Open Journal of Stomatology, 2011, 1, 195-201
doi:10.4236/ojst.2011.14030 Published Online December 2011 (http://www.SciRP.org/journal/ojst/
OJST
).
Published Online December 2011 in SciRes. http://www.scirp.org/journal/OJST
Radiation-inducible promoter mediated PUMA gene in the the
treatment of Tca8113
Dong-Sheng Yu1, We i Zhao1, Jia-Xian Luo1, Bing Liu1, Xiao-Lin Wu2
1Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China;
2Department of Stomatology, The Conde S. Januário Hospital Centre, Macao, China.
Email: gdyudongsheng@yahoo.com.cn
Received 12 August 2011; revised 20 September 2011; accepted 3 October 2011.
ABSTRACT
Objective: The aim of this study is to explore the
therapeutic effect of the PUMA gene mediated by ra-
diation-inducible promoters in the treatment of ton-
gue squamous cell carcinoma. Methods: Recombi-
nant pcDNA3.1 (+)/E-PUMA was constructed, in
which the PUMA gene was mediated by a synthetic
radiation inducible promoter. The recombined plas-
mids were transfected into the Tca8113 cell and xeno-
grafts of human tongue squamous carcinoma in na-
ked mice respectively. After 24 h, the tumors were tr-
eated with 3 Gy of irradiation to upregulate the PU-
MA gene expression. PUMA mRNA was detected by
RT-PCR. Proliferating cell nuclear antigen (PCNA)
and apoptosis were detected by immunohistochemi-
cal method and in situ end-labeling (ISEL) respecti-
vely. The data were analyzed using the SPSS11.0 soft-
ware package for chi-square test. Results: Compared
with the control group, the comparative survival rate
of Tca8113 cells in the PUMA/IR group was markedly
decreased and the xenografts were signifycantly sup-
pressed. Up-regulation of PUMA gene expression was
observed in the Tca8113 cells and in the xenografts
after irradiation. The apoptosis indices of the Tca-
8113 cells and xenograft with irradiation were mar-
kedly higher than those without irradiation. At the
same time, the proliferation indices of the Tca8113
cells and xenografts with irradiation were markedly
lower than those without irradiation. Conclusions:
radiation-inducible promoters can serve as molecular
switches to improve the expression of PUMA gene in
tongue squamous cell carcinoma both in vivo and in
vitro. Low-dose induction radiation can significantly
improve therapeutic efficiency.
Keywords: Blast; Graph Theory; Redundant Sequences;
CD-HIT
1. INTRODUCTION
The p53 up-regulated modulator of apoptosis (PUMA)
was newly discovered in 2001, which possessed power-
ful pro-apoptotic effect as a member of the Bcl-2 family
and the key protein during the apoptosis caused by ra-
diation [1-3]. Although its specific mechanism for in-
ducing apoptosis needs further explanation, the pro-
apoptotic function has achieved rather good results, be-
coming a promising new target in gene therapy [4-6].
Radiation-inducible promoters can regulate and control
the downstream therapeutic gene expression, a new stra-
tegy for radiation-gene therapy founded in recent years
[7,8]. It combines traditional radiation treatment with
gene therapy, which may reduce radiation dosage and in-
creases efficacy of gene therapy [9,10]. Human tongue
squamous cell carcinoma is the most common type of
neoplasm in the oral cavity [11,12]. Little is known
about the role of PUMA in the targeted treatment of hu-
man tongue squamous cell carcinoma. This research adopt-
ed radiation-inducible promoters to mediate PUMA gene
for treating human tongue squamous cell carcinoma in
vivo or in vitro, to observe the synergistic effect of radia-
tion and to explore a new policy for the targeted therapy
of tongue squamous cell carcinoma.
2. MATERIALS AND METHODS
2.1. Cells and Cell Culture
Tongue squamous carcinoma cells (China Center for Ty-
pe Culture Collection Wuhan University Hubei China)
were cultured in RPMI 1640 (GIBCO, Carlsbad, CA,
USA) culture medium supplemented with 10% fetal calf
serum (GIBCO), 50 U/ml penicillin G, and 50 µg/ml
streptomycin. Subculture occurred weekly (seeded at 5 ×
105 cells/ml), with feeding with complete medium twi-
ce a week. All cells were cultured in a humidified 5%
CO2 environment at 37˚C.
