Dual antiglioma action of metformin cell cycle arrest and mitochondria-dependent apoptosis.pdf

(1043 KB) Pobierz
C7080-4_OD07__2448
Cell. Mol. Life Sci. 64 (2007) 1290 – 1302
1420-682X/07/101290-13
DOI 10.1007/s00018-007-7080-4
Cellular and Molecular Life Sciences
Research Article
Dual antiglioma action of metformin: cell cycle arrest and
mitochondria-dependent apoptosis
A. Isakovic a , L. Harhaji b , D. Stevanovic c , Z. Markovic d , M. Sumarac-Dumanovic e , V. Starcevic c , D. Micic e, * and
V. Trajkovic f, *
a Institute of Biochemistry, School of Medicine, University of Belgrade, Belgrade (Serbia)
b Institute for Biological Research, Belgrade (Serbia)
c Institute of Physiology, School of Medicine, University of Belgrade, Belgrade (Serbia)
d Vinca Institute of Nuclear Sciences, Belgrade (Serbia)
e Institute of Endocrinology, Diabetes and Diseases of Metabolism, School of Medicine, University of Belgrade,
Dr Subotica 13, 11000 Belgrade (Serbia), Fax: +381 11 3065081, e-mail : micicd@eunet.yu
f Institute of Microbiology and Immunology, School of Medicine, University of Belgrade, Dr Subotica 1, 11000
Belgrade (Serbia), Fax: +381 11 265 7258, e-mail : vtrajkovic@eunet.yu
Received 14 February 2007; received after revision 26 March 2007; accepted 3 April 2007
Online First 20 April 2007
Abstract. The present study reports for the first time a
dual antiglioma effect of the well-known antidiabetic
drug metformin. In low-density cultures of the C6 rat
glioma cell line, metformin blocked the cell cycle
progression in G 0 /G 1 phase without inducing signifi-
cant cell death. In confluent C6 cultures, on the other
hand, metformin causedmassive induction of caspase-
dependent apoptosis associated with c-Jun N-terminal
kinase (JNK) activation, mitochondrial depolariza-
tion and oxidative stress. Metformin-triggered apop-
tosis was completely prevented by agents that block
mitochondrial permeability transition (cyclosporinA)
and oxygen radical production (N-acetylcisteine),
while the inhibitors of JNK activation (SP600125) or
glycolysis (sodium fluoride, iodoacetate) provided
partial protection. The antiglioma effect of metformin
was reduced by compound C, an inhibitor of AMP-
activated protein kinase (AMPK), and was mimicked
by the AMPK agonist AICAR. Similar effects were
observed in the human glioma cell line U251, while rat
primary astrocytes were completely resistant to the
antiproliferative and proapoptotic action of metfor-
min.
Keywords. Metformin, cancer, cell cycle, apoptosis, mitochondrial depolarization, oxidative stress, c-Jun
N-terminal kinase, AMP-activated protein kinase.
Introduction
glucoregulatory properties of metformin are mainly
attributed to reduced hepatic glucose production and
augmented glucose uptake by the peripheral tissues
[1] . More recent studies have documented the ability
of metformin to stimulate adenosine monophosphate-
activated protein kinase (AMPK) [1, 2] , an intra-
cellular energy sensor that is activated by rising AMP
and acts by switching on ATP-generating catabolic
Metformin (1-(diaminomethylidene)-3,3-dimethyl-
guanidine) is an antihyperglycaemic drug commonly
used for the management of type-2 diabetes [1] . The
* Corresponding authors.
128080651.007.png 128080651.008.png
 
Cell. Mol. Life Sci. Vol. 64, 2007
Research Article
1291
pathways while switching off ATP-requiring processes
[3] . Since AMPK inhibits hepatic glucose production
and stimulates muscle glucose uptake [1] , it is
potentially an ideal mediator of metformin action. In
contrast to the progress in understanding the mech-
anisms underlying the therapeutic effect of metfor-
min, its biological actions that are not directly linked
to antidiabetic activity have rarely been investigated.
