14
ORIGINAL PAPER Valproic Acid Induced Differentiation and Potentiated Efficacy of Taxol and Nanotaxol for Controlling Growth of Human Glioblastoma LN18 and T98G Cells Subhasree Roy Choudhury Surajit Karmakar Naren L. Banik Swapan K. Ray Received: 29 April 2011 / Revised: 11 July 2011 / Accepted: 14 July 2011 / Published online: 24 July 2011 Ó Springer Science+Business Media, LLC 2011 Abstract Glioblastoma shows poor response to current therapies and warrants new therapeutic strategies. We examined the efficacy of combination of valproic acid (VPA) and taxol (TX) or nanotaxol (NTX) in human glioblastoma LN18 and T98G cell lines. Cell differentia- tion was manifested in changes in morphological features and biochemical markers. Cell growth was controlled with down regulation of vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR), nuclear factor-kappa B (NF-jB), phospho-Akt (p-Akt), and multi- drug resistance (MDR) marker, indicating suppression of angiogenic, survival, and multi-drug resistance pathways. Cell cycle analysis showed that combination therapy (VPA and TX or NTX) increased the apoptotic sub G1 population and apoptosis was further confirmed by Annexin V-FITC/ PI binding assay and scanning electron microscopy. Combination therapy caused activation of caspase-8 and cleavage of Bid to tBid and increased Bax:Bcl-2 ratio and mitochondrial release of cytochrome c and apoptosis- inducing factor (AIF). Upregulation of calpain and casp- ases (caspase-9 and caspase-3) and substrate degradation were also detected in course of apoptosis. The combination of VPA and NTX most effectively controlled the growth of LN18 and T98G cells. Therefore, this combination of drugs can be used as an effective treatment for controlling growth of human glioblastoma cells. Keywords Apoptosis Á Glioblastoma Á Nanotaxol Á Taxol Á Valproic acid Introduction Glioblastoma is the most common and deadliest malignancy, accounting for nearly 40% of primary brain tumors in adults with a mean survival of less than 1 year following diagnosis [1]. It is a heterogenous and highly vascularized brain tumor characterized by poorly differentiated neoplastic astrocytes that avoid apoptosis due to P-glycoprotein (P-gp) mediated multi-drug resistance [2]. Development of new therapeutic strategies that induce differentiation and apoptosis, provide anti-angiogenic activity, and overcome multi-drug resis- tance is urgently warranted. Valproic acid (VPA), a histone deacetylase inhibitor, has been shown to induce differentiation and anti-angio- genic and anti-proliferative activities in several cancers, including both P-gp positive and negative cell lines [3]. The multimodal efficacy of VPA with other anti-cancer drugs could be useful for successful treatment of glio- blastoma. While the anti-cancer efficacy of taxol (TX) is well established in ovarian, head-and-neck, bladder, breast, and lung cancers [4], recently TX has also been shown to provide potent therapeutic effects in experimental brain tumor [5, 6]. Because of its low aqueous solubility, TX is less per- meable to the blood–brain-barrier (BBB) [7]. Besides, the commercial formulation of 6 mg/ml (7 mM) TX in 1:1 mixture of the solvents Cremophor EL Ò (CrEL, poly- oxyethylated castor oil) and ethanol has systemic toxicity, side effects, and short time stability, and is associated with hypersensitivity reactions [8]. TX is a substrate for the P-gp S. Roy Choudhury Á S. Karmakar Á S. K. Ray (&) Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA e-mail: [email protected] N. L. Banik Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA 123 Neurochem Res (2011) 36:2292–2305 DOI 10.1007/s11064-011-0554-7

Valproic Acid Induced Differentiation and Potentiated

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Valproic Acid Induced Differentiation and Potentiated

ORIGINAL PAPER

Valproic Acid Induced Differentiation and Potentiated Efficacyof Taxol and Nanotaxol for Controlling Growth of HumanGlioblastoma LN18 and T98G Cells

Subhasree Roy Choudhury • Surajit Karmakar •

Naren L. Banik • Swapan K. Ray

Received: 29 April 2011 / Revised: 11 July 2011 / Accepted: 14 July 2011 / Published online: 24 July 2011

� Springer Science+Business Media, LLC 2011

Abstract Glioblastoma shows poor response to current

therapies and warrants new therapeutic strategies. We

examined the efficacy of combination of valproic acid

(VPA) and taxol (TX) or nanotaxol (NTX) in human

glioblastoma LN18 and T98G cell lines. Cell differentia-

tion was manifested in changes in morphological features

and biochemical markers. Cell growth was controlled with

down regulation of vascular endothelial growth factor

(VEGF), epidermal growth factor receptor (EGFR), nuclear

factor-kappa B (NF-jB), phospho-Akt (p-Akt), and multi-

drug resistance (MDR) marker, indicating suppression of

angiogenic, survival, and multi-drug resistance pathways.

Cell cycle analysis showed that combination therapy (VPA

and TX or NTX) increased the apoptotic sub G1 population

and apoptosis was further confirmed by Annexin V-FITC/

PI binding assay and scanning electron microscopy.

Combination therapy caused activation of caspase-8 and

cleavage of Bid to tBid and increased Bax:Bcl-2 ratio and

mitochondrial release of cytochrome c and apoptosis-

inducing factor (AIF). Upregulation of calpain and casp-

ases (caspase-9 and caspase-3) and substrate degradation

were also detected in course of apoptosis. The combination

of VPA and NTX most effectively controlled the growth of

LN18 and T98G cells. Therefore, this combination of drugs

can be used as an effective treatment for controlling growth

of human glioblastoma cells.

Keywords Apoptosis � Glioblastoma � Nanotaxol �Taxol � Valproic acid

Introduction

Glioblastoma is the most common and deadliest malignancy,

accounting for nearly 40% of primary brain tumors in adults

with a mean survival of less than 1 year following diagnosis

[1]. It is a heterogenous and highly vascularized brain tumor

characterized by poorly differentiated neoplastic astrocytes

that avoid apoptosis due to P-glycoprotein (P-gp) mediated

multi-drug resistance [2]. Development of new therapeutic

strategies that induce differentiation and apoptosis, provide

anti-angiogenic activity, and overcome multi-drug resis-

tance is urgently warranted.

Valproic acid (VPA), a histone deacetylase inhibitor,

has been shown to induce differentiation and anti-angio-

genic and anti-proliferative activities in several cancers,

including both P-gp positive and negative cell lines [3].

The multimodal efficacy of VPA with other anti-cancer

drugs could be useful for successful treatment of glio-

blastoma. While the anti-cancer efficacy of taxol (TX) is

well established in ovarian, head-and-neck, bladder, breast,

and lung cancers [4], recently TX has also been shown to

provide potent therapeutic effects in experimental brain

tumor [5, 6].

Because of its low aqueous solubility, TX is less per-

meable to the blood–brain-barrier (BBB) [7]. Besides, the

commercial formulation of 6 mg/ml (7 mM) TX in 1:1

mixture of the solvents Cremophor EL� (CrEL, poly-

oxyethylated castor oil) and ethanol has systemic toxicity,

side effects, and short time stability, and is associated with

hypersensitivity reactions [8]. TX is a substrate for the P-gp

S. Roy Choudhury � S. Karmakar � S. K. Ray (&)

Department of Pathology, Microbiology, and Immunology,

University of South Carolina School of Medicine, Columbia,

SC 29209, USA

e-mail: [email protected]

N. L. Banik

Department of Neurosciences, Medical University

of South Carolina, Charleston, SC 29425, USA

123

Neurochem Res (2011) 36:2292–2305

DOI 10.1007/s11064-011-0554-7

Page 2: Valproic Acid Induced Differentiation and Potentiated

[9] and co-administration of the P-gp inhibitor PSC833

[10] or HM30181A [11] with TX has shown significant

therapeutic efficacy in brain tumor.

