ArticlePDF Available

Photodynamic therapy-mediated oxidative stress can induce expression of heat shock proteins

Authors:

Abstract and Figures

Photodynamic therapy (PDT) is an experimental cancer therapy inducing tumor tissue damage via photosensitizer-mediated oxidative cytotoxicity. A previous report indicates that oxidative stress induced by hydrogen peroxide or menadione activates the heat shock transcription factor in mouse cells but does not result in either increased transcription or translation of heat shock proteins (HSPs). Our study documents that photosensitizer-mediated oxidative stress can activate the heat shock factor as well as increase HSP-70 mRNA and protein levels in mouse RIF-1 cells. The cellular heat shock response after PDT varied for the different photosensitizers being examined. Treatments using either a chlorin (mono-L-aspartyl chlorin-e6)- or purpurin (tin etio-purpurin)-based sensitizer induced HSP-70 expression, whereas identical photosensitization conditions with a porphyrin (Photofrin)-based sensitizer failed to induce a cellular HSP response. These sensitizers, which generate singlet oxygen as the primary oxidant during photosensitization, were used in experiments under isoeffective treatment conditions. HSP-70 expression after photosensitization was associated with the concomitant induction of thermotolerance in PDT-treated cells. Interestingly, reverse transcription-PCR demonstrated that in vivo PDT treatments of RIF-1 tumors induce expression of HSP-70 for all photosensitizers including Photofrin. These results indicate that photosensitizer-generated singlet oxygen exposure can induce in vitro and in vivo HSP-70 expression, and that specific subcellular targets of PDT (which can differ for various sensitizers) are determinants for HSP-70 activation after oxidative stress.
Content may be subject to copyright.
1996;56:2355-2360. Published online May 1, 1996.Cancer Res
Charles J. Gomer, Stefan W. Ryter, Angela Ferrario, et al.
Expression of Heat Shock Proteins
Photodynamic Therapy-mediated Oxidative Stress Can Induce
Updated Version http://cancerres.aacrjournals.org/content/56/10/2355
Access the most recent version of this article at:
Citing Articles http://cancerres.aacrjournals.org/content/56/10/2355#related-urls
This article has been cited by 14 HighWire-hosted articles. Access the articles at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and .pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions .permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
American Association for Cancer Research Copyright © 1996 on July 14, 2011cancerres.aacrjournals.orgDownloaded from
[CANCER RESEARCH 56. 2355-2360, May 15. 1996]
Photodynamic Therapy-mediated Oxidative Stress Can Induce Expression of Heat
Shock Proteins1
Charles J. Corner,2 Stefan W. Ryter,3 Angela Ferrano, Natalie Rucker, Sam Wong, and Anita M. R. Fisher
Clayton Ocular Oncology Center, Children* Hospital Los Angeles, and Departments of Pediatrics ¡C.J. G.I. Radiation Oncology ¡C.J. G.¡,and Molecular Pharmacology and
Toxicology ¡S.R., C. J. C.], University of Southern California, Los Angeles, California 90027
ABSTRACT
Photodynamic therapy (PDT) is an experimental cancer therapy induc
ing tumor tissue damage via photosensitizer-mediated oxidative cytotox-
icity. A previous report indicates that oxidative stress induced by hydro
gen peroxide or menadione activates the heat shock transcription factor in
mouse cells but does not result in either increased transcription or trans
lation of heat shock proteins (HSPs). Our study documents that photo
sensitizer-mediated oxidative stress can activate the heat shock factor as
well as increase HSP-70 mRNA and protein levels in mouse RIF-1 cells.
The cellular heat shock response after PDT varied for the different
photosensitizers being examined. Treatments using either a chlorin
(mono-L-aspartyl chlorin-e6)- or purpurin (tin etio-purpurin)-based sen-
sitizer induced HSP-70 expression, whereas identical photosensitization
conditions with a porphyrin (Photofrin)-based sensitizer failed to induce a
cellular HSP response. These sensitizers, which generate singlet oxygen as
the primary oxidant during photosensitization, were used in experiments
under isoeffective treatment conditions. HSP-70 expression after photo
sensitization was associated with the concomitant induction of thermotol-
erance in PDT-treated cells. Interestingly, reverse transcription-PCR
demonstrated that in vivo PDT treatments of RIF-1 tumors induce ex
pression of HSP-70 for all photosensitizers including Photofrin. These
results indicate that photosensitizer-generated singlet oxygen exposure
can induce in vitro and in vivo HSP-70 expression, and that specific
subcellular targets of PDT (which can differ for various sensitizers) are
determinants for HSP-70 activation after oxidative stress.
INTRODUCTION
PDT4 is an experimental treatment for malignant and benign dis
eases (1, 2). Tumors of the bronchus, bladder, esophagus, head and
neck, brain, and skin are being treated with PDT in clinical trials (3,
4). Phase I and II studies evaluating PDT for psoriasis and age-related
macular degeneration are also under way (5). Most PDT clinical trials
use a porphyrin-based photosensitizer, PH-II (1). However, several
PDT clinical trials are now using second-generation photosensitizers
involving chlorin-, purpurin-, benzoporphyrin-, and phthalocyanine-
based derivatives (6, 7). The primary oxidant associated with these
photosensitization reactions is singlet oxygen, which is generated
during a Type II photochemical reaction (8). Photosensitization oxi
dizes cellular macromolecules including lipids, proteins, and nucleic
acids, and PDT damages a variety of subcellular structures (1, 9).
Cytotoxicity involves both programmed cell death as well as cellular
necrosis (10-12).
Received 1/12/96; accepted 3/19/96.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance with
18 U.S.C. Section 1734 solely to indicate this fact.
' This research was performed in conjunction with the Clayton Foundation for Re
search and was supported in part by USPHS Grant R37-CA-31230 from NIH and Office
of Naval Research Grant N000014-91-J-4047 from the Department of Defense.
2 To whom requests for reprints should be addressed, at Childrens Hospital Los
Angeles, Mail Stop 67, 4650 Sunset Boulevard. Los Angeles. CA 90027.
3 Present address: Swiss Institute for Experimental Cancer Research, 155 Chemin des
Boveresses, Epalinges, CH-1066, Switzerland.
4 The abbreviations used are: PDT, photodynamic therapy; GAPDH, glyceraldehyde-
3-phosphate dehydrogenase; HSE, heat shock element; HSF. heat shock factor; HSP, heat
shock protein; NPe6, mono-L-aspartyl chlorin-e6; PH-II, Photofrin porfimer sodium;
RIF-1, radiation-induced fibrosarcoma; RT-PCR, reverse transcription PCR; SnET2, tin
etio-purpurin.
Oxidative stress induced by porphyrin-mediated photosensitization
increases expression of early-response genes (c-fos, c-jun, c-myc,
egr-\) as well as the cellular stress proteins glucose-regulated protein
and heme oxygenase (13-15). Signaling pathways involved in PDT-
mediated gene activation have not been defined, but it appears that
protein phosphorylation is involved (16, 17). A recent study using
murine cells reports that oxidative stress mediated by hydrogen per
oxide or menadione activates HSF but does not result in the accumu
lation of HSP-70 mRNA or HSP (18). In the present study, we
examined whether different photosensitizers, which function via com
mon photochemical pathways to generate singlet oxygen, can induce
HSP-70 expression and concomitant cellular thermotolerance. HSPs
represent a highly conserved family of constitutively expressed and
stress-induced cellular proteins ranging in size from A/r 20,000 to
more than 110,000 (19, 20). These molecular chaperones play a
central role in the binding of denatured proteins, and in the folding
and/or cytoplasmic translocation of nascent proteins (21, 22). Induc-
ible HSPs are also associated with the development of thermotoler
ance or transient resistance to heat stress (23-25). A variety of
exogenous stress conditions, including exposure to heat, arsenite,
amino acid analogues, heavy metals, and ethanol, can induce HSPs
(19, 26). Results of our study demonstrate that cellular PDT can
induce HSP-70 expression but that photosensitizers (with similar
photochemical pathways but differing subcellular localization prop
erties) exhibit varying degrees of HSP-70 induction. Interestingly, in
vivo PDT of RIF-1 tumors using the porphyrin, chlorin, or purpurin
photosensitizers all resulted in increased expression of HSP-70
mRNA.
