACS analysis demonstrated
that treatment of either VCaP or LN-
CaP cells with androgens increased in-
tracellular ROS levels (Supplemental
Fig. 9). By adapting the CM2-
H2DCF-DA assay, we used basic fluo-
rescence microscopy to more rapidly
and easily assess relative ROS levels.
Using this approach, we demonstrated
that VCaP cells treated for 3 d with
R1881 have elevated ROS levels, an ef-
fect that could be blocked by the anti-
oxidant TEMPOL (Fig. 7A). Further-
more, treatment of prostate cancer
cells with either of the antioxidants
TEMPOL (Fig. 7B) or N-acetyl cys-
teine (Supplemental Fig. 10) was suffi-
cient to block androgen-mediated au-
tophagy. This corresponded with the
FIG. 5. Inducible expression of ATG7-targeting shRNA inhibits androgen-mediated lipid
accumulation and cell growth. A, Diagram of the pINDUCER11 construct used in this study. B
ability of antioxidants to block andro-
and C, To characterize the LNCaP ATG7 shRNA stable cells, cells were treated for 2 d ⫾ 800 gen-mediated lipid accumulation (Fig.
ng/ml Dox and subjected to fluorescence microscopy (B) or Western blot analysis (C). D, TG 7C). Importantly, blocking ROS pro-
levels were quantitated as described in Fig. 4 but using LNCaP ATG7 shRNA-inducible stable
duction, although having minimal ef-
cells cotreated for 3 d ⫾ Dox and ⫾ R1881. Results are expressed as TG levels normalized to
cell numbers ⫹ SE (n ⫽ 2). *, Significant changes from double vehicle-treated cells. E, Stable fect on basal cell growth, inhibited an-
cells were again cotreated but for 7 d followed by quantification of cell numbers using the drogen-mediated cell growth (Fig. 7D),
assay described in Fig. 1. Results are expressed as mean relative cell number ⫹ SE (n ⫽ 2). **, consistent with what has been demon-
Significant change (P ⬍ 0.01) from R1881 alone-treated cells.
strated before in LNCaP cells (48, 58,
76, 77). Taken together, these data
out R1881. Consistent with what others have shown (41– suggest that androgens increase cell growth, in part, by
43, 47, 74), knockdown of FAS severely impaired AR’s activating a complex metabolic shift that is signaled by
ability to increase intracellular fat levels (Fig. 6A) and cell the elevation of intracellular ROS levels.
growth (Fig. 6B). Despite the similarities to the knock-
down of ATG7, knockdown of FAS did not impact au-
tophagy as assessed by the Western blot analysis of Discussion
LC3BII levels (Fig. 6C). These findings suggest that au-
tophagy can promote intracellular lipid accumulation by The work presented here indicates that androgens, via
a mechanism that is at least partially independent of FAS AR, utilize autophagy to promote prostate cancer cell
activity. growth. Initially, these data were counterintuitive to us
because autophagy is usually considered to be a catabolic
Androgens promote autophagy, in part, through process that is activated during times of stress (78). This
the production of ROS contrasts with the established roles of androgens as ana-
Although we established the importance of autophagy bolic steroids. Indeed, previous studies have reported that
for androgen-mediated prostate cancer cell growth, we androgens can block autophagy in either LNCaP cells or
still did not understand how androgens increased au- PC-3 cells transiently transfected with AR (62, 79, 80).
tophagy. Previous work by Scherz-Shouval et al. (75) However, these studies looked at more rapid androgenic
demonstrated that elevated ROS levels led to increased effects under different conditions, during hypoxia or
autophagy. Given that androgens can increase ROS in complete serum starvation, conditions where androgens
prostate cancer cells to a level that potentiates cell growth do not optimally promote cell growth. Conversely, our
(48, 58, 76, 77), we decided to explore whether andro- data clearly indicate in multiple cellular models that when
gens could stimulate autophagy by increasing the intra- androgens promote cell growth, autophagy is activated.
cellular levels of ROS. Consistent with what others have Further, autophagy has been recently shown by multiple
Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 11

reported that androgens could block serum starvation-


induced autophagy at up to 24 h, longer treatments (i.e.
72 h) that increased cell survival also increased au-
tophagy. These findings indicate that when the extracel-
lular environment can provide a necessary level of sup-
port, androgen-treated cancer cells can utilize autophagy
for their benefit. Ultimately, we think that cancer cells,
not surprisingly, have the ability to adapt to their envi-
ronment. In the context of prostatic diseases, androgens
activate signaling pathways that facilitate the cell’s poten-
tial to use diverse nutrients, building blocks, and energy
sources for growth and survival. Androgen-mediated au-
tophagy may be one way in which a prostate cell can
redirect its efforts away from nonessential processes to-
ward processes it deems more necessary like growth
and/or survival.