2.2. Animals
BALB/c naked mice were provided by the Experimen-
D.-S. Yu et al. / Open Journal of Stomatology 1 (2011) 195-201
196
tal Animal Center of Sun Yat-sen University. Animal ex-
periments were performed with the permission from the
Animal Ethical Commission of Sun Yat-sen University,
China.
2.3. pcDNA (+)–3.1/E-PUMA Construction
All restriction and modifying enzymes were supplied by
Promega or Roche and were used according to the ma-
nufacturer’s instructions. DNA isolation and purification
were carried out using the appropriate kit from Qiagen
(Crawley, UK). Nhe I and Hind III were used for conduct-
ing double digestion of pMD18-T-E, producing DNA frag-
ments containing radiation inducible promoter sequences.
The six-CArG elements enhancer (E6) was cloned using
the complementary single-strand-edoligodeoxyribo-nu-
cleotides pair: 5’-AGATCTGC TAGC(CCATATAA-GG)6-
TCGCGAA-3’ and 5’-T CGCGA(CCTTATATGG)6GC-
TAGCAGATCTA-3’, which were synthesized by the Son-
gon Company, Shanghai, China. Linkers were produced
by mixing 0.05 nmol of each oligonucleotide in 5 µl vo-
lumes, heating them at 55˚C for 5 min and then cooling
them to room temperature. They were inserted into PU-
MA upstream of the plasmid pcDNA (+)3.1-PUMA, ob-
taining the eukaryotic expression plasmid pcDNA (+)3.1-
E-PUMA,which carries the radiation inducible promo-
ter mediated PUMA gene.
2.4. The Killing Effect of PUMA Gene Mediated
by Radiation-Inducible Promoter in Vitro
Grouping. Tca8113 cells were divided into four groups
as follows:
Control group: no intervention; PUMA group: PUMA
gene transfection only; IR group: irradiation only; PU-
MA/IR group: PUMA gene transfection and irradiation.
Transfection of DNA. Tca8113 cells were transfected
with Lipofectamine (GIBCO) by modifying the manu-
facturer’s procedure based on our previous study [7,8].
Tca8113 cells (2 × 105) were seeded into a six-well plate
at 2 × 105 cells per well, and conventionally maintained
for 24 h. A total of 24 wells of Tca8113 cells were pre-
pared to be transfected. Two micrograms of plasmid
DNA (10 µL) was mixed with 90 µL of serum-free me-
dium. This mixture was then incubated with a mixture of
15 µL lipofectamine (2 mg/mL) and 85 µL serum-free
medium for 45 min at room temperature. Subsequently,
the DNA-Lipofectamine complex (200 µL) was added to
each well.
Radiation procedure. After transfection for 24 h, the
transfected cells were irradiated by linear accelerator
(Primvs, Siemens, Germany) at a dose of 3 Gy. We cho-
se the radiation dose of 3 Gy based on our previous re-
search [8]. The monolayers of the transfected cells in the
control group were also mock-irradiated and subse qu-
ently treated identically as with the irradiated monolay-
ers of transfected cells.
Microculture tetrazolium assay. A micro-culture te-
trazolium (MTT) was used to assess cell viability. All ex-
periments were performed in triplicate. The results were
expressed as percentages of the control, which was con-
sidered 100%.