MATERIALS AND METHODS
Drugs. PH-II porfimer sodium was provided by Quadra Logics, Inc. (Van
couver, British Columbia, Canada); NPe6 was provided by Porphyrin Products
(Logan, UT); and SnET2 was provided by PDT Systems (Santa Barbara, CA).
Working solutions were 2.5 mg/ml for PH-II and NPe6 and 1 mg/ml for
SnET2.
Cells and Cell Culture. Murine RIF cells (RIF-1) were used in all exper
iments (27). Cells were grown in RPMI 1640 supplemented with 15% PCS and
antibiotics. The cells were plated into plastic Petri dishes before experiments
involving clonogenic survival, mRNA analysis, or thermotolerance evaluation
and into plastic T-75 flasks before mobility shift assays, protein synthesis
analysis, and Western analysis. The plating efficiency for untreated cells
ranged from 50 to 60%. Single-treatment photosensitization experiments were
performed as reported previously (28). Briefly, cells were incubated in the dark
with a photosensitizer (25 Mg/ml for PH-II and NPe6 or 0.75 /xg/ml for SnET2)
at 37°Cfor 16 h in RPMI 1640 supplemented with 5% FCS. After incubation,
the cells were incubated for 30 min in fresh RPMI 1640 supplemented with
10% PCS and then exposed to visible light. Cells incubated with PH-II were
exposed to broad-spectrum (570-650 nm) red light (0.35 mW/cm2, 315 J/m2)
generated by a parallel series of 30-W fluorescent bulbs (29). NPe6-treated
cells were exposed to 664 nm light (2 mW/cm2, 3400 J/m2) generated by an
argon ion laser-pumped dye laser, and SnET2-treated cells were exposed to
660 nm light (2 mW/cm2, 1600 J/m2) generated by the same laser. After light
treatments, cells were re-fed with complete growth media and incubated at
37°C.Cytotoxicity was determined 8 days after treatment by clonogenic assay.
Survival levels for PDT experiments involving the three photosensitizers
ranged from 25 to 39% for RNA and protein analysis and from 12 to 41% for
2355
American Association for Cancer Research Copyright © 1996 on July 14, 2011cancerres.aacrjournals.orgDownloaded from
PHOTOSENSITIZATION INDUCES HSPs
HSE binding analysis. Hyperthermia involved exposing cells to 45°Cin a
temperature-controlled water bath. Induction and measurement of PDT-medi-
ated thermotolerance included first treating cells with PDT followed at varying
time intervals (0-24 h) with heat at 45°Cfor 45 min. Heat-induced thermo
tolerance involved a priming heat dose (45°Cfor 20 min) followed at varying
time intervals (0-24 h) with heat at 45°Cfor 45 min. Cells were trypsinized
and replated for survival analysis using colony formation at the completion of
thermotolerance experiments (26).
Mobility Shift Assays. Binding of nuclear protein to the mouse HSE was
analyzed using an electrophoretic mobility shift assay as described previously
(30). Briefly, cells were harvested using solutions containing protease inhibi
tors (0.2 IHM phenylmethylsulfonyl fluoride, l /UMleupeptin, and 2 fiM pep-
statin) and phosphatase inhibitors (2 mM Na orthovanadate and 5 mM NaF).
Cells were washed in PBS, scraped from dishes, incubated in hypotonie buffer
[10 mM HEPES (pH 7.9), 1.5 mM MgCU, 10 mM KC1,0.5 mM DTT] for 30 min
and then lysed by Dounce homogenization. Nuclei were isolated and extracted
first in a low-salt buffer [20 mM HEPES (pH 7.9). 25% glycerol. 1.5 mM
MgCk 20 mM KC1, 0.2 mM EDTA] and then in high-salt buffer [20 mM
HEPES (pH 7.9), 25% glycerol, 1.5 mM MgCl2. 1.4 M KC1, 0.2 mM EDTA].
Extracts were centrifuged for 30 min at 25,000 x g and resulting supernatants
dialyzed overnight against a buffer containing 20 mM HEPES (pH 7.9), 20%
glycerol. 100 mM KG, and 0.2 mM EDTA. A double-stranded oligonucleotide
corresponding to the proximal HSE from the mouse HSP-70 promoter (-321
to -350; 5'AGACGCGAAACTGCTGGAAGATTCCTGGCC3') was used
for DNA binding. The probe was 5' end labeled with 12P using T4 polynu-
cleotide kinase. Nuclear protein (5 fig) was added to a mixture containing 2 /ng
poly(dl-dC) and 10,000 cpm of "P-labeled HSE oligonucleotide. For compet
itive analysis, 100-fold excess of cold HSE was added to a duplicate of each
reaction. Resulting mixtures were incubated at 37°Cfor 30 min. DNA-protein
complexes were resolved by electrophoresis on a 4% polyacrylamide gel (30).
mRNA Slot Blot Analysis. Total cellular RNA was collected using a
guanidinium isothiocyanate phenol chloroform extraction procedure (31).
RNA (8 fig) was blotted onto a nylon filter using positive pressure and UV
cross-linked. The filter was then incubated overnight in hybridization solution
(50% formamide, 10% dextran sulfate, l M NaCl, 1% SDS, and 2 mg salmon
sperm DNA) containing <2P-labeled DNA probes encoding for HSP-70 or
a-tubulin. Blots were then washed in a solution containing 0.IX SSC (SSC
containing 0.1% SDS) and 0.1% SDS for 30 min at 25°Cand then for an
additional 30 min at 42°Cfor a-tubulin or at 65°Cfor HSP-70. Blots were then
exposed to X-ray film, washed, and rehybridized. Relative RNA levels were
determined by densitometric scanning of the resulting autoradiograms.
Protein Synthesis and Western Immunoblot Analysis. Protein synthesis
profiles were analyzed as described previously (32). Cells were labeled for 1
h with 30 fiCi/ml rran.v-[15S)methionine (ICN Biomedicals. Inc., Irvine. CA)
in methionine-free media at selected times after the various photosensitization
treatments described above. Cells were then dissolved in a SDS loading buffer,
and lysates containing equal amounts of radioactivity were size separated using
PAGE (15). Resulting autoradiograms were then quantified by scanning den-
sitometry. Documentation of HSP-70 levels after PDT was obtained by West
ern immunoblot analysis. Treated cells were lysed in sample buffer as de
scribed previously and separated by electrophoresis on a 10% polyacrylamide
gel (28). The proteins were then transferred to nitrocellulose paper and incu
bated for 2 h at 37°Cin 10% nonfat milk. The resulting blots were then
incubated with a mouse monoclonal antibody specific for inducible HSP-70
(SPA-810: StressGen Biotechnologies Corp.. Victoria. British Columbia, Can
ada; Ref. 28). The resulting antigen-antibody complexes were visualized with
an alkaline phosphatase-linked avidin/biotin detection system (Vectastain;
Vector Laboratories. Burlingame, CA).
