Anda di halaman 1dari 11

Nanotechnology

Nanotechnology 30 (2019) 055101 (11pp) https://doi.org/10.1088/1361-6528/aaedd5

Use of DNA-generated gold nanoparticles to


radiosensitize and eradicate radioresistant
glioma stem cells
Tatsuki Kunoh 1,2,4,7, Tsutomu Shimura3,7, Tomonari Kasai1,2,5,7 ,
Syuji Matsumoto1,2,6, Hafizah Mahmud2, Apriliana Cahya Khayrani2,
Masaharu Seno1,2 , Hitoshi Kunoh1,2 and Jun Takada1,2
1
Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology
Agency (JST), 3-1-1 Tsushima-naka, Kita-ku, Okayama, 700-8530, Japan
2
Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-naka, Kita-
ku, Okayama, 700-8530, Japan
3
Department of Environmental Health, National Institute of Public Health, 2-3-6 Minami, Wako, Saitama,
351-0197, Japan

E-mail: kuno.tatsuki.gb@u.tsukuba.ac.jp, simura.t.aa@niph.go.jp and kasaitnr@stf.teu.ac.jp

Received 26 August 2018, revised 25 October 2018


Accepted for publication 2 November 2018
Published 30 November 2018

Abstract
The surface reactivity of gold nanoparticles (AuNPs) is receiving attention as a radiosensitizer of
cancer cells for radiation therapy and/or as a drug carrier to target cells. This study demonstrates
the potential of DNA-AuNPs (prepared by mixing calf thymus DNA with HAuCl4 solution) as a
radiosensitizer of human glioma cells that have cancer stem cell (CSC)-like properties, to reduce
their survival. CSC-like U251MG-P1 cells and their parental glioblastoma U251MG cells are
treated with a prepared DNA-AuNP colloid. The radiosensitivity of the resultant AuNP-
associated cells are significantly enhanced. To reveal the mechanism by which survival is
reduced, the generation of reactive oxygen species (ROS), apoptosis induction, or DNA damage
in the cells is assayed using the fluorescent dye DCFDA, annexin V-FITC/PI, and foci formation
of γ-H2AX, respectively. X-ray irradiation with administration of AuNPs overcomes the
radioresistance of U251MG-P1 cells. It does not induce ROS generation or apoptosis in the cells
but enhances the number of abnormal nuclei with abundant γ-H2AX foci, which is judged as cell
death by mitotic catastrophe. The AuNP association with the cells effectively induces mitotic
catastrophe in x-ray-irradiated CSC-like cells, implicating that DNA-AuNPs might be a
promising tool to develop an efficient radiosensitizer against CSC.
Keywords: DNA-generated AuNPs, x-ray-irradiation, radiosensitizer, glioma stem cells, mitotic
catastrophe

(Some figures may appear in colour only in the online journal)

1. Introduction
4
Present address: Faculty of Life and Environmental Sciences, University of Nanoparticles are at the leading edge of nanotechnology, defined
Tsukuba, Tsukuba, Ibaraki, 305-8572, Japan.
5
Present address: School of Bioscience and Biotechnology, Tokyo
as the study of structures in the range of 1–100 nm in size [1, 2].
University of Technology, 1404-1 Katakuramachi, Hachioji, Tokyo 192- Due to their distinctive physicochemical and electrical proper-
0982, Japan. ties, nanoparticles attract the attention of scientists and engineers
6
Present address: Nanotechnology Hub, Kyoto University, Yoshida-
Honmachi, Sakyo-ku, Kyoto 606-8501, Japan.
in biomedical, optical, catalytic, and electronic fields [1, 2]. To
7 date, a number of metal nanoparticles composed of gold, silver,
Authors to whom any correspondence should be addressed.

