+ All Categories
Home > Documents > The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell...

The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell...

Date post: 20-Aug-2016
Category:
Upload: moshe
View: 214 times
Download: 0 times
Share this document with a friend
19
ORIGINAL PAPER The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death Luba Shargorodsky Leo Veenman Beatriz Caballero Yelena Pe’er Svetlana Leschiner Julia Bode Moshe Gavish Published online: 28 April 2012 Ó Springer Science+Business Media, LLC 2012 Abstract Various studies have shown that several lethal agents induce cell death via the mitochondrial 18 kDa Translocator Protein (TSPO). In this study we tested the possibility that nitric oxide (NO) is the signaling compo- nent inducing the TSPO to initiate cell death process. Cell viability assays included Trypan blue uptake, propidium iodide uptake, lactate dehydrogenase release, and DNA fragmentation. These assays showed that application of the specific TSPO ligand PK 11195 reduced these parameters for the lethal effects of the NO donor sodium nitroprusside (SNP) by 41, 27, 40, and 42 %, respectively. TSPO silencing by siRNA also reduced the measured lethal effects of SNP by 50 % for all of these four assays. With 2,3-bis[2-methoxy-4-nitro-5-sulphophenyl]-2H-tetra- zolium-5-carboxyanilide (XTT) changes in metabolic activity were detected. PK 11195 and TSPO knockdown fully prevented the reductions in XTT signal otherwise induced by SNP. Collapse of the mitochondrial membrane potential was studied with the aid of JC-1 (5,5 0 ,6,6 0 -tetra- chloro-1,1 0 ,3,3 0 -tetraethyl-benzimidazolylcarbocyanine chloride). PK 11195 and TSPO knockdown reduced, respectively by 36 and 100 %, the incidence of collapse of the mitochondrial membrane potential otherwise induced by SNP. 10-N-Nonyl-Acridine Orange (NAO) was used to detect mitochondrial reactive oxygen species generation due to SNP. PK 11195 and TSPO knockdown reduced this effect of SNP by 65 and 100 %, respectively. SNP did not affect TSPO protein expression and binding characteristics, and also did not cause TSPO S-nitrosylation. However, b-actin and various other proteins (not further defined) were S-nitrosylated. In conclusion, TSPO is required for the lethal and metabolic effects of the NO donor SNP, but TSPO itself is not S-nitrosylated. Keywords 18 kDa mitochondrial Translocator Protein (TSPO) Nitric oxide (NO) Protein S-nitrosylation Apoptosis Reactive oxygen species (ROS) Abbreviations DWm Mitochondrial membrane potential ANOVA Analysis of variance ANT Adenine nucleotide transporter B max Maximal binding capacity BSA Bovine serum albumin BST Biotin-switch technique CAPON A cytosolic adaptor and scaffold protein associated with nitric oxide synthase CBR Central-type benzodiazepine receptor CCCP Carbonyl cyanide 3-chlorophenylhydrazone CysNO S-Nitrosocysteine Dexras1 A member of the Ras family of small G-proteins DMEM Dulbecco’s Modification of Eagle’s Medium EDT Ethylenediaminetetraacetic acid eNOS Endothelial NO synthase Leo Veenman and Luba Shargorodsky contributed equally. L. Shargorodsky L. Veenman B. Caballero Y. Pe’er S. Leschiner M. Gavish (&) Department of Molecular Pharmacology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Ephron Street, Bat-Galim, P.O.B. 9649, 31096 Haifa, Israel e-mail: [email protected] J. Bode Abteilung Pa ¨diatrie I, Zentrum Kinderheilkunde und Jugendmedizin, Universita ¨tsmedizin Go ¨ttingen, Go ¨ttingen, Germany 123 Apoptosis (2012) 17:647–665 DOI 10.1007/s10495-012-0725-2
Transcript
Page 1: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

ORIGINAL PAPER

The nitric oxide donor sodium nitroprusside requires the 18 kDaTranslocator Protein to induce cell death

Luba Shargorodsky • Leo Veenman •

Beatriz Caballero • Yelena Pe’er • Svetlana Leschiner •

Julia Bode • Moshe Gavish

Published online: 28 April 2012

� Springer Science+Business Media, LLC 2012

Abstract Various studies have shown that several lethal

agents induce cell death via the mitochondrial 18 kDa

Translocator Protein (TSPO). In this study we tested the

possibility that nitric oxide (NO) is the signaling compo-

nent inducing the TSPO to initiate cell death process. Cell

viability assays included Trypan blue uptake, propidium

iodide uptake, lactate dehydrogenase release, and DNA

fragmentation. These assays showed that application of the

specific TSPO ligand PK 11195 reduced these parameters

for the lethal effects of the NO donor sodium nitroprusside

(SNP) by 41, 27, 40, and 42 %, respectively. TSPO

silencing by siRNA also reduced the measured lethal

effects of SNP by 50 % for all of these four assays. With

2,3-bis[2-methoxy-4-nitro-5-sulphophenyl]-2H-tetra-

zolium-5-carboxyanilide (XTT) changes in metabolic

activity were detected. PK 11195 and TSPO knockdown

fully prevented the reductions in XTT signal otherwise

induced by SNP. Collapse of the mitochondrial membrane

potential was studied with the aid of JC-1 (5,50,6,60-tetra-

chloro-1,10,3,30-tetraethyl-benzimidazolylcarbocyanine

chloride). PK 11195 and TSPO knockdown reduced,

respectively by 36 and 100 %, the incidence of collapse of

the mitochondrial membrane potential otherwise induced

by SNP. 10-N-Nonyl-Acridine Orange (NAO) was used to

detect mitochondrial reactive oxygen species generation

due to SNP. PK 11195 and TSPO knockdown reduced this

effect of SNP by 65 and 100 %, respectively. SNP did not

affect TSPO protein expression and binding characteristics,

and also did not cause TSPO S-nitrosylation. However,

b-actin and various other proteins (not further defined)

were S-nitrosylated. In conclusion, TSPO is required for

the lethal and metabolic effects of the NO donor SNP, but

TSPO itself is not S-nitrosylated.

Keywords 18 kDa mitochondrial Translocator Protein

(TSPO) � Nitric oxide (NO) � Protein S-nitrosylation �Apoptosis � Reactive oxygen species (ROS)

Abbreviations

DWm Mitochondrial membrane potential

ANOVA Analysis of variance

ANT Adenine nucleotide transporter

Bmax Maximal binding capacity

BSA Bovine serum albumin

BST Biotin-switch technique

CAPON A cytosolic adaptor and scaffold protein

associated with nitric oxide synthase

CBR Central-type benzodiazepine receptor

CCCP Carbonyl cyanide

3-chlorophenylhydrazone

CysNO S-Nitrosocysteine

Dexras1 A member of the Ras family of small

G-proteins

DMEM Dulbecco’s Modification of Eagle’s

Medium

EDT Ethylenediaminetetraacetic acid

eNOS Endothelial NO synthase

Leo Veenman and Luba Shargorodsky contributed equally.