Many attempts have been made to reformulate TX-

bound nanoparticles, which are natural or synthetic poly-

mers of lipids or metal containing vehicles of dimension

\100 nm and hold promise as the effective drug delivery

carriers across the BBB [12]. ABI-007, also known as

‘Abraxane’, is a Cremophor-free, albumin-bound 130-nm

nanoparticle form of TX that has shown anti-cancer

activity in human lung (H522), breast (MX-1), ovarian

(SK-OV-3), prostate (PC-3), and colon (HT29) tumor xe-

nografts [13]. The delivery of TX as Abraxane has been

found to overcome insoluble nature and solvent related

toxicities of TX [14]. The clinical potency of Abraxane in

metastatic breast cancer [15] and advanced non-small cell

lung cancer patients is well established [14], but the ther-

apeutic potency of Abraxane or nanotaxol (NTX) in glio-

blastoma has not yet been explored.

In this study, we investigated the anti-tumor efficacy of

VPA, TX, or NTX alone and in combination in multi-drug

resistant glioblastoma LN18 (with overexpression of P-gp)

and T98G (with less expression of P-gp) cell lines [2, 16–

19]. The combination of VPA and TX or NTX was found

to markedly down regulate several survival signals and

angiogenesis markers, induce differentiation, and activate

mitochondria-dependent pathways for apoptosis in both

LN18 and T98G cell lines.

Materials and Methods

Reagents

VPA and primary b-actin IgG antibody (monoclonal clone

AC-15) were obtained from Sigma Chemical (St. Louis,

MO). TX (paclitaxel) was procured from Bristol-Myers

Squibb (Princeton, NJ). NTX (Abraxane) was obtained

from Abraxis Oncology (Schaumburg, IL). The colori-

metric MTT assay kit was procured from Millipore

Chemicon (Temecula, CA). AnnexinV-fluorescein isothi-

ocyanate (FITC)/propidium iodide (PI) kit for detection of

apoptosis was purchased from BD Biosciences (San Jose,

CA). Wright staining kit was bought from Fisher Scientific

(Middletown, VA). Other primary IgG antibodies were

obtained from Santa Cruz Biotechnology (Santa Cruz, CA).

Depending on the primary antibody, the secondary anti-

body used was either horseradish peroxidase (HRP)-con-

jugated goat anti-mouse IgG or HRP-conjugated goat anti-

rabbit IgG (ICN Biomedicals, Aurora, OH). Enhanced

Chemiluminescence (ECL) Plus reagent was purchased

from GE Healthcare Bio-Sciences (Piscataway, NJ).

Cell Culture and Treatments

Human glioblastoma LN18 and T98G cell lines were

purchased from the American Type Culture Collection

(ATCC, Manassas, VA). Both LN18 and T98G cells

were separately grown in monolayer to sub-confluency in

75-cm2 flasks containing 10 ml of DMEM and RPMI

1640 (Mediatech, Herndon, VA), respectively, and sup-

plemented with 10% fetal bovine serum (FBS) (Invitro-

gen, Carlsbad, CA), 1% penicillin, and 1% streptomycin

(GIBCO, Grand Island, NY) in a fully-humidified incu-

bator containing 5% CO2 and 95% air at 37�C. Cells

were allowed to starve for 24 h in RPMI 1640 or

DMEM with 2% FBS, 1% penicillin, and 1% strepto-

mycin and thereafter maintained in this low-serum con-

dition during the treatments. Freshly prepared stock

solutions of VPA, TX, and NTX were made in serum-

free medium just prior to treatment. Dose–response

studies were carried out to determine the suitable doses

of the drugs for the inhibition of cell growth, induction

of differentiation, and cell death. Cells were treated with

0.5, 1, 2, or 4 mM of VPA for 72 h followed by treat-

ment with either 50 nM TX or 50 nM NTX for 24 h

alone and in combination.

MTT Assay

The differential sensitivities of LN18 and T98G cells to

VPA, TX, or NTX either alone or in combination were

determined using the 3-[4,5-dimethylthiazole-2-yl]-2,5-

diphenyltetrazolium bromide (MTT)-based colorimetric

assay. We used different doses of VPA to induce differ-

entiation whereas a fix dose of either TX or NTX to induce

apoptosis. The rationale was to reduce cell proliferation

with induction of differentiation and increase apoptosis

using combination of drugs in glioblastoma cells. For MTT

assay, both cell lines were first exposed to 0.5, 1, 2, and

4 mM VPA for 72 h. At 48 h post-exposure of VPA, cells

were treated with 50 nM TX or NTX for another 24 h

alone and in combination with VPA (total treatment for

72 h). Cells were then exposed to MTT reagent for 4 h at

37�C after which isopropanol was used to dissolve the

formazan crystals. The optical density (OD) was recorded

at 570 nm in a microplate reader. Percent growth inhibition

was determined as [1-(OD of treated cells/OD of control

cells)]9 100. All experiments were performed in tripli-

cates. Cell viability data were analyzed using Compusyn

software (ComboSyn, Paramus, NJ) to generate a combi-

nation index (CI). Conventionally, CI [ 1 indicates

antagonism, CI = 1 indicates additive effect, and CI \ 1

indicates synergism [20].

Neurochem Res (2011) 36:2292–2305 2293

123

Page 3: Valproic Acid Induced Differentiation and Potentiated

Wright Staining for Examination of Morphological

Features of Apoptosis

Both LN18 and T98G cells were treated with 2 mM VPA

for 72 h as monotherapy and at 48 h post-exposure of

VPA, 50 nM TX or NTX was added alone separately or in

combination with VPA for another 24 h (total treatment for

72 h). The cells (adherent and non-adherent) from each

treatment were centrifuged and sedimented on the culture

dish and washed twice with phosphate-buffered saline

(PBS, pH 7.4). The cells were fixed with 95% (v/v) ethanol

and dried before Wright staining. The morphology of

apoptotic cells were detected under the light microscope.

Cells were considered apoptotic with characteristic features

such as chromatin condensation, cell-volume shrinkage,

and membrane-bound apoptotic bodies [18].

Cell Cycle Analysis

LN18 and T98G cells were seeded in a 6-well plate at a

density of 1 9 105 cells per well and incubated for 24 h in

low-serum medium prior to addition of drugs. Cells were

untreated (control, CTL) and treated with 2 mM VPA for

72 h as monotherapy and at 48 h post-exposure of VPA,

50 nM TX or NTX was added alone separately or in

combination with VPA for another 24 h (total treatment for

72 h). Cells were trypsinized and harvested by centrifu-

gation. Flow cytometric analysis of cell cycle was per-

formed by staining of permeabilized cells with PI for DNA

content. Cells were re-suspended in PBS, fixed with 70%

ethanol, labeled with staining solution (0.05 mg/ml PI,

2 mg/ml RNase A, 0.1% TritonX-100 in PBS), and incu-

bated for 30 min at room temperature (RT) in darkness.

DNA content was then analyzed using an Epics XL-MCL

Flow Cytometer (Beckman Coulter, Fullerton, CA), as we

reported recently [21].

Flow Cytometric Analysis of Apoptosis

The percentage of cells undergoing apoptosis was quanta-

tively determined by Annexin V-FITC/PI binding assay.