0957-4484/19/055101+11$33.00 1 © 2018 IOP Publishing Ltd Printed in the UK


Nanotechnology 30 (2019) 055101 T Kunoh et al

copper, and platinum have been synthesized by physical and [36]. Since their ability to reconstitute tumors is closely
chemical methods [3–7]. However, these conventional methods associated with the treatment resistance and tumor recurrence,
often have problems such as being costly, environmentally energetic studies to focus CSCs would open effective ther-
hazardous, or otherwise toxic [8]. Thus, biological production apeutic approaches to treat and suppress various types of
based on the principles of green chemistry have been explored. cancer [34–36]. Therefore, in this study we primarily focused
For example, extracts from organisms (brown algae, lemongrass, to study whether use of DNA-generated AuNPs [19] con-
tea, and fruits), human and microbial living cells, and biomo- tributes to abrogate radioresistance, one of CSCs properties.
lecules such as proteins and sugars have been used to create gold Herein, we first examined the cytotoxicity of DNA-
nanoparticles (hereafter AuNPs) [9–18]. Our previous study generated AuNPs in human U251MG-P1 cells (the derivative
demonstrated that incubation of DNA in HAuCl4 solution suc- of its parental U251MG cells) that have CSC properties.
cessfully led to form spherical AuNPs with a size of approxi- Subsequently, the radiation sensitivity of the cells and their
mately 5 nm in diameter (hereafter DNA-generated AuNPs) parental cancer cells, U251MG cells, associated or unasso-
through oxidation of guanine moiety of DNA, implicating the ciated with AuNPs, was compared to verify whether the
production of Au-DNA complex [19]. AuNPs association conferred any potential for radio-
Several lines of evidence revealed that size, shape, and sensitization to the cells. Finally, we explored ways to
surface functionalization of AuNPs are key factors regarding influence the survival rates of these cells.
their internalization by human cells [18]. DNA-AuNPs have
the ability to enter a wide variety of cell types [18]. Inter-
estingly, large DNA fragments are more readily incorporated 2. Results
into cancer cells such as liver, lung, and breast cancer cells
than into normal cells [20]. Moreover, AuNPs delivered to 2.1. Microscopic analyses of colloidal DNA-AuNPs and
cancer cells successfully reduced x-ray irradiation dose assessment of toxicity to human cells
required for DNA damage dependent cell death due to effi-
When calf thymus DNA was incubated in HAuCl4 solution,
cient generation of photoelectrons and Auger electrons upon
size-controlled nanoparticles were formed in this mixture
x-ray irradiation of AuNPs [21]. For such medical applica-
(hereafter referred to as colloid), as reported previously [19].
tions, generally AuNPs are functionalized with multiple tar-
The average diameter of the basic AuNPs prepared by the
geting molecules such as ligands, antibodies, and aptamers
present condition was ca. 4.5 nm on average [19]. The larger
[18, 22, 23]. Once these modified AuNPs are delivered to
particles, aggregations of the basic AuNPs, existed on and
target cells and tissues, they can readily act as an effective
near the surface of DNA molecules (figure 1(a), left). More-
radiation sensitizer upon x-ray irradiation [24–26]. In aqueous
over, apart from the aggregated DNA, a number of similar
conditions, x-ray irradiation of AuNPs causes water ioniz-
aggregates were scattered on the carbon grid (figure 1(a),
ation to result in formation of reactive oxygen species (ROS),
right). As discussed below, solitary-looking AuNPs apart
hydroxyl radicals (·OH) and superoxide anions (O2-) [27]. In
from the aggregated DNA in the TEM images are supposed to
mammalian cells into which AuNPs have been incorporated, associate with non-aggregated DNA molecules. However,
ROS is also generated intracellularly upon x-ray irradiation even in highly magnified TEM images, it was difficult to
[25, 28]. Radiation-induced ROS generation oxidatively determine how these AuNPs were associated with aggregated
damages cellular components such as DNA, RNA, proteins, or non-aggregated DNA molecules. We therefore lump all
and lipids, which may trigger cell death [29, 30]. Indeed, the these particles as ‘DNA-AuNPs’ hereafter.
extent of DNA damage from x-ray irradiation influences the The particle density of the DNA-AuNPs in the colloid
eventual cell fate, e.g. cell cycle arrest, DNA repair, and/or was estimated using liquid TEM. Again, the aggregates of
apoptotic pathway activation [31]. These earlier findings led DNA-AuNPs (30–40 nm in diameter) were dispersed nearly
us to fascinate a possibility of using DNA-generated AuNPs homogeneously in the colloid (figure 1(b)). On average, 533
for medical applications such as cancer radiotherapy without particles were distinguished in 2.5×2.5×0.2 μm3 of the
further surface modifications. However, we do not yet know colloid image; thus, the density of DNA-AuNPs in the pre-
for certain whether these AuNPs are toxic to living organisms pared colloid was estimated as ca. 8.8×1011 μl−1.
or are safe for medical applications on human cells. We next examined whether the colloid was toxic to
Increasing evidence shows that many types of tumors U251MG-P1 cells. The colloid was serially diluted with the
contain a subpopulation of cells called cancer stem cells culture medium, added to well cultures of U251MG-P1 cells,
(hereafter CSCs) [32–34]. Similar to normal pluripotent stem and incubated for 24 h. Subsequently, the number of cells in
cells, CSCs have characteristic properties such as self- each well was determined using the MTT assay. The relative
renewal, multi-differentiation, resistance to radiotherapy, and cell viability against that of the colloid-untreated control
cytotoxic chemotherapy due to their dormant state, pre- culture was slightly higher than 100% when the particle
ferential activation of the DNA damage response, efficient density was lower („1.5×1010 particles μl−1), but was only
DNA repair machinery, and resistance to apoptosis [34, 35]. ca. 60% when particle density was higher (…2.9×1011
In addition, CSCs show ROS resistance by low intracellular particles μl−1) (figure 1(c)); thus, the low density treatment
ROS concentrations due to up-regulation of ROS scavengers with DNA-AuNPs was not toxic to the U251MG-P1 cells.

2
Nanotechnology 30 (2019) 055101 T Kunoh et al

Figure 1. Microscopic analyses of colloidal DNA-AuNPs and verification of their toxicity to human cells. (a) TEM images of air-dried DNA-
AuNP colloid on aggregated DNA (left) and carbon grid frame (right), respectively. (b) Liquid TEM image of DNA-AuNP colloid, showing
nearly homogeneous distribution of electron-dense nanoparticles. (c) Relative viability of U251MG-P1 cells after incubation with serially
diluted DNA-AuNP colloids against that of PBS-treated control cells (MTT assay). (d) DIC images of U251MG-P1 cells treated with PBS
(left) and with DNA-AuNP colloid (right) after the silver enhancement stain for AuNPs. Note that dark brown particles (corresponding to
AuNPs) were associated with the latter cells only. Some particles were incorporated intracellularly, judging from the focal plane.

3
Nanotechnology 30 (2019) 055101 T Kunoh et al

Figure 2. Sensitivity of DNA-AuNP colloid-treated and -untreated U251MG and U251MG-P1 cells to x-ray irradiation. Error
bars=standard deviations. (a) Survival of U251MG (circle) and U251MG-P1 (square) cells treated with PBS after x-ray irradiation.
Survival of U251MG cells was notably lower than that of U251MG-P1 cells at 1–5 Gy. (b) Survival of U251MG (circle) and U251MG-P1
(square) cells treated with DNA-AuNP colloid after x-ray irradiation. Survival of U251MG cells was comparable to that of U251MG-P1 cells
at 1–5 Gy. Note that x-ray resistance, the CSC-like property, of U251MG-P1 cells was higher than that of their parental U251MG cells (a).
Importantly, this property was abrogated when the cells were associated with DNA-AuNPs (b). (c) DIC images of U251MG-P1 cells treated
with PBS (left) and with DNA-AuNP colloid (right two) after silver enhancement stain of AuNPs. Note that localization of AuNPs was not
notably affected by x-ray irradiation.