L. Shargorodsky � L. Veenman � B. Caballero � Y. Pe’er �S. Leschiner � M. Gavish (&)

Department of Molecular Pharmacology, Faculty of Medicine,

Rappaport Family Institute for Research in the Medical Sciences,

Technion-Israel Institute of Technology, Ephron Street,

Bat-Galim, P.O.B. 9649, 31096 Haifa, Israel

e-mail: [email protected]

J. Bode

Abteilung Padiatrie I, Zentrum Kinderheilkunde und

Jugendmedizin, Universitatsmedizin Gottingen, Gottingen,

Germany

123

Apoptosis (2012) 17:647–665

DOI 10.1007/s10495-012-0725-2

Page 2: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

ErPC3 Erucylphosphohomocholine

FACS Fluorescence assisted cell counting

FBS Fetal bovine serum

GAPDH Glyceraldehyde-3-phosphate

dehydrogenase

GSNO S-Nitroso-L-glutathione

HEN buffer 100 mM Hepes, 1 mM EDTA, 0.1 mM

neocuproine

HENS buffer HEN containing 1 % SDS

HEPES 4-(2-Hydroxyethyl)-1-

piperazineethanesulfonic acid

HRP Horse radish peroxidase

HS Horse serum

iNOS Inducible NO synthase

JC-1 5,50,6,60-Tetrachloro-1,10,3,30-tetraethyl-

benzimidazolylcarbocyanine chloride

Kd Equilibrium dissociation constant

Ki Binding affinity of the inhibitor

(in displacement assays)

LDH Lactate dehydrogenase

MEM-EAGLE Minimum Essential Medium Eagle

MMTS Methylmethane thiosulfonate

NAO 10-N-Nonyl-Acridine Orange

nNOS Neuronal NO synthase

NO Nitric oxide

NOS Nitric oxide synthase

PAP7 PBR associated protein 7

PBS Phosphate buffered saline

PC12 A cell line derived from a

pheochromocytoma of the rat adrenal

medulla

PI Propidium iodide

pk 10 A protein of 10 kDa MW

PK 11195 1-(2-Chlorophenyl-N-methyl-1-

methylpropyl)-3-isoquinoline-carbox-

amide

PRAX-1 PBR associated protein 1

Protein-SNO S-Nitrosylated proteins

RNS Reactive nitrogen species

Ro5 4864 7-Chloro-5-(4-chlorophenyl)-1,3-

dihydro-1-methyl-2H-1,4-benzodiazepin-

2-one

ROS Reactive oxygen species

RT-PCR Real time polymerase chain reaction

Scr Sham control scrambled siRNA cells

SD Standard deviation

SDS Sodium dodecyl sulfate

SDS-PAGE SDS-polyacrylamide gel

siRNA cells U118MG TSPO knockdown cells

SNO S-Nitrosothiol

SNO-RAC SNO precipitation using resin-assisted

capture

SNP Sodium nitroprusside

Tris Tris(hydroxymethyl)aminomethane

U118MG Human glioblastoma cell line

TSPO Mitochondrial 18 kDa Translocator

Protein

VDAC Voltage dependent anion channel

XTT 2,3-Bis[2-methoxy-4-nitro-5-

sulphophenyl]-2H-tetrazolium-5-

carboxyanilide

Introduction

The 18 kDa mitochondrial Translocator Protein (TSPO),

formerly known as peripheral-type benzodiazepine recep-

tor (PBR), can be found in peripheral tissues and in glial

cells in the brain [1–6]. The primary intracellular location

of TSPO is the outer mitochondrial membrane, while it can

also be associated with the cell nucleus and outer cell

membrane [7–11]. TSPO have been implicated in: apop-

tosis, steroidogenesis, oxidative stress, mitochondrial res-

piration, immune response, inflammation, glial activation,

ischemia, regulation of the mitochondrial membrane

potential (DWm), cell proliferation, cell differentiation,

tumorigenicity, and mental and neuropathological disor-

ders (for reviews, see [2–5, 11–15]). TSPO knockdown and

TSPO ligand studies have shown that the TSPO can induce

cell death via reactive oxygen species (ROS) generation,

cardiolipin oxidation, collapse of the DWm, and mito-

chondrial cytochrome c release [16–18]. Alternatively, it

has been suggested that TSPO may play a role in the

protection of cells from ROS damage [19, 20]. It was found

that various agents can induce cell death via activation of

the TSPO [16–18]. However, none of these agents in these

studies interacted directly with the TSPO [16–18]. Thus,

the question remained how these diverse agents can mod-

ulate TSPO function, i.e. which is the intermediate intra-

cellular factor that acts directly on the TSPO. One

candidate we considered was nitric oxide (NO), potentially

causing S-nitrosylation of the TSPO.

Like TSPO, NO is known to be involved in cell death

induction [21–26]. In healthy brain, NO is produced mainly

by neuronal NO synthase (nNOS) found in a small subset

of neurons, and endothelial NO synthase (eNOS) found in

endothelial cells [27]. In diseased brain, processes related

to damage such as hypoxia and pathogens activate induc-

ible NOS (iNOS) in microglia and astrocytes [28, 29]. It is

well known that NO can affect protein function via S-nit-

rosylation (by covalent attachment of NO to cysteine thi-

ols) [30, 31]. Aberrant S-nitrosylation is implicated in

tumorigenicity and neurodegeneration [32–34]. NO can

cause cell death by interference with various cellular

648 Apoptosis (2012) 17:647–665

123

Page 3: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

mechanisms: inhibition of the mitochondrial respiratory

chain; collapse of the DWm; inhibition of glyceraldehyde-

3-phosphate dehydrogenase (GAPDH); and generation of

ROS and reactive nitrogen species (RNS). Such processes

deplete the cell of ATP, leading to cell necrosis [35].

However, NO can also induce apoptosis, for example via:

(i) induction of p53; (ii) endoplasmic reticulum dysfunc-

tion; (iii) mitochondrial cytochrome c release; and (iv)

activation of MAP kinase pathways [35]. NO has also been

suggested to be involved in tumorigenicity and neurode-

generation [35–37]. NOS may be linked to the TSPO via

scaffolding proteins, including CAPON in association with

PBR associated protein 7 (PAP7, also known as acyl-CoA

binding domain containing 3, ACBD3) and Dexras1 [38,

39]. CAPON is a cytosolic adaptor and scaffold protein

associated with NOS. Dexras1 is a member of the Ras

family of small G-proteins. Thus, we assumed that NO may

act via the TSPO to induce cell death.

With the present study we sought to determine whether

the TSPO indeed is involved in the induction of cell death

due to application of NO donors, in particular, whether NO

donors may directly affect TSPO.