LN18 and T98G cells were seeded in 6-well plate at a

density of 1 9 105 cells per well and incubated for 24 h in

low-serum medium prior to addition of drugs. Cells were

untreated (CTL) and treated with 2 mM VPA for 72 h as

monotherapy and at 48 h post-exposure of VPA, 50 nM

TX or NTX was added alone separately or in combination

with VPA for another 24 h (total treatment for 72 h). After

treatments, both adherent and non-adherent cells were

harvested and double-stained using the Annexin V-FITC/PI

staining kit according to the manufacturer’s protocol. Cells

were washed with cold PBS, resuspended in 19 binding

buffer (0.1 M HEPES/NaOH, pH 7.4, 1.4 M NaCl, 25 mM

CaCl2), stained with Annexin V-FITC/PI, and incubated

for 15 min at RT in the darkness. Cells were analyzed for

Annexin V-stained apoptotic population using an Epics

XL-MCL Flow Cytometer (Beckman Coulter, Fullerton,

CA), as per our previous reports [20, 21].

Morphological Analysis by Scanning Electron

Microscopy

Both LN18 and T98G cells were grown to sub-confluency

on 13-mm round cover slip and incubated in low-serum

medium for 24 h before addition of drugs. Cells were either

untreated (CTL) and treated with 2 mM VPA for 72 h as

monotherapy and at 48 h post-exposure of VPA, 50 nM

TX or NTX was added alone separately or in combination

with VPA for another 24 h (total treatment for 72 h). After

the treatments, cells were fixed with 2.5% glutaraldehyde

in 4% paraformaldehyde in phosphate buffer (pH 7.4) for

1 h at RT. Cells were post-fixed in 1.0% osmium tetroxide

(OsO4) for 10 min and washed in 0.1 M phosphate buffer

(pH 7.2). Dehydration was done in ascending concentration

of ethanol. Cover slips were dried with the critical point

apparatus using liquid CO2 as transition fluid. The speci-

mens were cold sputter coated with gold and observed in a

JEOL 6,300 V Scanning Electron Microscope (SEM) at an

accelerating voltage of 15 kV, as we reported earlier [21].

Protein Extraction and Western Blotting

For Western blotting, total proteins were extracted fol-

lowing homogenization of cells, quantitated spectrophoto-

metrically, denatured in boiling water for 5 min, and

resolved by SDS-polyacryl amide gel electrophoresis and

then electroblotted to the polyvinylidene (PVDF) mem-

branes. The blots were incubated with a primary IgG

antibody followed by incubation with an alkaline HRP-

conjuaged secondary IgG antibody. Subsequently, specific

protein bands were detected by HRP/H2O2-catalyzed oxi-

dation of luminol in alkaline conditions using the ECL

system and autoradiography [18, 22].

Statistical Analysis

Results were analyzed using Minitab� 15 Statistical Soft-

ware (Minitab, State College, PA). Data were expressed as

mean ± standard error of mean (SEM) of separate exper-

iments (n C 3) and compared by one-way analysis of

variance (ANOVA) followed by Fisher’s post hoc test.

Significant difference between CTL and a treatment was

indicated by *P \ 0.05, or **P \ 0.001; and between TX

and VPA ? TX, or NTX and VPA ? NTX was indicated

by #P \ 0.05.

2294 Neurochem Res (2011) 36:2292–2305

123

Page 4: Valproic Acid Induced Differentiation and Potentiated

Results

Treatments Decreased Cell Viability

The growth inhibitory effect of VPA, TX, and NTX either

alone or in combination on LN18 and T98G cells were

determined (Fig. 1). VPA monotherapy was used at dif-

ferent concentrations such as 0.5, 1, 2, and 4 mM for 72 h

while 50 nM TX or 50 nM NTX was used alone for 24 h.

For combination studies, the anti-proliferative effect was

determined by treating both cell lines with VPA (0.5, 1, 2,

and 4 mM) for 48 h and then either 50 nM TX or 50 nM

NTX was added in presence of VPA for another 24 h (total

treatment for 72 h). VPA inhibited cell growth in both cell

lines (Fig. 1a). Treatment with VPA dose-dependently

decreased the cell viability in LN18 and T98G cells. The

percentages of cell viability in LN18 and T98G cells after

treatment with 50 nM TX or NTX for 24 h were also

decreased (Fig. 1a). Most importantly, treatment with

VPA ? TX, or VPA ? NTX showed significant reduction

in cell viability when compared with TX or NTX mono-

therapy (Fig. 1a).

We used a variety of doses of VPA but a fixed dose of

either TX or NTX and found that higher doses of combi-

nation of drugs showed more cell death than lower doses.

But our aim was to use particular doses of the drugs, which

would induce cellular differentiation, inhibit cell prolifer-

ation, and arrest cell cycle to trigger apoptosis. VPA

(2 mM) and TX or NTX (50 nM) alone showed the best

efficacy, VPA (2 mM) ? TX (50 nM) produced the best

synergistic efficacy (CI = 0.791 in LN18 cells and

CI = 0.98 in T98G cells), and VPA (2 mM) ? NTX (50

nM) also showed the best synergistic efficacy (CI = 0.45

in LN18 cells and CI = 0.49 in T98G cells) (Fig. 1a) and

therefore, these treatments were finally selected for further

studies for inducing differentiation and apoptosis.

Fig. 1 VPA, TX, and NTX alone and combination therapy decreased

cell viability and increased apoptotic features in LN18 and T98G

cells. Treatments: 0 control (CTL), 1 CTL (untreated) or monotherapy

(0.5 mM VPA, and 50 nM TX or NTX) or combination therapy

(0.5 mM VPA followed by either 50 nM TX or 50 nM NTX), 2 CTL

(untreated) or monotherapy (1 mM VPA, and either 50 nM TX or

50 nM NTX) or combination therapy (1 mM VPA followed by either

50 nM TX or 50 nM NTX), 3 CTL (untreated) or monotherapy

(2 mM VPA, and either 50 nM TX or 50 nM NTX) or combination

therapy (2 mM VPA followed by either 50 nM TX or 50 nM NTX),

and 4 CTL (untreated) or monotherapy (4 mM VPA, and either

50 nM TX or 50 nM NTX) or combination therapy (4 mM VPA

followed by either 50 nM TX or 50 nM NTX). Treatment schedule

with monotherapy: VPA for 72 h, TX or NTX for 24 h; and with

combination therapy: VPA for 48 h and then TX or NTX in presence

of VPA for 24 h (total treatment for 72 h). a MTT assay was used to

assess cell viability following monotherapy and combination therapy.

b Wright staining to evaluate the morphological features of differ-

entiation and apoptosis

Neurochem Res (2011) 36:2292–2305 2295

123

Page 5: Valproic Acid Induced Differentiation and Potentiated

Induction of Morphological and Biochemical Features

of Differentiation and Apoptosis

Wright staining revealed that exposure to VPA inhibited cell

proliferation and induced morphological features of differ-

entiation such as long and thin projections of the cells

(Fig. 1b). Western blotting showed the biochemical features

of astrocytic differentiation with upregulation of the glial

fibrillary acidic protein (GFAP) and down regulation of the

inhibitor of differentiation 2 (ID2) in both LN18 and T98G

cells (Fig. 2a, c, d). Uniform b-actin expression served as an

internal control. Increase in GFAP, an astrocyte-specific

intermediate filament protein, is a biochemical marker of

differentiation in glioblastomas. The treatment with VPA ?