The delayed induction demonstrated here suggests an
indirect mechanism by which AR signaling enhances au-
tophagy. Recent work by Scherz-Shouval et al. (75) show-
ing that elevated ROS levels activate autophagy, com-
bined with the previous work of several groups
demonstrating that androgen-mediated ROS generation
promotes prostate cancer cell growth (48, 58, 76, 77),
provided the rationale for us to test whether androgenic
regulation of autophagy and subsequent cell growth re-
quired ROS signaling (Fig. 7). The regulation of ROS
levels in cells is a double-edged sword. Although too
much ROS can trigger apoptosis, moderate levels pro-
mote cell signaling activities that are needed for both pro-
liferation and survival (81). In our experiments, elevated
ROS levels contributed to the androgen-induced au-
tophagy, intracellular lipid accumulation, and cell growth
(Fig. 7), indicating a role for ROS as a signaling molecule.
While ROS were required for the induction of au-
tophagy, we cannot exclude the possibility that other ini-
FIG. 6. Autophagy-mediated lipid accumulation is downstream of tiating mechanisms exist. There are many modulators of
FAS. LNCaP cells were transfected with mock, a negative scramble
autophagy, some of which are regulated by androgens.
control (siControl) or three siRNAs targeting separate regions of FAS
mRNA. Cells were then treated for 3 d with vehicle or 10 nM R1881. A, For example, we have previously shown that androgens
Cells were assayed for intracellular TG levels as described in Fig. 4D. increase AMP-activated protein kinase (AMPK) signaling
Each sample was performed in triplicate, and results from a through the expression of calcium/calmodulin-dependent
representative experiment are shown. Results are expressed as TG
levels normalized to cell numbers ⫹ SE (n ⫽ 2). *, Significant changes kinase kinase ␤ (53). Because AMPK is a known positive
from mock-transfected cells. B, Cells were transfected with mock, a regulator of autophagy (82), our initial hypothesis was
control siRNA, or siRNAs targeting three separate regions of FAS and that androgens increased autophagy through the up-reg-
then treated for 7 d ⫾ R1881. Relative cell numbers were then
quantitated as described in Fig. 1. Each sample was performed in
ulation of the calcium/calmodulin-dependent kinase ki-
triplicate, and results from a representative experiment are shown. nase ␤-AMPK cascade. However, preliminary studies us-
Results are expressed as mean relative cell number ⫹ SE (n ⫽ 3). *, ing siRNAs targeting AMPK have yielded inconclusive
Significant changes from mock-transfected cells. C, Lysates from
results (data not shown). Veeramani et al. (77) have re-
LNCaP cells transfected and treated the same as in A were subjected to
Western blot analysis. A representative blot is shown (n ⫽ 4). ported that androgens can increase ROS levels and sub-
sequent prostate cancer cell proliferation through an AR
groups to be required for the progression of various types nongenomic mechanism. While androgens may function
of cancers (14 –27), supporting a progrowth role. It through membrane signaling, given the delayed onset of
should be also noted that although Bennett et al. (79) autophagy after treatment and the stereotypical AR phar-
12 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

the fact that LC3B is typically found in


discrete locations on the lipid droplet
membranes (Fig. 4C) (35, 65). How-
ever, until more dynamic labeling stud-
ies can be performed, this process re-
mains an enigma.
One of the other major questions in
the field is how does AR-mediated fat
accumulation promote cell growth. To
date, several explanations have been
proposed. However, a dearth of exper-
imental evidence has kept this issue
largely unsolved. One theory is that the
elevated levels of fat can help control
cellular redox levels (42). Others have
postulated that the fat droplets may
serve as reservoirs that can be tapped
into whenever the cell needs lipid
building blocks to make new mem-
FIG. 7. Androgens promote autophagy-mediated cell growth, in part, through elevating branes during cell division. Although
intracellular ROS levels. A, VCaP cells were cotreated for 3 d ⫾ 1 mM TEMPOL and ⫾ 10 nM this is an attractive hypothesis, in pros-
R1881. Cells were then assayed for ROS production using CM-H2DCF-DA-based fluorescence
microscopy analysis. B, VCaP cells were treated as in A and subjected to Western blot analysis tate cancer, typically only a fraction of
for LC3B and GAPDH (loading control). C, VCaP cells were treated as in A and B. Cells were the cells are actively proliferating at
then assayed for intracellular TG levels as described in Fig. 4D. Each sample was performed in any one time, indicating that this might
triplicate, and results from a representative experiment are shown. Results are expressed as
TG levels normalized to cell numbers ⫹ SE (n ⫽ 2). *, Significant changes from double vehicle-
be only a part of the answer (41). An
treated cells. D, VCaP cells were again cotreated but for 7 d and then assayed for relative cell alternative hypothesis is that the intra-
numbers as described in Fig. 1. Each sample was performed in triplicate, and results from a cellular lipid accumulation is needed
representative experiment are shown. Results are expressed as mean relative cell number ⫹ SE
for various aspects of signal transduc-
(n ⫽ 3). *, Significant changes from double vehicle-treated cells.
tion. Many secondary messengers,
such as prostaglandins and phosphati-
macology we see (Fig. 2, D and E), it may be premature to dylinositols, are derived from neutral lipid scaffolds. Fur-
say genomic AR signaling does not play any role in the ther, many of the posttranslational modifications (e.g.