Interestingly, two independent clinical studies have
recently shown that metformin may reduce the risk of
cancer in patients with type II diabetes [4, 5] . This is
consistent with the ability of metformin to suppress
both the spontaneous development of mammary
adenocarcinomas in HER-2/neu-transgenic mice [6]
and the induction of pancreatic cancer in high fat-fed
hamsters [7] . It is not clear, however, whether the
observed effects were due to a direct action of the drug
on tumor cells or resulted from its effects on insulin
metabolism. Namely, it is known that hyperinsuline-
mia and associated metabolic alterations, which are
corrected by metformin treatment, may play a role in
the onset of some malignancies [8] . The capacity of
metformin to block proliferation of human breast
cancer cells [9] or cause apoptotic cell death in a
mouse insulinoma cell line [10] argues in favor of
direct antitumor action, but the underlying mecha-
nisms are not completely understood.
Gliomas are extremely aggressive neuroectodermal
tumors that represent the most common primary
malignancy in human central nervous system [11] .
Gliomas are incurable in most of the cases, and it has
been postulated that their relative resistance to
apoptosis may contribute to chemotherapy and radi-
ation resistance [11] . Cell motility apparently contrib-
utes to the invasive phenotype of malignant gliomas,
and interference with cell motility by different strat-
egies results in increased susceptibility of glioma to
apoptosis [11] . Interestingly, a recent study revealed
the ability of metformin to inhibit in vitro migration of
malignant glioma cells [12] , indicating its potential
usefulness in antiglioma therapy. Accordingly, glioma
cells express both AMPKa1 and AMPKa2 [13] , the
putative targets of metformin intracellular action, and
pharmacological activation or overexpression of
AMPK has been shown to reduce glioma cell growth
[14] . However, the effects of metformin on glioma cell
proliferation and viability have not been explored so
far.
The present study demonstrates for the first time that
metformin induces cell cycle arrest as well as mito-
chondrial depolarization- and oxidative stress-de-
pendent apoptosis in cultured glioma cells. Impor-
tantly, primary astrocytes were almost completely
resistant to the antiproliferative/cytotoxic effects of
the drug.
Materials and methods
Cell cultures. All chemicals were from Sigma (St. Louis, MO)
unless specifically stated. The rat glioma cell line C6 and the human
glioma cell line U251 were kindly donated by Dr. Pedro Tranque
(Universidad de Castilla-La Mancha, Albacete, Spain). Primary
astrocytes were isolated from brains of newborn Albino Oxford
rats as previously described [15]. The tumor cell lines and
astrocytes were maintained at 378C in a humidified atmosphere
with 5%CO 2 in aHepes (20 mM)-bufferedRPMI 1640 cell culture
medium supplemented with 5% fetal calf serum, 2 mM L-
glutamine, 10 mM sodium pyruvate, penicillin/streptomycin and
10 mM glucose. The cells were prepared for experiments using the
conventional trypsinization procedure with trypsin/EDTA. For the
measurement of cell number/mitochondrial activity or flow
cytometric analysis of the cell cycle in low-density cell cultures,
cells were incubated in 96-well flat-bottom plates (5 10 3 cells per
well) or 24-well flat-bottom plates (5 10 4 cells per well),
respectively. For the enzyme-linked immunosorbent assay
(ELISA)/Chou-Talalay analysis or cell viability/flow cytometric
examination of apoptotic events in high-density cell cultures, cells
were incubated in 96-well (2 10 4 cells per well) or 24-well (2 10 5
cells per well) flat-bottomplates, respectively. Cells were rested for
24 h when high-density cell cultures reached confluence and
treated with metformin hydrochloride (99.9%; Hemofarm,
Vrsac, Serbia) and/or different agents, as described in Results
and Figure legends. Suspension of nanocrystalline C 60 fullerene
( n -C 60 ) was prepared as previously described [16].
Determination of cell number and mitochondrial activity. The
number of adherent, viable cells was assessed using a crystal violet
assay, while mitochondrial dehydrogenase activity, as another
indicator of cell viability, was determined by mitochondria-
dependent reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-
tetrazolium bromide (MTT) to formazan. Both tests were per-
formed exactly as previously described [17] . The results are
presented as relative increase compared to the control value
(untreated cells) arbitrarily set to 1 or as % of control value.