Methods

Materials

S-Nitroso-L-glutathione (GSNO) and S-Nitrosylated Pro-

tein Detection Assay Kits were from Cayman Chemical

Company (Ann Arbor, MI). Various cell culture materials,

Trypan blue, cell proliferation kits (XTT based), and EZ-

ECL Chemiluminescence Detection Kits were from Beit

Haemek—Biological Industries (Israel). Dulbecco’s phos-

phate buffered saline (PBS) with CaCl2 and MgCl2, bovine

serum albumin (BSA), sodium nitroprusside dehydrate

(SNP), clonazepam, 7-chloro-5-(4-chlorophenyl)-1,3-dihy-

dro-1-methyl-2H-1,4-benzodiazepin-2-one (Ro5 4864),

1-(2-chlorophenyl-N-methyl-1-methylpropyl)-3-isoquino-

line-carbox-amide (PK 11195), 10-N-Nonyl-Acridine

Orange (NAO), propidium iodide (PI), carbonyl cyanide

3-chlorophenylhydrazone (CCCP), anti-b-actin antibody

(mouse anti-human), anti-VDAC antibody (mouse anti-

human), sodium ascorbate, sodium nitrite, L-cysteine,

methylmethane thiosulfonate (MMTS) and Ponceau

staining solution were from Sigma-Aldrich (Rehovot,

Israel). Cytotoxicity Detection Kit (LDH kit), Cell Death

Detection ELISAPLUS Kit (Cell Death Kit), Protease

inhibitor (Complete) were from Roche (Mannheim, Ger-

many). Bradford solution and Coomassie blue, were from

Bio-Rad (Munich, Germany). [3H]PK 11195 was from

New England Nuclear (Boston, MA). CytoScint was

purchased from MP Biomedicals (Irvine, CA). Whatman

GF/C filters were obtained from Tamar (Mevaseret Zion,

Israel). Nitrocellulose membranes were from Hybond

ECL, Amersham Biosciences (Buckinghamshire, Eng-

land). Dried milk powder was from Carnation (Glendale,

CA). Anti-human TSPO anti-serum from rabbit was pre-

pared in our laboratory [17]. Anti-VDAC and anti-b-actin

antibodies were from Sigma, Rehovot, Israel. The sec-

ondary antibodies (anti-rabbit and anti-mouse—IgG

linked to horseradish peroxidase) were from Healthcare,

Amersham (Buckinghamshire, England). Western-

BrightTM-ECL HRP western blotting detection kit was

purchased from Advansta (Menlo Park, CA). Kodak

BioMax MR Film was obtained from Scientific Imaging

Film (Chalon-sur-Saone, France). The fluorescent dye

5,50,6,60-tetrachloro-1,10,3,30tetraethylbenzimidazolylcarb

ocyanine iodide (JC-1) was from Calbiochem (Merck,

Darmstadt, Germany). Thiopropyl Sepharose 6B was

purchased from GE Healthcare (Amersham, Bucking-

hamshire, England).

Cell cultures and experimental conditions

The human glioblastoma cell line, U118MG, and the rat

pheochromocytoma cell line, PC12, were cultured at 37 �C

in an atmosphere of 5 % CO2 and 90 % relative humidity.

These tumor cells (U118MG) and neuronal like cells (PC12)

were chosen, because cell death is an important mechanism

in tumorigenicity and neuronal death due to disease and

injury. PC12 cells were incubated in DMEM supplemented

with 8 % fetal bovine serum (FBS), 8 % heat-inactivated

horse serum (HS), 1 % glutamine, and 0.1 % penicillin

(100,000 units/ml) ? streptomycin (100 mg/ml) solution.

U118MG cells were cultured in MEM-EAGLE supple-

mented with 10 % FBS, 2 % glutamine and 0.05 % genta-

mycin (50 mg/ml). U118MG TSPO knockdown cells

(siRNA cells), and their sham control scrambled siRNA

cells (Scr) prepared in our laboratory, were cultured in

MEM-EAGLE supplemented with 10 % FCS, 2 % gluta-

mine and 0.05 % hygromycin (30 lg/ml) [18]. Western blot

and real time polymerase chain reaction (RT-PCR) were

applied routinely to determine the persistence of the TSPO

knockdown, see below.

For 2,3-bis[2-methoxy-4-nitro-5-sulphophenyl]-2H-tetra-

zolium-5-carboxyanilide (XTT inner salt) and lactate

dehydrogenase (LDH) assays, the cells were grown in 96

well-plates for 24 h (2 9 104 cells seeded per well). For

S-nitrosylation assays by resin-assisted capture (SNO-RAC)

cells were grown in 10 cm Petri dishes (seeded 1 9 106 cells

per dish) for 96 h reaching 70–80 % confluence. For all other

experiments 1.25 9 105 cells were seeded in 6 well plates

and grown for 72 h until reaching 60–70 % confluence. For

RT-PCR analysis as verification for TSPO knockdown,

1 9 104 cells were seeded in 75 cm2 cell culture flasks

Apoptosis (2012) 17:647–665 649

123

Page 4: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

(Sarstedt, Nuembrecht, Germany) in triplicates and cul-

tured for 2 days (until they reached 90 % confluence).

Then, floating and trypsinized adherent cells were col-

lected, as described further below. For each assay applying

NO donors, culture medium of the cells was replaced with

new medium with reduced serum levels (i.e. 0.5 % FBS for

U118MG; 0.5 % FBS and 0.5 % HS for PC12), and the

required cell culture conditions were applied, including

incubation with the appropriate compounds (Table 1). To

assay the effects of the NO donor SNP on cell viability, the

cells were incubated in the dark for 24 h in the appropriate

medium and then collected for the different analyses. In

addition, to determine S-nitrosylation of proteins by SNP

application, cells were incubated with NO donors for

5 min, 10 min, 15 min, 30 min, 60 min, and 24 h, as

required. The incubations included the NO donors SNP and

S-nitrosoglutathione (GSNO) at different concentrations

(500, 750, and 1000 lM), as described in previous studies

[40–43]. For a positive control of S-nitrosylation of TSPO,

also the NO donor S-Nitrosocysteine (CysNO) was applied

(1 mM for 10 min), as described previously [44]. The

vehicle for SNP, GSNO, and CysNO was PBS (Biological

Industries) (1 % of the total volume of medium). In addi-

tion, the cells received the TSPO specific ligands PK 11195

(25 lM), with and without NO donors. All the ligands were

applied 1 h before the NO donor and then also together

with the NO donor. For the negative control of the ligand

applications, we used the vehicle (medium ? 1 % ethanol,

i.e. vehicle control). Positive controls were applied as

required for the various assays. The concentration of PK

11195 applied to study TSPO related effects were as

described previously [17]. SNP, GSNO, and CysNO were

applied according to methods described previously [44–

47]. After NO donor and TSPO ligand application we

assayed cell death, including apoptosis, mitochondrial

activity, mitochondrial ROS generation, DWm, TSPO

expression and binding characteristics, and assays for

protein S-nitrosylation as described in detail below.