RNA preparation and RT-PCR. After IR for 48 h,
total mRNA from transfected Tca8113 cells was ex-
tracted using an RNeasy Mini Kit (Qiagen, USA) ac-
cording to the manufacturer’s in structions. RT-PCR was
carried out using a reverse transcription kit (Promega,
USA) and human β-actin gene was used as the internal
control. The PUMA gene primers were as follows: 5’-T-
GTCGAATAAACGCTTTACAAAC-3’ (forward) and 5’-
AACGTTTGTAATG ATGG CTTCTG-3’ (reverse). The
β-actin primers were as follows: 5’-GGTCGGAGTCA-
ACGGATTTGGTCG-3’ (forward) and 5’-CCTCCGAC-
GCCTGCTTCAC CAC-3’ (reverse). The transcript lev-
els were normalized according to transcription of the β-
actin internal control, and the products were resolved by
agarose gel electrophoresis. The intensity was quantified
by image-analysis software (NIH Image; Apple, Cuper-
tino, CA, USA).
Detection of apoptosis by flow cytometry. After trans-
fection for 48 h, both adherent and floating cells were
harvested and centrifuged. The cell pellets were washed
with phosphate-buffered saline (PBS) and then fixed
with overnight with 70% ethanol. After incubation in
phosphate-citric acid, the cells were resuspended in PBS
containing propidium iodide (10 µg/mL) and RNase (10
µg/mL) .The cells were quantified with a FACScan flow
cytometer (Becton Dickinson). Salmon sperm DNA (ss-
DNA) was used as the DNA toxicity control.
2.5. Tumor Inhibition Effect in Vivo
Establishment of transplantable tumor model. The
freeze-stored Tca8113 cells were revived in an incubator
with 5% CO2 at 37˚C, and subcultured in RPMI-1640
containing 100 mL/L fetal bovine serum. The trypsin
method was utilized to collect the cancer cells. Naked
mice 6 to 8 weeks old were given subcutaneous inject-
tions at 6 × 106 inoculated tumor cells for each mouse.
When the diameters of transplanted tumor reached 8 mm,
the treatments were begun.
Plasmid transfection and radiation therapy. The
mice with transplanted tumors were randomly divided
into four groups with 10 mice for each group: (A) con-
trol group, (B) transfection group, (C) radiation group,
and (D) transfection and radiation group. Then, 2 μg of
the plasmid pcDNA3.1 (+)/E-PUMA was dissolved into
100 μL of serum-free RPMI-1640 culture medium. At
the same time, 80 μL serumfree RPMI-1640 culture me-
C
opyright © 2011 SciRes. OJST
D.-S. Yu et al. / Open Journal of Stomatology 1 (2011) 195-201 197
dium was used to dilute 20 μL Lipofectamine. Both of
them were mixed and shaken well, and another 800 μL
of serum-free RPMI-1640 culture medium was added
into the mixture. This prepared miscible liquid was used
for intratumoral injection (100 μL/mouse). After 24 h
transfection, groups C and D were exposed to 3 Gy of
radiation via a linear accelerator.
Description of tumor growth curve. Vernier caliper was
used to measure maximum of the tumors and maximum
transverse diameter of tumors (b) every four days, and
the maximum tumor size of multiple tumors were re-
corded. Tumor size was calculated using the formula V =
1/2(a × b2). On the 20th day of transfection, the animals
were euthanized and tumor growth curves were con-
structed.
RNA preparation and RT-PCR. After transfection
for 96 h, the naked mice were sacrificed and the tumor
tissues were subsequently collected. RT-PCR was em-
ployed as in the in vitro experiment described above.
Detect PCNA expression. The proliferating cell nu-
clear antigen (PCNA) was detected with immunohisto-
chemical kit (Boster, Wuhan, China) according to the
manufacturer’s protocol: Briefly, the tumor specimens
were conventionally fixed, embedded, sectioned, depar-
affinized, incubated for 15 min at RT in 3% H2O2 diluted
in methanol; add diluted primary antibody and incubate
overnight at 4˚C; The sections were then washed three
times with buffer solution, secondary antibodies (rabbit
anti-rat monoclonal antibodies 1/75 dilution) were added,
and the specimens were incubated for 30 min at room
temperature. ABC reagent was added and the sections
were incubated for 30 min and immersed in hematoxylin
for 90 s for counterstaining. Primary antibodies were
replaced with PBS for the blank control whereas oral
mucosal cells were used for the positive control.