regulation of autophagy. Certainly, as our understanding palmitoylation and myristoylation) that are required for
of the regulation of autophagy advances, it may be worth- signal transduction cascades are lipid based. For example,
while to reexamine earlier microarray signatures to deter- the phosphoinositol 3-kinase-Akt signaling pathway,
mine any additional points of AR regulation. which plays a major role in prostate cancers, requires
We demonstrate here that androgen-mediated au- these lipid modifications to properly organize itself at the
tophagy promotes the accumulation of intracellular fats. plasma membrane and thus ensure efficient signaling
Although androgens are known to accumulate intracellu- (83). Additionally, androgens have been demonstrated to
lar fats through the expression of several enzymes in- potentiate prostate cancer cell proliferation by increasing
volved in de novo lipogenesis (36, 45– 48), it appears that the expression of squalene synthase, a lipogenic enzyme
autophagy may function independently of these tran- that contributes to membrane lipid raft formation (84).
scriptional events. Recent work in the field has demon- Another intriguing option is that the fat depots provide
strated a bona fide role for autophagy in fat depot forma- precursors for the production of androgens. Several stud-
tion. Adipose-specific atg5 or atg7 knockout mice are ies have indicated that one of the causes of castrate-resis-
lean and resistant to type 2 diabetes (31–34). In these tant prostate cancer is the up-regulation of enzymes in-
mice, it appears there is a severe defect in normal fat cell volved in local steroid hormone production (85– 87).
differentiation. Further, the MAP1-LC3 conjugation sys- Thus, even though circulating levels of androgens are neg-
tem is required for lipid droplet formation in both hepa- ligible following hormone therapy, the intratumoral con-
tocytes and cardiomyocytes (35). It is possible that the centration of androgens remains high. While the andro-
membrane-forming capabilities of autophagy are used by gen-mediated lipid droplets are thought to be mainly TGs
the cell to help form the lipid depots. This is supported by due to the known AR-regulated lipogenic genes, the neu-
Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 13

which ones are benign bystanders. Our


understanding of these signaling path-
ways will ultimately yield new thera-
peutic targets. Thus, as our work indi-
cates, in addition to exploring the use
of antioxidants and inhibitors of fatty
acid synthesis, autophagy may offer a
new therapeutic target for AR-driven
prostatic diseases.

Acknowledgments
We thank members of the Frigo Laboratory
for technical advice, helpful suggestions,
and critical comments on this study and
manuscript. We especially thank Dr. Ana
Maria Cuervo (Albert Einstein College of
Medicine, Bronx, New York, NY) for ad-
vice on various autophagy approaches. We
FIG. 8. Proposed model of how androgen-mediated autophagy promotes prostate cell
growth. Androgens, via AR, promote intracellular lipid accumulation through 1) increasing the also thank Dr. Thomas Westbrook (Baylor
expression of enzymes involved in de novo lipogenesis (e.g. FAS, ACC, and ACL) and 2) College of Medicine, Houston, TX) for
elevating intracellular ROS levels. The increased ROS levels stimulate lipid accumulation providing the pINDUCER system plasmids
through an autophagy-mediated mechanism that is not fully understood but is downstream/ and advice; Dr. Herman Dierick (Baylor
independent of FAS. The intracellular fat depots can then ultimately promote prostate cell
College of Medicine) for help with the
growth and/or survival through potentially altering multiple aspects of cell biology and
metabolism. pGIPZ constructs; Dr. Charles Sawyers
(Memorial Sloan-Kettering Cancer Center,
New York, NY) for providing the LAPC4
tral lipid stains used here could also be detecting increases cells; Dr. Brian Altman (University of Pennsylvania, Philadel-
in cholesterol levels. Finally, one emerging possibility to phia, PA) and Dr. Tso-Pang Yao (Duke University) for provid-
explain the role of fat droplets in cell growth is the au- ing the GFP-LC3 retroviral construct (Altman) and helpful dis-
tophagy-mediated breakdown of fat droplets. This pro- cussions regarding autophagy; Dr. Fei Su (University of
cess, termed lipophagy, can yield free fatty acids that enter Houston) and the Duke Comprehensive Cancer Center Flow
Cytometry Shared Resource for help with cell sorting; both the
␤-oxidation (65, 66, 70), which is an important energy-
Duke University Department of Pathology’s Electron Micros-
producing process in the prostate (88 –92). This suggests copy Core and MD Anderson Cancer Center High Resolution
that lipophagy may provide an alternative fuel source for Electron Microscopy Facility (Houston, TX) for help with TEM
cells. Although inhibition of autophagy led to an overall and immunogold TEM, respectively; and Dr. Sean Hartig (Bay-
decrease in lipid levels, the opposite of what one would lor College of Medicine) for imaging advice and, along with Dr.
expect if high rates of lipophagy were occurring, it is Chin-Yo Lin (University of Houston), critical reading of this
plausible that some of the newly mobilized fats were re- manuscript.
directed and used for fatty acid oxidation, an avenue of
Address all correspondence and requests for reprints to:
research we are currently exploring. Daniel E. Frigo, Center for Nuclear Receptors and Cell Signal-
The data presented here suggest that AR signaling, via ing, Department of Biology and Biochemistry, University of
a ROS signal, promotes the autophagy-mediated mobili- Houston, 3605 Cullen Blvd, Houston, Texas 77204. E-mail:
zation of intracellular lipid depots that are then utilized by frigo@uh.edu.