Cell cycle and apoptosis analysis. The cell cycle was analyzed by
measuring the amount of propidium iodide (PI)-labeled DNA in
ethanol-fixed cells, exactly as previously described [18]. Apoptotic
and necrotic cell death were analyzed by double staining with
fluoresceinisothiocyanate (FITC)-conjugated annexin Vand PI, in
which annexin V bound to the apoptotic cells with exposed
phosphatidylserine, while PI labeled the necrotic cells with
membrane damage. Staining was performed according to the
manufacturers instructions (BD Pharmingen, San Diego, CA).
The green (FL1) and red (FL2) fluorescence of annexin/PI-stained
live cells and PI-stained fixed cells was analyzed with a FACSCa-
libur flow cytometer (BD, Heidelberg, Germany) using a peak
fluorescence gate to exclude cell aggregates during cell cycle
analysis. The numbers of viable (annexin /PI ), apoptotic (annex-
in + /PI ) and necrotic (annexin + /PI + ) cells as well as the proportion
of cells in different cell cycle phases were determined with
CellQuest Pro software (BD). DNA fragmentation, as another
marker of apoptosis, was determined during cell cycle analysis by
counting the hypodiploid cells in the sub-G 0 /G 1 compartment [18].
Caspase activation. Activation of caspases was measured by flow
cytometry after labeling the cells with a cell-permeable, FITC-
conjugated pan-caspase inhibitor (ApoStat; R&D Systems, Min-
neapolis, MN) according to the manufacturers instructions.
Increase in green fluorescence (FL1) is a measure of caspase
activitywithin individual cells of the treated population. The results
are expressed as the percentage of cells containing active caspases.
Measurement of reactive oxygen species (ROS). Intracellular
production of ROS was determined by measuring the intensity of
green fluorescence emitted by the redox-sensitive dye dihydro-
rhodamine 123 (DHR; Invitrogen, Paisley, UK), which was added
to cell cultures (1 mM) at the beginning of treatment. At the end of
incubation, cells were detached by trypsinization and washed in
PBS, and the green fluorescence (FL1) of DHR-stained cells was
analyzed using a FACSCalibur flow cytometer.
1292 A. Isakovic et al.
Antiglioma action of metformin
Mitochondrial depolarization. The mitochondrial depolarization
was assessed using DePsipher (R&D Systems), a lipophilic cation
susceptible to changes inmitochondrial membrane potential. It has
the property of aggregating upon membrane polarization, forming
an orange-red fluorescent compound. If the potential is disturbed,
the dye can not access the transmembrane space and remains or
reverts to its green monomeric form. The cells were stained with
DePsipher as described by the manufacturer, and the green
monomer and the red aggregates were detected by flow cytometry.
The results are presented as a green/red fluorescence ratio
(geomean FL1/FL2), an increase of which reflects mitochondrial
depolarization.
Cell-based ELISA for mitogen-activated protein kinases (MAPK)
and glial fibrillary acidic protein (GFAP). A cell-based ELISAwas
used to measure activation of the MAPK family members p38
MAPK, extracellular signal-regulated kinase (ERK) and c-Jun-N-
terminal kinase (JNK), as well as activation of transcription factor
nuclear factor-kB(NF-kB) and expression of the astrocyte marker
GFAP. TheELISAwas performed using the appropriate antibodies
exactly as previously described [19]. The results are presented
relative to the control value, which was arbitrarily set to 1.
Mathematical analysis of cytotoxic interactions. To analyze the
type (additive, synergistic or antagonistic) of metformin interaction
with H 2 O 2 or n -C 60 , cells were treated with different doses of each
agent alone and appropriate combinations. Cell viability was
assessed using a crystal violet assay. The values of the combination
index, reflecting additive (=1), synergistic ( < 1) or antagonistic
( > 1) interactions, were calculated according to the method
designed by Chou and Talalay [20].
Statistics. The statistical significance of differences was analyzed by
t -test or ANOVA followed by the Student-Newman-Keuls test. A
value of p < 0.05 was considered significant.
violet (Fig. 1D) andMTT tests (not shown). Microscopic
examination revealed a dramatic change in the mor-
phology of metformin-treated glioma cells, which be-
came larger and adopted a spindle-like shape with
markedly elongated fine tapering processes (Fig. 1E).