To examine how DNA-AuNPs behave when the colloid rates of U251MG and U251MG-P1 cells were compared after
encounters with U251MG-P1 cells, we incubated the cells treatment with x-rays followed by PBS relative to those of the
with the colloid (1.5×1010 DNA-AuNPs μl−1) or with PBS non-irradiated control cells. The survival rate of U251MG-P1
as the control. DNA-AuNPs in these mixtures were sensitized cells was higher than that of U251MG cells (figure 2(a)) at all
by the silver enhancement method (figure 1(d)). Dark brown doses, showing that the U251MG-P1 cells were apparently
particles were observed in all of the cells incubated with the more resistant to x-ray irradiation, probably due to their CSC-
colloid (figure 1(d), right), but not in the control cells like properties.
(figure 1(d), left). Judging from the focal plane in which these We next examined whether the association of these cells
particles were observed, some of the particles were likely to with AuNPs influenced their survival rate after x-ray irra-
be inside the cells, while others were on the cell surface diation. At first, we confirmed visually that x-ray irradiation
(figure 1(d)). Thus, from the optical images, we could not did not at least localize AuNPs (figure 2(c)). The colony assay
definitely conclude that all of the DNA-AuNPs had been after x-ray irradiation showed no difference between the
incorporated into the cells. Therefore, to avoid confusion, we survival rate of both AuNPs-associated U251MG and
hereafter call these cells as ‘AuNP-associated’ cells. U251MG-P1 cells at all radiation doses (1–5 Gy)
(figure 2(b)). Thus, the radioresistant phenotype of U251MG-
P1 cells disappeared by administration with AuNPs
2.2. Sensitivity of AuNPs-associated U251MG-P1 cells to x-ray
(figures 2(a), (b)). In addition, the survival rate of AuNPs-
irradiation
associated U251MG-P1 cells (figure 2(b)) was significantly
As described above, U251MG-P1 cells originated from a (P<0.05) lower than even that of the PBS-treated U251MG
primary culture from the tumor of a mouse that had been cells (figure 2(a)). From these results, we concluded that
injected with a spheroid-forming population of human brain association with AuNPs efficiently abrogated the x-ray
glioblastoma U251MG cells. In a colony assay, the survival radiation resistance related to the CSC-like properties,

4
Nanotechnology 30 (2019) 055101 T Kunoh et al

activating apoptotic pathways. To determine whether apop-


tosis induction was enhanced in AuNP-associated U251MG-
P1 cells after x-ray irradiation, we used FACS analysis after
staining with annexin V-FITC and propidium iodide (PI) [37].
In the analytical diagram in figure 4(a), zones B1, B2, B3, and
B4 correspond to populations of necrotic cells, apoptotic cells
at the late stage, vital cells, and apoptotic cells at the early
stage, respectively. When the percentage of apoptotic cells in
zones B2 and B4 was compared against the total analyzed
cells in all zones (figure 4(b)), regardless of the irradiation
doses, an average of 3%–5% of the PBS-treated control cells
were apoptotic, whereas 7%–8% the AuNPs-associated cells
were apoptotic on average (figure 4(b)). However, the sta-
tistical analysis revealed that the percentage of apoptotic cells
between these two cell groups did not differ significantly at
any dose (P>0.05). Thus, these results led us to conclude
Figure 3. Influence of the AuNP association and x-ray irradiation on
ROS levels as reflected by relative fluorescence in DNA-AuNP- or that association with DNA-AuNPs did not enhance the x-ray
PBS-treated U251MG and U251MG-P1 cells at 24 h after x-ray irradiation-dependent apoptotic cell death in U251MG-P1
irradiation (FACS analysis of DCFDA-stained cells). In PBS-treated cells.
U251MG cells, ROS generation was enhanced by irradiation, but
none was generated in PBS-treated U251MG-P1 cells. ROS
generation in AuNP-associated U251MG-P1 cells was lower than in 2.5. Formation of abnormal nuclei representing mitotic
PBS-treated cells. ROS generation in both types of AuNP-associated catastrophe in AuNP-associated cells after x-ray irradiation
cells was reduced by 5 Gy irradiation. Error bars=standard
deviations. Double asterisks=significant differences at P<0.01. Since x-ray irradiation is known to cause DNA damage [31],
we used γ-H2AX staining to monitor DNA double-strand
breaks (hereafter DSBs) to determine whether AuNPs affec-
probably due to cell death such as apoptosis induction and
ted DNA repair in U251MG and U251MG-P1 cells [38].
mitotic catastrophe.
Faint γ-H2AX staining was observed in non-irradiated PBS-
treated U251MG and U251MG-P1 cells (figures 5(a), (c),
2.3. Influence of the AuNP association and x-ray irradiation on upper left, respectively). However, in both cells, intense
ROS levels in U251MG and U251MG-P1 cells fluorescence representing the remaining DSBs was detected
The dose-dependent reduction of survival rates of U251MG and 24 h after 5 Gy irradiation (figures 5(a), (c), lower left,
U251MG-P1 after x-ray irradiation (figure 2) suggested that the respectively). Similar florescence detection was obtained in
irradiation caused adverse effects that were enhanced by the both x-ray-irradiated and non-irradiated AuNPs-associated
association of these cells with AuNPs. In PBS-treated U251MG U251MG and U251MG-P1 cells (figures 5(b), (d), respec-
cells, the relative fluorescence (=ROS generation) was sig- tively). Here, cells with intense fluorescence, representing
nificantly (P<0.01) enhanced by x-ray irradiation at 5 Gy, serious DNA damage, were regarded as γ-H2AX-positive,
while ROS generation was unaffected in PBS-treated U251MG- and counted among U251MG and U251MG-P1 cells that
P1 cells even after 5 Gy irradiation (figure 3). In addition, ROS were PBS-treated or AuNPs-associated before x-ray irradia-
generation in AuNPs-associated U251MG-P1 cells was sig- tion. As shown in figure 5(e), the percentage of positive cells
nificantly (P<0.01) lower than that in the control PBS-treated increased to ca. 60% in PBS-treated and AuNPs-associated
cells (figure 3). Furthermore, ROS generation after 5 Gy irra- U251MG after x-ray irradiation. Overall, the incidence of the
diation was significantly (P<0.01) lower in AuNP-associated positive cells was significantly lower (P<0.01) in U251MG-
than PBS-treated U251MG cells. These results suggest that the P1 cells than in U251MG cells. An earlier report [39] that
ROS generation in both groups of cells could be suppressed only CSC cells can efficiently self-repair DNA damage caused by
when the cells are associated with the AuNPs. The additional x-ray irradiation and/or AuNPs association may account for
result that ROS generation in the AuNPs-associated U251MG such a difference. However, the percentage was ca. 40% after
and U251MG-P1 cells was significantly (P<0.01) reduced after irradiation in PBS-treated U251MG-P1 cells, but increased to
irradiation at 5 Gy relative to that in both PBS-treated cells ca. 60% in AuNPs-associated U251MG-P1 cells after irra-
(figure 3) led us to conclude that ROS generation was unable to diation (figure 5(e)). Thus, AuNPs are effective to abrogate
account for the reduced survival of AuNP-associated U251MG- efficient repair of x-ray irradiation-induced DSBs in CSC-like
P1 cells after irradiation. U251MG-P1 cells.
The right columns in figures 5(a)–(d) show nuclei stained
2.4. Influence of the AuNP association and x-ray irradiation on
with Hoechst. Note that small fragmented DNAs (=micro-
nuclei) with γ-H2AX staining were detected in both AuNP-
apoptosis induction in U251MG and U251MG-P1 cells
associated U251MG and U251MG-P1 cells after x-ray irra-
As described above [29–31], x-ray irradiation adversely diation (figures 5(b), (d), lower right, respectively, arrows).
affects human cells by arresting the cell cycle and/or Although multinucleated cells were evident among both