TX caused upregulation of GFAP expression in T98G cells

only whereas VPA ? NTX significantly increased GFAP

expression in both LN18 and T98G cells, when compared

with control or monotherapy (Fig. 2a, c, d).

Inhibition of Angiogenic, Survival, and Multi-Drug

Resistance Markers

Vascular endothelial growth factor (VEGF) is a major

regulatory factor in tumor angiogenesis and predominantly

Fig. 2 Combination therapy

(either VPA ? TX or

VPA ? NTX) induced

differentiation and inhibited

angiogenic, survival, and multi-

drug resistance factors.

Treatments: control (CTL);

2 mM of VPA, 50 nM TX,

50 nM NTX, VPA

(2 mM) ? TX (50 nM), VPA

(2 mM) ? NTX (50 nM).

Treatment schedule with

monotherapy: VPA for 72 h,

TX or NTX for 24 h; and with

combination therapy: VPA for

48 h and then TX or NTX in

presence of VPA for 24 h (total

treatment for 72 h). a Western

blotting showed uniform level

of 42 kD b-actin, upregulation

of glial fibrillary acidic protein

(GFAP), and down regulation of

inhibitor of differentiation

factor 2 (ID2) following

combination therapy.

b Representative Western blots

showed levels of VEGF, EGFR,

NF-jB, total Akt, p-Akt, and

MDR (P-gp). c Densitometric

analysis of percent change in

protein levels in LN18 cells.

d Densitometric analysis of

percent change in protein levels

in T98G cells. Combination

therapy inhibited angiogenic,

survival, and multi-drug

resistance markers

2296 Neurochem Res (2011) 36:2292–2305

123

Page 6: Valproic Acid Induced Differentiation and Potentiated

overexpressed in glioblastoma [23]. Western blotting was

used to examine any effect of the single or combinatorial

treatment on VEGF expression. Both single and combina-

tion therapies were found to potently inhibit VEGF

expression in both LN18 and T98G cells (Fig. 2b–d).

Another genetic alteration associated with glioblastoma

is amplification of epidermal growth factor receptor

(EGFR) gene, resulting in overexpression of EGFR protein.

EGFR amplification has been shown to increase glioblas-

toma proliferation and invasion [24]. We assessed expres-

sion of EGFR by Western blotting. Use of VPA ? TX was

found to significantly reduce EGFR expression whereas

VPA ? NTX totally abolished EGFR expression in both

LN18 and T98G cell lines (Fig. 2b), compared with all

other treatments (Fig. 2c, d).

The levels of several anti-apoptotic proteins were

determined by Western blotting (Fig. 2b). Treatment with

the combination of drugs (either VPA ? TX or VPA ?

NTX) showed dramatic down regulation of the survival

factors such as NF-jB, total-Akt, and p-Akt in both LN18

and T98G cell lines (Fig. 2b). Treatment with VPA ?

NTX most effectively inhibited expression of the survival

factors (e.g., NF-jB, total-Akt, and p-Akt) in both cell lines

(Fig. 2c, d).

Untreated LN18 cells (harboring overexpression of

P-gp) showed high level of P-gp expression. Treatment

with 2 mM VPA alone and especially combination of drugs

(either VPA ? TX or VPA ? NTX) remarkably down

regulated the P-gp expression in LN18 cell line (Fig. 2b, c).

No detectable level of P-gp was observed in untreated

T98G cells (harboring very little expression of P-gp). Use

of VPA ? NTX was more effective than that of

VPA ? TX in down regulating MDR expression.

Flow Cytometric Analysis of Cell Cycle

Flow cytometric analysis was performed to examine the

cell cycle distribution patterns in LN18 and T98G cells

following treatments (Fig. 3). Flow cytometric data

revealed that treatment with VPA caused a small increase

in sub G1 phase, a decrease in G0/G1 phase, and an

increase in G2/M population in both cell lines. Treatment

with TX showed a significant increase in sub G1 phase and

down regulated G0/G1 population with a significant

increase in G2/M phase in both LN18 and T98G cell lines.

A significant increase in sub G1 phase and a decrease in

both G0/G1 and G2/M populations in both cell lines

(Fig. 3a) were seen due to treatment with NTX. Combi-

nation therapy (either VPA ? TX or VPA ? NTX) most

effectively increased the sub G1 apoptotic phase. Notably,

the use of VPA ? NTX caused more apoptosis than the use

of VPA ? TX in both cell lines (Fig. 3b).

Fig. 3 Cell cycle analysis by

flow cytometry. Histograms

display DNA content (x axis, PI

fluorescence) versus counts

(y axis). Treatments: control

(CTL); 2 mM VPA, 50 nM TX,

50 nM NTX, VPA

(2 mM) ? TX (50 nM), VPA

(2 mM) ? NTX (50 nM).

Treatment schedule with

monotherapy: VPA for 72 h,

TX or NTX for 24 h; and with

combination therapy: VPA for

48 h and then TX or NTX in

presence of VPA for 24 h (total

treatment for 72 h). a Cell cycle

phase distribution was shown by

histograms. b Bar diagram

representation of cells in sub G1

phase

Neurochem Res (2011) 36:2292–2305 2297

123

Page 7: Valproic Acid Induced Differentiation and Potentiated

Flow Cytometric Detection of Apoptosis by Annexin

V-FITC/PI Binding Assay

Annexin V-FITC/PI double-labeling technique was used to

understand the nature of cell death (apoptotic or necrotic)

after the treatments (Fig. 4). Monotherapy with VPA, TX,

and NTX induced apoptosis in LN18 and T98G cells

(Fig. 4a). But the combination therapy showed a significant

increase in apoptosis, when compared with either treatment

alone. Flow cytometric data revealed that combination

therapy (either VPA ? TX or VPA ? NTX) most signifi-

cantly increased apoptosis in both glioblastoma cell lines

(Fig. 4b). However, use of VPA ? NTX showed greater

induction of apoptosis than the use of VPA ? TX in both

cell lines. Annexin V-FITC/PI staining confirmed the

induction of cell death through phosphatidylserine exter-

nalization and indicated the mode of cell death was

apoptosis and not necrosis.

Scanning Electron Microscopy to Detect Fine

Morphological Changes

Scanning electron microscopy of both LN18 and T98G

cells after treatment with VPA showed heterogeneity of

cytoplasmic prolongations and microextensions, including

blebs and filopodia formation (Fig. 5). On the other hand,

combination therapy (either VPA ? TX or VPA ? NTX)

most effectively induced certain features of apoptosis

Fig. 4 Flow cytometric detection of apoptosis. Treatments: control

(CTL); 2 mM VPA, 50 nM TX, 50 nM NTX, VPA (2 mM) ? TX

(50 nM), VPA (2 mM) ? NTX (50 nM). Treatment schedule with

monotherapy: VPA for 72 h, TX or NTX for 24 h; and with

combination therapy: VPA for 48 h and then TX or NTX in presence

of VPA for 24 h (total treatment for 72 h). a Double parameter dot-

plots show FITC-fluorescence (x axis) versus PI-fluorescence (y axis).

Quadrants: lower left, normal live cells (PI negative and Annexin V

negative); lower right, early apoptotic cell (PI negative and Annexin

V positve); upper right, late apoptotic or necrotic cells (PI positive

and Annexin V positive); and upper left, mechanically injured cells

(PI positive and Annexin V negative). b Bar diagram representation

of apoptosis. Combination therapy significantly induced apoptosis in

both LN18 and T98G cells

2298 Neurochem Res (2011) 36:2292–2305

123

Page 8: Valproic Acid Induced Differentiation and Potentiated

including membrane blebbing, perforations, loss of mem-

brane integrity, nuclear fragmentations, cell shrinkage, and

formation of apoptotic bodies (Fig. 5).

