You are on page 1of 11

Journal of Neurochemistry, 2007, 103, 569–579 doi:10.1111/j.1471-4159.2007.04775.

ATP-sensitive potassium channel opener iptakalim protects against


MPP+-induced astrocytic apoptosis via mitochondria and
mitogen-activated protein kinase signal pathways

Shu Zhang,1 Fang Zhou,1 Jian-Hua Ding, Xi-Qiao Zhou, Xiu-Lan Sun and Gang Hu
Laboratory of Neuropharmacology, Institute of Neurosciences, Nanjing Medical University, Nanjing, Jiangsu, P. R. China

Abstract protective effect of IPT on MPP+-induced astrocytic apoptosis.


Inhibition of astrocytic apoptosis has been regarded as a novel Furthermore, IPT could also activate ERK/MAPK and main-
prospective strategy for treating neurodegenerative disorders tain increased phospho-ERK1/2 level after MPP+ exposure.
such as Parkinson’s disease. In the present study, we dem- Taken together, these findings reveal for the first time that IPT
onstrated that iptakalim (IPT), an ATP-sensitive potassium protects against MPP+-induced astrocytic apoptosis via inhi-
channel (KATP channel) opener, exerted protective effect on bition of mitochondria apoptotic pathway and regulating the
MPP+-induced astrocytic apoptosis, which was reversed by MAPK signal transduction pathways by opening mitochondrial
selective mitochondrial KATP channel blocker 5-hydroxyde- ATP-sensitive potassium (mitoKATP) channels in astrocytes.
canoate. Further study revealed that IPT inhibited glutathione And targeting KATP channels expressed in astrocytes may
(GSH) depletion, mitochondrial membrane potential loss and provide a novel therapeutic strategy for neurodegenerative
subsequent release of pro-apoptotic factors (cytochrome c disorders.
and apoptosis-inducing factor (AIF), and c-Jun NH2-terminal Keywords: apoptosis, astrocyte, ATP-sensitive potassium
kinase/mitogen-activated protein kinases (MAPK) phos- channel opener, iptakalim, mitochondria, mitogen-activated
phorylation induced by MPP+. Meanwhile, extracellular signal- protein kinase.
regulated kinase (ERK) 1/2 inhibitor PD98059 inhibited the J. Neurochem. (2007) 103, 569–579.

Astrocytes, the major non-neuronal cells in the brain, have may lead to the development of novel therapeutic strategies
been implicated in the segregation, maintenance, and support for neurodegenerative disorders (Takuma et al. 2004). And
of neurons, including clearance and release of extracellular the drugs that protect against astrocytic apoptosis are
glutamate (Anderson and Swanson 2000; Mazzanti et al. promising neuroprotectants.
2001), scavenging oxygen free radicals (Chen et al. 2001), ATP-sensitive potassium (KATP) channels, which link cell
homeostatic maintenance of extracellular ionic environment metabolism to its membrane potential, belong to a class of
and pH (Amiry-Moghaddam and Ottersen 2003; Simard and
Nedergaard 2004), provision of metabolic substrates for Received January 16, 2007; revised manuscript received March 15,
neurons (Kasischke et al. 2004), and coupling of cerebral 2007; accepted May 30, 2007.
blood flow to neuronal activity (Takano et al. 2006). Address correspondence and reprint requests to Gang Hu, Laboratory
Accordingly, astrocyte dysfunction and even dysregulation of Neuropharmacology, Institute of Neurosciences, Nanjing Medical
of astrocyte-specific functions can critically influence neur- University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, P. R. China.
E-mail: ghu@njmu.edu.cn
onal survival (Seifert et al. 2006). Traditionally, necrosis was 1
These authors contributed equally to this work.
believed to be the predominant mechanism of astrocytes Abbreviations used: DYm, mitochondrial membrane potential; 5-HD,
death in brain injury models, but increasing evidence showed 5-hydroxydecanoate; AIF, apoptosis-inducing factor; DZ, diazoxide;
that astrocytic apoptosis may contribute to pathogenesis of ERK, extracellular signal-regulated kinase; IPT, iptakalim; JNK, c-Jun
many neurodegenerative disorders such as Alzheimer’s NH2-terminal kinase; KATP channels, ATP-sensitive potassium channels;
KCO, KATP channel opener; Kir, inwardly rectifying potassium subunit;
disease and Parkinson’s disease (PD) (Kobayashi et al. MAPK, mitogen-activated protein kinase; PBS, phosphate-buffered
2002; Szydlowska et al. 2006). Thus, the advance in the saline; PD, Parkinson’s disease; PI, propidium iodide; SUR, sulfonylurea
knowledge of molecular regulation of astrocytic apoptosis receptors.

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579 569
570 S. Zhang et al.