This work was supported by National Institutes of Health
the cell through various ways to increase cell growth (Fig. Grants K01DK084205 and R03DK092434 (to D.E.F.),
8). This is important because these findings begin to link P30EY007551 (to A.R.B.), and R01CA139818 (to D.P.M.),
together multiple aspects of AR-mediated prostate cell and by a grant from the Texas Emerging Technology Fund.
biology. Despite the known importance of AR signaling Disclosure Summary: The authors have nothing to disclose.
in the prostate, the mechanisms by which AR manifests its
biological actions and how these are dysregulated during References
disease progression are poorly understood. As a result, it
1. Gao W, Dalton JT 2007 Expanding the therapeutic use of andro-
has become increasingly important to identify which AR- gens via selective androgen receptor modulators (SARMs). Drug
regulated signaling networks actually drive the disease vs. Discov Today 12:241–248
14 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

2. Culig Z, Bartsch G 2006 Androgen axis in prostate cancer. J Cell 22. Lv L, Li D, Zhao D, Lin R, Chu Y, Zhang H, Zha Z, Liu Y, Li Z, Xu
Biochem 99:373–381 Y, Wang G, Huang Y, Xiong Y, Guan KL, Lei QY 2011 Acetylation
3. Levine B, Kroemer G 2008 Autophagy in the pathogenesis of dis- targets the M2 isoform of pyruvate kinase for degradation through
ease. Cell 132:27– 42 chaperone-mediated autophagy and promotes tumor growth. Mol
4. Jiang M, Fernandez S, Jerome WG, He Y, Yu X, Cai H, Boone B, Yi Cell 42:719 –730
Y, Magnuson MA, Roy-Burman P, Matusik RJ, Shappell SB, Hay- 23. Pavlides S, Tsirigos A, Migneco G, Whitaker-Menezes D, Chia-
ward SW 2010 Disruption of PPAR␥ signaling results in mouse varina B, Flomenberg N, Frank PG, Casimiro MC, Wang C, Pestell
prostatic intraepithelial neoplasia involving active autophagy. Cell RG, Martinez-Outschoorn UE, Howell A, Sotgia F, Lisanti MP
Death Differ 17:469 – 481 2010 The autophagic tumor stroma model of cancer: role of oxi-
5. Mizushima N, Levine B 2010 Autophagy in mammalian develop- dative stress and ketone production in fueling tumor cell metabo-
ment and differentiation. Nat Cell Biol 12:823– 830 lism. Cell Cycle 9:3485–3505
6. Rabinowitz JD, White E 2010 Autophagy and metabolism. Science 24. Sotelo J, Briceño E, López-González MA 2006 Adding chloroquine
330:1344 –1348 to conventional treatment for glioblastoma multiforme: a random-
7. Kondo Y, Kanzawa T, Sawaya R, Kondo S 2005 The role of au- ized, double-blind, placebo-controlled trial. Ann Intern Med 144:
tophagy in cancer development and response to therapy. Nat Rev 337–343
Cancer 5:726 –734 25. Wei H, Wei S, Gan B, Peng X, Zou W, Guan JL 2011 Suppression
8. Mathew R, Karantza-Wadsworth V, White E 2007 Role of au- of autophagy by FIP200 deletion inhibits mammary tumorigenesis.
tophagy in cancer. Nat Rev Cancer 7:961–967 Genes Dev 25:1510 –1527
9. Hippert MM, O’Toole PS, Thorburn A 2006 Autophagy in cancer: 26. Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H, Bause A, Li
good, bad, or both? Cancer Res 66:9349 –9351 Y, Stommel JM, Dell’antonio G, Mautner J, Tonon G, Haigis M,
10. Yue Z, Jin S, Yang C, Levine AJ, Heintz N 2003 Beclin 1, an Shirihai OS, Doglioni C, Bardeesy N, Kimmelman AC 2011 Pan-
autophagy gene essential for early embryonic development, is a creatic cancers require autophagy for tumor growth. Genes Dev
haploinsufficient tumor suppressor. Proc Natl Acad Sci USA 100: 25:717–729
15077–15082 27. Lazova R, Camp RL, Klump V, Siddiqui SF, Amaravadi RK,
11. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, Troxel A, Rosen J, Pawelek JM 2012 Punctate LC3B expression is a common feature
Eskelinen EL, Mizushima N, Ohsumi Y, Cattoretti G, Levine B of solid tumors and associated with proliferation, metastasis, and
2003 Promotion of tumorigenesis by heterozygous disruption of the poor outcome. Clin Cancer Res 18:370 –379
beclin 1 autophagy gene. J Clin Invest 112:1809 –1820 28. White E, DiPaola RS 2009 The double-edged sword of autophagy
12. Feng Z, Zhang H, Levine AJ, Jin S 2005 The coordinate regulation modulation in cancer. Clin Cancer Res 15:5308 –5316