Activation of Caspase-8 and Proteolytic Cleavage

of Bid to tBid

In order to determine the changes in apoptosis related

proteins in both LN18 and T98G cells following treat-

ments, we performed Western blotting (Fig. 6). Uniform

b-actin expression served as a loading control. Compared

with control and monotherapy, treatment with VPA ?

NTX caused substantial increase in the 20 kD active cas-

pase-8 fragment in both cell lines. Active caspase-8

cleaved Bid as a substrate for generation of tBid (Fig. 6a)

and its translocation to mitochondria for induction of cell

death. Thus, this combination therapy was capable of

inducing apoptosis partly via extrinsic pathway in both

LN18 and T98G cell lines.

Induction of Apoptosis with an Increase in Bax:Bcl-2

Ratio

The Bcl-2 family of proteins plays a key role in regulation

of mitochondrial permeability during apoptosis via intrinsic

pathway. The levels of expression of pro-apoptotic protein

Bax and anti-apoptotic protein Bcl-2 were examined by

Western blotting to determine the changes in Bax:Bcl-2

ratio (Fig. 6b). Expression of b-actin, considered as a

loading control, was uniform in all treatment groups. Cells

treated with TX, NTX, and VPA ? TX showed an increase

in Bax expression and decrease in Bcl-2 expression in both

cell lines (Fig. 6c). Use of VPA ? NTX most dramatically

increased Bax expression and decreased Bcl-2 expression,

when compared with control or single treatments. Combi-

nation therapy significantly increased the Bax:Bcl-2 ratio in

both cell lines. An increase in the Bax:Bcl-2 ratio can

trigger release of several pro-apoptotic molecules from

mitochondria for activation of intrinsic caspase cascade for

apoptosis.

Activation of Mitochondrial Pathway of Apoptosis

An alteration in the permeability of the outer mitochondrial

membrane causes the release of various pro-apoptotic

molecules, including cytochrome c and AIF, which pro-

mote the mitochondrial pathway of apoptosis (Fig. 7). We

used b-actin as cytosolic loading control and expression

was almost uniform in all treatment groups. Cells treated

with TX, NTX, VPA ? TX, or VPA ? NTX showed an

increase in cytosolic levels of cytochrome c in both LN18

and T98G cell lines (Fig. 7a). Single treatment with VPA

showed upregulation of cytosolic cytochrome c in T98G

cells but not in LN18 cells (Fig. 7a). Mitochondrial release

of cytochrome c is a well known pre-requisite for forma-

tion of apoptosome and activation of caspases for apopto-

sis. Treatment with NTX alone or VPA ? NTX generated

the cytosolic cleaved AIF fragments in both LN18 and

T98G cells (Fig. 7a) and thereby facilitated apoptosis.

Upregulation of Cysteine Proteases

Western blotting was used to determine expression of

proteases such as calpain and caspase-9 in both LN18 and

T98G cells following single (VPA, TX, and NTX) and

combination (either VPA ? TX or VPA ? NTX) treat-

ments (Fig. 7b). Level of b-actin expression remained

uniform in all treatments and was used as an internal

control. Single or combination therapy increased expres-

sion of calpain, a Ca2?-dependent cysteine protease, and

also expression and activation of caspase-9 in both glio-

blastoma cell lines (Fig. 7b). Further, activation of caspase-

Fig. 5 Scanning electron photomicrographs of LN18 and T98G cells.

Treatments: control (CTL); 2 mM VPA, 50 nM TX, 50 nM NTX,

VPA (2 mM) ? TX (50 nM), VPA (2 mM) ? NTX (50 nM). Treat-

ment schedule with monotherapy: VPA for 72 h, TX or NTX for

24 h; and with combination therapy: VPA for 48 h and then TX or

NTX in presence of VPA for 24 h (total treatment for 72 h). Without

any treatment (CTL) both cell lines showed normal morphology; VPA

treatment induced differentiation with filopodia and extensive cyto-

plasmic prolongation; TX, VPA ? TX, NTX, and VPA ? NTX

treatments induced apoptotic features such as membrane blebbing,

perforation, and disruption of membrane integrity (magnification

93000)

Neurochem Res (2011) 36:2292–2305 2299

123

Page 9: Valproic Acid Induced Differentiation and Potentiated

3 was detected following combination therapy in both cell

lines (Fig. 7b). Caspase-3, the major executioner caspase

[25], causes cleavage of cellular substrates to promote

morphological and biochemical features of apoptosis.

Increase in Proteolysis

Calpain and caspase-3 can be simultaneously activated

for apoptosis in glioblastoma cells [18]. We found the

increases in calpain and caspase-3 activities in the forma-

tion of calpain-specific 145 kD spectrin breakdown prod-

ucts (SBDP) and caspase-3 specific 120 kD SBDP,

respectively, in both LN18 and T98G cells following

combination therapy (Fig. 7c).

The increase in caspase-3 activity was further examined

in the cleavage of inhibitor of caspase-3 activated DNase

(ICAD) (Fig. 7c) that would then release CAD from the

CAD/ICAD complex followed by CAD translocation to the

nucleus for DNA fragmentation. Our results showed that

single therapy (VPA, TX, and NTX) or combination ther-

apy (either VPA ? TX or VPA ? NTX) markedly caused

cleavage of ICAD in LN18 and T98G cells (Fig. 7c). Thus,

degradation of ICAD further confirmed an increase in

caspase-3 activity in apoptosis.

Taken together, the results of our investigation showed

induction of differentiation, suppression of angiogenesis,

survival, and multi-drug resistance markers, activation of

proteolytic pathways, and cleavage of specific substrates

for apoptosis in both LN18 and T98G cells following

combination therapy (either VPA ? TX or VPA ? NTX).

Based on our experimental results, we provided a sche-

matic model to show the sequence of molecular events in

these pathways after the treatments of human glioblastoma

cells (Fig. 8).

Fig. 6 Western blotting to

examine activation of extrinsic

pathway and Bax:Bcl-2 ratio in

LN18 and T98G cells.

Treatments: control (CTL);

2 mM VPA, 50 nM TX, 50 nM

NTX, VPA (2 mM) ? TX

(50 nM), VPA (2 mM) ? NTX

(50 nM). Treatment schedule

with monotherapy: VPA for

72 h, TX or NTX for 24 h; and

with combination therapy: VPA

for 48 h and then TX or NTX in

presence of VPA for 24 h (total

treatment for 72 h).

a Representative Western blots

showed expression of 42 kD

b-actin, activation of caspase-8

in the generation of 20 kD

active fragment, and 22 kD Bid

cleavage to 15 kD tBid.