inwardly rectifying potassium channels that are widely Materials and methods
distributed in the brain (Dunn-Meynell et al. 1998; Zawar
et al. 1999). Functional KATP channels are heteroctameric Astrocyte cell culture
complexes, consisting of discrete pore-forming (inwardly Rat primary astrocytes were prepared according to previously
rectifying potassium subunit; Kir6.1/Kir6.2) and regulatory described protocol with slight modifications (Hamprecht and Loffler
subunits (sulfonylurea receptors; SUR1/SUR2). The chan- 1985). Briefly, tissues from whole brains of postnatal (P1–P2)
nels are usually closed in normal conditions, but are activated Sprague–Dawley (SD) rats were triturated and then cells were plated
rapidly in response to decrease in intracellular ATP/ADP on poly-D-lysine pre-coated cell culture flasks in Dulbecco’s
ratio under metabolic stress, which results in hyperpolariza- modified Eagle’s medium containing 10% fetal calf serum, 100 U/
tion of the cell membrane and limitation of Ca2+ influx, thus mL penicillin and 100 lg/mL streptomycin. Cultures were main-
blocking subsequent neurotoxic biochemical cascades (Seino tained at 37C in a humidified atmosphere of 5% CO2/95% air. After
reaching a confluent monolayer of glial cells (10–14 days),
and Miki 2003). Emerging evidence including from our
microglia were separated from astrocytes by shaking off for 5 h at
laboratory has revealed a novel neuronal protective role of
100 rpm. The enriched astrocytes were >96% positive for glial
KATP channel openers (KCOs) in PD models both in vivo and fibrillary acidic protein as assessed by immunocytochemical
in vitro (Tai et al. 2003; Wang et al. 2005; Yang et al. 2004, staining.
2006b). Therefore, KCOs that can freely cross the blood-
brain barrier are considered as potential neuroprotectants GSx assay
(Wang et al. 2006; Yang et al. 2005). Accumulated evidence GSH was assayed as total glutathione (GSx), which was the sum of
shows that modulation of KATP channels expressed in the reduced and oxidized forms [GSH+2·glutathione disulfide
neurons exerts neuroprotective effect (Liss et al. 2005; (GSSG)], by a modified enzymatic recycling method of Tietze
Nakagawa et al. 2005). Nevertheless, little is known about (Tietze 1969). Astrocytes were washed in phosphate-buffered saline
the role of regulating astrocytic KATP channels in neurode- (PBS), dissolved in 200 lL PBS buffer (0.05 mmol/L edetic acid;
0.05% Triton-X 100, pH 8.0), and centrifuged to remove protein.
generative disorders.
According to the Bradford method (Kruger 1994), a volume of
Iptakalim (IPT) is a lipophilic para-amino compound with
50 lL of supernatant was used to assess protein by using bovine
low molecular weight, which can freely cross the blood-brain serum albumin as a standard. A volume of 100 lL of supernatant
barrier and has been demonstrated to be a novel KCO by was neutralized with 50 lL of a 5% (w/v) salicylsalicylic acid
pharmacological, electrophysiological, and biochemical stud- solution. The solution was centrifuged, and then the GSH content
ies, and receptor binding test (Wang 2003; Wang et al. 2004; was determined after the addition of 4 lL 10 mmol/L b-NADPH,
Xie et al. 2005). Our previous studies provided compelling 3 lL 10 mmol/L 5, 5¢- dithio-bis-2-nitrobenzoic acid, 2 kU/L GSH
supports for the neuroprotective effects of IPT on behavioral reductase. The GSH content was determined by kinetic measure-
recovery and neuronal survival in various animal models ment of the absorbance changes at 412 nm for 5 min and calculated
such as stroke and PD, as well as in cultured cells (Wang by comparison with standards.
et al. 2004, 2005; Yang et al. 2004). IPT is not only a useful
Determination of protein
pharmacological tool for KATP channel investigation, but it
The cells were dissolved in 1% dodecylsulfate in the presence of
also serves as a novel, high-potent, low-toxic, therapeutic
0.1 mol/L NaOH. The amount of protein was determined by
agent that protects brain neurons against a variety of applying the method of Bradford assay (Kruger 1994) using bovine
neurodegenerative diseases (Wu et al. 2005). As KATP serum albumin as the standard.
channels are expressed in astrocytes (Thomzig et al. 2001),
whether IPT exerts neuroprotective effects via regulating
Morphological assessment and quantification of apoptotic
KATP channels in astrocytes needs to be further investigated.
astrocytes
The neurotoxin MPP+, a high affinity inhibitor of
mitochondrial complex I, which is metabolite formed by Hoechst staining
monoamine oxidase-B oxidation of MPTP in astrocytes To quantify apoptotic astrocytes, astrocytic monolayer was fixed and
(Tipton and Singer 1993), can induce selective dopaminer- stained with Hoechst 33324 (Sigma, St Louis, MO, USA). The
morphological features of apoptosis (cell shrinkage, chromatin
gic neuronal degeneration in substantia nigra (Chiueh and
condensation, and fragmentation) were monitored by fluorescence
Rauhala 1998). In addition, MPP+ is a common neurotoxin
microscopy (Olympus BX 60, Tokyo, Japan). At least 400 cells
used to explore the alteration of astrocytic function (Henze from 12 randomly selected fields per dish were counted, and each
et al. 2005; Tripanichkul et al. 2006). And KATP channel is treatment was performed in triplicate.
considered as a potential downstream target of mitochond-
rial complex I inhibition (Liss et al. 2005). Therefore, in the Flow cytometry
present study, we try to investigate whether IPT offers Astrocytic apoptosis was estimated using the Annexin-V Fluoresc-
protective effect on MPP+-induced astrocytic apoptosis by ein (FITC) apoptosis detection kit (Oncogene Research Products,
opening KATP channels and to elucidate the anti-apoptosis San Diego, CA, USA) according to the kit instructions. The cell
mechanisms of IPT. samples were analyzed in a flow cytometry apparatus (Becton

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
Iptakalim inhibits astrocytic apoptosis 571

Dickinson FACSVantage SE, San Jose, CA, USA). Annexin V binds using a Bio-Rad miniprotein-III wet transfer unit. The membranes
to phosphatidylserine that is translocated during apoptosis from the were incubated with 5% bovine serum albumin dissolved in TBST
inner to the outer leaflet of the plasma membrane. Live cells with (pH 7.5, 10 mmol/L Tris-HCl, 150 mmol/L NaCl, and 0.1%
intact membranes are distinguished by their ability to exclude Tween-20) at 25C for 1 h, washed three times and incubated with
propidium iodide (PI), which readily penetrates dead or damaged different antibodies [c-Jun NH2-terminal kinase (JNK), phosphor-
cells. Dual analysis was introduced using a quadrant dot plot, in JNK, extracellular signal-regulated kinase (ERK), and phosphor-
which necrotic cells were identified as single PI-positive, early ERK at 1 : 1000]. The membranes were washed three times with
apoptotic cells were annexin V-FITC-positive only, and cells in late TBST buffer and incubated with the secondary antibody (1 : 2000)
apoptosis were recognized as double-positive for annexin V-FITC for 1 h followed by four washings. Signal detection was performed
and PI. Cells that stained negative for both annexin V-FITC and PI with an enhanced chemiluminescence kit.
were classified as live cells. Finally, the number of cells in each
category was expressed as a percentage of the total number of Statistical analysis
stained cells counted. All values were expressed as mean ± SEM. The significance of the
difference between control and samples treated with various drugs
Measurement of astrocyte mitochondrial membrane potential was determined by one-way ANOVA followed by the post hoc least
Astrocyte mitochondrial membrane potential (DYm) was assessed significant difference test. Differences were considered significant at
with the fluorescent probe JC-1. At 490 nm, cells with depolarized p < 0.05.
mitochondria contained JC-1 predominantly in monomeric form and
fluoresced green. Cells with polarized mitochondria predominantly
contained JC-1 in aggregate form, and show fluoresced red-orange. Results
Astrocytes were incubated with 5 lmol/L of JC-1 (Molecular
Probes, Eugene, OR, USA) for 10 min at 37C, washed, and placed
IPT inhibits MPP+-induced astrocytic apoptosis
on a thermostatted stage at 37C. Fluorescent images were
To explore whether IPT could inhibit MPP+-induced astro-
visualized by a Nikcon Optical TE2000-S inverted fluorescence
microscope with excitation at 490 nm and emission at >520 nm. cytic apoptosis, the nuclear staining with Hoechst 33342 in
Astrocytes with polarized mitochondria were seen with distinct the MPP+-treated astrocytes was observed. As showed in
mitochondria fluorescing red-orange, and, in astrocytes with Fig. 1, untreated astrocytes exhibited regular and round-
depolarized mitochondria, the cytoplasm and mitochondria appeared shaped nuclei. In contrast, the condensation and fragmenta-
green. Acquired signal was analyzed with image-analysis software tion of nuclei, characteristic of apoptotic cells, was evident in
(Simple PCI; Compix Inc., Cranberry Township, PA, USA). A astrocytes treated with different concentrations of MPP+ for
minimum of six fields were selected and average intensity for each 48 h (Fig. 1a). Pretreatment with 1, 10, or 100 lmol/L IPT
region was quantified. The ratio of J-aggregate to JC-1 monomer reduced the MPP+-induced astrocytic apoptosis in a concen-
intensity for each region was calculated. A decrease in this ratio was tration-dependent manner. Diazoxide (DZ), a selective mit-
interpreted as loss of DYm, whereas an increase in the ratio was
ochondrial KATP (mitoKATP) channel opener, had the similar
interpreted as gain in DYm.
effect to IPT with a maximum inhibition at 100 lmol/L.
Detection of mitochondrial cytochrome c and AIF levels Pretreatment with selective mitoKATP channel blocker
107 astrocytes were washed with ice-cold phosphate-buffered 5-hydroxydecanoate (5-HD, 250 lmol/L) could abolish the
saline, and cells were resuspended for 15 min on ice in inhibitory effect of IPT (10 lmol/L) or DZ (100 lmol/L) on
permeabilization buffer containing 75 mmol/L NaCl, 1 mmol/L MPP+-induced astrocytic apoptosis (Fig. 1b). However,
NaH2PO4, 250 mmol/L sucrose, 1 mmol/L phenylmethylsulfonyl sarcolemmal KCO P1075 (1, 10, 100 lmol/L) failed to
fluoride, additional protease inhibitors, and 0.05% digitonin. inhibit MPP+-induced astrocytic apoptosis (p > 0.05) (data
Following a centrifugation step at 800 g at 4C for 10 min, the not shown).
supernatant was separated from the pellet consisting of cellular The results of flowcytometry further confirmed the
debris. The crude mitochondrial pellet was collected by centrif- anti-apoptotic effects of IPT on MPP+-induced astrocytic
ugation at 13 000 g at 4C for 10 min. Mitochondrial proteins
apoptosis (Fig. 1c). Annexin-V binding and PI uptake were
(50 lg) were then subjected to immunoblot analysis using a
detected in astrocytes. The portion of annexin-V positive
monoclonal Cytochrome c (1 : 1000) and AIF antibody (1 : 1000)
as described below.
cells increased from 9% in control cells to 29% in the cells
treated with MPP+ for 48 h. The treatment of IPT (10 lmol/L)
Western blotting or DZ (100 lmol/L) 30 min prior to MPP+ addition induced a
Cells were washed twice with ice-cold PBS and homogenized in decrease in the portion of apoptotic cells from 29% in cells
200 lL lysis buffer. After incubation for 20 min on ice, cell treated with MPP+ to 11% and 10%, respectively. And these
lysates were centrifuged (10 000 g for 10 min at 4C) and protein effects were abolished by mitoKATP channel blocker 5-HD
concentration in the extracts was determined by the Bradford (250 lmol/L), suggesting that IPT could inhibit MPP+-
assay (Kruger 1994). Proteins in cell extracts were denatured with induced astrocytic apoptosis through opening mitoKATP
sodium dodecyl sulphate sample buffer and separated by 10% channels.
SDS-PAGE. Proteins were transferred to nitrocellulose membranes