In Vivo Treatment and RT-PCR Analysis of RIF-1 Tumors. Female
C3H/HeJ mice (8-12 weeks old) were injected s.c. in the right flank with IO4
RIF-1 cells. PDT- and laser-induced hyperthermia treatments were started
when the largest diameter of the resulting tumors reached 6 mm. For PDT,
mice received an i.v. injection of one of the three photosensitizers at a dose of
either 5 mg/kg (PH-II and NPe6) or 1.5 mg/kg (SnET2). PDT light treatments
were then started either 6 (NPe6) or 24 h (PH-1I and SnET2) after injection. An
argon ion-pumped dye laser tuned to either 630 (PH-II), 660 (SnET2). or 664
nm (NPe6) was used for in vivo PDT treatments. Light was delivered to tumor
surfaces via a quartz fiber microlens delivery system, and power outputs were
measured with a power meter (33). Total light doses of 200 J/cirr together with
power densities of 75 mW/cnr were used for PDT treatments. Tumor hyper
thermia treatments involved a 20-min exposure to 44.5-45.0°C. which was
achieved using 810 nm diode ¡aserlight delivered at a power density of 220
mW/cnr. These treatment conditions were expected to induce comparable but
noncurative tumoricidal responses. Animals were sacrificed 6 h after the
various treatments, and tumors were collected for RT-PCR analysis. Total
RNA was isolated from control and treated tumors using a guanidinium
isothiocyanate phenol chloroform extraction (31). RT-PCR was performed
using the Gene Amp RNA PCR kit (Perkin-Elmer, Norwalk. CT). RNA (0.5
jiig) was reverse transcribed and amplified for 25 cycles (30 s denaturation at
94°C,30 s annealing at 65°C, and I min extension at 72°C).Primers to
both HSP-70 (5'CCAACGGCATCCTGAACG3f and 5TCCTTGTCGGC-
CAGCGTG3') and GAPDH (5'TGAAGGTCGGTGTGAACGGATTTGGC3'
and 5'CATGTAGGCCATGAGGTCCACCAC3') were added simultaneously
to reaction tubes for amplification. The resulting cDNA products (a 324-bp
HSP-70 fragment and a 983-bp GAPDH fragment) were size separated on a
1.5% agarose gel containing ethidium bromide and photographed under UV
illumination.
RESULTS
PDT-mediated Oxidative Stress Induces HSF Binding to HSE.
The classical heat shock response involves transcriptional induction of
heat shock genes via activation of the HSF (34). Electrophoretic
mobility shift assays demonstrated that cellular exposure to PDT-
mediated oxidative stress activated HSF binding to HSE as shown
previously after hydrogen peroxide or menadione treatments (18). Fig.
\A shows nuclear protein binding to mouse HSE for cells exposed to
NPe6-mediated PDT as well as for cells exposed to heat. The slower-
migrating, higher-molecular-weight band (Fig. \A, arrow) corre
sponds to inducible HSF binding to HSE, whereas the faster-migrat
ing, lower-molecular-weight band corresponds to the constitutive
HSE binding activity (35). Protein binding specificity to HSE was
confirmed in competition assays by including excess cold HSE in
duplicate reactions. HSF binding activity induced by NPe6-mediated
PDT was observed within 20 min of treatment and remained elevated
for 5 h (data not shown). SnET2-mediated PDT and dark incubation
of SnET2 also resulted in specific HSE binding activity, as shown in
Fig. Iß.Interestingly. PH-II-mediated PDT was associated with min
imal binding activity of HSF to HSE as shown in Fig. 1C.
PDT Can Produce a Transient Increase in HSP-70 mRNA and
Protein. mRNA levels of HSP-70 were evaluated by slot blot anal
ysis after in vitro photosensitization conditions. Fig. 2 shows relative
HSP-70 mRNA levels in RIF-1 cells (expressed as a ratio of HSP-70
versus a-tubulin) as a function of time after PDT. Increases in HSP-70
mRNA were observed after NPe6- and SnET2-mediated PDT. Max
imum mRNA levels were observed by 4 h posttreatment with a return
to background levels by 10 h. PH-II-mediated PDT did not induce an
increase in HSP-70 mRNA. Confirming results were obtained when
mRNA levels were analyzed using RT-PCR (data not shown).
Stress Proteins Are Expressed after PDT-mediated Oxidative
Stress. Synthesis of stress proteins after PDT-mediated oxidative
stress was examined using cellular incorporation of [15S]methionine
followed by SDS-PAGE and densitometric analysis. Fig. 3 shows the
kinetics of HSP-70 protein synthesis induced by isoeffective PDT
treatments for the three classes of photosensitizer. Significant HSP-70
synthesis was observed after NPe6-mediated PDT, with the maximal
level of synthesis occurring 4-8 h after treatment. Protein synthesis
levels returned to background levels within 24 h of PDT treatment.
PH-II-mediated PDT did not elicit HSP-70 synthesis after the 16-h
photosensitizer incubation protocol. An intermediate level of HSP-70
protein synthesis was also observed after SnET2-mediated PDT, and
both the induction and decay kinetics were similar to that observed for
NPe6 treatment conditions.
2356
American Association for Cancer Research Copyright © 1996 on July 14, 2011cancerres.aacrjournals.orgDownloaded from
PHOTOSENSITIZATION INDUCES HSPs
Time after POT BTime after PDT
g Time after PDT
>l1I
Fig. I. Eleclrophorelic mobility shift analysis showing specific HSE binding activity in protein extracts from RIF-1 cells. Nuclear extracts were prepared from control cells and cells
treated with either heat, photosensitizer alone, or photosensitizer after PDT. Nuclear protein was collected l h after heat treatment and at either 20 min or l h after PDT using NPeo
(A), SnET2 (ß),or PH-II (C). Lanes marked "free HSE, " reaction mixtures without proteins. Lanes marked " + ," addition of excess cold HSE oligonucleotide (12.5 ng). Arrow, HSF
specific binding activity to HSE.
24
Fig. 2. Kinetics of HSP-70 mRNA expression after PDT using PH-II. SnET2, or NPe6.
Densitometric ratios of HSP-70 veriw.v a-tubulin were obtained by scanning autoradio-
grams resulting from RNA slot blots hybridized with *2P-labeled cDNA probes first for
HSP-70 and then for a-tubulin.
Stress protein levels at various time intervals after cellular PDT
were examined by Western immunoblot analysis. Fig. 4 shows the
results of Western immunoblots using an antibody reactive against
inducible HSP-70. Cell exposure to heat served as a positive control
condition for evaluating induction of HSP-70. HSP-70 expression was
observed after NPe6- and SnET2-mediated PDT. Inducible HSP lev
els remained elevated 48 h after PDT. The increased HSP levels were
consistent with the protein synthesis data shown in Fig. 3. Specifi
cally, NPe6-mediated PDT was associated with the highest expression
of HSP-70, whereas PH-II-mediated PDT did not induce HSP-70
expression.
PDT Oxidative Stress Induces Thermotolerance. Thermotoler-
ance is a transient resistance to a subsequent heat treatment after a
priming exposure to heat (20). The thermotolerance phenotype is
correlated with HSP expression, and these proteins appear to play a
pivotal role in heat resistance (23-25). Fig. 5A shows the development
of thermotolerance when heat is used as a priming condition. A single
heat dose of 45°Cfor 45 min produces a 0.1% survival level. How
ever, when this heat dose is preceded by a priming heat treatment
(45°Cfor 20 min) that induces HSPs, the effective cytotoxicity of the
45°C45-min treatment is decreased significantly. These results are in
agreement with previous data evaluating thermotolerance (25). Fig. 5,
B and C, shows results from thermotolerance experiments when the
priming stress treatment was either NPe6- or SnET2-mediated PDT,
respectively. Thermotolerance was induced after PDT treatments,
which produced HSP expression. A priming oxidative stress treatment
using NPe6- or SnET2-mediated PDT resulted in decreased cellular
.2
«
e
(O
C
1
a.
Q.
V)
X
o
I
o
ce
2-
1.5-
1-
0.5-
SnET2
NPe6
PH-II
16 24 32 40 48
Time after PDT (h)
Fig. 3. Kinetics of HSP-70 protein synthesis following PDT using PH-II. SnET2. or
NPe6. Densitometric ratios of HSP-70 versus actin were obtained by scanning autoradio-
grams from SDS-PAGE of lysates from ira«.i-[wS|methionine-labeled RIF cells.