5
Nanotechnology 30 (2019) 055101 T Kunoh et al

Figure 4. Incidence of apoptotic cells among AuNP-associated and PBS-treated U251MG-P1 cells after x-ray irradiation (FACS analysis).
(a) Distribution of PBS-treated cells (upper two) and AuNP-associated cells (lower two) in the respective zones after x-ray irradiation (B1,
necrotic cells; B2, late-stage apoptotic cells; B3, viable cells; B4 zones, early-stage apoptotic cells). (b) Incidence of apoptotic cells (total
number of cells in zones B2 and B4) in the PBS-treated and AuNP-associated cells after x-ray irradiation. Error bars=standard deviations.
Overlapping of all standard error bars suggests no difference among incidences (P>0.05).

AuNP-associated U251MG and U251MG-P1 cells after x-ray We subsequently examined the radiation sensitivity of
irradiation (figure 5(f)), the number of multinucleated cells in the U251MG-P1 and U251MG cells, associated or unasso-
U251MG-P1 cells differed significantly (P<0.01) from the ciated with AuNPs, and the potential of the AuNP association
control. to enable radiosensitization to the cells. The U251MG-P1
cells were known to have CSC-like properties such as higher
expression of CD44 (a marker for CSCs) and aberrant acti-
vation of principal pluripotency genes (OCT3/4, SOX2,
3. Discussion
KLF4, and Nanog) in comparison with those of the parent
The TEM image of the DNA-AuNPs colloid showed aggre- U251MG cells [41, 42]. As expected, the U251MG-P1 cells
gates of multiple electron-dense nanoparticles (=AuNPs with were thus apparently more resistant to x-ray irradiation
∼5 nm diameter) on or near the surface of aggregated DNA (figure 2(a)), being consistent with other CSCs [34, 35].
(figure 1(a), left). In addition, some AuNPs seemed to exist Based on these results, we subsequently examined the
solitarily apart from aggregated DNA without coagulation radiation sensitivity of the AuNP-associated or -unassociated
(figure 1(a), right). Therefore, for future investigation of U251MG-P1 and U251MG cells and found that the survival
practical therapeutic techniques, it is important to elucidate rate of both AuNP-associated U251MG and U251MG-P1
whether and/or how the association of DNA molecules with cells was close enough at all radiation doses (1–5 Gy)
the surface of these particles contribute to enhance radio- (figure 2(b)). Thus, the association of U251MG-P1 cells with
sensitization activity for CSC cells. AuNPs efficiently abrogated the radiation x-ray resistance
Notably, the treatment with a relatively low density of related to CSC-like properties. We infer that such effects
AuNPs in the colloid („1.5×1010 particles μl−1) was not could be related to AuNP-mediated ROS generation in the
toxic to target human cells (figure 1(c)), although we do not x-ray-irradiated cells [43]. In addition, radiation-induced ROS
know why the higher density of DNA-AuNPs adversely generation damages cellular components, which may trigger
affected the cells. Results were comparable to those of human cell death [29, 30]. Contrary to what we expected, the
cells after treatment with AuNPs that had been surface-coated experimental results (figure 3) did not show that ROS gen-
with glucose or cetyltrimethylammonium bromide (CTAB) eration accounted for the reduced survival of AuNP-asso-
[36, 40]. As demonstrated in figure 1(d), at least some of the ciated U251MG-P1 cells after irradiation. Notably, ROS
particles were likely to be inside the cells, while others might generation was apparently lower in U251MG-P1 than
have localized on the cell surface. U251MG cells, regardless of the AuNP association and