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
572 S. Zhang et al.

(a)
Con. MPP+ 50µmol/L MPP+ 100µmol/L

MPP+ 200µmol/L MPP+ 400µmol/L MPP+ 800µmol/L

(b)

(c)

Fig. 1 Iptakalim (IPT) inhibits MPP+-induced astrocytic apoptosis. were identified as single PI-positive, early apoptotic cells were Annexin
The morphological features of apoptosis were monitored by fluores- V-FITC-positive only, and cells in late apoptosis were recognized as
cence microscopy after staining with Hoechst 33342. Scale bar: double-positive for Annexin V-FITC and PI. Cells that stained negative
50 lm. (a) MPP+ (50–800 lm) induced astrocytic apoptosis in a for both Annexin V-FITC and PI were classified as live cells. The data
concentration-dependent manner when incubated for 48 h. (b) Pre- presented as mean ± SEM of four individual experiments. *p £ 0.05;
treatment with IPT (10 lmol/L) or diazoxide (DZ) (100 lmol/L) for **p £ 0.01, compared with untreated group. #p £ 0.01; ##p £ 0.001,
30 min inhibited MPP+ (200 lmol/L)-induced astrocytic apoptosis, compared with MPP+-treated group. +p £ 0.05; ++p £ 0.01, compared
which was abolished by 5-hydroxydecanoate (5-HD; 250 lmol/L). (c) with both IPT and MPP+-treated group. $p £ 0.05; $$p £ 0.01, com-
And the apoptosis rate was measured by flowcytometry analysis after pared with both DZ and MPP+-treated group.
staining with Annexin V-FITC and propidium iodide (PI). Necrotic cells

IPT suppresses MPP+-induced endogenous glutathione MitoKCO DZ also increased the GSx level to 115%, 124%,
depletion in astrocytes 152%, and 154% of control at the concentration of 10, 50,
GSH is known to protect proteins from oxidation by 100, and 200 lmol/L in astrocytes (Fig. 2b). The GSx level
conjugating with oxidized thiol groups, and depletion of was decreased to 40% of control after incubation of the
GSH may be responsible for inhibition of complex I activity astrocytes with MPP+ (200 lmol/L) for 48 h. But MPP+
via thiol oxidation (Jha et al. 2000). To search into the failed to decrease the GSx level at low concentration
relationship between endogenous glutathione and MPP+- (<25 lmol/L) (Fig. 2c). Either IPT (10 lmol/L) or DZ
induced cell damage, the GSx level of astrocyte was (100 lmol/L) could suppress MPP+-induced endogenous
measured. As demonstrated in Fig. 2a, the GSx level was GSx depletion and increase GSx level by 49% and 43% of
significantly increased by 35%, 67%, and 69% of control control respectively, which was abolished by mitoKATP
after exposure to IPT 1, 10, and 100 lmol/L respectively. channel blocker 5-HD (250 lmol/L) (Fig. 2d). These results

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
Iptakalim inhibits astrocytic apoptosis 573

(a) (b)

(c) (d)

Fig. 2 Iptakalim (IPT) or diazoxide (DZ) suppresses MPP+-induced (100 lmol/L) for 30 min prior to MPP+ (200 lmol/L) exposure for 48 h.
endogenous glutathione depletion in astrocytes. (a) Incubation with The endogenous glutathione depletion induced by MPP+ was sup-
increasing concentration of IPT (0.01–100 lmol/L) or (b) DZ (1– pressed. The data presented as mean ± SEM of four individual
200 lmol/L) for 48 h increased the intracellular GSx levels in astro- experiments. *p £ 0.05; **p £ 0.01, compared with untreated group.
cytes. (c) Exposure to MPP+ (5–200 lmol/L) for 48 h decreased #
p £ 0.01, compared with MPP+-treated group. +p £ 0.05, compared
intracellular GSx levels in a concentration-dependent manner in ast- with both IPT and MPP+-treated group. $p £ 0.05, compared with both
rocytes. (d) Astrocytes were treated with IPT (10 lmol/L) or DZ DZ and MPP+-treated group.