These morphological changes, however, were not asso-
ciated with glioma cell differentiation toward mature
astrocytes, as expression of the astrocyte marker GFAP
in C6 cells actually decreased after metformin treatment
to 61 5.9% of the value observed in untreated cells
( n =3, p < 0.05). Flow cytometric analysis showed that
metformin-treated glioma cells display higher FSC
values in comparison with control cells (Fig. 1F), thus
confirming the observed increase in cell size. Finally, the
proportion of cells in the G 0 /G 1 cell cycle phase was
significantly increased in metformin-treated glioma
cultures, while apoptotic cells with fragmented DNA
(sub-G 0 /G 1 ) could not be observed (Fig. 1G). Therefore,
the antiglioma effect of metformin is mainly a conse-
quence of cell cycle arrest. In primary astrocytes,
however, neither cell cycle alterations nor associated
morphological changes were discernible (data not
shown).
Metformin induces caspase-dependent apoptosis in
confluent glioma cells. We next investigated whether
metformin could affect confluent glioma cells, expecting
a weaker effect due to the reduced proliferation rate of
confluent cells. Surprisingly, the number of initially
confluent glioma cells after 48 h of incubation with
metformin (4 mM) was reduced to < 30% of control
values, which could not be attributed solely to a
proliferation block. Metformin-treated C6 cells lost
their polygonal morphology and became smaller, with
granular appearance and poorly defined margins
(Fig. 2A). Flow cytometric analysis revealed that met-
formin caused a large increase in numbers of apoptotic
(annexin + PI ) glioma cells, while an increase in numbers
of annexin + PI + cells, which presumably underwent
secondary necrosis, was less pronounced (Fig. 2A).
Primary astrocytes, on the other hand, completely
preserved their normal morphology in the presence of
metformin and were largely resistant to its proapoptotic
action (Fig. 2B). Accordingly, a significant time-depend-
ent activation of caspases, apoptosis-executing cysteine
proteases with aspartate specificity, was observed in
metformin-treated C6 cells (Fig. 2C), while only a
marginal caspase induction was seen in primary astro-
cytes under the same cultivating conditions (Fig. 2D).
The increases in apoptotic cell number, caspase activa-
tion and DNA fragmentation were all efficiently pre-
vented by the pan-caspase inhibitor zVAD-fmk and the
protein synthesis inhibitor cycloheximide (CHX)
(Fig. 2E), thus confirming that metformin toxicity to
confluent glioma cells was due to induction of caspase-
Results
Metformin blocks proliferation of low-density glioma
cells. To assess the effect of metformin on glioma cell
proliferation, C6 and U251 glioma cells were seeded at
low density and their numbers determined by crystal
violet assay during 4-day incubation period. Metformin
inhibited the increase in glioma cell number in a dose-
dependent manner (Fig. 1A, C), and the highest con-
centration of the drug (8 mM) completely blocked the
proliferation of glioma cells. Similar results were ob-
tained with the MTTassay for mitochondrial respiration
(Fig. 1B). However, the values of mitochondrial respi-
ration per cell (expressed asMTT/crystal violet ratio) did
not significantly change in metformin-treated glioma
cultures (Fig. 1B), indicating that the mitochondrial
function was not affected. Therefore, the observed
reduction in cell number was due to an antiproliferative
rather than a cytotoxic effect of the drug. This was
consistent with the complete absence of apoptotic
(annexin + ) cells in glioma cell cultures exposed to
metformin (data not shown). The antiproliferative effect
was apparently reversible, as glioma cells incubated with
metformin (8 mM) for 48 h regained their proliferative
capacity after removal of the drug (data not shown). In
contrast to their transformed counterparts, rat primary
astrocytes were completely resistant to the antiprolifer-
ative action of metformin, as demonstrated by the crystal
Cell. Mol. Life Sci. Vol. 64, 2007
Research Article
1293
Figure 1. Metformin blocks proliferation of low-density
glioma cells. (A–D) Low-density C6 cells (A, B), U251
cells (C) or primary astrocytes (D) were incubated with
metformin, and cell number (crystal violet, CV) or
mitochondrial activity (MTT) was measured at the
indicated time points. A representative of three experi-
ments is presented, and the data are mean values of
triplicate observations (SD < 15% of the mean; * p < 0.05
refers to untreated cells). (E–G) After 96 h of treatment
with metformin (4 mM), the morphology of C6 cells was
examined by light microscopy (E), and cell size (F) and
cell cycle phase (G) were analyzed using flow cytometry.