6
Nanotechnology 30 (2019) 055101 T Kunoh et al

Figure 5. Incidence of DNA double-strand breaks (DSBs) and abnormal nuclei caused by x-ray irradiation as detected by foci formation of
γ-H2AX and Hoechist staining, respectively. (a) Weak (left upper) and intense (left lower) fluorescence representing DSBs in PBS-treated
U251MG cells before (0 Gy) and after (5 Gy) irradiation, respectively. Images in the right column show normal nuclei before and after x-ray
irradiation. (b) DSBs (left) and nuclei (right) in AuNP-treated U251MG cells before and after x-ray irradiation. Note the micronuclei that
formed after irradiation (right lower, arrows). (c), (d) DSBs (left) and nuclei (right) in PBS-treated and AuNP-associated U251MG-P1 cells,
respectively, before and after irradiation. Note that γ-H2AX-positive micronuclei were formed after irradiation (right lower, arrows).
(e) Percentage of intensely fluorescing cells (γ-H2AX-positive cells) among PBS-treated and AuNP-associated U251MG and U251MG-P1
cells before and after irradiation. (f) Incidence of abnormal nuclei accompanying micronuclei in PBS-treated and AuNP-associated U251MG
and U251MG-P1 cells before and after irradiation. Note that the percentage significantly (P<0.05) increased in PBS-treated U251MG-P1
cells after irradiation and especially (P<0.01) in AuNP-associated U251MG and U251MG-P1 cells. Error bars=standard deviations.
Double and single asterisks=significant differences at P<0.01 and <0.05, respectively.

7
Nanotechnology 30 (2019) 055101 T Kunoh et al

-unassociation of the cells (figure 3) as supported by an earlier AuNPs-associated CSC cells (U251MG-P1 cells), the exper-
paper [44]. Thus, the activity of free radical scavenging sys- imental outcomes lead us to expect that DNA-generated AuNPs
tems might be increased in the PBS-treated U251MG-P1 are a promising tool for radiotherapy treatment of cancerous cells
cells. Resistance to ROS was also reported in other CSCs due and tissues.
to up-regulation of ROS scavengers [35]. Interestingly, the
incidence of intense γ-H2AX-positive fluorescence, which
represents serious DNA damage (= DSBs), was lower in 4. Conclusion
U251MG-P1 cells than in U251MG cells. CSC cells can
efficiently self-repair DNA damage caused by x-ray irradia- In this study, we initially found that the association with
tion and/or association with AuNPs [39], which may account DNA-AuNPs (which were incorporated into and/or attached
for the weaker fluorescence in U251MG-P1 cells. to cells) were not toxic to radioresistant glioma stem cells
Because the extent of DNA damage from x-ray irradia- with CSCs-properties. Then we found that the pretreatment of
tion influenced cell cycle arrest, DNA repair, and/or apop- the cells with such AuNPs abrogated radioresistance of the
totic pathway activation [31], we next examined whether CSC-like cells. The subsequent cell cycle analyses and
x-ray irradiation-induced apoptosis in U251MG-P1 cells. The fluorescence microscopy revealed that such a suppression of
FACS analysis, which distinguished dead (necrotic or apop- the radioresistance of CSC-like cells was due to an increase in
totic) and vital AuNPs-associated and PBS-treated control the number of abnormal nuclei with abundant γ-H2AX foci (a
U251MG-P1 cells after x-ray-irradiation failed to show any marker of DNA double-strand breaks), which was judged to
significant differences in the incidence of apoptosis at any be cell death by mitotic catastrophe. Results led us to consider
dose (figure 4). Thus, association with DNA-AuNPs did not that DNA-generated AuNPs could be a promising material to
enhance x-ray irradiation-dependent apoptotic cell death in enhance radiosensitization of CSCs-like glioma stem cells.
U251MG-P1 cells. We would like to emphasize novel merits to using the
Finally, we found that the incidence of abnormal cells present DNA-generated AuNPs: (i) preparation of the DNA-
with fragmented DNA (=micronuclei) increased in both AuNPs colloid is quite simple (just incubation of DNA in
AuNPs-associated U251MG and U251MG-P1 cells after HAuCl4 solution at room temperature for 16 h, followed by
x-ray irradiation (figure 5). This phenomenon was more neutralization in PBS buffer) [19], (ii) spherical DNA-gen-
apparent in U251MG-P1 cells. The abnormal nuclei closely erated AuNPs (∼5 nm in diameter) are readily associated with
resemble those in dead cells due to mitotic catastrophe cancer cells inculuding CSC-like glioma stem cells without
[39, 45, 46]. DSBs detected by γ-H2AX staining were evident using any transfection agent and interact with their chromo-
in cells undergoing mitotic catastrophe. This may reflect that some DNA, as expected from an earlier report [20]. The
remaining DSBs cause mitotic failure by premature or inap- development of the novel, practical, efficient technique to
propriate entry of cells into mitosis due to defective cell cycle abrogate radioresistance of CSC-like cells are still under
checkpoints [45]. On the basis of these results, we consider intensive investigation based on the present outcome.
that the CSC-like properties of U251MG-P1 cells were
abrogated by association with AuNPs through mitotic cata-
strophe rather than ROS generation and apoptosis induction, 5. Experimental procedure
thus reducing survival after x-ray irradiation. The intracellular
location of AuNPs is known to affect radiation enhancement. 5.1. Cell culture
Particularly, AuNPs attached to DNA have a greater impact
U251MG-P1 cells were obtained as a primary culture from a
than an AuNP in other locations [47]. Therefore, the defor- tumor-bearing mouse that had been injected with a spheroid-
mation of nuclei due to the AuNPs association (figure 5) most forming population of human glioblastoma U251MG cells
likely reflects that DNA-AuNPs which were incorporated into [41]. These cells were cultured in DMEM high glucose
the target cells directly or indirectly interacted with chromo- medium (Sigma-Aldrich, St. Louis, MO, USA) supplemented
some DNA, although further detailed studies are required for with 10% fetal bovine serum (FBS) (Thermo Fisher Scien-
a definitive conclusion. tific, Waltham, MA, USA) and 100 U ml–1 penicillin/strep-
As mentioned above, CSC cells are known to be strongly tomycin (P/S) (Wako Pure Chemical Industries, Osaka,
resistant to x-ray irradiation, which often poses a problem for Japan) at 37 °C in a 5% CO2 incubator.
cancer therapy [34]. However, the present findings demonstrated
that the association of these cells with DNA-generated AuNPs
5.2. Preparation of DNA-AuNP colloid using calf thymus DNA
reduced viability of U251MG-P1 cells after x-ray irradiation,
plausibly through DSBs, which are critical lesions that can result As described previously [19], 0.5 ml of 100 mM Au(III)
in cell death or a wide variety of genetic alterations including chloride solution (pH 2.0), which had been prepared by dis-
large- or small-scale deletions, heterozygosity loss, translocations, solving HAuCl4 (Sigma-Aldrich) in ultrapure water (UPW),
and chromosome loss [48]. DSBs often lead to mitotic cata- was added to 3.5 ml of calf thymus DNA (Sigma-Aldrich)
strophe [45, 46]. Although the present study failed to explain the solution (final concentrations of 2.5–7.5 mg ml−1) in 15 ml
mechanism underlying this mitotic catastrophe in x-ray-irradiated conical tubes and incubated on a shaking mixer (SHM-2002,