Collection of material

Material was collected for various complementary methods

used to assay cell death as a consequence of NO donor

application. For LDH assays, assaying cell death/viability

[48], 96 well plates were centrifuged (2509g, 10 min) and

cell free supernatant was collected for further analysis. For

XTT assays, assaying metabolic activity/proliferation/and/

or cell viability [49–51], cells were left in their original 96

well plates. For application of the S-Nitrosylated Protein

Detection Assay Kit to assay S-nitrosylation of proteins in

general, cells were collected with the wash buffer supplied

by the kit of Cayman Chemical Company. To determine

specific S-nitrosylation of TSPO, VDAC and b-actin, as

well as S-nitrosylation of proteins in general, cells were

rinsed with cold PBS, and then incubated for 30 min at

4 �C with shaking in lysis buffer (PBS, 0.1 % SDS, 1 %

triton X-100 plus protease inhibitors), and then the cells

were collected by scraping to optimize protein yield.

For all other assays, including RT-PCR, western blot,

binding analysis, and all of the flow cytometry assays [PI,

10-N-Nonyl-Acridine Orange (NAO) and 5,50,6,60-tetra-

chloro-1,10,3,30-tetraethyl-benzimidazolylcarbocyanine

chloride (JC-1)], culture medium was collected and the

cells were trypsinized (3 min, at 37 �C). After trypsiniza-

tion and collection, the cell suspensions were centrifuged

(2009g, 10 min at 4 �C), then the cell pellets were washed

in 1 ml of PBS, and centrifuged again. For RT-PCR cell

were pelleted by centrifugation 10009g, 7 min, 20 �C. The

final pellets were immediately processed for the assays in

question. For TSPO binding assays, the pellets were snap-

frozen in liquid nitrogen and stored at -70 �C until further

use. For western blot analysis of TSPO protein levels and

for assays of protein S-nitrosylation, cell pellets were lysed

and the lysate stored at -70 �C until further use.

General protein measurement

Protein levels, as required for binding assays and western

blot, were measured according to Bradford [52], using BSA

as a standard.

TSPO binding

To detect changes in TSPO binding characteristics after

application of the NO donor SNP, binding assays with

[3H]PK 11195 were performed. The cell pellets stored at

-70 �C were thawed and homogenized in 1 ml of PBS

using a Kinematika Polytron (setting 6) for 10 s. Binding

of [3H]PK 11195 to membranes of the various cell lines

was conducted as previously described [16–18]. The

Table 1 Scheme for NO donor and TSPO ligand application

Group Vehicle control NO donor TSPO ligand NO donor ? TSPO ligand

Treatment Medium ? 1 % Ethanol Medium ? 1 %

Ethanol ? SNP or GSNO

Medium ? 1 %

Ethanol ? PK 11195

Medium ? 1 % Ethanol ? SNP or

GSNO ? PK 11195

NO donors: SNP sodium nitroprusside, GSNO S-nitrosoglutathione

650 Apoptosis (2012) 17:647–665

123

Page 5: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

reaction mixture contained 400 ll of the homogenized

membranes in question (40 lg protein) and 25 ll of [3H]PK

11195 solution (0.18–6 nM final concentration) in the

absence (total binding) or presence (non-specific binding) of

75 ll unlabeled PK 11195 (10 lM final concentration). After

incubation for 90 min at 4 �C, the samples were vacuum

filtered through Whatman GF/C filters, washed three times

with 4 ml of 50 mM potassium phosphate buffer (pH 7.4).

Then the filters were placed in vials containing 4 ml of

CytoScint (MP Biomedicals). Radioactivity was counted

after 12 h with the 1600 CA Tri-Carb liquid scintillation

analyzer. The binding density (Bmax) and equilibrium dis-

sociation constant (Kd) were determined by Scatchard anal-

ysis of saturation curves of [3H]PK 11195 binding.

Displacement assays were applied to study whether

application of SNP could interfere directly with TSPO

binding to its ligand [3H]PK 11195. For these displacement

studies, rat kidney membranes homogenates were used as

previously described [53]. The reaction mixture contained

400 ll of homogenized kidney membranes (5 mg of

homogenate/ml) and 25 ll of [3H]PK 11195 (6 nM final

concentration) in the absence (total binding) or presence of

various concentrations (from 10-9 to 10-4 M) of SNP.

After incubation for 60 min at 4 �C, samples were filtered

under vacuum over Whatman GF/B filters and washed

three times with 5 ml of 50 mM Tris–HCl buffer, pH 7.4.

The filters were incubated in CytoScintTM and, after 12 h,

radioactivity was counted with the liquid scintillation

analyzer. Ki values were calculated by the equation

Ki = IC50/(1 ? C/Kd), where C = [3H] PK 11195 con-

centration, IC50 = concentration causing 50 % inhibition

of [3H]PK 11195 binding and Kd = 2 nM (from Scatchard

analysis of [3H]PK 11195 binding to kidney membranes).

Western blot

This analysis was done in order to assess relative protein

levels of TSPO after application of the NO donors, in

particular SNP, as well as to verify the stability of the

TSPO knockdown. Furthermore, western blot was applied

to determine the relative amount of S-nitrosylated proteins,

including S-nitrosylated TSPO after application of SNP

and CysNO. Collected samples with equal amount of

protein (10 lg protein/lane) were prepared in 29 sample

buffer (0.125 M 2-amino-2-(hydroxymethyl) propane-1,3-

diol (Tris)–HCl, pH 6.8, glycerol (20% v/v), SDS (sodium

dodecyl sulfate) (4% w/v), 0.14 M b-mercaptoethanol, and

bromophenol blue (0.005% w/v). The SDS–polyacryl-

amide gels were run and analyzed as described previously

[18, 54]. Rabbit anti-human TSPO antiserum 1:1,000 pre-

pared in our laboratory was used [17]. Mouse anti-human

VDAC antibody 1:5,000 was also used, and mouse anti-

human b-actin antibody 1:5,000 was used as a loading

reference. The primary antibodies were labeled with

IgG secondary antibody linked to horseradish peroxidase

(anti-rabbit and also anti-mouse IgG 1:5,000). Binding

of antibodies to their antigens was detected with the

WesternBrightTM-ECL western blotting detection kit (Advansta,

Menlo Park, CA). Labeling was captured on X-Omat blue XB-1

Kodak scientific Imaging Film. The results were analyzed using

densitometry.

RT-PCR

Obtained pellets were used for RNA extraction (RNeasy�

Mini Kit, Qiagen, Germany). The concentrations of RNAs

were determined by NanoDropTM measurement (Nano-

Drop Technologies, Wilmington, DE). Reverse transcrip-

tion of RNA was performed with iSript cDNA Synthesis

Kit (BioRad, Hercules, CA), according to the instructions

of the manufacturer. RT-PCR was performed using 10 ng

target cDNA and 3 pmol of each primer in SYBR-Green-

Mix (SYBR Green, Applied Biosystems, Foster City, CA).