of the p53 and mTOR pathways in cells. Proc Natl Acad Sci USA 29. Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu
102:8204 – 8209 M, Metzger D, Reggiani C, Schiaffino S, Sandri M 2009 Autophagy
13. Arico S, Petiot A, Bauvy C, Dubbelhuis PF, Meijer AJ, Codogno P, is required to maintain muscle mass. Cell Metab 10:507–515
Ogier-Denis E 2001 The tumor suppressor PTEN positively regu- 30. Masiero E, Sandri M 2010 Autophagy inhibition induces atrophy
lates macroautophagy by inhibiting the phosphatidylinositol 3-ki- and myopathy in adult skeletal muscles. Autophagy 6:307–309
nase/protein kinase B pathway. J Biol Chem 276:35243–35246 31. Goldman S, Zhang Y, Jin S 2010 Autophagy and adipogenesis:
14. Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, implications in obesity and type II diabetes. Autophagy 6:179 –181
Thomas-Tikhonenko A, Thompson CB 2007 Autophagy inhibi- 32. Zhang Y, Goldman S, Baerga R, Zhao Y, Komatsu M, Jin S 2009
tion enhances therapy-induced apoptosis in a Myc-induced model Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice
of lymphoma. J Clin Invest 117:326 –336 reveals a role in adipogenesis. Proc Natl Acad Sci USA 106:19860 –
15. Bauvy C, Gane P, Arico S, Codogno P, Ogier-Denis E 2001 Au- 19865
tophagy delays sulindac sulfide-induced apoptosis in the human 33. Baerga R, Zhang Y, Chen PH, Goldman S, Jin S 2009 Targeted
intestinal colon cancer cell line HT-29. Exp Cell Res 268:139 –149 deletion of autophagy-related 5 (atg5) impairs adipogenesis in a
16. Boya P, González-Polo RA, Casares N, Perfettini JL, Dessen P, cellular model and in mice. Autophagy 5:1118 –1130
Larochette N, Métivier D, Meley D, Souquere S, Yoshimori T, 34. Singh R, Xiang Y, Wang Y, Baikati K, Cuervo AM, Luu YK, Tang
Pierron G, Codogno P, Kroemer G 2005 Inhibition of macroau- Y, Pessin JE, Schwartz GJ, Czaja MJ 2009 Autophagy regulates
tophagy triggers apoptosis. Mol Cell Biol 25:1025–1040 adipose mass and differentiation in mice. J Clin Invest 119:3329 –
17. Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli-Uzun- 3339
bas G, Kamphorst JJ, Chen G, Lemons JM, Karantza V, Coller HA, 35. Shibata M, Yoshimura K, Furuya N, Koike M, Ueno T, Komatsu
Dipaola RS, Gelinas C, Rabinowitz JD, White E 2011 Activated M, Arai H, Tanaka K, Kominami E, Uchiyama Y 2009 The MAP1-
Ras requires autophagy to maintain oxidative metabolism and tu- LC3 conjugation system is involved in lipid droplet formation.
morigenesis. Genes Dev 25:460 – 470 Biochem Biophys Res Commun 382:419 – 423
18. Kim MJ, Woo SJ, Yoon CH, Lee JS, An S, Choi YH, Hwang SG, 36. Swinnen JV, Brusselmans K, Verhoeven G 2006 Increased lipogen-
Yoon G, Lee SJ 2011 Involvement of autophagy in oncogenic K- esis in cancer cells: new players, novel targets. Curr Opin Clin Nutr
Ras-induced malignant cell transformation. J Biol Chem 286: Metab Care 9:358 –365
12924 –12932 37. Fritz V, Benfodda Z, Rodier G, Henriquet C, Iborra F, Avancès C,
19. Li J, Hou N, Faried A, Tsutsumi S, Takeuchi T, Kuwano H 2009 Allory Y, de la Taille A, Culine S, Blancou H, Cristol JP, Michel F,
Inhibition of autophagy by 3-MA enhances the effect of 5-FU-in- Sardet C, Fajas L 2010 Abrogation of de novo lipogenesis by
duced apoptosis in colon cancer cells. Ann Surg Oncol 16:761–771 stearoyl-CoA desaturase 1 inhibition interferes with oncogenic sig-
20. Liang J, Shao SH, Xu ZX, Hennessy B, Ding Z, Larrea M, Kondo naling and blocks prostate cancer progression in mice. Mol Cancer
S, Dumont DJ, Gutterman JU, Walker CL, Slingerland JM, Mills Ther 9:1740 –1754
GB 2007 The energy sensing LKB1-AMPK pathway regulates 38. Rysman E, Brusselmans K, Scheys K, Timmermans L, Derua R,
p27(kip1) phosphorylation mediating the decision to enter au- Munck S, Van Veldhoven PP, Waltregny D, Daniëls VW, Machiels
tophagy or apoptosis. Nat Cell Biol 9:218 –224 J, Vanderhoydonc F, Smans K, Waelkens E, Verhoeven G, Swinnen
21. Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, Kondo S, Kondo Y, JV 2010 De novo lipogenesis protects cancer cells from free radicals
Yu Y, Mills GB, Liao WS, Bast Jr RC 2008 The tumor suppressor and chemotherapeutics by promoting membrane lipid saturation.