Densitometric analysis of

percent changes in protein

levels in LN18 and T98G cells.

b Representative Western blots

showed expression of 21 kD

Bax, and 26 kD Bcl-2.

c Densitometric analysis

showed the Bax:Bcl-2 ratio

2300 Neurochem Res (2011) 36:2292–2305

123

Page 10: Valproic Acid Induced Differentiation and Potentiated

Discussion

In this investigation, we explored the efficacy of the

combination therapy (either VPA ? TX or VPA ? NTX)

in two multi-drug resistant glioblastoma cell lines LN18

(increased P-gp expression) [2] and T98G (reduced P-gp

expression) [16, 17]. This is the first report on a novel

combination therapy (either VPA ? TX or VPA ? NTX)

for human glioblastoma LN18 and T98G cells. Earlier

findings reported that VPA reduced cell proliferation,

induced cytotoxicity in human glioblastoma cells, and

increased GFAP expression for astrocytic differentiation

[26]. To observe the therapeutic effects, most of the pre-

clinical cell culture studies used a high dose of VPA that

could develop new derivatives of VPA having more potent

activity in lower dose than the parent VPA. The current

study for the first time demonstrated that either VPA ? TX

or VPA ? NTX down regulated ID2 expression in both

cell lines. The ID family of helix-loop-helix proteins is

involved in cellular proliferation and angiogenesis and

Fig. 7 Western blotting for

determination of mitochondrial

release of pro-apoptotic factors,

activation of proteases, and

substrate cleavage in LN18 and

T98G cells. Treatments: control

(CTL); 2 mM VPA, 50 nM TX,

50 nM NTX, VPA

(2 mM) ? TX (50 nM), VPA

(2 mM) ? NTX (50 nM).

Treatment schedule with

monotherapy: VPA for 72 h,

TX or NTX for 24 h; and with

combination therapy: VPA for

48 h and then TX or NTX in

presence of VPA for 24 h (total

treatment for 72 h).

a Representative Western blots

showed levels of 42 kD b-actin,

15 kD cytochrome c, and 67 kD

AIF. Densitometric analysis of

percent changes in protein

levels in LN18 and T98G cells.

b Representative Western blots

showed levels of 80 kD calpain,

35 kD caspase-9 active

fragment, and 20 kD caspase-3

active fragment. Densitometric

analysis of percent changes in

protein levels in LN18 and

T98G cells. c Representative

Western blots showed levels of

145 SBDP, 120 kD SBDP, and

40 kD ICAD fragment.

Combination therapy (either

VPA ? TX or VPA ? NTX)

activated calpain and caspase

mediated proteolysis leading to

apoptosis in LN18 and T98G

cells

Neurochem Res (2011) 36:2292–2305 2301

123

Page 11: Valproic Acid Induced Differentiation and Potentiated

inhibition of ID protein indicates anti-proliferative and

anti-angiogenic activities [27].

The VEGF family and its receptors are known to act as the

central signaling pathway in glioblastoma angiogenesis [23].

Amplification of EGFR gene often occurs in glioblastoma

resulting in overexpression of EGFR, a transmembrane

tyrosine kinase receptor responsible for glioblastoma pro-

liferation and invasion [24]. Our combination therapy using

VPA ? TX or VPA ? NTX showed potent anti-angiogenic

activity by down regulating expression of VEGF and EGFR.

VPA is a potent inhibitor of tumor angiogenesis and caused

significant decrease in VEGF expression in colon adeno-

carcinoma cell line [28]. TX down regulates VEGF expres-

sion in LN18 cells [29] and other glioblastoma U138MG and

U251MG cells [30]. Our combination therapy showed the

anti-angiogenic potential by down regulating expression of

VEGF and EGFR in glioblastoma cells. A previous study

reported synergistic efficacy of combination of pegylated-

interferon-alpha (PEG-IFN-a) and TX in glioblastoma

U87MG cells due to inhibition of proliferation and angio-

genesis [31]. Similarly, a Phase II clinical trial of TX and

topotecan with filgrastim (also known as G-CSF) showed

therapeutic effects in glioblastoma patients [32], indicating

importance of TX in the combination therapy to control

growth of glioblastoma.

Aberrant or overexpressed Akt signaling is a major

event in glioblastoma. Phosphorylation of Akt (p-Akt)

results in activation of Akt kinase activity, which has high

potential to deregulate cell cycle, induce cell proliferation,

avoid apoptosis, and stimulate cell survival through

upregulation of NF-jB [33]. So, NF-jB down regulation

leads to apoptosis [34]. Combination therapy using

VPA ? TX and especially VPA ? NTX showed down

regulation of survival signals and anti-apoptotic factors due

to suppressed expression of total-Akt, p-Akt, and NF-jB in

both LN18 and T98G cells. TX in combination with all-

trans retinoic acid decreased expression of p-Akt and NF-

jB to promote apoptosis in U87MG xenografts [6],

implicating the importance of combination therapy for

suppression of survival pathways in glioblastoma.

Down regulation of P-gp expression was observed in both

LN18 and T98G cells following combination therapy, indi-

cating VPA could work as an inhibitor of P-gp to facilitate

TX and NTX induced cytotoxicity by overcoming multi-

drug resistance. Our study is in agreement with the previous

reports that demonstrate Elacridar [35] and Valspodar [10],

the strong inhibitors of P-gp, are highly effective in

increasing the delivery of TX across the BBB and signifi-

cantly reducing tumor volume in glioblastoma xenografts.

Recently, an oral combination therapy using the P-gp

inhibitor HM30181A and TX significantly regressed tumor

volume by inhibiting TX efflux in animal model of glio-

blastoma [11], thereby lending support to the possibility of

either VPA ? TX or VPA ? NTX as the potential combi-

nation therapy for treatment of glioblastoma.

Flow cytometric analysis of the cell cycle revealed that

monotherapy with VPA or TX caused accumulation of

cells in sub G1 and G2/M phases in both LN18 and T98G

cells. These findings are in line with earlier studies showing

that VPA in LN18 and U251MG cell lines [26] and TX in

T98G and LN229 cell lines caused G2/M arrest [36]. Our

combination therapy using VPA ? TX and especially

VPA ? NTX showed an immense increase in sub G1

phase and distinctly induction of apoptosis. The maximum

AnnexinV-FITC binding without PI staining in both cell

lines following combination therapy suggested that the

mode of cell death was apoptosis and not necrosis. No

report on effects of NTX in glioblastoma is yet available,

but a recent report on the combination of TX and ceramide

in nanoemulsion formulation showed significant apoptosis

in glioblastoma U118MG cell line [37]. Our scanning

electron microscopy identified ultrastuctural features of

differentiation with extensive cytoplasmic prolongation

and filopodia formation in both LN18 and T98G cells due

Fig. 8 Schematic presentation of molecular components for apopto-

sis in human glioblastoma cells. Extrinsic death receptor and intrinsic

mitochondrial (both caspase-dependent and caspase-independent)

pathways were involved in apoptosis in human glioblastoma LN18

and T98G cells. Combination therapy with VPA ? TX and most

remarkably with VPA ? NTX induced differentiation, inhibited

angiogenic and survival factors, down regulated multi-drug resis-

tance, and causes activation of calpain and caspase-3 for induction of

apoptosis

2302 Neurochem Res (2011) 36:2292–2305

123

Joseph George
Rectangle
Page 12: Valproic Acid Induced Differentiation and Potentiated

to VPA treatment, supporting a previous finding where

VPA treatment induced stellate and flattened morphologi-

cal appearances in glioblastoma cells [26]. The combina-

tion therapy (either VPA ? TX or VPA ? NTX) showed

characteristic features of apoptosis including membrane

asymmetry and blebbing, cell shrinkage, and apoptotic

body formation in both LN18 and T98G cells.

Treatment with either VPA ? TX or VPA ? NTX was

found to activate caspase-8 for Bid cleavage, indicating

activation of extrinsic or death receptor pathway for

apoptosis. Earlier studies reported that VPA alone [38] and

in combination with IFN-c [39] induced apoptosis through

activation of caspase-8, Bid cleavage, and alterations in

expression of Bax and Bcl-2. TX alone [6, 30, 40] or in

combination with all-trans retinoic acid [41] caused cas-

pase-8 activation for Bid cleavage to generate tBid and

trigger apoptosis. Caspase-8 mediated cleavage of Bid

provides a link between death receptor and mitochondrial

pathways of apoptosis. An increase in the Bax:Bcl-2 ratio

is a key determinant step for inducing the release of several

pro-apoptotic molecules from the mitochondria. In our

current study, VPA ? TX and more importantly VPA ?