2357
American Association for Cancer Research Copyright © 1996 on July 14, 2011cancerres.aacrjournals.orgDownloaded from
PHOTOSENSmZATION INDUCES HSPs
hr after PDT
o,
O 0) 2
U T O 4 8 12 24 48
NPe6 P —¿ HSP-70
SnET2 _
PH-II HSP-70
A time
45°(20 min) > 45°(45 min)
100r
/20 mins
<—45°/45mins
.001
Fig. 4. Western immunoblot analysis shows HSP-70 expression after NPeó- and
SnET2-mediated PDT bul not after PH-II-mediated PDT. Treated cells were examined
from 4 to 48 h after photosensitization. Heat-treated cells (45"C for 20 min) served as a
positive control and were analyzed 6 h after treatment. Protein samples for NPe6 and
SnET2 treatment'- were run on one gel. and the upper heat treatment represents the
positive control for both groups. Drug-alone samples were collected immediately after the
16-h photosensitizer incubation, and samples labeled control were also collected at this
time.
sensitivity to a subsequent heat treatment. Kinetics of PDT-induced
thermotolerance were similar to those observed for heat. The combi
nation of PH-II-mediated PDT and heat did not result in surviving
cells, and this precluded analysis of thermotolerance under the con
ditions used in this study. Therefore, overexpression of HSPs after
PDT correlated with the induction of thermotolerance.
PDT Treatment of RIF Tumors Induces HSP-70 mRNA. Ex
pression of HSP-70 mRNA in PDT-treated tumor tissue was exam
ined using RT-PCR and results are shown in Fig. 6. Primers to
GAPDH were included to evaluate loading profiles. Control condi
tions and light-alone treatments did not induce HSP-70 mRNA. How
ever, all PDT treatments as well as heat resulted in detectable HSP-70
expression. PH-II mediated PDT was as efficient as NPe6- or SnET2-
mediated PDT at inducing in vivo expression of HSP-70 mRNA.
DISCUSSION
Our study demonstrates that PDT-mediated oxidative stress can
activate HSF binding activity as well as induce transcriptional and
translational production of HSP-70. Therefore, photosensitizer-gener-
ated reactive oxygen species such as singlet oxygen can be added to
the list of exogenous insults that activate cellular HSPs (20). Growing
evidence indicates that HSP-70 plays a major role in thermotolerance
(23), and PDT conditions that induced a cellular HSP response also
elicited thermotolerance. The question of whether heat-induced ther
motolerance confers cellular resistance to PDT has not been addressed
in the current study. Interestingly, the cellular HSP-70 response after
photosensitization varied depending on the specific sensitizer being
examined. Photosensitization involving an extended incubation with
either a chlorin (NPe6)- or purpurin (SnET2)-based sensitizer induced
HSP-70 expression, whereas isoeffective photosensitization condi
tions with a porphyrin (PH-II)-based sensitizer failed to activate HSF
binding or induce HSP expression. The fact that PH-II-mediated
photosensitization did not induce an in vitro HSP-70 response sug
gests that random singlet oxygen exposure to cells in culture is not
2358
B
O 3 6 9 121518212427
Hours Between Treatments
Atime
NPe6-PDT > 45°(45 min)
100
_ 10
re
o>
o
mean + S.E. (n=3)
.01
.001
.0001 •¿
<-NPe6 PDT
/45 min
0 3 6 9 121518212427
Hours Between Treatments
Atime
SnET2-PDT > 45°(45 min)
100
«1Ö
'E ,
9
I 1
U>
» .01
.001«
mean + SE (n=3) <—SnET2 PDT
/45 mins
12 15 18 21 24 27
Hours Between Treatments
Fig. 5. Expression of thermotolerance in RIF-1 cells treated with either hyperthermia
(A) or PDT-mediated oxidative stress (B and C). Cells were first treated with a priming
dose of either heat (A} or PDT using NPe6 (B) or SnET2 (C) and then incubated at 37°C
for the indicated time intervals before being challenged with heat. Survival levels resulting
from the individual treatments are indicated by arrows on the right ordinale. Data are
means; bars, S.E.
American Association for Cancer Research Copyright © 1996 on July 14, 2011cancerres.aacrjournals.orgDownloaded from
PHOTOSF.NSmZATION INDUCES HSPs
GAPDH
HSP-70
Fig. 6. Reverse transcriplion-PCR analysis of HSP-70 mRNA expression in RIF
tumors growing in C3H mice. Total RNA was isolated from control tumors and tumors
exposed to light alone (660 nm); photosensitizer alone: heat; or PDT using PH-II, NPe6,
or SnET2. The 983-bp fragment for GAPDH was coamplified with the 324-bp HSP-70
fragment.
sufficient by itself to activate the heat shock response. Rather, our data
suggest that specific subcellular targets of photosensitization are as
sociated with inducing HSP-70 expression. Sites of sensitizer local
ization define cellular PDT targets, whereas actual damage is medi
ated primarily by the Type II photochemical generation of singlet
oxygen (1-3). Estimates of singlet oxygen quantum yields for the
three sensitizers used in our study range from 0.4 to 0.6 (36, 37). The
minimal diffusion distance of singlet oxygen in cells (0.1-0.2 ¿im)
implies that subcellular damage will occur close to the sites of
sensitizer localization (38). Targets identified for PH-II-mediated
PDT are the plasma membrane and mitochondria, whereas targets
identified for NPe6- and SnET2-mediated PDT are the lysosomes and
plasma membrane (39-41). Differences in lysosomal versus mito
chondria! injury could therefore play a role in initiating the HSP
response after PDT. Studies using photosensitizers with selective
subcellular targets such as the mitochondria! localizing rhodamine
derivatives (42) or the lysosomal localizing Nile blue derivatives (43)
may be beneficial in identifying subcellular targets involved in the
cellular heat shock response.
In vitro patterns of HSP-70 expression after PH-II photosensitiza
tion were not predictive of the in vivo stress responses induced by
PH-II-mediated PDT. Interestingly, RT-PCR demonstrated that in
vivo PDT treatments of RIF-1 tumors were associated with expression
of HSP-70 mRNA for all photosensitizers tested, including PH-II.
Therefore, secondary effects of photosensitization in addition to direct
subcellular oxidation may play a role in the heat shock response
observed after in vivo PDT. For example, cellular photosensitization
activates phospholipase A2, which in turn hydrolyzes fatty acids from
membrane phospholipids and generates arachidonic acid (2, 11).
Thromboxane, a product of arachidonic acid metabolism, is released
in rats immediately after PH-II-mediated PDT (2, 44). Interestingly, it
has been reported recently that cellular exposure to arachidonic acid
induces a full heat shock response in mammalian cells (45). HSP-70
transcription occurs in an arachidonic acid dose-dependent manner
and involves DNA binding activity of HSF. Therefore, in vivo PDT
treatments that produce arachidonic acid via a cascade involving
phospholipase A2 activation may elicit a stress response.
Finally, little is known about the actual in vivo targets of PDT and
this is especially true for the growing number of second-generation
compounds. Our study confirms that inducible stress protein expres
sion occurs after PDT, and this observation may be useful in the
development of in vivo molecular markers of PDT targets. Conven
tional histological studies provide information regarding generalized
areas of PDT damage arising hours or days after treatment (2).
However, questions still remain regarding which specific targets
within the treated tumor volume are initially damaged by PDT.
Likewise, little is known about the uniformity of the initial PDT
response or how actual sensitizer localization profiles correspond to
tissue damage profiles. Our data suggest that HSP-70 expression can
be used as a biomarker for sites of in vivo PDT-mediated oxidative
damage using porphyrin-, chlorin-, or purpurin-based photosensitiz
ers. In situ hybridization and immunohistochemical analysis using
stress protein probes and antibodies may provide selective and sensi
tive methods for documenting in vivo profiles of initial in vivo targets
of PDT or sites of indirect arachidonic acid-mediated responses.
Combining profiles of photosensitizer localization using fluorescence
analysis with PDT-mediated HSP-70 expression profiles may there
fore produce a unique opportunity to correlate localization sites with
PDT targets.
REFERENCES
10.
12.
13
14.
15,
16.
17
19.
20.
22.
23.
24.