gene ARHI regulates autophagy and tumor dormancy in human Cancer Res 70:8117– 8126
ovarian cancer cells. J Clin Invest 118:3917–3929 39. Liu Y, Zuckier LS, Ghesani NV 2010 Dominant uptake of fatty acid
Mol Endocrinol, February 2013, 27(2):0000 – 0000 mend.endojournals.org 15

over glucose by prostate cells: a potential new diagnostic and ther- nutrients but can lead to Chop-dependent induction of Bim to sen-
apeutic approach. Anticancer Res 30:369 –374 sitize growth factor-deprived cells to apoptosis. Mol Biol Cell 20:
40. Zha S, Ferdinandusse S, Hicks JL, Denis S, Dunn TA, Wanders RJ, 1180 –1191
Luo J, De Marzo AM, Isaacs WB 2005 Peroxisomal branched chain 57. Meerbrey KL, Hu G, Kessler JD, Roarty K, Li MZ, Fang JE, Her-
fatty acid ␤-oxidation pathway is upregulated in prostate cancer. schkowitz JI, Burrows AE, Ciccia A, Sun T, Schmitt EM, Bernardi
Prostate 63:316 –323 RJ, Fu X, Bland CS, Cooper TA, Schiff R, Rosen JM, Westbrook
41. Flavin R, Zadra G, Loda M 2011 Metabolic alterations and tar- TF, Elledge SJ 2011 The pINDUCER lentiviral toolkit for inducible
geted therapies in prostate cancer. J Pathol 223:283–294 RNA interference in vitro and in vivo. Proc Natl Acad Sci USA
42. Baron A, Migita T, Tang D, Loda M 2004 Fatty acid synthase: a 108:3665–3670
metabolic oncogene in prostate cancer? J Cell Biochem 91:47–53 58. Veeramani S, Yuan TC, Lin FF, Lin MF 2008 Mitochondrial redox
43. Migita T, Ruiz S, Fornari A, Fiorentino M, Priolo C, Zadra G, signaling by p66Shc is involved in regulating androgenic growth
Inazuka F, Grisanzio C, Palescandolo E, Shin E, Fiore C, Xie W, stimulation of human prostate cancer cells. Oncogene 27:5057–
Kung AL, Febbo PG, Subramanian A, Mucci L, Ma J, Signoretti S, 5068
Stampfer M, Hahn WC, Finn S, Loda M 2009 Fatty acid synthase: 59. Kumar B, Koul S, Khandrika L, Meacham RB, Koul HK 2008
a metabolic enzyme and candidate oncogene in prostate cancer. Oxidative stress is inherent in prostate cancer cells and is required
J Natl Cancer Inst 101:519 –532 for aggressive phenotype. Cancer Res 68:1777–1785
44. Pflug BR, Pecher SM, Brink AW, Nelson JB, Foster BA 2003 In- 60. Dolado I, Swat A, Ajenjo N, De Vita G, Cuadrado A, Nebreda AR
creased fatty acid synthase expression and activity during progres- 2007 p38␣ MAP kinase as a sensor of reactive oxygen species in
sion of prostate cancer in the TRAMP model. Prostate 57:245–254 tumorigenesis. Cancer Cell 11:191–205
45. Hatzivassiliou G, Zhao F, Bauer DE, Andreadis C, Shaw AN, 61. Knudsen KE, Scher HI 2009 Starving the addiction: new opportu-
Dhanak D, Hingorani SR, Tuveson DA, Thompson CB 2005 ATP nities for durable suppression of AR signaling in prostate cancer.
citrate lyase inhibition can suppress tumor cell growth. Cancer Cell Clin Cancer Res 15:4792– 4798
8:311–321 62. Chhipa RR, Wu Y, Ip C 2011 AMPK-mediated autophagy is a
46. Beckers A, Organe S, Timmermans L, Scheys K, Peeters A, Brus- survival mechanism in androgen-dependent prostate cancer cells
selmans K, Verhoeven G, Swinnen JV 2007 Chemical inhibition of subjected to androgen deprivation and hypoxia. Cell Signal 23:
acetyl-CoA carboxylase induces growth arrest and cytotoxicity se- 1466 –1472
lectively in cancer cells. Cancer Res 67:8180 – 8187 63. Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-
47. Swinnen JV, Heemers H, van de Sande T, de Schrijver E, Brussel- Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-
mans K, Heyns W, Verhoeven G 2004 Androgens, lipogenesis and Ghiso JA, Ahn HJ, Ait-Mohamed O, Ait-Si-Ali S, Akematsu T,
prostate cancer. J Steroid Biochem Mol Biol 92:273–279 Akira S, Al-Younes HM, Al-Zeer MA, Albert ML, Albin RL,
48. Lin H, Lu JP, Laflamme P, Qiao S, Shayegan B, Bryskin I, Monardo Alegre-Abarrategui J, Aleo MF, Alirezaei M, Almasan A, Almonte-
L, Wilson BC, Singh G, Pinthus JH 2010 Inter-related in vitro Becerril M, Amano A, et al. 2012 Guidelines for the use and inter-
effects of androgens, fatty acids and oxidative stress in prostate pretation of assays for monitoring autophagy. Autophagy 8:445–
cancer: a mechanistic model supporting prevention strategies. Int J 544