demonstrate that IPT can inhibit MPP+-induced endogenous levels, and these effects were abolished by mitoKATP
GSx depletion through opening mitoKATP channels. channel blocker 5-HD (250 lmol/L) (Fig. 3d). These data
indicate that IPT can prevent MPP+-induced DYm loss and
IPT prevents MPP+-induced DYm loss and pro-apoptotic subsequent releasing pro-apoptotic factor in astrocytes by
factor release in astrocytes opening mitoKATP channels.
As DYm loss, a consequence of mitochondrial membrane
permeability, is detected in several models of apoptosis, IPT inhibits MPP+-induced JNK phosphorylation in
molecular probe JC-1 was used to detect the effect of IPT on astrocytes
MPP+-induced astrocyte DYm change. Astrocytes exposed to Increasing evidence shows that JNK plays an important role in
MPP+ (200 lmol/L) for 6 h displayed a loss of DYm apoptosis (Lotharius et al. 2005). So we investigated whether
(Fig. 3a), indicated by fluorescence of JC-1 shifted from IPT could affect MPP+-induced JNK phosphorylation in
red-orange to greenish yellow. Pretreatment with 10 lmol/L astrocytes. Treatment with MPP+ (200 lmol/L) led to a rapid
IPT or 100 lmol/L DZ could prevent mitochondria from loss and transient phosphorylation of JNK with the peak levels of
of DYm induced by MPP+, which was abolished by addition phospho-JNK occurring at 30 min, indicating that JNK
of 5-HD (250 lmol/L) (Fig. 3b). signaling pathway is activated in response to MPP+ treatment
The effect of IPT on MPP+-induced change of astrocyte in astrocytes (Fig. 4a). Next, we detected whether IPT could
mitochondrial membrane permeability was assessed by regulate MPP+-induced JNK phosphorylation at 30 min time
measuring mitochondrial pro-apoptotic factors (cytochrome point. As shown in Fig. 4b, pretreatment with IPT (10 lmol/L)
c and AIF) levels. Pretreatment with MPP+ (200 lmol/L) or DZ (100 lmol/L) reduced MPP+-induced increase of
markedly decreased mitochondrial cytochrome c and AIF phospho-JNK by over 40%. And these effects were abolished
levels in astrocytes, implying that MPP+ increased the release by mitoKATP channel blocker 5-HD (250 lmol/L). These
of cytochrome c and AIF from mitochondria and subse- results indicate that IPT can inhibit MPP+-induced JNK
quently increased the AIF nuclear levels and cytochrome c phosphorylation via opening mitoKATP channels.
cytosolic levels, respectively. Cytochrome c release occurred
as early as 3 h after MPP+ exposure, whereas AIF release ERK activation is involved in the anti-apoptotic effects of
occurred in a delayed fashion (Fig. 3c). Either IPT (10 lmol/ IPT on astrocytes
L) or DZ (100 lmol/L) significantly depressed the MPP+- Extracellular signal-regulated kinase/mitogen-activated pro-
induced the decrease of mitochondrial cytochrome c and AIF tein kinases (MAPK) pathway has been demonstrated to be

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
574 S. Zhang et al.

(a)

Fig. 3 Iptakalim (IPT) alleviates MPP+-in-


duced mitochondrial membrane potential
loss and pro-apoptotic factor release in
(b) astrocytes. Quantification of mitochondrial
membrane potential expressed as a ratio of
J-aggregate to JC-1 monomer (red : green)
fluorescence intensity. Scale bar: 100 lm.
(a) Astrocytes were treated for various
periods of time up to 8 h with MPP+
(200 lmol/L), and the mitochondrial mem-
brane potential was measured. (b) Pre-
treatment with IPT (10 lmol/L) or diazoxide
(DZ; 100 lmol/L) for 30 min prior to MPP+
(200 lmol/L) exposure for 6 h suppressed
(c) MPP+-induced mitochondrial depolariza-
tion. And 5-hydroxydecanoate (5-HD;
250 lmol/L) blocked these effects. (c)
Western blot analysis of mitochondrial AIF
and cytochrome c level. Astrocytes were
exposed to MPP+ (200 lmol/L) for various
periods of time up to 24 h. *p £ 0.05, com-
pared with AIF control groups. #p £ 0.05;
##
p £ 0.01, compared with cytochrome c
control groups. (d) IPT (10 lmol/L) or DZ
(100 lmol/L) inhibited MPP+-induced de-
crease of cytochrome c and AIF levels in
astrocytic mitochondria. Western blots at
the top of each panel are from a typical
(d) experiment. Bar graphs are the quantified
results expressed as mean ± SEM of rel-
ative cytochrome c or AIF levels from four
independent experiments. Results are rep-
resentative of three separate experiments
and each bar indicates the mean ± SEM.
*p £ 0.05; **p £ 0.01, compared with un-
treated group. #p £ 0.05; ##p £ 0.01, com-
pared with MPP+-treated group. +p £ 0.05,
compared with both IPT and MPP+-treated
group. $p £ 0.05, compared with both DZ
and MPP+-treated group.

involved in anti-apoptotic pathways (De Girolamo and Billett astrocytes exposed to KCOs and /or MPP+. Enhanced level
2006; Yue et al. 2000). In present study, ERK1/2 inhibitor of phospho-ERK1/2 was detected 15 min after exposure to
PD98059 (50 lmol/L) had no effect on the normal condition IPT (10 lmol/L) or DZ (100 lmol/L), which sustained for at
and MPP+-induced astrocytic apoptosis. However, in the least 4 h (Fig. 5b). Exposure of astrocytes to MPP+ alone
presence of PD98059, the protective effects of IPT (10 lmol/ also increased phospho-ERK1/2 level within 15 min, but it
L) and DZ (100 lmol/L) on MPP+-induced astrocytic subsided to control level at 2 h and decreased to below
apoptosis were inhibited (Fig. 5a). These results suggest that control level at 4 h (Fig. 5c). However, both IPT and DZ
ERK/MAPK pathway might be involved in the anti- could maintain MPP+-induced increase of phospho-ERK1/2
apoptotic effects of IPT on astrocytes. To confirm it, we levels for at least 4 h, which was completely abolished by the
performed the western blot analysis of phospho-ERK1/2 in ERK1/2 inhibitor PD98059 (Fig. 5d). These results show

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
Iptakalim inhibits astrocytic apoptosis 575