The reaction was carried out at a final volume of 10 ll in

384 well optical plates. For each sample triplicates were

performed and water was taken for negative control. 18S

was chosen as internal standard as genes whose expression

should not be changed by treatment of the cells. The

reaction was carried out at the ABI PRISM 7900HT Fast

Real Time PCR System (Applied Biosystems, Life Tech-

nologies Corporation, Carlsbad, CA) with 40 cycles of 15 s

at 95 �C and one minute at 60 �C followed by the disso-

ciation measurement.

Housekeeper sequence for RT-PCR primer:

18S Forward: 50-AACTTTCGATGGTAGTCGCCG-30

18S Reverse 50-GGATGTGGTAGCCGTTTCTCAG-30

TSPO sequence for RT-PCR primer:

TSPO_Forward: 50-TTGGAGGAAGGCCTATTCACT-30

TSPO_Reverse: 50-GCCGCATTCACCTTGAAGA-30

Data analysis was performed using SDS2.4 software.

Viability assays

Viability assays were performed on normal U118MG cells

(wild type), PC12 cells (wild type), U118MG TSPO

knockdown cells (siRNA) and their background control,

i.e. U118MG Scr cells to determine the effects of the NO

donors and the TSPO ligand PK 11195. Trypan blue and PI

uptake defined dead cells were analyzed as described pre-

viously [18]. LDH release into the cell culture medium was

determined using a Cytotoxicity Detection Kit (Roche) as

described by the manufacturer. Evaluation of cytotoxicity

was calculated according to the formula suggested by the

Apoptosis (2012) 17:647–665 651

123

Page 6: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

manufacturer (Roche): % LDH released = [(A - B)/(C -

B)] 9 100; where A = LDH activity in the culture medium

of cells treated with NO donor; B = LDH activity of cul-

ture medium from untreated cells; and C = LDH activity

after total cell lysis with 1 % Triton.

Apoptosis

It is known that one of the TSPO functions is regulation of

apoptotic rates [12, 14, 16–18, 55–57]. Thus, we wanted to

determine whether our treatments with NO donors affect

this TSPO function. Cell morphology, including rounding

and blebbing analyzed microscopically was indicative of

the occurrence of apoptosis, as described previously [17,

18]. DNA fragmentation indicative of apoptotic cell death

was assayed with the Cell Death Detection ELISAPLUS

(Cell Death Kit) from Roche Applied Science (Mannheim,

Germany), as described previously [17, 18].

Mitochondrial and metabolic activity assays

Several parameters of metabolic activity and mitochondrial

activity/dysfunction following the various treatments were

assayed. The XTT assay is based on reduction of XTT

(normally yellow in PBS) by mitochondrial dehydrogen-

ases of viable cells, while extracellular reduction of XTT

may involve NADH coenzyme Q-dependent ferricyanide

reductases and a ubiquinone-dependent plasma membrane

electron transport chain [58]. We measured reduction of

XTT by means of the Cell proliferation-XTT based assay

kit (Biological Industries), as described by the manufac-

turer. To assay incidence of collapse of the DWm, JC-1 was

used as described previously [17, 18, 57].

Oxidative stress

Oxidative stress localized to mitochondria was assessed by

fluorescence measurement of NAO. NAO is able to bind

with high affinity to non-oxidized cardiolipin in a 2:1 ratio.

In the case of oxidized cardiolipin, NAO has been reported

to bind cardiolipin with a decreased affinity reflected by

lower levels of green fluorescence [13, 18, 59]. The NAO-

based assay gives clear results regarding the role of TSPO

in ROS generation [14]. Other assays attempting to relate

TSPO to oxidative stress at best only presented equivocal

results in our hands [14].

Protein S-nitrosylation

The ability of SNP in our paradigm to S-nitrosylate pro-

teins in general was investigated with the S-Nitrosylated

Protein Detection Assay Kit (Cayman) according to the

instructions of the manufacturer. Using this method, free

SH groups are first blocked with buffer A containing

blocking reagent, as supplied by the kit) and any S-NO

bonds left present in the sample are then cleaved with

Buffer B containing reducing and labeling reagents, sup-

plied by the kit. Biotinylation of the newly formed SH

groups provides the basis for visualization by streptavidin-

based colorimetric detection (applying S-nitrosylation

detection reagent I, including HRP, provided with the kit)

after running SDS-PAGE gels and transfer of the proteins

to membranes. For the detection of S-nitrosylated proteins,

the membranes were incubated overnight at 4 �C with

S-nitrosylation detection reagent I, 1:75. HRP signal

indicative of S-nitrosylation was detected with the EZ-

ECL-detection reagent while using the X-Omat blue XB-1

Kodak scientific Imaging Film.

S-nitrosylation of specific proteins was assayed by bio-

tin switch assay for protein S-nitrosothiols (SNOs), and

precipitation using resin-assisted capture (SNO-RAC) [44,

47]. Briefly, control wild type U118MG cells as well as

cells treated with 1 mM SNP (5, 10 and 15 min) were lysed

as described above. Then, 1 mg of lysed protein was

diluted in 1.25 ml final volume of HEN buffer (100 mM

Hepes, 1 mM EDTA, 0.1 mM neocuproine) containing

2.5 % SDS and 0.1 % MMTS. (Protein thiols are blocked

with MMTS). This ‘‘blocking’’ reaction was performed at

50 �C for 30 min with frequent agitation. After blocking,

proteins were precipitated with three volumes of cold

acetone and incubated at -20 �C for 20 min. The mixture

was then centrifuged at 20009g for 5 min, washed three

times with 70 % acetone and re-suspended in HENS buffer

(HEN containing 1 % SDS). This material was added to

50 ll of thiol-reactive resin slurry (Thiopropyl Sepharose

6B) in the presence of 20 mM sodium ascorbate (ascorbate

converts SNOs to free thiols). Following rotation in the

dark overnight, the resin was washed 4 9 with 1 ml HENS

buffer, and then 2 9 with 1 ml HENS/10 buffer (HENS

diluted 1:10). Nascent thiols are covalently trapped with

the resin-bound 2- or 4-pyridyl disulfides. Captured pro-

teins were eluted with 40 ll HENS/10 containing 100 mM

2-mercaptoethanol for 20 min at room temperature, and

30 ll each eluant mixed with 10 ll of 29 sample buffer to

be used for SDS-PAGE. Positive and negative controls

were also included in the SDS-PAGE. For positive con-

trols, cells were treated with 1 mM of the robust S-nitro-

sylation promoter CysNO (instead of SNP) for 10 min at

37 �C and 5 % CO2 as described previously [44, 47]. Non-

ascorbate samples were prepared in parallel with the SNO-

RAC assays to serve as negative controls for each sample

to avoid false positives. Specific anti-bodies were applied

to the proteins transferred to the membranes to detect

S-nitrosylated forms of TSPO, VDAC, and b-actin, as

described in the section western blot. In addition to the

immunoblots of the membranes, we also applied

652 Apoptosis (2012) 17:647–665

123

Page 7: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

Coomassie blue staining to the SDS-PAGE gels with sol-

ubilized proteins collected before pull down of S-nitrosy-

lated proteins (starting material), as well as after pull down

of S-nitrosylated proteins (Protein-SNO). Coomassie blue

staining of gels with S-nitrosylated proteins collected after

pull down was done to determine the range of molecular

weights of S-nitrosylated proteins by SNP treatment, in

comparison to the controls.