Oncol 37:761–766 64. Mizushima N, Yoshimori T, Levine B 2010 Methods in mamma-
49. Frigo DE, McDonnell DP 2008 Differential effects of prostate can- lian autophagy research. Cell 140:313–326
cer therapeutics on neuroendocrine transdifferentiation. Mol Can- 65. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M,
cer Ther 7:659 – 669 Tanaka K, Cuervo AM, Czaja MJ 2009 Autophagy regulates lipid
50. Kimura S, Noda T, Yoshimori T 2007 Dissection of the autopha- metabolism. Nature 458:1131–1135
gosome maturation process by a novel reporter protein, tandem 66. Sinha RA, You SH, Zhou J, Siddique MM, Bay BH, Zhu X, Prival-
fluorescent-tagged LC3. Autophagy 3:452– 460 sky ML, Cheng SY, Stevens RD, Summers SA, Newgard CB, Lazar
51. Dwyer MA, Joseph JD, Wade HE, Eaton ML, Kunder RS, Kazmin MA, Yen PM 2012 Thyroid hormone stimulates hepatic lipid ca-
D, Chang CY, McDonnell DP 2010 WNT11 expression is induced tabolism via activation of autophagy. J Clin Invest 122:2428 –2438
by estrogen-related receptor ␣ and ␤-catenin and acts in an auto- 67. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda
crine manner to increase cancer cell migration. Cancer Res 70: T, Kominami E, Ohsumi Y, Yoshimori T 2000 LC3, a mammalian
9298 –9308 homologue of yeast Apg8p, is localized in autophagosome mem-
52. Sherk AB, Frigo DE, Schnackenberg CG, Bray JD, Laping NJ, branes after processing. EMBO J 19:5720 –5728
Hammond M, Patterson J, Thompson SK, Kazmin D, Norris JD, 68. Tanida I, Minematsu-Ikeguchi N, Ueno T, Kominami E 2005 Lys-
McDonnell DP 2008 Development of a small molecule serum and osomal turnover, but not a cellular level, of endogenous LC3 is a
glucocorticoid-regulated kinase 1 antagonist and its evaluation as a marker for autophagy. Autophagy 1:84 –91
prostate cancer therapeutic. Cancer Res 68:1–9 69. Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H,
53. Frigo DE, Howe MK, Wittmann BM, Brunner AM, Cushman I, Øvervatn A, Bjørkøy G, Johansen T 2007 p62/SQSTM1 binds
Wang Q, Brown M, Means AR, McDonnell DP 2011 CaM kinase directly to Atg8/LC3 to facilitate degradation of ubiquitinated pro-
kinase ␤-mediated activation of the growth regulatory kinase tein aggregates by autophagy. J Biol Chem 282:24131–24145
AMPK is required for androgen-dependent migration of prostate 70. Heaton NS, Randall G 2010 Dengue virus-induced autophagy reg-
cancer cells. Cancer Res 71:528 –537 ulates lipid metabolism. Cell Host Microbe 8:422– 432
54. Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, 71. Le Lay S, Briand N, Blouin CM, Chateau D, Prado C, Lasnier F, Le
Askew DS, Baba M, Baehrecke EH, Bahr BA, Ballabio A, Bamber Liepvre X, Hajduch E, Dugail I 2010 The lipoatrophic caveolin-1
BA, Bassham DC, Bergamini E, Bi X, Biard-Piechaczyk M, Blum deficient mouse model reveals autophagy in mature adipocytes.
JS, Bredesen DE, Brodsky JL, Brumell JH, Brunk UT, Bursch W, Autophagy 6:754 –763
Camougrand N, Cebollero E, Cecconi F, Chen Y, et al. 2008 Guide- 72. Shin KJ, Wall EA, Zavzavadjian JR, Santat LA, Liu J, Hwang JI,
lines for the use and interpretation of assays for monitoring au- Rebres R, Roach T, Seaman W, Simon MI, Fraser ID 2006 A single
tophagy in higher eukaryotes. Autophagy 4:151–175 lentiviral vector platform for microRNA-based conditional RNA
55. Mizushima N, Yoshimori T 2007 How to interpret LC3 immuno- interference and coordinated transgene expression. Proc Natl Acad
blotting. Autophagy 3:542–545 Sci USA 103:13759 –13764
56. Altman BJ, Wofford JA, Zhao Y, Coloff JL, Ferguson EC, Wieman 73. Heemers H, Maes B, Foufelle F, Heyns W, Verhoeven G, Swinnen
HL, Day AE, Ilkayeva O, Rathmell JC 2009 Autophagy provides JV 2001 Androgens stimulate lipogenic gene expression in prostate
16 Shi et al. Autophagy Increases Prostate Cell Growth Mol Endocrinol, February 2013, 27(2):0000 – 0000

cancer cells by activation of the sterol regulatory element-binding of autophagy-related molecules and processes (2nd edition). Au-
protein cleavage activating protein/sterol regulatory element-bind- tophagy 7:1273–1294
ing protein pathway. Mol Endocrinol 15:1817–1828 83. Sarker D, Reid AH, Yap TA, de Bono JS 2009 Targeting the PI3K/