8
Nanotechnology 30 (2019) 055101 T Kunoh et al

LMS, Tokyo, Japan) at room temperature for 16 h. The (MTT) (Sigma-Aldrich). The cells that had been incubated
resultant DNA-AuNP colloid was neutralized by adding with the DNA-AuNP colloid or the control PBS were treated
1.0 ml of 20-fold concentrated phosphate-buffered saline with 5 mg ml−1 MTT in PBS (final concentration of
(PBS, pH 7.4) to adjust pH to ca. 7.0 (hereafter referred to as 1 mg ml−1) and incubated at 37 °C for 5 h. Formazan crystals
DNA-AuNP colloid). formed in these mixtures during the incubation were dis-
solved in 10% SDS containing 20 mM HCl at 37 °C over-
5.3. Transmission electron microscopy night. Formazan formation, which depends on the metabolic
activity of vital cells, is frequently used as an indicator of cell
To confirm the characteristics of the DNA-AuNP colloid, we viability [49]. Thus, using a spectrophotometer, we compared
prepared and viewed the DNA-AuNP colloids with electron the optical density at 570 nm (peak absorbance for formazan)
microscopy as follows. For TEM observation, the DNA- of the suspension of colloid-treated cells with that of the PBS-
AuNP colloid was diluted 200–1000-fold with 99.5% ethanol. treated control cells. The toxicity was evaluated by the rela-
The diluted specimens were dropped onto carbon grids tive cell viability.
(Nisshin-EM, Tokyo, Japan), air-dried, then viewed with a
TEM (JEOL 2100 F, Tokyo, Japan). To determine the particle 5.7. Colony assay
density in the DNA-AuNPs colloid, we used a Si-based
microchannel sample holder for liquid TEM called a K-kit Effects of x-ray irradiation on the colony-forming ability of
(Bio MA-Tek, Hsinchu, Taiwan) according to the manu- test cells were examined by seeding 1 cm dishes (Thermo
facturer’s protocol. The K-kit is a sealable carrier with a Fisher Scientific) with 1×103 cells of U251MG or
liquid microchannel (window: thickness=100 nm, width= U251MG-P1, incubating the dishes at 37 °C in a 5% CO2
24 μm; length=300 μm). Briefly, the DNA-AuNP colloid incubator for 16 h, then irradiating the cells with x-rays as
was diluted 20 fold and placed in the K-kit to impregnate gaps described above. After an additional 16 h incubation, the cells
(gap size 0.2 μm) in the K-kit. Then the kit was tightly sealed, were subcultured (cell density: 1×103 cells per new dish)
set on the TEM specimen holder, and inserted into the TEM and incubated for a week, followed by fixation with 99%
column, then observed according to routine methods. The ethanol and then staining with Giemsa stain solution (Milli-
electron-dense particles in 10 squares (2.5 μm×2.5 μm) in pore, Billerica, MA, USA). After air-drying, colonies con-
TEM images were counted. Theoretically, almost all particles sisting of more than 50 cells were counted. The survival rate
in the 0.2 μm gap should appear in these squares; thus, the was evaluated as the relative colony number compared with
density of AuNPs per microliter of the colloid was estimated that of the non-irradiated control cells.
on the basis of the counted number.
5.8. Sensitization of AuNPs associated with human cells
5.4. Preparation of DNA-AuNP-associated human cells
To ascertain whether AuNPs were associated with human
The U251MG-P1 cells were seeded at 5×10 cells in 100 μl
3 cells, AuNPs-associated U251MG-P1 cells were sensitized
of the above culture medium and incubated for 24 h. Subse- using a silver enhancement method for optical microscopic
quently, 50 μl of the DNA-AuNP colloid, which was serially observation. After 24 h incubation with the DNA-AuNP
diluted with the culture medium (9.1×109–5.8×1011 par- colloid, the U251MG-P1 cells were trypsinized and fixed with
ticles μl−1) was added to the above cell cultures and incu- 2.5% glutaraldehyde at 4 °C overnight. After washed once in
bated for an additional 24 h. UPW, the cells were adhered to SurePath Precoat slides
(Becton, Dickinson and Company, Franklin Lakes, NJ, USA),
5.5. X-ray irradiation then washed three times with 99.5% ethanol. They were then
incubated with a Silver Enhancement Kit (BBI Solutions,
To determine the x-ray resistance of the AuNP-associated Cardiff, UK) for approximately 10 min at room temperature
cells, the U251MG and U251MG-P1 cells, which had been until the sensitized AuNPs were seen as dark brown spots
incubated with the DNA-AuNP colloid or the control PBS for using an optical microscope. Subsequently, the cells were
>24 h were irradiated with x-rays using a 150 kVp x-ray washed by UPW to stop the enhancing reaction, stained with
generator (model MBR-1505R2, Hitachi) through a 0.5 mm Kernechtrot solution (Sigma-Aldrich) to visualize nuclei, and
Cu and 0.1 mm Al filters at a dose of 0.7 Gy min−1. For observed with differential interference contrast (DIC) optics
reference, the routinely cultured cells that were not incubated and a light microscope using an objective lens (x20, NA0.40)
with the DNA-AuNP colloid were also irradiated the same (BX51 System Microscope, Olympus, Tokyo, Japan).
way. These cells were then subjected to the following colony
assay. 5.9. FACS analysis