Statistical analysis

Sufficiently large experimental and control groups (n C 4)

were used for each assay. Results are expressed as

means ± SD. One-way analysis of variance (ANOVA)

was performed. Bartlett’s test for homogeneity of variance

was done. When appropriate, Kruskal–Wallis non-para-

metric ANOVA, was performed. For parametric post hoc

analysis Tukey–Kramer was applied. The non-parametric

Mann–Whitney or Dunn’s multiple analysis comparison

were used as post hoc tests, depending on the sample sizes

(for n \ 5 Mann–Whitney was used exclusively). When

comparing only between two groups, depending on the

significant difference between the SDs, Mann–Whitney or

Student’s t test were performed as appropriate. p \ 0.05

was considered statistically significant. The program used

for statistical analysis was Instat version-2 (GraphPad

Software, San Diego, CA).

Results

The two different NO donors, GSNO (500 lM) and SNP

(1,000 lM) strongly enhanced DNA fragmentation levels

measured with the Cell Death Kit indicative of apoptosis in

the two cell lines, U118MG (human) and PC12 (rat)

(Fig. 1a, b). In contrast to SNP, applying 1,000 lM of

GSNO to PC12 cells caused full cell detachment and

rounding of the cells, while apoptotic levels appeared lower

than observed with 500 lM (data not shown). Thus, as the

effects of SNP were well controllable, reliable, and well

distinguishable both from negative control as well as

positive control (Fig. 1c–i), SNP was the NO donor of

choice of the present study. Cell viability of U118MG and

PC12 was also assayed with the LDH kit after application

of SNP (500 lM and 1,000 lM) (Fig. 1c, d). In particular,

applying 1,000 lM SNP on PC12 and U118MG cells

resulted in a twofold and fivefold increase in cell death,

respectively, compared to vehicle control. Compared to the

positive control (1 % Triton treatment causing 100 % cell

death), applying 1,000 lM of SNP to PC12 and U118MG

cells achieved 49 and 53 % cell death, respectively

(Fig. 1c, d). 500 lM SNP was not toxic at all in PC12

cells, but still caused a fourfold increase in cell death in

U118MG cells, compared to vehicle control, i.e. attaining

about 42 % cell death compared to the positive control

(Fig. 1c, d). Assaying cell viability with Trypan blue dye

showed that SNP at 1,000 lM caused 26 and 46 % cell

death of the total cell population in PC12 and in U118MG

cells, respectively (Fig. 1e, f). The vehicle treated controls

only displayed 4 and 5 % cell death, respectively. Mea-

suring cell viability applying PI uptake with the aid of

FACS, also showed that U118MG cells are sensitive to

SNP. For this PI assay, U118MG cells were treated with

three different concentrations of SNP (500, 750, and

1000 lM) (Fig. 1g–i). Representative effects of SNP

(1,000 lM) treatment on cell death and its control detected

with PI in U118MG cells are presented in Fig. 1g, h. In

these examples, in the control only 6 % of the cells present

PI inclusion (Fig. 1g), while in the SNP treated cells 44 %

of the cells present PI inclusion (Fig. 1h). In Fig. 1i is

shown that average cell death levels were 8 % in vehicle

treated cell cultures as determined with the PI analysis.

SNP causes cell death in a dose-dependent manner

(Fig. 1i). As with the other assays, the PI inclusion showed

that 1,000 lM of SNP caused robust cell death of the total

cell population of U118MG (average of 46 %). Thus,

comparing these effects of SNP and GSNO makes clear

that U118MG cells exposed to 1,000 lM of SNP provide

the most clear cut results amenable to further assays. As

mentioned above, 1,000 lM of GSNO did not provide well

interpretable results. We also took into consideration that

the U118MG cell line was more sensitive to SNP than the

PC12 cell line (Fig. 1c, b). Therefore, we continued

working with the U118MG cells, applying the most

effective concentration of SNP (1,000 lM). We chose this

dose of SNP resulting in a level of cell death (46 %) that

would allow for up-regulation as well as down-regulation

of cell death.

Figure 2a shows that the application of 1,000 lM of the

NO donor SNP to U118MG cells causes a significant

decrease in signal of XTT reduction (by 71 %) indicative

of compromised metabolic activity (and/or a reduction in

cell number). This is in accord with the viability assays

showing reduced cell viability after SNP exposure (46 %

cell death, Fig. 1f). Furthermore, JC-1 assays with FACS

indicate significantly enhanced incidence of collapse of the

DWm by SNP, i.e. in U118MG cells exposed to 1,000 lM

of SNP maintenance of the DWm was only 30 % of that

observed in vehicle control (Fig. 2b).

As hypothesized in the ‘‘Introduction’’, we found that

the specific TSPO ligand PK 11195 attenuates cell death

induced by the NO donor SNP and this reduction in cell

death by PK 11195 is significant (n = 4, p \ 0.05), as

observed by Trypan blue uptake (Fig. 3a). In particular,

U118MG cells treated with 1,000 lM of SNP showed

46 % cell death, but when 25 lM of PK 11195 was added

Apoptosis (2012) 17:647–665 653

123

Page 8: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

to SNP treated cells, the cell death rate went down to 27 %.

In the vehicle control groups the cell death levels were 5 %

of the total population. Similar to the effects observed with

Trypan blue, PI uptake indicated that PK 11195 added to

the SNP-treated cells significantly reduced cell death rate

from 44 to 32 % (Fig. 3b). In the vehicle control cell death

levels were 6 % as determined with PI inclusion.