(a) The present study showed that, as a novel KCO, IPT


possessed similar protective effects to classic mitoKCO DZ
on MPP+-induced astrocytic apoptosis in vitro. And this anti-
apoptotic effect of IPT could be abolished by selective
mitoKATP channel blocker 5-HD. The protective effect of IPT
on MPP+-induced astrocytic apoptosis was further confirmed
by flow cytometry analysis. However, sarcolemmal KCO
P1075 failed to protect against MPP+-induced astrocytic
apoptosis. These findings suggest that IPT inhibit astrocytic
apoptosis through opening mitoKATP channels in astrocytes.
KCOs, especial mitochondria KCOs, can provide protective
(b) effects for neurons and neuroblast against cell damage
induced by ischemia, trauma, and toxic reagents (such as
rotenone and MPP+) (Hu et al. 2005; Nakagawa et al. 2005;
Yang et al. 2006a). Moreover, Bajgar demonstrated that the
amount of mitoKATP channels located in brain cells is at least
six fold higher than that in heart cells (Bajgar et al. 2001),
indicating that mitoKATP channels play essential role in the
functions of central nervous system. Thus, mitoKATP chan-
nels may be an important molecular target for the regulation
of astrocytic functions. It was found that IPT exhibited
marked neuroprotective effect on rotenone-induced PD
model (Yang et al. 2005, 2006b). However, opening the
KATP channels, which were composed of Kir6.2 and SUR1
Fig. 4 Iptakalim (IPT) or diazoxide (DZ) inhibits MPP+-induced c-Jun subunits, in dopamine neurons exhibited an unexpected role
NH2-terminal kinase (JNK) phosphorylation in astrocytes. (a) Astro- in promoting neurodegenerative process of PD (Deutch and
cytes were exposed to MPP+ (200 lmol/L) for the indicated times. (b) Winder 2006; Liss et al. 2005). Our previous study found
Astrocytes were pretreated with IPT (10 lmol/L) or DZ (100 lmol/L) that IPT failed to open KATP channels in rat substantia nigra
for 30 min and then exposed to MPP+ (200 lmol/L) for another
pars compacta DA neurons (Wu et al. 2006). Thus, IPT may
30 min. Western blots at the top of each panel are from a typical
offer protective effects on PD model through opening KATP
experiment. Bar graphs are the quantified results expressed as
channels composed of Kir6.1 and SUR1/SUR2 subunits in
mean ± SEM of relative phospho-JNK levels from four independent
experiments. *p £ 0.05; **p £ 0.01, compared with untreated group.
astrocytes (Thomzig et al. 2001).
#
p £ 0.01, compared with MPP+-treated group. +p £ 0.05, compared The involvement of oxidative stress in MPP+-induced
with both IPT and MPP+-treated group. $p £ 0.05, compared with both dopaminergic neuronal apoptosis, as demonstrated by an
DZ and MPP+-treated group. increase in the product of reactive oxygen species and/or a
decrease in antioxidants, has been described (Chun et al.
2001; Kaul et al. 2003). And it has been demonstrated that
that IPT inhibits MPP+-induced astrocytic apoptosis via oxidative stress might originate in astrocytes rather than in
activating ERK/MAPK signal pathway. neurons (Makarov et al. 2006; Sagara et al. 1993). However,
whether a similar cytotoxic effect of MPP+ occurs in
astrocytes is unclear. The present study showed that MPP+
Discussion
could decrease endogenous glutathione level in astrocytes.
Although much of the work on neuroprotection has focused Glutathione, the major antioxidant in cells, has been shown
on improving the survival of neurons, a prominent concom- to be neuroprotective both in vivo and in vitro (Cho et al.
itant effect of brain insults is the damage of glia, in particular 2003). Astrocytes surrounding dopaminergic neurons in the
astrocytes. Increasing evidence implies that astrocytic apop- brain may have a close relation with the selective vulnerab-
tosis participates in many neurodegenerative disorders ility of these neurons by scavenging reactive oxygen species
(Benjelloun et al. 1998; Franz et al. 2002; Szydlowska et al. and releasing CysGly, which is the precursor of GSH
2006), and astrocytic dysfunction may further contribute to synthesis in neurons. Hence, alteration in astrocytic gluta-
neuronal loss and the overall pathology (Seifert et al. 2006). thione level may be an important contributor to the
Inhibition of astrocytic apoptosis is an essential neuropro- pathogenesis of neurodegenerative disease such as PD
tective strategy (Takuma et al. 2004). Therefore, the drugs (Jenner and Olanow 1998). The results of present study
that can inhibit astrocytic apoptosis may offer protective reveal that treatment with IPT alone could significantly
effects on neurodegenerative disorders such as PD. increase the endogenous glutathione level in astrocytes. IPT

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
576 S. Zhang et al.

(a)

(b)

Fig. 5 Extracellular signal-regulated kinase


(ERK) pathway mediates the anti-apoptotic
effect of iptakalim (IPT) on astrocytes. (a)
Pretreatment with PD98059 (50 lmol/L) for
30 min inhibited the anti-apoptotic effects of
IPT (10 lmol/L) or diazoxide (DZ;
100 lmol/L) on astrocytes. (b) Western blot
analysis of astrocytes stained for phospho-
ERK showed that IPT (10 lmol/L) or DZ
(100 lmol/L) quickly induced a sustained
activation of ERK1/2, lasting at least 4 h. (c)
MPP+ (200 lmol/L) alone activated ERK1/2
(c)
for only the first 2 h after exposure. (d) IPT
(10 lmol/L) or DZ (100 lmol/L) maintained
the increased phospho-ERK1/2 levels for at
least 4 h after MPP+ exposure, and this
effect was abolished by PD98059 (50 lmol/
L). The data presented as mean ± SEM of
four individual experiments. *p £ 0.05;
**p £ 0.01, compared with untreated group.
#
p £ 0.05; ##p £ 0.01, compared with
MPP+-treated group. +p £ 0.01, compared
with both IPT and MPP+ treated group.
$
p £ 0.01, compared with both DZ and
MPP+ treated group.

also inhibited the glutathione depletion induced by MPP+, glutamate anti-porter Xc-, leading to glutathione depletion
which was abolished by mitoKATP channel blocker 5-HD. (Cho and Bannai 1990). Because IPT and MPP+ share a
We found that both IPT and DZ could increase astrocytic common mechanism at regulating astrocytic glutamate
glutamate uptake (Zhang and Hu 2004). And MPP+ inhibited uptake, we proposed that IPT might reverse the inhibitory
glutamate uptake and subsequently increased extracellular effect of MPP+ on glutathione level through enhancing the
glutamate levels in astrocytes, which was reversed by IPT glutamate uptake, reducing extracellular glutamate levels and
(Hu et al. 2005). MPP+ could also decrease glutathione level subsequently alleviating the inhibitory effect on cystine-
in astrocytes (McNaught and Jenner 2000). As glutamate is a glutamate anti-porter in astrocytes. As glutathione partici-
precursor for glutathione, the inhibitory effect of MPP+ on pates in the regulation of apoptosis (Hall 1999), IPT may
glutamate uptake may lead to the reduction of glutathione prevent astrocytic apoptosis through increasing intracellular
level in astrocytes. Thus, we inferred that IPT-induced the glutathione level.
increase of glutamate uptake could result in enhanced Furthermore, the anti-apoptotic mechanisms of IPT were
production of glutathione. In addition, the current explan- explored. GSH depletion is directly responsible for the noted
ation for glutamate-mediated toxicity is due to oxidative decreases in complex I activity and the subsequent mito-
glutamate toxicity besides excitotoxicity. This mechanism chondria dysfunction (Jha et al. 2000). Mitochondrial dys-
involves inhibition of cystine transport by the cystine- function is a prominent feature in apoptosis, and release of