Gonier, C. J.. Rucker, N., Ferrano, A., and Wong. S. Principals and applications of
photodynamic therapy. Radiât.Res.. 120: 1-18. 1989.
Henderson. B. W. Photodynamic therapy: coming of age. Pholodermatology. 6:
200-211, 1989.
Fisher, A. M. R., Murphree, A. L.. and Gomer. C. J. Clinical and preclinical
photodynamic therapy. Lasers Surg. Med.. 17: 2-31. 1995.
Pass, H. I. Photodynamic therapy in oncology: mechanisms and clinical use. J. Nati.
Cancer Inst., 85: 145-157, 1993.
Levy, J. C. Photodynamic therapy. Trends Biotechnol., 13: 14-17, 1995.
Gomer. C. J. Preclinical examination of first and second generation photosensitizers
used in photodynamic therapy. Photochem. Photobiol.. 54: 1093-1107. 1991.
Marcus. S. L.. and Dugan. M. H. Global status of clinical photodynamic therapy: the
registration process for a new therapy. Lasers Surg. Med., 12: 318-324. 1992.
Weishaupt. K. R.. Gomer. C. J.. and Dougherty. T. J. Identification of singlet oxygen
as the cytotoxic agent and photoinactivation of a murine tumor. Cancer Res., 36:
2326-2329. 1976.
Moan, J.. and Berg. K. Photochemotherapy of cancer: experimental research. Photo
chem. Photobiol., 55: 931-948, 1992.
Agarwal. M. L.. Clay. M. E.. Harvey, EJ., Evans. H. H., Antunez. A. R.. and Oleinick,
N. L. Photodynamic therapy induces rapid cell death by apoptosis in L5178Y mouse
lymphoma cells. Cancer Res.. 51: 5993-5996. 1991.
Agarwal, M. L., Larkin, H.. Zaidi, S. I. A., Mukhtar, H.. and Oleinick. N. L.
Phospholipase activation triggers apoptosis in photosensitized mouse lymphoma
cells. Cancer Res., 53: 5897-5902. 1993.
Zaidi. S. I. A.. Oleinick. N. L.. Zaim, M. T.. and Mukhtar. H. Apoptosis during
photodynamic therapy induced ablation of RIF-1 tumors in C3H mice: electron
microscopy, histopathologic and biochemical evidence. Photochem. Photobiol.. 58:
771-776. 1993.
Luna. M. C.. Wong. S., and Gomer, C. J. Photodynamic therapy mediated induction
of early response genes. Cancer Res., 54: 1374-1380, 1994.
Gomer, C. J., Luna, M., Ferrano, A., and Rucker, N. Increased transcription and
translation of heme oxygenase in Chinese hamster fibroblasts following photody
namic stress or Photofrin II incubation. Photochem. Photobiol.. 53: 275-279. 1991.
Gomer. C. J.. Ferrarlo. A.. Rucker. N.. Wong. S.. and Lee. A. Glucose regulated
protein (GRP-78) induction and cellular resistance to oxidative stress mediated by
porphyrin photosensitization. Cancer Res.. 51: 6574-6579, 1991.
Oleinick. N. L.. He. J.. Xue. L-Y.. and Balasubramaniam. U. Photodynamic therapy
and apoptosis. Photochem. Photobiol., 61: 39S, 1995.
Dubbelman. T. M. A. R. Signalling induced by photodynamic therapy. Photochem.
Photobiol., 61: 31S, 1995.
Bruce, J. L., Price. B. D., Coleman. C. N.. and Calderwood. S. K. Oxidative injury
rapidly activates the heal shock transcription factor but fails to increase levels of heat
shock proteins. Cancer Res.. 53: 12-15. 1993.
Subjeck, J. R.. and Shyy. T-T. Stress protein systems of mammalian cells. Am J.
Physiol.. 250: 1-17. 1986.
Lindquist. S.. and Craig. E. A. The heat shock proteins. Ann Rev. Genet.. 22:
631-677. 1988.
Beckmann. R. P., Mizzen. L. A., and Welch. W. J. Interaction of Hsp 70 with newly
synthesized proteins: implications for protein folding and assembly. Science (Wash
ington DC). 248: 850-854. 1990.
Frydman. J.. Nimmesgern. E.. Ohtsuka, K., and Hartl, F. U. Folding of nascent
polypeplide chains in a high molecular weight mass assembly with molecular chap-
erones. Nature (Lond.), 370: 111-117, 1994.
Li, G. C., Li, L., Liu, R. Y., Rehman, M., and Lee, W. M. F. Heat shock protein hsp70
protects cells from thermal stress even after deletion of its ATP-binding domain. Proc.
Nail. Acad. Sci. USA, 89: 2036-2040, 1992.
Landry. J.. Chretien, P.. Lambert. H.. Hickey, E.. and Weber. L. A. Heat shock
resistance conferred by expression of the human HSP-27 gene in rodent cells. J. Cell
Biol., 109: 7-15, 1989.
2359
American Association for Cancer Research Copyright © 1996 on July 14, 2011cancerres.aacrjournals.orgDownloaded from
PHOTOSENSITIZATION INDUCES HSPs
25. Riabowol, K. T., Mizzen. L. A., and Welch, W. J. Heat shock is lethal to fibroblasts
microinjecled with antibody against hsp-70. Science (Washington DC), 242: 433-
436. 1988.
26. Laszlo. A., and Li, G. C. Effect of amino acid analogs on the development of
thermotolerance and on thermotolerant cells. J. Cell. Physiol.. 154: 419-432. 1993.
27. Luna, M. C.. and Gomer. C. J. Isolation and initial characterization of mouse tumor
cells resistant to porphyrin-mediated photodynamic therapy. Cancer Res.. 51: 4243-
4249. 1991.
28. Fisher. A. M. R., Ferrano. A., and Gomer, C. J. Adriamycin resistance in Chinese
hamster fibroblasts following oxidative stress induced by photodynamic therapy.
Photochem. Photobiol.. 58: 581-588. 1993.
29. Gomer. C. J.. Rucker, N., Ferrano. A., and Murphree. A. L. Expression of potentially
lethal damage in Chinese hamster cells exposed to hematoporphyrin derivative
photodynamic therapy. Cancer Res.. 46: 3348-3352. 1986.
30. Ryter. S. W.. and Gomer. C. J. NFkB binding activity in mouse L12IO cells
following Photofrin II mediated photosensitization. Photochem. Photobiol., 58:
753-756. 1993.
31. Luna, M. C.. Ferrano, A.. Rucker. N., and Gomer. C. J. Decreased expression and
function of or-2 macroglobulin receptor/low density lipoprotein receptor-related pro
tein in photodynamic therapy-resistant mouse tumor cells. Cancer Res.. 55: 1820-
1823, 1995.
32. Li, L-J., Li, X., Ferrano, A.. Rucker, N.. Liu, E. S., Wong, S., Gomer, C. J.. and Lee,
A. S. Establishment of a Chinese hamster ovary cell line which expresses grp78
antisense transcripts and suppresses A23187 induction of both GRP78 and GRP94. J.
Cell Physiol., 153: 575-582, 1992.
33. Ferrano. A.. Kessel. D., and Gomer, C. J. Metabolic properties and photosensitizing
responsiveness of mono-L-aspartyl chlorin e6 in a mouse tumor model. Cancer Res..
52: 2890-2893. 1992.
34. Sistonen, L.. Sarge. K. D., and Morimoto. R. I. Human heat shock factors 1 and 2 are
differentially activated and can synergistically induce hsp-70 gene transcription. Mol.
Cell Biol.. 14: 2087-2099. 1994.
35. Liu. R. Y.. Kim, D.. Yang. S-H.. and Li. G. C. Dual control of heat shock response:
involvement of a constitutive heat shock element-binding factor. Proc. Nati. Acad.
Sci. USA, 90: 3078-3082, 1993.
36. Grossweiner, L. I. The Science of Photobiology, p. 43. Boca Raton. FL: CRC Press,
1994.