As expected, micrographs of the cells (Fig. 4) demon-

strated morphological signs of apoptosis induced by SNP,

including rounding and blebbing of cells (Fig. 4b), while in

vehicle treated cells the number of apoptotic cells was low

(Fig. 4a). With addition of PK 11195 to SNP exposed cells

there was a decrease in the number of the apoptotic cells in

comparison to those exposed to SNP without PK 11195

(Fig. 4c). In accord with the microscopic observations,

DNA fragmentation measured by the Cell Death Kit

demonstrated that addition of PK 11195 to SNP exposure,

in comparison to SNP alone, reduced DNA fragmentation

levels by 42 % (Fig. 5a). Also in this case, application of

PK 11195 significantly counteracted the decrease in

Fig. 1 Cell death induced by the NO donors GSNO and SNP in PC12

and U118MG cells. In (a) and (b) the y-axes present optical density

(OD) marking DNA fragmentation as an indicator for apoptosis levels

caused by GSNO (500 lM) and SNP (1,000 lM) applied for 24 h to

PC12 cells (a) and U118MG cells (b). In (c) and (d) is shown that

SNP (at 500 and 1,000 lM for 24 h) induces cell death in PC12 and

U118MG cell lines as measured by LDH release. The positive control

is cells treated with medium containing 1 % Triton which caused total

cell lysis. The y-axes in (c) and (d) mark the OD levels of the LDH

signal. In (e) and (f) Trypan blue uptake is used to indicate induction

of cell death by the NO donor SNP in PC12 cells (e) and in U118MG

cells (f). The y-axes in (e) and (f) mark the percentage of Trypan blue

positive cells from total population of cells. In (g), (h), and (i), PI

uptake indicates SNP induced cell death in U118MG cells. g,

h Representative FACS results of PI uptake: g vehicle treated cells;

h cells treated with SNP (1,000 lM for 24 h). M1 in (g) and

(h) represents the live cells and M2 represents the dead cells. i Cell

death induction of U118MG cells by different concentrations of the

NO donor SNP is shown, as determined by PI inclusion. The y-axis

marks the percentage of dead cells having taken up PI. The x-axis

marks SNP concentrations (0, 500, 750, and 1000 lM for 24 h).

*p \ 0.05, **p \ 0.01, and ***p \ 0.001 versus vehicle control.

(‘‘0’’ and ‘‘control’’ is vehicle control as indicated in the panels).##p \ 0.01 and ###p \ 0.001 versus SNP 500 lM (n = 6 for all

groups)

654 Apoptosis (2012) 17:647–665

123

Page 9: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

metabolic activity due to SNP in U118MG cells as mea-

sured with XTT, virtually restoring the normal level of

metabolic activity (Fig. 5b). SNP by itself caused a 73 %

reduction in metabolic activity. Assays of the collapse of

the DWm with the aid of JC-1 showed that the incidence of

DWm collapse with SNP alone was 72 %, while SNP in

combination with PK 11195 showed only 46 % incidence

of DWm collapse (Fig. 5c).

The role of TSPO in the lethal effects of SNP was fur-

ther checked by using TSPO knockdown. Viability assays

were performed on TSPO knockdown cells, i.e. TSPO

siRNA U118MG cells (siRNA) and on sham control cells,

i.e. scrambled control U118MG cells (Scr). siRNA cells

have only about 30 % of the TSPO protein levels observed

in Scr cells [18]. In accord with this previous report

applying immunoblot, the present study applying RT-PCR

assays showed that gene expression of the TSPO was

reduced to 20 % of the levels detected in Scr control cells

(n = 6, p \ 0.01, for each assay), as described also else-

where [60]. Our present results show that TSPO knock-

down counteracts cell lethality induced by 1,000 lM SNP

(Fig. 6). The Trypan blue assays showed that Scr cells

treated with 1,000 lM of SNP presented a more than

twofold increase in cell death compared to Scr vehicle

control, whereas cell death of siRNA cells was not sig-

nificantly increased (Fig. 6a). Similar results were found

with application of 750 lM of SNP [61]. Furthermore,

while the LDH assay showed a more than twofold increase

in cell death of SNP-treated Scr cells, compared to the Scr

control cells, only negligible effects of SNP were observed

in TSPO knockdown cells (Fig. 6b). The results of PI

uptake also showed that TSPO knockdown protects against

cell death induced by SNP. In particular, TSPO knockdown

caused a twofold reduction in cell death otherwise induced

by 1,000 lM of SNP, compared to sham control (Fig. 6c).

The results in Fig. 6d show that 1,000 lM of SNP caused a

twofold increase in DNA fragmentation levels in Scr cells,

compared to vehicle treated control, while in TSPO

knockdown cells (siRNA), no such effect could be dis-

cerned whatsoever (Fig. 6d).

Application of the NO donor SNP also had effects on

various aspects of mitochondrial function under TSPO

control assayed in this study (Fig. 7). Scr cells treated with

SNP show a significant reduction of 80 % in their meta-

bolic activity compared to their vehicle control, as detected

with the XTT assay (Fig. 7a). In contrast, TSPO knock-

down siRNA cells did not show any difference between

vehicle control and SNP treated cells in this respect

(Fig. 7a). Applying the JC-1 assay, Scr cells exposed to

SNP showed a significant reduction, of more than 50 %,

regarding the incidence of DWm collapse compared to their

vehicle control, while siRNA cells showed no significant

difference between SNP treatment and vehicle control,

Fig. 2 SNP affects mitochondrial function in U118MG cells. a SNP

(1,000 lM) causes a decrease in mitochondrial activity in U118MG

cells, as determined with XTT assays (n = 8). In (a) the y-axis

indicates OD levels as a measure for relative mitochondrial activity.

b SNP (1,000 lM) causes collapse of the DWm as measured by JC-1

fluorescence (n = 6). In (b) the y-axis indicates the drop in red/green

fluorescence ratio of SNP treated cells normalized to control (set at 1),

which is indicative for the incidence of collapse of the DWm.

***p \ 0.001 for SNP treatment versus vehicle control (control)

Fig. 3 PK 11195 reduces lethal effects of SNP. U118MG cells were

treated with 1,000 lM of SNP with or without 25 lM of PK 11195

(PK) for 24 h, or with vehicle (control). a Lethal effects of SNP and

protective effects of PK 11195 as measured by Trypan blue uptake.

The y-axis indicates % of cells that were Trypan blue positive per

sample (n = 4 for all groups in a). b The lethal effects of SNP and

protective effects of PK 11195 as measured by PI uptake. The y-axis

indicates the % of cells that were PI positive per sample (n = 6 for all

groups in b). *p \ 0.05 for SNP treatment versus vehicle control;#p \ 0.05 for SNP in combination with PK 11195 (SNP ? PK)

versus SNP alone (n = 6 for all groups)

Apoptosis (2012) 17:647–665 655

123

Page 10: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

regarding modulation of the DWm (Fig. 7b). SNP-treated

Scr cells showed a significant (70 %) increase in mito-

chondrial ROS generation (as assayed by NAO), as com-

pared to the Scr control cells, while TSPO knockdown

siRNA cells receiving the same treatment were not affected

(Fig. 7c). Representative examples of NAO assays are

shown in Fig. 7d. The data on metabolic activity, modu-

lation of the DWm, and mitochondrial ROS generation as a

consequence of SNP exposure (Fig. 7) are in accord with

the cell viability assays employing Trypan blue, PI, LDH,

and DNA fragmentation assays (Fig. 6).