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
Iptakalim inhibits astrocytic apoptosis 577

pro-apoptotic proteins (e.g., cytochrome c, an apoptosis- and Billett 2006; Yue et al. 2000). To demonstrate the role
inducing factor, and Endo G etc) from the mitochondrial of ERK/MAPK signal pathway in the protect effect of IPT
intermembrane space has been considered to be a critical on MPP+-induced astrocytic apoptosis, ERK1/2 inhibitor
event that occurs during apoptosis (Green and Reed 1998). PD98059 was used in this study. Inhibition of ERK/MAPK
Both cytochrome c and AIF are important for cell viability pathway by PD98059 failed to induce astrocytic apoptosis in
when they are located in mitochondria, but when either is the normal condition, suggesting that this pathway does not
released from the mitochondria, it activates death programs. play a major role in astrocytic survival. However, PD98059
Mitochondrial release of cytochrome c into the cytoplasm could inhibit the anti-apoptotic effect of IPT. Western
induces the formation of an oligomeric complex containing blotting analyses demonstrated that IPT could activate
cytochrome c and Apaf-1. This complex, called the apopto- ERK/MAPK and maintain increased phospho-ERK1/2 level
some, supports the catalytic activation of caspase-9, which after MPP+ exposure, suggesting that ERK/MAPK pathway
further cleaves and activates the effector caspase-3 resulting may be the downstream signal pathway of mitoKATP
in the subsequent degradation of cellular death substrates. channels in astrocytes. These results demonstrate that IPT
Thus, release of cytochrome c is a key step in the initiation of can protect against MPP+-induced astrocytic apoptosis
caspase-dependent apoptosis (Li et al. 1997). AIF induces through activating ERK/MAPK survival signal pathway.
caspase-independent cell death primarily. Following AIF Our results also showed that the selective mitoKCO DZ
translocation from the mitochondria to the nucleus, classic had the similar anti-apoptotic effects to IPT. DZ was
apoptotic features, such as phosphatidylserine exposure, originally developed as an antihypertensive agent, but was
partial chromatin condensation and nuclear condensation, found to induce hyperglycemia by reducing insulin secretion.
occur in the absence of caspase activation. AIF appears to The clinical use of DZ has been hampered by its lack of
play an important role in the acute neurotoxicity induced by potency and selectivity giving rise to side effects (Hansen
trauma, hypoglycemia, transient ischemia, and chronic 2006). In contrast, IPT possesses several advantages, such as
neurodegenerative diseases (Zhang et al. 2002; Zhu et al. free penetration through the blood-brain barrier and low-
2003; Wang et al. 2003). Therefore, therapeutic strategies toxic side-effects during systemic administration (Wu et al.
targeting both caspase-dependent and independent pathways 2005). Therefore, IPT is a promising neuroprotectants for
may be more protective against some toxic insults. The treating neurodegenerative disease.
present study demonstrated that IPT significantly inhibited In conclusion, the present study reveals for the first time
MPP+-induced loss of DYm and subsequent release of pro- that IPT protects against MPP+-induced astrocytic apoptosis
apoptotic protein (cytochrome c and AIF) from mitochondria via inhibition of mitochondria apoptotic pathway and regu-
in primary cultured astrocytes. These findings suggest that lating the MAPK signal transduction pathways (such as JNK,
IPT may protect against astrocytic apoptosis by inhibiting EERK/MAPK) by opening mitoKATP channels in astrocytes.
caspase-dependent and independent apoptosis pathways. These findings indicate that IPT, a novel and blood-brain
JNK is stress-activated MAPK and has been implicated in barrier permeable KCO, is a promising anti-apoptotic agent
various stress-mediated apoptosis such as nerve growth for protection of astrocytes. And targeting mitoKATP channels
factor withdrawal, excitotoxic stress and oxidative stress expressed in astrocytes may offer a novel therapeutic strategy
(Davis 2000; Dickens et al. 1997). And the level of phospho- for neurodegenerative diseases such as PD.
JNK also increases in substantia nigra of MPTP-treated mice
(Saporito et al. 2000). In present study, treatment with MPP+
Acknowledgements
led to a rapid and transient phosphorylation of JNK,
indicating that JNK signaling pathway is activated in These studies were supported in part by grants from the National
response to MPP+ treatment in astrocytes. And IPT could Natural Science Foundation of China (No.30625038 and No.
suppress MPP+-induced increase of phospho-JNK level, 30572172), the Key Project of Natural Science Foundation of
which was also abolished by mitoKATP channel blocker 5- Jiangsu Educational Department (No. 05KJA31014 and No.
06KJA31029), the Key Project of Jiangsu Health Department
HD. Thus, the results in the present study demonstrate that
(No.K200501), and Specialized Research Fund for the Doctoral
IPT may exert anti-apoptotic effect via suppressing mito-
Program of Higher Education of China (No. 20040312004).
chondria and JNK/MAPK apoptotic pathways.
Potential drugs that prevent apoptosis are mainly divided
into two groups: inhibitors of apoptotic signal pathways and References
stimulators of survival signal pathways (Takuma et al. 2004). Amiry-Moghaddam M. and Ottersen O. P. (2003) The molecular basis of
The ERK/MAPK signal pathway is required for survival water transport in the brain. Nat. Rev. Neurosci. 4, 991–1001.
signaling in response to growth factors in non-cardiomyo- Anderson C. M. and Swanson R. A. (2000) Astrocyte glutamate trans-
cytic cells (Parrizas et al. 1997). However, several recent port: review of properties, regulation, and physiological functions.
Glia 32, 1–14.
studies have suggested that the ERK/MAPK signal pathway
participates in the regulation of cell apoptosis (De Girolamo

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
578 S. Zhang et al.