Proliferation indices (PIs) were calculated by observ-
ing the proportion of positively staining cells. Staining
was considered positive when the cell nuclei had been
stained brown. The number of positive cells and total
observed cells was counted under light microscopy
(×400 magnification) with a 10 × 10 grid. The observers
randomly selected distinct fields during three separate
evaluations in each slide. Ten sections were used to
evaluate every group. PIs were calculated using the fol-
lowing formula: PI = (positive ly staining cells/total ob-
served cells) × 100%.
In situ end-labeling (ISEL) detects apoptosis. Apop-
tosis was detected by in situ end-labeling (ISEL) with an
apoptosis labeling kit (Boster, Wuhan, China) according
to the manufacturer’s protocol. Tumor specimens were
conventionally fixed, embedded, sectioned, and deparaf-
finized. Endogenous peroxidases were then blocked by
immersing the slides in 3% H2O2 for 30 min at room
temperature. The slides were incubated in ISEL solution
in an 18˚C ice bath for 2 h. ABC solution was then ap-
plied and the slides were incubated at room temperature
for 30 min. DAB was subsequently applied for 5 min at
room temperature. Counterstaining was done by immer-
sion in hematoxylin for 60 s. Finally, the slides were
rehydrated and topped with coverslips. For the blank
control, the DIG-dUTP was replaced with TBS solution,
whereas oral mucosal cells were used in the positive
control.The calculation of AI is the same as that of PI.
2.6. Statistical Analysis
All experiments, as well as the measurements on each
experiment, were run in triplicate. The experimental data
are presented as means ±s.d. Statistical analyses were
performed by a one-way ANOVA and chi-square test us-
ing SPSS software 11.0 (SPSS Inc., Chicago, IL, USA).
A P value of 0.05 was considered statistically signify-
cant.
3. RESULTS
3.1. Inhibition of Tca8113 Growth Cells in Vitro
The variability of Tca8113 cells was detected by MTT
assay. Figure 1 shows that Tca8113 cells grew rapidly in
the blank control group. The Tca8113 cells in the PUMA
group and the IR group were significantly inhibited com-
pared with the control group. Furthermore, the growth of
Tca8113 cells in the PUMA/IR group was significantly
more inhibited than in any other group.
3.2. Expression of PUMA mRNA in Transfected
Tca8113 Cells
After IR for 48 h, the PUMA expression was detected by
RT-PCR. Figure 2 shows that the mRNA levels in the
PUMA/IR group (0.782 ± 0.035) were significantly
Figure 1. Tca8113 cell growth in vitro. The
growth of Tca8113 cells in the PUMA/IR
group was significantly inhibited compared
with any other group. There was significant
difference between the PUMA group and
the PUMA/IR group.
C
opyright © 2011 SciRes. OJST
D.-S. Yu et al. / Open Journal of Stomatology 1 (2011) 195-201
198
1900 bp
750 bp
Figure 2. PUMA transcription after trans-
fection into Tca8113 cells. Lines M, DNA
maker; Line 1, product of β-actin (750
bp); Line 2, product of PUMA gene in
the PUMA/IR group; Line 3, product of
PUMA gene in the PUMA group.
higher than those in the PUMA group (0.527 ± 0.013, P
< 0.01). This result suggests that the synthetic radiation
inducible promoters combined with IR up-regulated
PUMA gene expression in the transfected Tca8113 cells.
3.3. FACScan Analysis of Cells Apoptosis
After IR for 48 h, the percentage of cell apoptosis in the
PUMA/IR group was about (26.32 ± 2.32)%, which is
significantly higher than the (16.65 ± 1.44)% in the
PUMA group (P < 0.05) and the (9.76 ± 0.85)% in the IR
group (P < 0.01) (Figure 3). These results indicate that
IR marked enhanced the anticancer ability of PUMA ge-
ne therapy in vitro.
3.4. Tumor Gr owth Curve
The transplanted tumor growth curves of the naked mice
from different groups are shown in Figure 4. PUMA
gene transfection and 3Gy irradiation exposure have cer-
tain inhibitory effects on tumor growth, but are obvi-
ously lower than that of the radiation gene therapy.