37. Spikes. J. Chlorins as photosensitizers in biology and medicine. J. Photochem.
Photobiol. B Biol.. 6: 259-274. 1990.
38. Moan. J. On the diffusion length of singlet oxygen in cells and tissue. J. Photochem.
Photobiol. B Biol.. 6: 343-344, 1990.
39. Kessel. D. Use of fluorescent probes for characterizing sites of photodamage. The
Spectrum (Bowling Green State University), 6: 3-5, 1993.
40. Roberts, W, G., Liaw, L-H.. and Berns. M. W. In vitro Photosensitization. An electron
microscopy study of cellular destruction with mono-L-aspartyl chlorin e6 and Pho-
tofnn. Laser Surg. Med.. 9: 102-108. 1989.
41. Kessel. D. Sites of photosensitization by derivatives of hematoporphyrin. Photochem.
Photobiol.. 44: 489-494, 1986.
42. Johnson. L. V.. Walsh. M. L.. and Chen. L. B. Localization of mitochondria in living
cells with rhodamine 123. Proc. Nati. Acad. Sci. USA. 77: 990-994, 1980.
43. Lin, C-W.. Shulok, J. R.. Kirley, S. D.. Cincona, L.. and Foley. J. W. Lysosomal
localization and mechanism of uptake of Nile blue photosensitizers in tumor cells.
Cancer Res., 51: 2710-2719. 1991.
44. Pingar, V. H., Wieman, J.. and Doak, K. W. Role of thromboxane and prostacyclin
release on photodynamic therapy induced tumor destruction. Cancer Res., 50: 2599-
2603. 1990.
45. Jurivivh, D. A.. Sistonen, L.. Sarge. K. D.. and Morimoto, R. I. Arachidonate is a
potent modulator of human heat shock gene transcription. Proc. Nati. Acad. Sci. USA,
91: 2280-2284. 1994.
2360
American Association for Cancer Research Copyright © 1996 on July 14, 2011cancerres.aacrjournals.orgDownloaded from
... Several factors limit the efficacy of PTT and/or PDT. A few of these include hypoxia-induced resistance, inability to treat deep tumors, and aggregation-caused quenching in PDT [53][54][55][56]. For PTT, factors like restricted tissue permeating ability and the ablation of normal tissues when high-power laser light is used may adversely affect the efficiency Figure 1. ...
... Several factors limit the efficacy of PTT and/or PDT. A few of these include hypoxiainduced resistance, inability to treat deep tumors, and aggregation-caused quenching in PDT [53][54][55][56]. For PTT, factors like restricted tissue permeating ability and the ablation of normal tissues when high-power laser light is used may adversely affect the efficiency of PTT [57][58][59][60]. ...
Article
Full-text available
Phototherapies induced by photoactive nanomaterials have inspired and accentuated the importance of nanomedicine in cancer therapy in recent years. During these light-activated cancer therapies, a nanoagent can produce heat and cytotoxic reactive oxygen species by absorption of light energy for photothermal therapy (PTT) and photodynamic therapy (PDT). However, PTT is limited by the self-protective nature of cells, with upregulated production of heat shock proteins (HSP) under mild hyperthermia, which also influences PDT. To reduce HSP production in cancer cells and to enhance PTT/PDT, small HSP inhibitors that can competitively bind at the ATP-binding site of an HSP could be employed. Alternatively, reducing intracellular glucose concentration can also decrease ATP production from the metabolic pathways and downregulate HSP production from glucose deprivation. Other than reversing the thermal resistance of cancer cells for mild-temperature PTT, an HSP inhibitor can also be integrated into functionalized nanomaterials to alleviate tumor hypoxia and enhance the efficacy of PDT. Furthermore, the co-delivery of a small-molecule drug for direct HSP inhibition and a chemotherapeutic drug can integrate enhanced PTT/PDT with chemotherapy (CT). On the other hand, delivering a glucose-deprivation agent like glucose oxidase (GOx) can indirectly inhibit HSP and boost the efficacy of PTT/PDT while combining these therapies with cancer starvation therapy (ST). In this review, we intend to discuss different nanomaterial-based approaches that can inhibit HSP production via ATP regulation and their uses in PTT/PDT and cancer combination therapy such as CT and ST.
... Traditional photodynamic therapy (PDT), which uses an external light source to excite photosensitizers in tumors and generate reactive oxygen species (ROS), can induce the expression of tumor antigens or cell death immunogens for adjuvant antitumor effect. [7][8][9][10][11] Due to the limited light penetration in tissue, the application is confined to lesions that are locally accessible to direct light stimulation. Previously, we and others reported a phenomenon where radionuclides capable of emitting Cerenkov radiation can interact with photosensitizers to generate cytotoxic ROS for cancer treatment. ...
Preprint
Full-text available
Interactions of light-sensitive drugs and materials with Cerenkov radiation-emitting radiopharmaceuticals generate cytotoxic reactive oxygen species (ROS) to inhibit localized and disseminated cancer progression, but the cell death mechanisms underlying this radionuclide stimulated dynamic therapy (RaST) remain elusive. Using ROS-regenerative nanophotosensitizers coated with a tumor-targeting transferrin-titanocene complex (TiO2-TC-Tf) and radiolabeled 2-fluorodeoxyglucose (18FDG), we found that adherent dying cells maintained metabolic activity with increased membrane permeabilization. Mechanistic assessment of these cells revealed that RaST activated the expression of RIPK-1 and RIPK-3, which mediate necroptosis cell death. Subsequent recruitment of the nuclear factors kappa B and the executioner mixed lineage kinase domain-like pseudo kinase (MLKL) triggered plasma membrane permeabilization and pore formation, respectively, followed by the release of cytokines and immunogenic damage-associated molecular patterns (DAMPs). In immune-deficient breast cancer models with adequate stroma and growth factors that recapitulate the human tumor microenvironment, RaST failed to inhibit tumor progression and the ensuing lung metastasis. A similar aggressive tumor model in immunocompetent mice responded to RaST, achieving a remarkable partial response (PR) and complete response (CR) with no evidence of lung metastasis, suggesting active immune system engagement. RaST recruited antitumor CD11b+, CD11c+, and CD8b+ effector immune cells after initiating dual immunogenic apoptosis and necroptosis cell death pathways in responding tumors in vivo. Over time, cancer cells upregulated the expression of negative immune regulating cytokine (TGF-β) and soluble immune checkpoints (sICP) to challenge RaST effect in the CR mice. Using a signal-amplifying cancer-imaging agent, LS301, we identified latent minimal residual disseminated tumors in the lymph nodes (LNs) of the CR group. Despite increased protumor immunogens in the CR mice, RaST prevented cancer relapse and metastasis through dynamic redistribution of ROS-regenerative TiO¬2 from bones at the early treatment stage to the spleen and LNs, maintaining active immunity against cancer progression and migration. This study reveals the immune-mechanistic underpinnings of RaST-mediated antitumor immune response and highlights immunogenic reprogramming of tumors in response to RaST. Overcoming apoptosis resistance through complementary necroptosis activation paves the way for strategic drug combinations to improve cancer treatment.
... HSPs play a significant role in cellular proliferation, differentiation, and carcinogenesis. PDT generates transient levels of reactive oxygen species (ROS) [40], which upregulates the levels of HSPs [57][58][59][60]. Based on these results, HSPs may exhibit complex cytoprotective mechanisms to influence the efficacy of PDT. ...