NTX caused significant upregulation of Bax with con-

comitant down regulation of Bcl-2 in both LN18 and T98G

cells to cause an increase in the Bax:Bcl-2 ratio, which

appeared in line with previous studies where TX induced

apoptosis by phosphorylation and inactivation of Bcl-2 [42]

and caused upregulation of Bax and down regulation of

Bcl-2 in LN18 cells [29] and also in U138MG and

U251MG cells [30].

Translocation of tBid and upregulation of Bax are

associated with mitochondrial release of cytochrome c in

the cytosol [18], which is a prerequisite for formation of

apoptosome leading to activation of caspase-9 and caspase-

3 for apoptosis [43]. Previously, TX alone or in combina-

tion with all-trans retinoic acid was reported to dramati-

cally increase the level of cytosolic cytochrome c in LN18

cells as well as in T98G and U87MG xenografts [1, 6, 29].

Mitochondria can also induce caspase-independent apop-

tosis through release of AIF into the cytosol [44]. In this

study, we showed that combination therapy with

VPA ? TX and more importantly VPA ? NTX increased

the expression of active fragment of AIF in the cytosol.

Both tBid and Bax translocation to mitochondria can

change mitochondrial outer membrane permeabilization,

resulting in the release of cytochrome c in the cytosol for

activation of caspases. AIF is a mitochondrial membrane

integral protein that can be cleaved by active calpain for

formation of a soluble apoptogenic form to exert caspase-

independent apoptosis [45].

In our study, VPA ? NTX was more effective than

VPA ? TX to increase the expression of caspase-9, cal-

pain, and caspase-3. Calpain and caspase-3 activities

(shown in the formation of 145 kD SBDP and 120 kD

SBDP, respectively) and also caspase-3 mediated ICAD

cleavage were found in both LN18 and T98G cell lines.

The activation of these proteases and their substrate

cleavage leading to apoptosis were previously reported

in glioblastoma LN18 [26, 29, 46], U138MG and U251MG

cells [30], and T98G and U87MG xenografts [1, 6, 29].

Taken together, results from the current investigation

showed that VPA ? TX and most notably VPA ? NTX

induced differentiation, suppressed angiogenic and survival

factors, inhibited multi-drug resistance, and activated cal-

pain and caspase proteolytic cascades for induction of

apoptosis in human glioblastoma cells (Fig. 8). Thus, the

combination of VPA ? NTX could be further explored as

a potential therapeutic strategy for treatment of glioblas-

toma in preclinical as well as in clinical settings in the near

future.

Acknowledgments This investigation was supported in part by the

NS-57811 and NS-62327 grants from the National Institutes of Health

and the SCIRF-11-002 grant from the State of South Carolina.

References

1. Narayana A, Leibel SA (2004) Primary and metastatic brain

tumors in adults. In: Leibel SA, Phillips TL (eds) Textbook of

radiation oncology. Elsevier, Philadelphia, pp 463–495

2. Bahr O, Rieger J, Duffner F, Meyermann R, Weller M, Wick W

(2003) P-glycoprotein and multidrug resistance-associated pro-

tein mediate specific patterns of multidrug resistance in malignant

glioma cell lines, but not in primary glioma cells. Brain Pathol

13:482–494

3. Tang R, Faussat AM, Majdak P, Perrot JY, Chaoui D, Legrand O,

Marie JP (2004) Valproic acid inhibits proliferation and induces

apoptosis in acute myeloid leukemia cells expressing P-gp and

MRP1. Leukemia 18:1246–1255

4. Rowinsky EK, Wright M, Monsarrat B, Lesser GJ, Donehower

RC (1993) Taxol: pharmacology, metabolism and clinical

implications. Cancer Surv 17:283–304

5. Karmakar S, Banik NL, Patel SJ, Ray SK (2007) Combination of

all-trans retinoic acid and taxol regressed glioblastoma T98G

xenografts in nude mice. Apoptosis 12:2077–2087

6. Karmakar S, Banik NL, Ray SK (2008) Combination of all-trans

retinoic acid and taxol-induced differentiation and apoptosis in

human glioblastoma U87MG xenografts in nude mice. Cancer

112:596–607

7. Prados MD, Schold SC, Spence AM, Berger MS, McAllister LD,

Mehta MP, Gilbert MR, Fulton D, Kuhn J, Lamborn K, Rector

DJ, Chang SM (1996) Phase II study of paclitaxel in patients with

recurrent malignant glioma. J Clin Oncol 14:2316–2321

8. Weiss RB, Donehower RC, Wiernik PH, Ohnuma T, Gralla RJ,

Trump DL et al (1990) Hypersensitivity reactions from taxol.

J Clin Oncol 8:1263–1268

9. Gallo JM, Li S, Guo P, Reed K, Ma J (2003) The effect of

P-glycoprotein on Taxol brain and brain tumor distribution in

mice. Cancer Res 63:5114–5117

10. Fellner S, Bauer B, Miller DS, Schaffri M, Fankhanel M, Spruss

T et al (2002) Transport of paclitaxel (Taxol) across the blood-

brain barrier in vitro and in vivo. J Clin Invest 110:1309–1318

Neurochem Res (2011) 36:2292–2305 2303

123

Page 13: Valproic Acid Induced Differentiation and Potentiated

11. Joo KM, Park K, Kong DS, Song SY, Kim MH, Lee GS et al

(2008) Oral taxol chemotherapy for brain tumors: ideal combi-

nation treatment of taxol and P-glycoprotein inhibitor. Oncol Rep

19:17–23

12. Koziara JM, Whisman TR, Tseng MT, Mumper RJ (2006) In vivo

efficacy of novel taxol nanoparticles in taxol-resistant human

colorectal tumors. J Cont Rel 112:312–319

13. Desai N, Trieu V, Yao Z, Louie L, Ci S, Yang A et al (2006)

Increased antitumor activity, intratumor taxol concentrations, and

endothelial cell transport of cremophor-free, albumin-bound

taxol, ABI-007, compared with cremophor-based taxol. Clin

Cancer Res 12:1317–1324

14. Green MR, Manikhas GM, Orlov S, Afanasyev B, Makhson AM,

Bhar P, Hawkins MJ (2006) Abraxane, a novel Cremophor-free,

albumin-bound particle form of taxol for the treatment of

advanced non-small-cell lung cancer. Ann Oncol 17:1263–1268

15. Gradishar WJ, Tjulandin S, Davidson N, Shaw H, Desai N, Bhar

P, Hawkins M, O’Shaughnessy J (2005) Phase III trial of nano-

particle albumin-bound taxol compared with polyethylated castor

oil-based taxol in women with breast cancer. J Clin Oncol

23:7794–7803

16. Abe T, Hasegawa S, Taniguchi K, Yokomizo A, Kuwano T, Ono

M et al (1994) Possible involvement of multidrug-resistance-

associated protein (MRP) gene expression in spontaneous drug

resistance to vincristine, etoposide and adriamycin in human

glioma cells. Int J Cancer 58:860–864

17. Rieger L, Rieger J, Winter S, Streffer J, Esser P, Dichgans J,

Meyermann R, Weller M (2000) Evidence for a constitutive,

verapamil-sensitive, non-P-glycoprotein multidrug resistance

phenotype in malignant glioma that is unaltered by radioche-

motherapy in vivo. Acta Neuropathol 99:555–562

18. Karmakar S, Weinberg MS, Banik NL, Patel SJ, Ray SK (2006)

Activation of multiple molecular mechanisms for apoptosis in

human malignant glioblastoma T98G and U87MG cells treated

with sulforaphane. Neuroscience 141:1265–1280

19. Dhandapani KM, Mahesh VB, Brann DW (2007) Curcumin

suppresses growth and chemoresistance of human glioblastoma

cells via AP-1 and NF-jB transcription factors. J Neurochem

102:522–538

20. Karmakar S, Davis KA, Choudhury SR, Deeconda A, Banik NL,

Ray SK (2009) Bcl-2 inhibitor and apigenin worked synergisti-

cally in human malignant neuroblastoma cell lines and increased

apoptosis with activation of extrinsic and intrinsic pathways.