0
2
4
6
8
10
12
14
16
18
ContrPUMA IRPUMA/IR
Apoptosis rate (%
)
Figure 3. Cells apoptosis in different group were analyzed by
flow cytometry 48 h after irradiation. The apoptosis index was
highest (26.32 ± 2.32)% in the PUMAIR group, which was
significantly higher than (16.65 ± 1.44) in the PUMA group (P
< 0.05) and (9.76 ± 0.85)% in the IR group (P < 0.01).
Figure 4. The growth curves of tumors in naked
mice. Tumor growth was fast in the blank con-
trol group, but significantly depressed in the IR,
PUMA and PUMAIR groups. Furthermore,
there was a significant difference between the
PUMA group and PUMA/IR group.
3.5. PUMA mRNA Expression in the Xenografts
PUMA gene mRNA expression is merely detected in the
control group and the IR group. The PUMA gene frag-
ment was detected in both the PUMA group and the
PUMA/IR group (Figure 5), but the band intensity of
the former is significantly higher than that of the latter.
Computer image analyzer scans of the electrophoretic
band grey values were compared with the β-actin grey
value and their ratios are shown in Ta b l e 1 . The differ-
ences between the two groups are significant (P < 0.05).
Radiation treatment increases PUMA expression level.
3.6. PCNA Staining and ISEL Labeling Results
The PCNA staining and ISEL labeling results are shown
in Figures 6(a)-(d) and Figure 6(A)-(D), whereas the PI
and AI results are in Table 2.
4. DISCUSSION
Apoptosis is a gene-determined active cell death. It plays
a key role in the normal development of multi-cellular
organisms. The occurrence of tumor is closely linked with
2000 bp
750 bp
1000 bp
500 bp
250 bp
M 1 2 3 4 5
Figure 5. Agarose gel electrophoresis of RT-
PCR products M: DNA marker; 1: control group;
2: radiation group; 3: tranfection and radiation
group; 4: tranfection group.
C
opyright © 2011 SciRes. OJST
D.-S. Yu et al. / Open Journal of Stomatology 1 (2011) 195-201
Copyright © 2011 SciRes.
199
Table 1. Comparison of grey level of agarose gel electrophore-
sis.
Group Mean (χ ± s)
PUMA group 0.826 ± 0.081
PUMA/IR group 1.031 ± 0.096
Table 2. The AIs and PIs in the different groups (χ ± s%).
Group PI (%) AI (%)
control 43.54 ± 6.42 8.76 ± 1.51
IR 35.26 ± 5.31a 12.33 ± 3.11a
PUMA 33.44 ± 4.32a 4.76 ± 4.12a
PUMA/IR 25.68 ± 5.43ab 28.98 ± 5.22ab
aCompared with control, P < 0.05; bCompared with PUMA, P < 0.05.
abnormal apoptosis [13-15]. Although many p53-regu-
lated genes participate in apoptosis, PUMA is currently
considered one of the key genes in the process [1,2,16].
PUMA belongs to the BH-3 subfamily of the bcl-2 pro-
tein family. The BH-3 structural domain and its position
in the mitochondria are indispensable in the induction of
apoptosis and inhibition of cell growth [1,17]. PUMA in-
duces apoptosis through p53-dependent and non-p53 de-
pendent pathways. Differing from other target spots, PU-
MA not only regulates expression through common p53
responsive elements located in its promoter, but also in-
duces expression by non-p53 dependent mechanisms,
such as glucocorticoids, removal of growth factors, ki-
nase inhibitors and phorbol esters [18,19]. While study-
ing the mechanism of myoblast apoptosis during the dif-
ferentiation and development process of the skeletal mu-
scle, Shaltouki et al. [20] found that PUMA can directly
mediate the release of cytochrome C through a non-p53-
dependent pathway, thereby activating Caspase-9 and
causing myoblast apoptosis. The function of PUMA is
unaffected by p53 expression. In experiments by Wang
[21], there was no correlation between p53 expression
and the inhibition of proliferation and the induction of
apoptosis in esophageal cancer cell lines, including KY-
SE-150, KYSE-410, KY SE-510, and YES-2. Exoge-
nous transfection of PUMA into human melanoma cell
systems causes apoptosis regardless of p53 expression.