74. Heemers H, Vanderhoydonc F, Roskams T, Shechter I, Heyns W, AKT pathway for the treatment of prostate cancer. Clin Cancer Res
Verhoeven G, Swinnen JV 2003 Androgens stimulate coordinated 15:4799 – 4805
84. Brusselmans K, Timmermans L, Van de Sande T, Van Veldhoven
lipogenic gene expression in normal target tissues in vivo. Mol Cell
PP, Guan G, Shechter I, Claessens F, Verhoeven G, Swinnen JV
Endocrinol 205:21–31
2007 Squalene synthase, a determinant of Raft-associated choles-
75. Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L, Elazar Z 2007 terol and modulator of cancer cell proliferation. J Biol Chem 282:
Reactive oxygen species are essential for autophagy and specifically 18777–18785
regulate the activity of Atg4. EMBO J 26:1749 –1760 85. Stanbrough M, Bubley GJ, Ross K, Golub TR, Rubin MA, Penning
76. Huang WC, Li X, Liu J, Lin J, Chung LW 2012 Activation of TM, Febbo PG, Balk SP 2006 Increased expression of genes con-
androgen receptor, lipogenesis, and oxidative stress converged by verting adrenal androgens to testosterone in androgen-independent
SREBP-1 is responsible for regulating growth and progression of prostate cancer. Cancer Res 66:2815–2825
prostate cancer cells. Mol Cancer Res 10:133–142 86. Mostaghel EA, Page ST, Lin DW, Fazli L, Coleman IM, True LD,
77. Veeramani S, Chou YW, Lin FC, Muniyan S, Lin FF, Kumar S, Xie Knudsen B, Hess DL, Nelson CC, Matsumoto AM, Bremner WJ,
Y, Lele SM, Tu Y, Lin MF 2012 Reactive oxygen species induced by Gleave ME, Nelson PS 2007 Intraprostatic androgens and andro-
p66Shc longevity protein mediate nongenomic androgen action via gen-regulated gene expression persist after testosterone suppres-
sion: therapeutic implications for castration-resistant prostate can-
tyrosine phosphorylation signaling to enhance tumorigenicity of
cer. Cancer Res 67:5033–5041
prostate cancer cells. Free Radic Biol Med 53:95–108
87. Locke JA, Guns ES, Lubik AA, Adomat HH, Hendy SC, Wood CA,
78. Mizushima N, Levine B, Cuervo AM, Klionsky DJ 2008 Autophagy Ettinger SL, Gleave ME, Nelson CC 2008 Androgen levels increase
fights disease through cellular self-digestion. Nature 451:1069 – by intratumoral de novo steroidogenesis during progression of cas-
1075 tration-resistant prostate cancer. Cancer Res 68:6407– 6415
79. Bennett HL, Fleming JT, O’Prey J, Ryan KM, Leung HY 2010 88. Kotzerke J, Volkmer BG, Neumaier B, Gschwend JE, Hautmann
Androgens modulate autophagy and cell death via regulation of the RE, Reske SN 2002 Carbon-11 acetate positron emission tomog-
endoplasmic reticulum chaperone glucose-regulated protein 78/BiP raphy can detect local recurrence of prostate cancer. Eur J Nucl
in prostate cancer cells. Cell Death Dis 1:e72 Med Mol Imaging 29:1380 –1384
80. Li M, Jiang X, Liu D, Na Y, Gao GF, Xi Z 2008 Autophagy protects 89. Liu Y 2006 Fatty acid oxidation is a dominant bioenergetic path-
LNCaP cells under androgen deprivation conditions. Autophagy way in prostate cancer. Prostate Cancer Prostatic Dis 9:230 –234
4:54 – 60 90. Oyama N, Akino H, Kanamaru H, Suzuki Y, Muramoto S,
Yonekura Y, Sadato N, Yamamoto K, Okada K 2002 11C-acetate
81. Ray PD, Huang BW, Tsuji Y 2012 Reactive oxygen species (ROS)
PET imaging of prostate cancer. J Nucl Med 43:181–186
homeostasis and redox regulation in cellular signaling. Cell Signal
91. Oyama N, Miller TR, Dehdashti F, Siegel BA, Fischer KC, Michal-
24:981–990 ski JM, Kibel AS, Andriole GL, Picus J, Welch MJ 2003 11C-acetate
82. Klionsky DJ, Baehrecke EH, Brumell JH, Chu CT, Codogno P, PET imaging of prostate cancer: detection of recurrent disease at
Cuervo AM, Debnath J, Deretic V, Elazar Z, Eskelinen EL, Fink- PSA relapse. J Nucl Med 44:549 –555
beiner S, Fueyo-Margareto J, Gewirtz D, Jäättelä M, Kroemer G, 92. Pinthus JH, Lu JP, Bidaisee LA, Lin H, Bryskine I, Gupta RS, Singh
Levine B, Melia TJ, Mizushima N, Rubinsztein DC, Simonsen A, G 2007 Androgen-dependent regulation of medium and long chain
Thorburn A, Thumm M, Tooze SA 2011 A comprehensive glossary fatty acids uptake in prostate cancer. Prostate 67:1330 –1338

You might also like