Article
Background The aim of this study was to identify changes in gene expression before and after 5-aminolevulinic acid-mediated photodynamic therapy (5-ALA-PDT) and to investigate the potential mechanism of 5-ALA-PDT based on ribonucleic acid sequencing (RNA-Seq) analysis.Methods Secondary hyperparathyroidism (SHPT) primary cells were isolated from surgically excised specimens and exposed to laser light. The transcription profiles of SHPT primary cells were identified through RNA-Seq. Differentially expressed genes (DEGs) were identified. Enrichment of functions and signaling pathway analysis were performed based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Quantitative real-time polymerase chain reaction (RT-qPCR) and western blot analysis were used to validate genes based on RNA-Seq results.ResultsIn total, 1320 DEGs were identified, of which 1019 genes were upregulated and 301 genes were downregulated. GO and KEGG pathway analyses identified significantly enriched pathways in DEGs, including TGF beta in extracellular matrix (ECM), negative regulation of triglyceride biosynthetic process, protein heterodimerization activity, systemic lupus erythematosus, ECM–receptor interaction, focal adhesion and protein digestion and absorption. Protein–protein interaction (PPI) network analyses identified potential heat shock protein (HSP) interactions among the DEGs. Eight HSP genes were also identified that were most likely involved in 5-ALA-PDT, which were further validated by RT-qPCR and western blotting.Conclusions The findings of this descriptive study reveal changes in the transcriptome profile during 5-ALA-PDT, suggesting that gene expression and mutation, signaling pathways, and the molecular network are altered in SHPT primary cells. The above findings provide new insight for further studies on the mechanisms underlying 5-ALA-PDT in SHPT.Graphical abstract
... Besides producing ROS molecules, the depletion of intracellular glutathione induces cellular oxidative stress which is regarded as the main reason for cytotoxicity. The depletion of glutathione may be due to chelation of cadmium ions reported in the study [39][40][41][42][43]. ...
... It is well documented that enhanced expression of 72 kDa heat shock protein (Hsp72), an inducible form of the 70 kDa Hsps, reduces tissue injury and improves cell survival in response to stress stimuli. In addition to heat (Shimizu et al., 1997;Maloyan et al., 1999;Demirel et al., 2001;Dehbi et al., 2010), several stressful conditions, including hypoxia (Benjamin et al., 1990;Tokyol et al., 2005;Lee et al., 2016;Wang et al., 2019), UV radiation (Ohnishi et al., 1996), oxidative stress (Gomer et al., 1996), exercise training (Skidmore et al., 1995;Demirel et al., 2001) and, among others have been shown to stimulate the expression of Hsp72 in various cell and tissue types. ...
Article
Full-text available
Akin, Senay, Metin Bastug, Ridvan Colak, Hakan Ficicilar, Betul Simten Saglam, Nazan S. Kosar, and Haydar Demirel. Possible adaptation of the adrenal gland Hsp72 expression to hypoxic stress. High Alt Med Biol. 00:000-000, 2021. Background: Adrenal glands play a central role in the general response to stress and controlling wholebody homeostasis. One of the most severe environmental stresses encountered by high-altitude climbers is hypoxia. Since the 72 kDa heat shock protein (Hsp72) has a critical role in cellular homeostasis, regulation of Hsp72 in adrenal glands seems to be crucial for maintaining cellular integrity of the gland and sustaining an adequate whole-body stress response in a hypoxic environment. Therefore, this study investigated if 15 days of hypoxia results in the induction of Hsp72 in adrenal glands. In addition, we examined whether heat treatment had any effect on adrenal Hsp72 expression to hypoxia, as cellular and systemic physiological cross-adaptation was suggested between heat stress and hypoxic stress. Materials and Methods: Male 4-month-old Wistar rats were randomly assigned to one of the four experimental groups (n = 8 each group): (1) control (C), (2) heat treatment (15H), (3) heat treatment and 15 days of normobaric hypoxia (15HHp), and (4) 15 days of normobaric hypoxia (15Hp). Three one-hour heat treatment sessions at 41°C were applied on the first two days before hypoxic exposure and on the day 7. Hypoxic exposure was consisting of normobaric hypoxia containing 9.7% O2. Results: Fifteen days of hypoxia did not increase the adrenal Hsp72 levels (p = 0.99). Furthermore, when hypoxia was added to the heat treatment, heat-related increases in adrenal Hsp72 levels disappeared. Adrenal weight to body weight ratio was not different among groups (p = 0.11). Plasma corticosterone levels were significantly lower in all experimental groups compared with control (p < 0.05), and addition of hypoxia resulted in further significant reduction of the plasma corticosterone levels (C > 15H>15HHp >15Hp; p < 0.05). Conclusions: These data demonstrate the adaptation of the adrenal gland to 15-day chronic normobaric hypoxic stress as well as possible cross-adaptation between heat and hypoxic stress in the adrenal gland.
Article
Full-text available
Photodynamic therapy (PDT) is an anti-tumor treatment administered for the elimination of early-stage malignancies and the palliation of symptoms in patients with late-stage tumors, which involves the activation of a photosensitizer (PS) using light of a specific wavelength, which also generates singlet oxygen and other reactive oxygen species (ROS) that cause tumor cell death. Several mechanisms are involved in the protective responses to PDT including the expression of chaperone/heat shock proteins (HSPs). The HSPs are a family of proteins that are induced by cells in response to exposure to stressful conditions. In the last few decades, it has been discovered that HSPs can play an important role in cell survival, due to the fact that they are responsible for many cytoprotective mechanisms. These proteins have different functions depending on their intracellular or extracellular location. In general, intracellular HSPs have been related to an anti-apoptotic function and recently, HSP-induced autophagy has shown to have a protective role in these chaperones. In contrast, extracellular HSPs or membrane-bound HSPs mediate immunological functions. In the present article, we attempt to review the current knowledge concerning the role of HSPs in the outcome of PDT in relation to autophagy and apoptosis mediated-resistance to photodynamic treatment. We will also discuss how certain PDT protocols optimally stimulate the immune system through HSPs.
Article
Full-text available
Photodynamic therapy is an alternative treatment mainly for cancer but also for bacterial infections. This treatment dates back to 1900 when a German medical school graduate Oscar Raab found a photodynamic effect while doing research for his doctoral dissertation with Professor Hermann von Tappeiner. Unexpectedly, Raab revealed that the toxicity of acridine on paramecium depends on the intensity of light in his laboratory. Photodynamic therapy is therefore based on the administration of a photosensitizer with subsequent light irradiation within the absorption maxima of this substance followed by reactive oxygen species formation and finally cell death. Although this treatment is not a novelty, there is an endeavor for various modifications to the therapy. For example, selectivity and efficiency of the photosensitizer, as well as irradiation with various types of light sources are still being modified to improve final results of the photodynamic therapy. The main aim of this review is to summarize anticancer and antibacterial modifications, namely various compounds, approaches, and techniques, to enhance the effectiveness of photodynamic therapy.
Chapter
Photodynamic therapy (PDT), a minimally-invasive therapeutic strategy, comprises a topical or systemic administration of a photosensitizer followed by selective irradiation of the target lesion. PDT is a promising alternative approach for cancer therapy which can directly destroy local tumor cells due to the release of cytotoxic reactive oxygen species at the tumor site. Importantly, PDT also induces immunological cell death, triggering an acute immune response that further potentiate PDT-mediated cell death. Nevertheless, some regulatory immune mechanisms can arise, limiting the immune response induced by PDT. The combination with immunotherapeutic approaches, like immune checkpoint inhibitors, is therefore necessary to reduce the immunosuppression at the tumor microenvironment and potentiate the antitumor immunity. Here, we explored the recent advances in the combinatory PDT immunotherapy and its current limitations for clinical translation.
Article
Photodynamic therapy (PDT) is a promising modality against various cancers including squamous cell carcinoma (SCC) with which the induction of apoptosis is an effective mechanism. Here, we initially describe the preclinical activity of 5-ethylamino-9-diethylaminobenzo [a] phenoselenazinium(EtNBSe)-mediated PDT treatment in SCC. Results of our studies suggest that EtNBSe-PDT provokes a cellular state of endoplasmic reticulum (ER) stress triggering the PERK/ eIF2α signaling pathway and induces the appearance of apoptosis in A431 cells at the meantime. With ER stress inhibitor 4-PBA or eIF2α inhibitor ISRIB, suppressing the EtNBSe-PDT induced ER stress substantially promotes apoptosis of A431 cells. Furthermore, we demonstrate that ATF4, whose expression is ER-stress-inducible and elevated in response to the PERK/eIF2α signaling pathway activation, contributes to cytoprotection against EtNBSe-PDT induced apoptosis. In a mouse model bearing A431 cells, EtNBSe shows intense phototoxicity and when associated with decreased ER stress, EtNBSe-PDT ameliorates tumor growth. Taken together, our study reveals an antagonistic activity of ER stress against EtNBSe-PDT treatment via inhibiting apoptosis in A431 cells. With further development, these results provide a proof-of-concept that downregulation of ER stress response has a therapeutic potential to improve EtNBSe-PDT sensitivity in SCC patients via the promotion of induced apoptosis.