Hideaki reported that extensive apoptosis occurred in the
expression mutation and wild characteristic of four PU-
MA-transfected cell lines [22]. Wang also found that the
inhibition of proliferation and induction of apoptosis by
Ad-PUMA in esophageal cancer cell lines were better
than that of Ad-p53 [21]. Apoptosis induction by PUMA
is independent of p53 expression, and its effect is better
than that of p53, which makes it a potential target for
gene therapy.
PUMA is a key protein in apoptosis, and radiation
damage can result in PUMA activation, leading to apop-
tosis through a series of complex mechanisms [23] Al-
though its specific induction mechanism requires further
explanation, the application of the pro-apoptotic function
of p53 in anti-tumor experiments has achieved fairly
Figure 6. Expression of PCNA and apoptosis cells in different group tumors. Expression of
PCNA (SABC, ×400) in the: (a) control group; (b) IR group; (c) PUMA group; (d)
PUMA/IR group. Apoptotic cells (ISEL, ×400) in the: (A) control group; (B) IR radiation
group; (C) PUMA group; (D) PUMA/IR group.
OJST
D.-S. Yu et al. / Open Journal of Stomatology 1 (2011) 195-201
200
good results. Yu et al. found that PUMA gene transfec-
tion could enhance the sensitivity of lung cancer cells to
radiotherapy [24].
There may be serious side effects in patients receiving
radiotherapy. Decreasing the occurrence of these side ef-
fects will be beneficial for cancer patients. PUMA gene
transfection can promote the apoptosis of tumor cells
and enhance its sensitivity to radiotherapy [25,26]. Cur-
rently, methods to increase PUMA-targeted expression
in tumors are the largest obstacle in the application of
PUMA in clinical practice. A new radiation-gene strat-
egy that uses radiation inducible promoters that stimu-
late downstream gene expression to construct control
and regulation systems for therapeutic gene expression
has been adopted in the current research [7,27,28]. This
system is a typical exogenous regulation mechanism that
can selectively kill tumor cells while protecting normal
structures from damage [7,29]. With the development of
radiotherapy techniques such as intensity modulation
radiated therapy and three-dimensional conformal radio
therapy, the radiation range can be adjusted to the target
areas by regulating the output dosage of various areas in
the radiation field [30,31].
To educe synergistic effects by integrating various
therapeutic strategies together is a trend in current gene
therapy [31,32]. In the current study, we demonstrated
that PUMA gene expression in tongue squamous cell
carcinoma was significantly upregulated at the transcri-
ptional level by treatment with radiation-inducible pro-
moters following 3 Gy of IR. We also proved that the
combination of PUMA gene therapy system with 3 Gy
of IR could significantly inhibit the growth of tumor
cells by promoting apoptosis and reducing proliferation
in vivo and in vitro. Although these results are not yet
applicable for the treatment of humans, they indicate that
radiation inducible promoters can serve as molecular
switches for regulating the expression of the PUMA ge-
ne and that low-dose radiation can significantly improve
therapeutic targeting efficiency in treatment of tongue
squamous cell carcinoma.
5. ACKNOWLEDGEMENTS
This study was supported by the National Natural Sciences Foundation
of China (Grant No. 3097 3340), the Guangdong Sciences and Tech-
nology Project (Grant No. 2008B030301113 and 2011B050400030),
and the Guangdong Natural Sciences Foundation (Grant No. 915100-
890100 0187 and S2011020003247).