Representative photographs and histograms are present-
ed. The numbers in (G) are mean SD values from three
independent experiments (* p < 0.05).
mediated, protein synthesis-dependent apoptosis. Re-
sults similar to those presented in Fig. 2A, C and E were
also obtained with U251 glioma cells (not shown).
decrease in the amount of red aggregates (Fig. 3A), a
pattern that is characteristic for disruption of mitochon-
drial membrane potential. This was accompanied by
significant generation of ROS, as concluded from a
green fluorescence shift that was readily observable in
metformin-treated C6 cells stained with the redox-
sensitive dye DHR (Fig. 3B). Results similar to those
presentedinFig.3AandBwerealsoobtainedwithU251
cells (data not shown). Cyclosporin A (CsA), an
inhibitor the mitochondrial permeability transition
(MPT) that precedes mitochondrial depolarization
[21] , and a well-known antioxidant agent, N-acetylcys-
teine (NAC) [22] , both prevented caspase activation,
DNA fragmentation and apoptotic cell death in C6
cultures (Fig. 3C). A time course analysis revealed that
Metformin-induced apoptosis is associated with mito-
chondrial depolarization and oxidative stress. To f u r t he r
elucidate the mechanisms underlying metformin-in-
duced apoptosis in confluent glioma cells, we assessed
if metformin could trigger mitochondrial depolarization
and production of ROS, two events that are frequently
associated with initiation of the apoptotic cascade.
Staining of C6 cells with the mitochondria-binding
fluorescent dye DePsipher revealed that metformin
treatment for 24 h caused an increase in the concen-
tration of the green monomer form with a concomitant
128080651.009.png 128080651.001.png 128080651.002.png 128080651.003.png 128080651.004.png 128080651.005.png
1294 A. Isakovic et al.
Antiglioma action of metformin
Figure 2. Metformin induces caspase-dependent
apoptosis in confluent glioma cells. (A–D) Confluent
C6 cells (A, C) or confluent primary astrocytes (B, D)
were incubated with metformin (4 mM). (E) Con-
fluent C6 cells were incubated with metformin
(4 mM) in the absence or presence of the pan-
caspase inhibitor zVAD-fmk (50 mM) or protein
synthesis blocker cycloheximide (CHX; 5 mg/mL).
Cell morphology (A, B), apoptosis (A, B, E; the
upper left quadrant in A, B contained < 5% cells)
and DNA fragmentation (E) were analyzed after
48 h using light microscopy or flow cytometry.
Caspase activation (C–E) was assessed at the indi-
cated time points (C) or after 36 h (D, E) by flow
cytometry. Representative photographs, dot plots
and histograms are presented. The data in (E) are
mean values SD from three separate experiments
(* p < 0.05 refers to both control and zVAD-fmk/
CHX-treated cells).
metformin-induced mitochondrial depolarization pre-
ceded ROS production by several hours (Fig. 3D).
Moreover, mitochondrial depolarization triggered by
metformin was actually associated with a decrease in
intracellular ROS levels (Fig. 3D), thus excluding the
possibility that disruption of mitochondrial membrane
potential was caused by oxidative stress. This was further
confirmed by the inability of either NAC or CHX to
prevent mitochondrial depolarization in glioma cells
(Fig. 3E), even though they efficiently reduced metfor-
min-induced ROS generation (Fig. 3F). On the other
hand, CsA readily hyperpolarized mitochondrial mem-
branes in metformin-treated C6 cells (Fig. 3E) but failed
to prevent production of ROS (Fig. 3F). Moreover, CsA
itself markedly increased ROS generation in C6 cells
even in the absence of metformin (Fig. 3F). These data
indicate that mitochondrial depolarization and oxidative
stress both participate in metformin-triggered apoptosis
of glioma cells but are apparently induced by partly
independent mechanisms. In accordance with the ob-
served resistance to metformin-induced apoptosis
(Fig. 2B, D), primary astrocytes did not display signifi-
cant mitochondrial depolarization or increased ROS
production following metformin treatment (not shown).
128080651.006.png
Zgłoś jeśli naruszono regulamin