Bajgar R., Seetharaman S., Kowaltowski A. J., Garlid K. D. and Paucek Jenner P. and Olanow C. W. (1998) Understanding cell death in Par-
P. (2001) Identification and properties of a novel intracellular kinson’s disease. Ann. Neurol. 44, 72–84.
(mitochondrial) ATP-sensitive potassium channel in brain. J. Biol. Jha N., Jurma O., Lalli G., Liu Y., Pettus E. H., Greenamyre J. T., Liu R. M.,
Chem. 276, 33369–33374. Forman H. I. and Andersen J. K. (2000) Glutathione depletion in
Benjelloun N., Menard A., Charriaut-Marlangue C., Mokhtari K., Perron PC12 results in selective inhibition of mitochondrial complex I
H., Hauw J. J. and Rieger F. (1998) Case report: DNA fragmen- activity. Implications for Parkinson’s disease. J. Biol. Chem. 275,
tation in glial cells in a cerebral biopsy from a multiple sclerosis 26096–26101.
patient. Cell Mol. Biol. 44, 579–583. Kasischke K. A., Vishwasrao H. D., Fisher P. J., Zipfel W. R. and
Chen Y., Vartiainen N. E., Ying W., Chan P. H., Koistinaho J. and Webb W. W. (2004) Neural activity triggers neuronal oxidative
Swanson R. A. (2001) Astrocytes protect neurons from nitric oxide metabolism followed by astrocytic glycolysis. Science 305, 99–
toxicity by a glutathione-dependent mechanism. J. Neurochem. 77, 103.
1601–1610. Kaul S., Kanthasamy A., Kitazawa M., Anantharam V. and Kanthasamy
Chiueh C. C. and Rauhala P. (1998) Free radicals and MPTP-induced A. G. (2003) Caspase-3 dependent proteolytic activation of protein
selective destruction of substantia nigra compacta neurons. Adv. kinase C delta mediates and regulates 1-methyl-4-phenylpyridi-
Pharmacol. 42, 796–800. nium (MPP+)-induced apoptotic cell death in dopaminergic cells:
Cho Y. and Bannai S. (1990) Uptake of glutamate and cysteine in C-6 relevance to oxidative stress in dopaminergic degeneration. Eur. J.
glioma cells and in cultured astrocytes. J. Neurochem. 55, 2091– Neurosci. 18, 1387–1401.
2097. Kobayashi K., Hayashi M., Nakano H., Fukutani Y., Sasaki K., Shi-
Cho I. H., Im J. Y., Kim D., Kim K. S., Lee J. K. and Han P. L. (2003) mazaki M. and Koshino Y. (2002) Apoptosis of astrocytes with
Protective effects of extracellular glutathione against Zn2+-in- enhanced lysosomal activity and oligodendrocytes in white matter
duced cell death in vitro and in vivo. J. Neurosci. Res. 74, 736– lesions in Alzheimer’s disease. Neuropathol. Appl. Neurobiol. 28,
743. 238–251.
Chun H. S., Gibson G. E., DeGiorgio L. A., Zhang H., Kidd V. J. and Kruger N. J. (1994) The Bradford method for protein quantitation.
Son J. H. (2001) Dopaminergic cell death induced by MPP(+), Methods Mol. Biol. 32, 9–15.
oxidant and specific neurotoxicants shares the common molecular Li P., Nijhawan D., Budihardjo I., Srinivasula S. M., Ahmad M., Al-
mechanism. J. Neurochem. 76, 1010–1021. nemri E. S. and Wang X. (1997) Cytochrome c and dATP-
Davis R. J. (2000) Signal transduction by the JNK group of MAP dependent formation of Apaf-1/caspase-9 complex initiates an
kinases. Cell 103, 239–252. apoptotic protease cascade. Cell 91, 479–489.
De Girolamo L. A. and Billett E. E. (2006) Role of extracellular-regu- Liss B., Haeckel O., Wildmann J., Miki T., Seino S. and Roeper J.
lated kinase and c-Jun NH2-terminal kinase in 1-methyl-4-phenyl- (2005) K-ATP channels promote the differential degeneration of
1,2,3,6-tetrahydropyridine-induced neurofilament phosphorylation. dopaminergic midbrain neurons. Nat. Neurosci. 8, 1742–1751.
J. Neurosci. Res. 83, 680–693. Lotharius J., Falsig J., van Beek J., Payne S., Dringen R., Brundin P. and
Deutch A. Y. and Winder D. G. (2006) A channel to neurodegeneration. Leist M. (2005) Progressive degeneration of human mesencephalic
Nat. Med. 12, 17–18. neuron-derived cells triggered by dopamine-dependent oxidative
Dickens M., Rogers J. S., Cavanagh J., Raitano A., Xia Z., Halpern J. R., stress is dependent on the mixed-lineage kinase pathway. J. Neu-
Greenberg M. E., Sawyers C. L. and Davis R. J. (1997) A cyto- rosci. 25, 6329–6342.
plasmic inhibitor of the JNK signal transduction pathway. Science Makarov P., Kropf S., Wiswedel I., Augustin W. and Schild L. (2006)
277, 693–696. Consumption of redox energy by glutathione metabolism contri-
Dunn-Meynell A. A., Rawson N. E. and Levin B. E. (1998) Distribution butes to hypoxia/ reoxygenation-induced injury in astrocytes. Mol.
and phenotype of neurons containing the ATP-sensitive K+ chan- Cell. Biochem. 286, 95–101.
nel in rat brain. Brain Res. 814, 41–54. Mazzanti M., Sul J. Y. and Haydon P. G. (2001) Glutamate on demand:
Franz G., Beer R., Intemann D. et al. (2002) Temporal and spatial profile astrocytes as a ready source. Neuroscientist 7, 396–405.
of Bid cleavage after experimental traumatic brain injury. J. Cerebr. McNaught K. St. P. and Jenner P. (2000) Extracellular accumulation of
Blood F. Met. 22, 951–958. nitric oxide, hydrogen peroxide, and glutamate in astrocytic cul-
Green D. R. and Reed J. C. (1998) Mitochondria and apoptosis. Science tures following glutathione depletion, complex I inhibition, and/or
281, 1309–1312. lipopolysaccharide-induced activation. Biochem. Pharmacol. 60,
Hall A. G. (1999) Review: the role of glutathione in the regulation of 979–988.
apoptosis. Eur. J. Clin. Invest. 29, 238–245. Nakagawa I., Alessandri B., Heimann A. and Kempski O. (2005)
Hamprecht B. and Loffler F. (1985) Primary glial cultures as a model for MitoKATP-channel opener protects against neuronal death in rat
studying hormone action. Methods Enzymol. 109, 341–345. venous ischemia. Neurosurgery 57, 334–340.
Hansen J. B. (2006) Towards selective Kir6.2/SUR1 potassium channel Parrizas M., Saltiel A. R. and LeRoith D. (1997) Insulin-like growth
openers, medicinal chemistry and therapeutic perspectives. Curr. factor 1 inhibits apoptosis using the phosphatidylinositol 3¢-kinase
Med. Chem. 13, 361–376. and mitogen-activated protein kinase pathways. J. Biol. Chem. 272,
Henze C., Hartmann A., Lescot T., Hirsch E. C. and Michel P. P. 154–161.
(2005) Proliferation of microglial cells induced by 1-methyl-4- Sagara J. I., Miura K. and Bannai S. (1993) Maintenance of neuronal
phenylpyridinium in mesencephalic cultures results from an glutathione by glial cells. J. Neurochem. 61, 1672–1676.
astrocyte-dependent mechanism: role of granulocyte macrophage Saporito M. S., Thomas B. A. and Scott R. W. (2000) MPTP activates
colony-stimulating factor. J. Neurochem. 95, 1069–1077. c-Jun NH(2)-terminal kinase (JNK) and its upstream regulatory
Hu L. F., Wang S., Shi X. R., Yao H. H., Sun Y. H., Ding J. H. and Hu G. kinase MKK4 in nigrostriatal neurons in vivo. J. Neurochem. 75,
(2005) ATP-sensitive potassium channel opener iptakalim protec- 1200–1208.
ted against the cytotoxicity of MPP+ on SH-SY5Y cells by Seifert G., Schilling K. and Steinhauser C. (2006) Astrocyte dysfunction
decreasing extracellular glutamate level. J. Neurochem. 94, 1570– in neurological disorders: a molecular perspective. Nat. Rev. Neu-
1579. rosci. 7, 194–206.