REFERENCES
[1] Nakano, K. and Vousden, K.H. (2001) PUMA, a novel
proapoptotic gene, is induced by p53. Molecular Cell,
7, 683-694. doi:10.1016/S1097-2765(01)00214-3
[2] Labi, V. and Villunger, A. (2010) PUMA-mediated
tumor suppression: A tale of two stories. Cell Cycle, 9,
4269-4275. doi:10.4161/cc.9.21.13666
[3] Avila, J.L., Grundmann, O., Burd, R., et al. (2009)
Radiation-induced salivary gland dysfunction results
from p53-dependent apoptosis. International Journal
of Radiation Oncology, Biology, Physics, 2, 523-529.
doi:10.1016/j.ijrobp.2008.09.036
[4] Chen, Y., Xiang, Y. and Lin, C. (2007) PUMA—A
promising tumor gene therapy target. Oncology Pro-
gress, 5, 282-285.
[5] Michalak, E.M., Vandenberg, C.J., Delbridg, A.R., et
al., (2010) Apoptosis-promoted tumorigenesis: Gamma-
irradiation-induced thymic lymph omagenesis requires.
Puma-driven leukocyte death Genes Dev, 24, 1608-
1613. doi:10.1101/gad.1940110
[6] Shao, L., Sun, Y., Zhang, Z., et al. (2010) Deletion of
proapoptotic PUMA selectively protects hematopo-
ietic stem and progenitor cells against high-dose ra-
diation. Blood, 23, 4707-4714.
doi:10.1182/blood-2009-10-248872
[7] Yu, D.-S., Zhao, W., Huang, H.-Z., et al. (2010) Syn-
thetic radiation-inducible promoters mediated HSV-
TK/GCV gene therapy in the treatment of oral squamous
cell carcinoma. Oral Diseases, 16, 445-452.
doi:10.1111/j.1601-0825.2010.01655.x
[8] Rao, G.V., Kumar, N.A., Gupta, R., et al. (2010) Ra-
diation-inducible silencing of uPA and uPAR in vitro
and in vivo in meningioma. International Journal of
Oncology, 4, 809-816.
[9] Nandi, S., Ulasov, I.V., Tyler, M.A., et al. (2008)
Low-dose radiation enhances surviving-mediated vi-
rotherapy against malignant glioma stem cells. Cancer
Research, 14, 5778-5784.
doi:10.1158/0008-5472.CAN-07-6441
[10] Soudry, E., Preis, M., Hod, R., et al. (2010) Squamous
cell carcinoma of the oral tongue in patients younger
than 30 years: Clinicopathologic features and outcome.
Clinical Otolaryngology, 4, 307-312.
doi:10.1111/j.1749-4486.2010.02164.x
[11] Zhang, T.H., Liu, H.C., Zhu, L.J., et al. (2011) Activa-
tion of notch signaling in human tongue carcinoma.
Journal of Oral Pathology & Medicine, 1, 37-45.
doi:10.1111/j.1600-0714.2010.00931.x
[12] Elias, S.T., Diniz, J., Almeida, R.S., et al. (2010) Cy-
totoxic effect of tobacco extracts on human oral
squamous cell carcinoma cell-line. Oral Oncology, 12,
869-873. doi:10.1016/j.oraloncology.2010.09.008
[13] Hajji, N. and Joseph, B. (2010) Epigenetic regulation
of cell life and death decisions and deregulation in
cancer. Eessays in Biochemistry, 1, 121-146.
doi:10.1042/bse0480121
[14] Kerr, J.F., Wyllie, A.H. and Currie, A.R. (1972) Apop-
tosis: A basic biological phenomenon with wide-
ranging implications in tissue kinetics. British Journal
of Cancer, 4, 239-257.
doi:10.1038/bjc.1972.33
[15] Qiu, W., Leibowitz, B., Zhang, L., et al. (2010)
Growth factors protect intestinal stem cells from radia-
tion-induced apoptosis by suppressing PUMA through the
PI3K/AKT/p53 axis. Oncogene, 11, 1622-1632.
doi:10.1038/onc.2009.451
C
opyright © 2011 SciRes. OJST
D.-S. Yu et al. / Open Journal of Stomatology 1 (2011) 195-201 201
[16] Chipuk, J.E. and Green, D.R. (2009) PUMA cooper-
ates with direct activator proteins to promote mito-
chondrial outer membrane permeabilization and apop-
tosis. Cell Cycle, 17, 2692-2696.