Article
Cancer therapy has undergone tremendous advancements in the past few years in terms of treatment modalities. The drawbacks of most of the therapies have encouraged researchers to get an insight of the complex chemical, biochemical and biological processes ongoing in the ever evolving cancer cells. The studies have led to an advent of reactive oxygen species mediated therapies to target and disrupt the cancer pathology. Photodynamic therapy (PDT) has emerged as a potent candidate for oxidative stress mediated non-invasive technique for rapid diagnosis and treatment of cancer. Towards this, biomacromolecules derived hybrid nanomaterials have contributed largely in the development of various therapeutics and theranostics for efficacious cancer management that can assist PDT. This review summarizes various hybrid biomaterials and advanced techniques that have been explored widely in the past few years for PDT application. The article also mentions some of the important in-vitro and in-vivo developments and observations explored by employing these materials for PDT application. The article also unravels the interactions of these materials at the biological interface and the probable mechanism that assist in generation of oxidative stress and subsequent cell death.
Article
Full-text available
Singlet oxygen, a metastable state of normal triplet oxygen, has been identified as the cytotoxic agent that is probably responsible for in vitro inactivation of TA-3 mouse mammary carcinoma cells following incorporation of hematoporphyrin and exposure to red light. This photodynamic inactivation can be completely inhibited by intracellular 1,3-diphenylisobenzofuran. This very efficient singlet oxygen trap is not toxic to the cells nor does it absorb the light responsible for hematoporphyrin activation. We have found that the singlet oxygen-trapping product, o-dibenzoylbenzene, is formed nearly quantitatively intracellularly when both the furan and hematoporphyrin are present during illumination but not when only the furan is present during illumination. The protective effect against photodynamic inactivation of the TA-3 cells afforded by 1,3-diphenylisobenzofuran coupled with the nearly quantitative formation of the singlet oxygen-trapping product indicates that singlet oxygen is the probable agent responsible for toxicity in this system.
Article
Full-text available
Retroviral-mediated gene transfer experiments show that rodent cells become heat resistant when stably and constitutively expressing a cloned human gene encoding an intact human 70-kDa heat shock protein (hsp70). Cells expressing higher levels of the hsp70 protein generally tolerate thermal stress better, whereas cells expressing either of two mutated hsp70-encoding genes, one with a 4-base pair out-of-frame deletion and one with an in-frame deletion of codons 438-618, are heat sensitive. These results provide strong evidence that expression of hsp70 leads directly to thermal tolerance. Surprisingly, cells expressing a mutant hsp70 of a human gene missing codons 120-428 are, nevertheless, heat resistant. Because the deleted region of this mutant contains the ATP-binding domain of human hsp70, this domain appears dispensable in the hsp70-mediated protection of cells from thermal stress.
Article
Full-text available
A mouse mammary tumor model was used to evaluate metabolic properties of the photosensitizer mono-L-aspartyl chlorin e6 (NPe6) and to determine the optimal time interval between drug administration and light treatment for effective photodynamic therapy (PDT). Photosensitizer metabolism was evaluated by comparing tissue distribution patterns of NPe6 having 14C atoms positioned on either the tetrapyrrole ring or on the aspartyl residue. High performance liquid chromatographic analysis of photosensitizer extracted from tumor tissue was also obtained as a function of time after drug administration. NPe6 distribution in tissue samples and pharmacological calculations of area under the curve were similar for both forms of [14]NPe6. Likewise, metabolic contaminants of NPe6 were not detected by high performance liquid chromatographic analysis following extraction of the photosensitizer from tumor tissue. Maximal in vivo PDT effectiveness was achieved when light treatments were started within 2 h of drug injection. PDT effectiveness was decreased by 50% when light treatments were initiated 6 h after drug injection and was abolished with a 12-h interval between NPe6 injection and light exposure. Responsiveness to NPe6-mediated PDT was correlated with photosensitizer levels in the plasma but not in tumor tissue. These results show that NPe6 was not metabolized following in vivo administration and that the responsiveness of NPe6 mediated PDT was associated with vascular clearance of the photosensitizer.
Article
Numerous photosensitizers with absorption peaks spanning the 600-800 nm "therapeutic window" have been and continue to be synthesized. Structural modifications of the dyes can then be made in order to improve tumor deliverability and retention. Chemical alterations can also enhance the yields of light generated reactive oxygen species. Utilization of lipoproteins, emulsions and antibody conjugates can enhance the selectivity of drug localization. Most cell types and subcellular structures are highly photosensitive and biochemical analysis indicates that cellular target sites associated with PDT correlate with photosensitizer location. In vivo data suggest that vascular and direct tumor cell damage as well as systemic and local immunological reactions are involved in PDT responsiveness. Additional mechanistic, synthetic and developmental studies are required in order to fully appreciate the potentials of PDT. However, continued enthusiasm and support for basic PDT research (as observed during the past 8 years) will depend to a large extent on the outcome of the current clinical trials.
Article
GRP78, a 78,000 dalton protein residing in the endoplasmic reticulum, is postulated to play important roles in protein folding and cell survival during calcium and other physiological stress. Here we describe the construction of an eukaryotic expression vector for the constitutive expression of grp78 antisense RNA and the creation of a CHO cell line, 78WO, which expresses high levels of the grp78 antisense RNA through amplification of the stably transfected antisense vector. We observed that whereas 78WO maintains a basal level of GRP78 similar to that of control cells, GRP78 is no longer inducible by A23187. The 78WO cells have undergone a compensatory increase in grp78 transcription such that the effects of antisense are cancelled out at the protein level under nonstressed conditions. In these same cells, GRP94, a 94,00 dalton ER protein, is also rendered noninducible by A23187. This provides the first evidence that the regulation of two ER proteins might be coupled such that the failure to induce GRP78 results in the down-regulation of GRP94. The 78WO cell line grows with a doubling time of about 26 hr and exhibits decreased tolerance to A23187, suggesting the GRPs contribute to cell viability under calcium stress. The establishment of this cell line, which can be stably maintained, will provide a useful tool for testing whether the induction of the GRPs is important for protein folding or transport and whether their enhanced synthesis is the cause or consequence of a variety of physiological adaptations.
Article
The clinical use of photodynamic therapy (PDT) has been ongoing for over a decade. However, attempts to apply for approval of the therapy from boards of health for general use began only in 1989. The unique nature of PDT and the resultant changes in the normal drug registration process, as well as steps which are being taken to approve PDT for the treatment of endobronchial lung cancer, superficial bladder cancer and esophageal cancer are described. The current clinical status of PDT in these indications is also reviewed.
Article
Porphyrin mediated photosensitization can enhance the transcription and translation of several oxidative stress genes. In this study, we report on the enhanced expression of the gene encoding for heme oxygenase in Chinese hamster fibroblasts by; (1) incubation in Photofrin II; (2) Photofrin II mediated photosensitization; and (3) photosensitization induced by Rose Bengal. Increased expression of heme oxygenase mRNA was accompanied by a concomitant increase in the synthesis of the 34 kDa heme oxygenase protein. Western blot analysis using antibody to heme oxygenase confirmed the immunoreactivity of the 34 kDa protein induced by Photofrin II and PDT. These results demonstrate that heme oxygenase can be activated by non-metalloporphyrins as well as by photosensitization associated with singlet oxygen mediated subcellular injury.