Biochem Biophys Res Commun 388:705–710

21. Roy Choudhury S, Karmakar S, Banik NL, Ray SK (2010)

Synergistic efficacy of sorafenib and genistein in growth inhibi-

tion by down regulating angiogenic and survival factors and

increasing apoptosis through upregulation of p53 and p21 in

malignant neuroblastoma cells having N-Myc amplification or

non-amplification. Invest New Drugs 28:812–824

22. Karmakar S, Choudhury SR, Banik NL, Ray SK (2010) Activa-

tion of multiple molecular mechanisms for increasing apoptosis

in human glioblastoma T98G xenograft. J Cancer Sci Ther

2:107–113

23. Knizetova P, Ehrmann J, Hlobilkova A, Vancova I, Kalita O,

Kolar Z, Bartek J (2008) Autocrine regulation of glioblastoma

cell cycle progression, viability and radioresistance through the

VEGF-VEGFR2 (KDR) interplay. Cell Cycle 7:2553–2561

24. Aldape KD, Ballman K, Furth A, Buckner JC, Giannini C, Burger

PC et al (2004) Immunohistochemical detection of EGFRvIII in

high malignancy grade astrocytomas and evaluation of prognostic

significance. J Neuropathol Exp Neurol 63:700–707

25. Sakahira H, Enari M, Nagata S (1998) Cleavage of CAD inhibitor

in CAD activation and DNA degradation during apoptosis. Nat-

ure 391:96–99

26. Das CM, Aguilera D, Vasquez H, Prasad P, Zhang M, Wolff JE,

Gopalakrishnan V (2007) Valproic acid induces p21 and topoi-

somerase-II (a/b) expression and synergistically enhances etopo-

side cytotoxicity in human glioblastoma cell lines. J Neurooncol

85:159–170

27. Selesniemi K, Reedy M, Gultice A, Guilbert LJ, Brown TLb

(2005) Transforming growth factor-b induces differentiation of

the labyrinthine trophoblast stem cell line SM10. Stem Cells Dev

14:697–711

28. Jones J, Bentas W, Blaheta RA, Makarevic J, Hudak L, Wedel S

et al (2008) Modulation of adhesion and growth of colon and

pancreatic cancer cells by the histone deacetylase inhibitor val-

proic acid. Int J Mol Med 22:293–299

29. Zhang R, Banik NL, Ray SK (2008) Differential sensitivity of

human glioblastoma LN18 (PTEN-positive) and A172 (PTEN-

negative) cells to Taxol for apoptosis. Brain Res 1239:216–225

30. George J, Banik NL, Ray SK (2009) Bcl-2 siRNA augments taxol

mediated apoptotic death in human glioblastoma U138MG and

U251MG cells. Neurochem Res 34:66–78

31. Son MJ, Song HS, Kim MH, Kim JT, Kang CM, Jeon JW et al

(2006) Synergistic effect and condition of pegylated interferon

alpha with paclitaxel on glioblastoma. Int J Oncol 28:1385–

1392

32. Pipas JM, Meyer LP, Rhodes CH, Cromwell LD, McDonnell CE,

Kingman LS et al (2005) A Phase II trial of paclitaxel and to-

potecan with filgrastim in patients with recurrent or refractory

glioblastoma multiforme or anaplastic astrocytoma. J Neurooncol

71:301–305

33. Pelloski CE, Lin E, Zhang L, Yung WK, Colman H, Liu JL et al

(2006) Prognostic associations of activated mitogen-activated

protein kinase and Akt pathways in glioblastoma. Clin Cancer

Res 12:3935–3941

34. Beg AA, Baltimore D (1996) An essential role for NFjB in

preventing TNF-a induced cell death. Science 274:782–784

35. Kemper EM, Boogerd W, Thuis I, Beijnen JH, van Tellingen O

(2004) Modulation of the blood-brain barrier in oncology: ther-

apeutic opportunities for the treatment of brain tumours? Cancer

Treat Rev 30:415–423

36. Roth W, Wagenknecht B, Grimmel C, Dichgans J, Weller M

(1998) Taxol-mediated augmentation of CD95 ligand-induced

apoptosis of human malignant glioma cells: association with bcl-

2 phosphorylation but neither activation of p53 nor G2/M cell

cycle arrest. Br J Cancer 77:404–411

37. Desai A, Vyas T, Amiji M (2008) Cytotoxicity and apoptosis

enhancement in brain tumor cells upon coadministration of taxol

and ceramide in nanoemulsion formulations. J Pharm Sci

97:2745–2756

38. Bokelmann I, Mahlknecht U (2008) Valproic acid sensitizes

chronic lymphocytic leukemia cells to apoptosis and restores the

balance between pro- and antiapoptotic proteins. Mol Med

14:20–27

39. Hacker S, Dittrich A, Mohr A, Schweitzer T, Rutkowski S,

Krauss J et al (2009) Histone deacetylase inhibitors cooperate

with IFN-gamma to restore caspase-8 expression and overcome

TRAIL resistance in cancers with silencing of caspase-8. Onco-

gene 28:3097–3110

40. Mielgo A, Torres VA, Clair K, Barbero S, Stupack DG (2009)

Paclitaxel promotes a caspase 8-mediated apoptosis through

death effector domain association with microtubules. Oncogene

28:3551–3562

41. Park SJ, Wu CH, Gordon JD, Zhong X, Emami A, Safa AR

(2004) Taxol induces caspase-10-dependent apoptosis. J Biol

Chem 279:51057–51067

42. Haldar S, Jena N, Croce CM (1995) Inactivation of Bcl-2 by

phosphorylation. Proc Natl Acad Sci USA 92:4507–4511

2304 Neurochem Res (2011) 36:2292–2305

123

Joseph George
Rectangle
Page 14: Valproic Acid Induced Differentiation and Potentiated

43. Kim R (2005) Recent advances in understanding the cell death

pathways activated by anticancer therapy. Cancer 103:1551–

1560

44. Cregan SP, Dawson VL, Slack RS (2004) Role of AIF in caspase

dependent and caspase-independent cell death. Oncogene 23:

2785–2796

45. Polster BM, Basanez G, Etxebarria A, Hardwick JM, Nicholls

DG (2005) Calpain I induces cleavage and release of apoptosis-

inducing factor from isolated mitochondria. J Biol Chem 280:

6447–6454

46. Catalano MG, Poli R, Pugliese M, Fortunati N, Boccuzzi G

(2007) Valproic acid enhances tubulin acetylation and apoptotic

activity of taxol on anaplastic thyroid cancer cell lines. Endocr

Relat Cancer 14:839–845

Neurochem Res (2011) 36:2292–2305 2305

123