doi:10.4161/cc.8.17.9412
[17] Yu, J. and Zhang, L. (2008) PUMA, a potent killer
with or without p53. Oncogene, 27, S71-S83.
doi:10.1038/onc.2009.45
[18] Hong, L.Z., Zhao, X.Y. and Zhang, H.L. (2010)
p53-mediated neuronal cell death in ischemic brain
injury. Neuroscience Bulletin, 3, 232-240.
doi:10.1007/s12264-010-1111-0
[19] Shaltouki, A., Freer, M., Mei, Y., et al. (2007) In-
creased expression of the pro-apoptotic Bcl2 family
member PUMA is required for mitochondrial release
of cytochrome C and the apoptosis associated with
skeletal myoblast differentiation. Apoptosis, 12, 2143-
2154. doi:10.1007/s10495-007-0135-z
[20] Wang, H., Qian, H., Yu, J., et al. (2006) Administra-
tion of PUMAadenovirus increases the sensitivity of
esophageal cancer cellsto anticancer drugs. Cancer
Biology & Therapy, 4, 380-389.
doi:10.4161/cbt.5.4.2477
[21] Hideaki, I., Takao, K. and Toru, M. (2005) Efficacy of
PUMA for malignant glioma cells regard less of the
p53 status. Human Gene Therapy, 6, 685-692.
[22] Vousden, K.H. (2005) Apotosis. p53 and PUMA: A
deadly duo. Science, 5741, 1685-1686.
doi:10.1126/science.1118232
[23] Yu, J., Yue, W., Wu, B., et al. (2006) PUMA sensitizes
lung cancer cells to chemotherapeutic agents and irra-
diation. Clinical Cancer Research, 9, 2928-2935.
doi:10.1158/1078-0432.CCR-05-2429
[24] Qiu, W., Carson-Walter, E.B., Liu, H., et al. (2008)
PUMA regulates intestinal progenitor cell radio-sen-
sitivity and gastrointestinal syndrome. Cell Stem Cell,
6, 576-583. doi:10.1016/j.stem.2008.03.009
[25] Sasano, N., Enomoto, A., Hosoi, Y., et al. (2010) Eda-
ravone, a known free radical scavenger, enhances
X-ray-induced apoptosis at low concentrations. Can-
cer Letters, 1, 52-57. doi:10.1016/j.canlet.2009.12.020
[26] Tsurushima, H., Yuan, X., Dillehay, L.E., et al. (2008)
Radiation-inducible caspase-8 gene therapy for ma-
lignant brain tumors. International Journal of Radia-
tion Oncology, Biology, Physics, 2, 517-525.
doi:10.1016/j.ijrobp.2008.02.002
[27] Marignol, L., Coffey, M., Hollywood, D., et al. (2007)
Radiation to control transgene expression in tumors.
Cancer Biology & Therapy, 7, 1005-1012.
doi:10.4161/cbt.6.7.4477
[28] Vilaboa, N. and Voellmy, R. (2006) Regulatable gene
expression systems for gene therapy. Current Gene
Therapy, 4, 421-38.
doi:10.2174/156652306777934829
[29] Bhide, S.A. and Nutting, C.M. (2010) Recent ad-
vances in radiotherapy. BMC Medicine, 8, 25-28.
doi:10.1186/1741-7015-8-25
[30] Brahme, A. and Lind, B.K. (2010) A systems biology
approach to radiation therapy optimization. Radiation
and Environmental Biophysics, 2, 111-124.
doi:10.1007/s00411-010-0268-2
[31] John-Aryankalayil, M., Palayoor, S.T., Cerna, D., et al.
(2010) Fractionated radiation therapy can induce a
molecular profile for therapeutic targeting. Radiation
Research, 4, 446-458. doi:10.1667/RR2105.1
[32] Marignol, L., Robson, T., McCarthy, H.O., et al. (2008)
The tissue plasminogen activator gene promoter: A
novel tool for radiogenic gene therapy of the prostate?
Journal of Gene Medicine, 9, 1032-1038.
doi:10.1002/jgm.1221
C
opyright © 2011 SciRes. OJST