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579
Iptakalim inhibits astrocytic apoptosis 579

Seino S. and Miki T. (2003) Physiological and pathophysiological roles of iptakalim on extracellular neurotransmitter levels in the striatum
ATP-sensitive K+ channels. Prog. Biophys. Mol. Biol. 81, 133–176. of unilateral 6-hydroxydopamine-lesioned rats. Neuropsycho-
Simard M. and Nedergaard M. (2004) The neurobiology of glia in the pharmacology 31, 933–940.
context of water and ion homeostasis. Neuroscience 129, 877–896. Wu J., Wakui M., Wang H. and Hu G. (2005) Iptakalim hydrochloride
Szydlowska K., Zawadzka M. and Kaminska B. (2006) Neuroprotectant and neuronal protection. Current Neuropharmacology 3, 1–9.
FK506 inhibits glutamate-induced apoptosis of astrocytes in vitro Wu J., Hu J., Chen Y. P. et al. (2006) Iptakalim modulates ATP-sensitive
and in vivo. J. Neurochem. 99, 965–975. K(+) channels in dopamine neurons from rat substantia nigra pars
Tai K. K., McCrossan Z. A. and Abbott G. W. (2003) Activation of mito- compacta. J. Pharmacol. Exp. Ther. 319, 155–164.
chondrial ATP-sensitive potassium channels increases cell viability Xie W., Wang H., Ding J., Wang H. and Hu G. (2005) Anti-proliferating
against rotenone-induced cell death. J. Neurochem. 84, 1193–1200. effect of iptakalim, a novel KATP channel opener, in cultured
Takano T., Tian G. F., Peng W., Lou N., Libionka W., Han X. and rabbit pulmonary arterial smooth muscle cells. Eur. J. Pharmacol.
Nedergaard M. (2006) Astrocyte-mediated control of cerebral 511, 81–87.
blood flow. Nat. Neurosci. 9, 260–267. Yang Y., Liu X., Ding J. H., Sun J., Long Y., Wang F., Yao H. H. and Hu G.
Takuma K., Baba A. and Matsuda T. (2004) Astrocyte apoptosis: (2004) Effects of iptakalim on rotenone-induced cytotoxicity and
implications for neuroprotection. Prog. Neurobiol. 72, 111–127. dopamine release from PC12 cells. Neurosci. Lett. 366, 53–57.
Thomzig A., Wenzel M., Karschin C., Eaton M. J., Skatchkov S. N., Yang Y., Liu X., Long Y. et al. (2005) Systematic administration of
Karschin A. and Veh R. W. (2001) Kir6.1 is the principal pore- iptakalim, an ATP-sensitive potassium channel opener, prevents
forming subunit of astrocyte but not neuronal plasma membrane rotenone-induced motor and neurochemical alterations in rats.
K-ATP channels. Mol. Cell. Neurosci. 18, 671–690. J. Neurosci. Res. 80, 442–449.
Tietze F. (1969) Enzymic method for quantitative determination of Yang J., Hu L. F., Liu X., Zhou F., Ding J. H. and Hu G. (2006a) Effects
nanogram amounts of total and oxidized glutathione applications to of iptakalim on extracellular glutamate and dopamine levels in the
mammalian blood and other tissues. Anal. Biochem. 27, 502–522. striatum of unilateral 6-hydroxydopamine-lesioned rats: a micro-
Tipton K. F. and Singer T. P. (1993) Advances in our understanding of dialysis study. Life Sci. 78, 1940–1944.
the mechanisms of the neurotoxicity of MPTP and related com- Yang Y., Liu X., Long Y. et al. (2006b) Activation of mitochondrial
pounds. J. Neurochem. 61, 1191–1206. ATP-sensitive potassium channels improves rotenone-related
Tripanichkul W., Sripanichkulchai K. and Finkelstein D. I. (2006) Es- motor and neurochemical alterations in rats. Int. J. Neuropsycho-
trogen down-regulates glial activation in male mice following pharmacology 9, 51–61.
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxication. Brain Yue T. L., Wang C., Gu J. L., Ma X. L., Kumar S., Lee J. C., Feuerstein
Res. 1084, 28–37. G. Z., Thomas H., Maleeff B. and Ohlstein E. H. (2000) Inhibition
Wang H. (2003) Pharmacological characteristics of the novel antihy- of extracellular signal-regulated kinase enhances Ischemia/Reox-
pertensive drug, iptakalim hydrochloride, and its molecular ygenation-induced apoptosis in cultured cardiac myocytes and
mechanisms. Drug Develop. Res. 58, 65–68. exaggerates reperfusion injury in isolated perfused heart. Circ. Res.
Wang X., Zhu S., Drozda M., Zhang W., Stavrovskaya I. G., Cattaneo E., 86, 692–699.
Ferrante R. J., Kristal B. S. and Friedlander R. M. (2003) Mino- Zawar C., Plant T. D., Schirra C., Konnerth A. and Neumcke B. (1999)
cycline inhibits caspase-independent and -dependent mitochondrial Cell-type specific expression of ATP-sensitive potassium channels
cell death pathways in models of Huntington’s disease. Proc. Natl in the rat hippocampus. J. Physiol. 514, 327–341.
Acad. Sci. USA 100, 10483–10487. Zhang Y. and Hu G. (2004) Iptakalim enhances astrocytic glutamate
Wang H., Zhang Y. L., Tang X. C., Feng H. S. and Hu G. (2004) uptake activity. Acta Pharmaceutica Sinica 39, 980–983.
Targeting ischemic stroke with a novel opener of ATP-sensitive Zhang X., Chen J., Graham S. H. et al. (2002) Intranuclear localization
potassium channels in the brain. Mol. Pharmacol. 66, 1160–1168. of apoptosis-inducing factor (AIF) and large scale DNA frag-
Wang S., Hu L. F., Yang Y., Ding J. H. and Hu G. (2005) Studies of mentation after traumatic brain injury in rats and in neuronal cul-
ATP-sensitive potassium channels on 6-hydroxydopamine and tures exposed to peroxynitrite. J. Neurochem. 82, 181–191.
haloperidol rat models of Parkinson’s disease implications for Zhu C., Qiu L., Wang X., Hallin U., Cande C., Kroemer G., Hagberg H.
treating Parkinson’s disease? Neuropharmacology 48, 984–992. and Blomgren K. (2003) Involvement of apoptosis-inducing factor
Wang S., Hu L. F., Zhang Y., Sun T., Sun Y. H., Liu S. Y., Ding J. H., in neuronal death after hypoxia-ischemia in the neonatal rat brain.
Wu J. and Hu G. (2006) Effects of systemic administration of J. Neurochem. 86, 306–317.

 2007 The Authors


Journal Compilation  2007 International Society for Neurochemistry, J. Neurochem. (2007) 103, 569–579

You might also like