Apoptotic and necrotic cells generated by x-ray irradiation


5.6. MTT assay
were identified and quantified using an Annexin V-FITC
To monitor toxicity of the DNA-AuNP colloid to human Apoptosis Detection Kit (Bio Vision, Mountain View, CA,
cells, we determined the viability of U251MG-P1 cells USA) and the manufacturer’s protocol. The x-ray-irradiated
associated with DNA-AuNPs using a cleavage assay with AuNPs-associated and PBS-treated control U251MG-P1 cells
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide were trypsinized, collected, and stained with the annexin

9
Nanotechnology 30 (2019) 055101 T Kunoh et al

V-FITC and PI. The annexin V-positive apoptotic cells and Research (17K19944) (TS) and for Scientific Research (C)
PI-positive necrotic cells were counted with a fluorescence- (16K07116) (T Kasai) by JSPS. This work was also sup-
activated cell sorting system (FACScan, Becton Dickinson, ported by the Program of the Network-type Joint Usage/
Mountainview, CA, USA). Research Center for Radiation Disaster Medical Science.
Furthermore, to examine the influence of the x-ray irra-
diation on ROS generation, the AuNPs-associated or PBS-
treated U251MG and U251MG-P1 cells were incubated at
ORCID iDs
37 °C in a 5% CO2 incubator for 24 h after the x-ray irra-
diation, then stained with 20 μM DCFDA (Sigma-Aldrich) in
Tatsuki Kunoh https://orcid.org/0000-0002-8423-2903
a minimum essential medium without serum for 30 min, then
Tomonari Kasai https://orcid.org/0000-0002-9346-6664
trypsinized, and collected as described earlier. ROS genera-
Masaharu Seno https://orcid.org/0000-0001-8547-6259
tion was quantified using the FACScan as described pre-
viously [49]. The intensity of the fluorescence due to ROS
generation in all cells was measured three times and expres-
sed as a mean with standard deviation relative to that of non- References
irradiated PBS-treated cells.
[1] Salata O V 2004 J. Nanobiotechnol. 2 3
5.10. Immunofluorescent staining [2] Murray C B, Kagan C R and Bawendi M G 2000 Annu. Rev.
Mater. Sci. 30 545
For detecting foci formation of γ-H2AX (representing sites of [3] Sengani M, Grumezescu A M and Rajeswari V D 2017
OpenNano 2 37
DNA double-strand breaks) after x-ray irradiation, U251MG
[4] Lu Q, Rosen J, Zhou Y, Hutchings G S, Kimmel Y C,
and U251MG-P1 cells cultured on cover glasses were fixed in Chen J and Gand Jiao F 2014 Nat Commun. 5 3242
4% v v–1 paraformaldehye for 10 min at room temperature, [5] Natsuki J, Natsuki T and Hashimoto Y 2015 Int. J. Mater. Sci.
then permeabilized using 0.5% Triton-X in PBS for 10 min Appl. 4 325
Then, the cells were washed in PBS and incubated in a [6] Stepanov A L, Golubev A N, Nikitin S I and Osin Y N 2014
Rev. Adv. Mater. Sci. 38 160
blocking solution (PBS supplemented with 5% BSA (Frac-
[7] Santra T S, Tseng F and Barik T K 2014 Am. J. Nano Res.
tion-V)) for 15 min. The cells were treated with a primary Appl. 2 5
antibody against γ-H2AX (1:500 dilution, Santa Cruz Bio- [8] Shankar S S, Rai A, Ankamwar B, Singh A, Ahmad A and
technology, Dallas, TX, USA) and then with a secondary Sastry M 2004 Nat. Mater. 3 482
antibody conjugated with Cy-3 (1:500 dilution, Jackson [9] Nune S K, Chanda N, Shukla R, Katti K, Kulkarni R R,
Thilakavathi S, Mekapothula S, Kannan R and Katti K V
ImmunoResearch Laboratories, West Grove, PA, USA). After
2009 J. Mater. Chem. 19 2912
the cells were counterstained with Hoechst 33258 (4 μg ml−1, [10] Liu B, Xie J, Lee J Y, Ting Y P and Chen J P 2005 J. Phys.
Vector Laboratories, Burlingame, CA USA) (hereafter, Chem. B 109 15256
Hoechst) for DNA, images were acquired using a light [11] Sujitha M V and Kannan S 2013 Spectrochim. Acta A 102 15
microscope equipped with a fluorescence lamp (Model BZ- [12] Anshup A, Venkataraman J S, Subramaniam C, Kumar R R,
Priya S, Kumar T R, Omkumar R V, John A and Pradeep T
X700, Keyence, Osaka, Japan).
2005 Langmuir 21 11562
[13] Johnston C W, Wyatt M A, Li X, Ibrahim A, Shuster J,
5.11. Statistical analyses Southam G and Magarvey N A 2013 Nat. Chem. Biol. 9 241
[14] Leng Y et al 2016 Sci. Rep. 6 28900
All experiments were done at least three times using inde- [15] Engelbrekt C, Sørensen K H, Zhang J, Welinder A C,
pendent samples. Variances among the values derived from Jensena P S and Ulstrupa J 2009 J. Mater. Chem. 19 7839
[16] Wei H, Li B, Du Y, Dong S and Wang E 2007 Chem. Mater.
each sample were analyzed with a one-way ANOVA using 19 2987
Excel. Dunnett’s test was then used to find significant dif- [17] Giljohann D A, Seferos D S, Daniel W L, Massich M D,
ferences among the means of values derived from three or Patel P C and Mirkin C A 2010 Angew. Chem. Int. Ed.
more independent samples. 49 3280
[18] Kunoh T, Takeda M, Matsumoto S, Suzuki I, Takano M,
Kunoh H and Takada J 2018 ACS Sustain. Chem. Eng.
6 364
Acknowledgments [19] Kong Y, Zhang X, Zhao Y, Xue Y and Zhang Y 2017 Biol.
Res. 50 2
We thank Ms Masumi Furutani, Ms Tomomi Tsukano, and [20] Zhang P, Qiao Y, Wang C, Ma L and Su M 2014 Nanoscale 6
10095
Mr Haruo Urata of the Central Research Laboratory at [21] Ghosh P, Han G, De M, Kim C K and Rotello V M 2008 Adv.
Okayama University Medical School for providing the Drug. Deliv. Rev. 60 1307
method for AuNPs sensitization in human cells. We [22] Dykman L A and Khlebtsov N G 2011 Acta Nat. 3 34
appreciate Dr Megumi Sasatani of Hiroshima University for [23] Liu C J et al 2008 Nanotechnology 19 295104
valuable comments. We also acknowledge Dr Beth E Hazen [24] Liu X, Liu Y, Zhang P, Jin X, Zheng X, Ye F, Chen W and
Li Q 2016 Int. J. Nanomed. 11 3517
for reviewing and editing the English in the manuscript. This [25] Kong T, Zeng J, Wang X, Yang X, Yang J, McQuarrie S,
study was financially supported by JST-CREST (2012–2017) McEwan A, Roa W, Chen J and Xing J Z 2008 Small 4 1537
(JT) by JST, and Grant-in-Aid for Challenging Exploratory [26] Misawa M and Takahashi J 2011 Nanomedicine 7 604

10
Nanotechnology 30 (2019) 055101 T Kunoh et al

[27] Retif P, Pinel S, Toussaint M, Frochot C, Chouikrat R, [40] Vaidyanath A, Mahmud H B, Khayrani A C, Oo A K K,
Bastogne T and Barberi-Heyob M 2015 Theranostics 5 1030 Seno A, Asakura M, Kasai T and Seno M 2017 J. Stem Cell
[28] Shimura T and Kunugita N 2016 Cell Cycle 15 1410 Res. Ther. 7 4
[29] Kehrer J P and Klotz L O 2015 Crit. Rev. Toxicol. 45 765 [41] Mahmud H et al 2018 Int. J. Mol. Sci. 19 659
[30] Nowsheen S and Yang E S 2012 Exp. Oncol. 34 243 [42] Klingberg H, Oddershede L B, Loeschner K, Larsen E H,
[31] Nair N et al 2015 Sci. Rep. 7 6838 Loft S and Møller P 2015 Toxicol. Res. 4 655
[32] Bao B, Ahmad A, Azmi A S, Ali S and Sarkar F H 2013 Curr. [43] Diehn M et al 2009 Nature 458 780
Protocols Pharmacol. 61 14.25.1–14 [44] Vakifahmetoglu H, Olsson M and Zhivotovsky B 2008 Cell
[33] Baek S J et al 2015 Oncol. Rep. 34 2233 Death Differ. 15 1153
[34] Shimura T, Noma N, Oikawa T, Ochiai Y, Kakuda S, [45] Castedo M, Perfettini J L, Roumier T, Andreau K,
Kuwahara Y, Takai Y, Takahashi A and Fukumoto M 2012 Medema R and Kroemer G 2004 Oncogene 23 2825
Oncogenesis 1 e12 [46] Shrestha S, Cooper L N, Andreev O A, Reshetnyak Y K and
[35] Baumann M, Krause M and Hill R 2009 Nat. Rev. Cancer 8 545 Antosh M P 2016 Jacobs J. Radiat. Oncol. 3 1
[36] Demchenko A P 2013 Cytotechnology 65 157 [47] Shrivastav M, De Haro L P and Nickoloff J A 2008 Cell Res.
[37] Mah L J, El-Osta A and Karagiannis T C Leukemia 24 679 18 134
[38] Zheng Y and Sanche L 2009 Radiat. Res. 172 114 [48] Mosmann T 1983 J. Immunol. Meth. 65 55
[39] Connor E E, Mwamuka J, Gole A, Murphy C J and Wyatt M D [49] Shimura T, Sasatani M, Kamiya K, Kawai H, Inaba Y and
2005 Small 3 325 Kunugita N 2016 Oncotarget 7 3559

11

Anda mungkin juga menyukai