In order to check whether the NO donor SNP, at a

concentration of 1,000 lM, had an effect on TSPO

expression, we performed binding analyses including

maximal binding capacity (Bmax) and the equilibrium dis-

sociation constant (Kd) of [3H]PK 11195 as well as western

blot analysis in normal U118MG cells. Binding analysis

showed that Bmax and Kd for SNP-treated cells did not

differ significantly from the control (Table 2). Also with

western blot we could not detect differences in TSPO

expression in U118MG cells exposed to 1,000 lM of SNP

for 24 h compared to vehicle treated control cells (Fig. 8).

Neither did PK 11195 exposure, or the combination of PK

11195 with SNP, affect TSPO protein expression (Fig. 8).

In addition, displacement experiments were performed on

rat kidney homogenates in order to evaluate potential

binding of SNP or NO to the TSPO. This assay showed that

SNP was not able to displace the TSPO ligand [3H] PK

11195 (Ki [ 10,000 nM). These data indicate that SNP

application does not affect TSPO binding characteristics or

expression in cell culture, and that SNP or NO generated by

SNP does not bind to TSPO.

The biotin-switch technique (BST) (Figs. 9, 10) was used

to assay if S-nitrosylation of TSPO by SNP is implicated in

the activation of its pro-apoptotic functions. For this pur-

pose, we performed immunodetection by applying our

TSPO specific antibody to western blots of S-nitrosylated

proteins after treatments with SNP and application of the

positive control (the robust S-nitrosylation promoter, CysNO

[44, 47] (Fig. 9a). First of all, total TSPO levels (starting

material) were not affected by CysNO and SNP (Figs. 8,

9b). Remarkably, S-nitrosylation of TSPO by SNP was not

detected (Fig. 9a). However, the positive control (CysNO)

did show S-nitrosylation of TSPO (Fig. 9a). Omission of

ascorbate from the reaction mixture demonstrated the lack of

false positivity (Fig. 9a). We also did not detect S-nitrosy-

lation by SNP of VDAC, which is closely associated with

the TSPO (Fig. 9c, d). However, VDAC was S-nitrosylated

by the positive control (CysNO) (Fig. 9c). To our knowl-

edge there are no other reports specifically dealing with

potential S-nitrosylation of VDAC, although it possesses

several cysteine residues [62].

In Fig. 10a we show the general patterns of molecular

weights of S-nitrosylated proteins after application of SNP

(1 mM) for different time periods (5, 10, and 15 min), and

also after application of the NO donor of the positive control

(1 mM of CysNO for 10 min), by Coomassie blue staining

of S-nitrosylated proteins run on a 12 % polyacrylamide gel,

in comparison to vehicle control, as described in ‘‘Meth-

ods’’. Various proteins of different molecular weights were

S-nitrosylated by SNP, as compared with vehicle control

(Fig. 10a), while total protein levels (starting material) were

not affected by the different treatments (Fig. 10b). Further-

more, we found that the cytoskeletal protein b-actin was one

of the proteins that could be S-nitrosylated by SNP

(Fig. 10c). In particular, treatment with SNP resulted in

45 % more S-nitrosylated b-actin than observed in the

vehicle control (p \ 0.01, n = 6) (Fig. 10c). S-nitrosylation

of b-actin by the positive control treatment (CysNO) was 2.8

fold more than detected in the vehicle control (p \ 0.001

versus vehicle control cells) (Fig. 10c). Total protein level of

b-actin (starting material) was not affected by SNP or in the

A B C

Fig. 4 Micrographs of effects of SNP treatment of U118MG cells.

The arrows indicate rounding and blebbing of cells. a Vehicle treated

cells; b cells treated with 1,000 lM of SNP for 24 h; c cells treated

with 1,000 lM of SNP in combination with 25 lM of PK 11195.

Compared to vehicle control treated cells (a), the NO donor SNP

causes abundant rounding and blebbing of cells and reduces the cell

density (b), which is attenuated by the TSPO ligand PK 11195 (c)

656 Apoptosis (2012) 17:647–665

123

Page 11: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

positive control (CysNO) (Fig. 10d). Also for the non-TSPO

proteins, omission of ascorbate from the reaction mixture

demonstrated the lack of false positivity for S-nitrosylation

(Fig. 10a). Noteworthy, we also used the S-Nitrosylated

Protein Detection Assay Kit (Cayman), as described in

‘‘Methods’’, and with this technique we also found S-nitro-

sylation of various proteins after 24 h of treatment with

SNP.

Discussion

Cell death, including apoptosis, induced by various agents

such as CoCl2, erucylphosphohomocholine (ErPC3), etc.,

Fig. 5 PK 11195 (PK) counteracts induction of cell death related

processes otherwise induced by SNP in U118MG cells. a PK 11195

reduces measured DNA fragmentation compared to treatment with

SNP alone. The y-axis indicates DNA fragmentation levels (repre-

sented in OD) (n = 6 for all groups in a). b PK 11195 attenuates

decrease in measured mitochondrial activity otherwise caused by

SNP, as measured by XTT assays. The y-axis indicates mitochondrial

activity as determined with XTT assay (represented in OD) (n = 8 for

all groups in b). c PK 11195 reduces DWm collapse induced by SNP

as measured by JC-1 fluorescence. The y-axis indicates normalized

red/green fluorescence ratio of JC-1, whose reduction is indicative of

DWm collapse (n = 6 for all groups in c). Control is vehicle control

as indicated in each panel. #p \ 0.05 versus SNP; **p \ 0.01 versus

control. ***p \ 0.001 versus vehicle control

Fig. 6 TSPO knockdown counteracts cell lethality induced by SNP

in U118MG cells. a Trypan blue uptake shows that SNP does not

induce cell death in TSPO knockdown cells (siRNA), but does induce

cell death in the control cells (Scr) cells. In (a) the y-axis indicates the

percentage of Trypan blue positive cells from total population of

cells. b LDH release also indicates that SNP causes cell death in Scr

cells, but not in siRNA cells. c PI uptake also shows that TSPO

knockdown (siRNA) protects against cell death induced by SNP. In

(c) the y-axis indicates the relative levels of dead cells having taken

up PI. The results are normalized to vehicle control, which was set as

100 % in both Scr and siRNA. d TSPO knockdown prevents DNA

fragmentation induced by SNP. The y-axis indicates DNA fragmen-

tation levels (OD) as an indicator for apoptosis. In (a), (b), and

(d) control is vehicle control, *p \ 0.05 versus Scr control.

***p \ 0.001 versus Scr (n = 6 for all groups)

Apoptosis (2012) 17:647–665 657

123

Page 12: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

includes activation of the TSPO leading to ROS generation

at mitochondrial levels, oxidation of cardiolipins, collapse

of the Dwm, caspase activation, and induction of DNA

fragmentation as components of the mitochondrial apop-

tosis cascade in human glioblastoma U118MG cells, as

described previously [13, 16–18, 63]. These are processes

that are also induced by NO [64–67]. Furthermore, nNOS

may be linked to the TSPO via the scaffolding protein

CAPON, which binds to the G protein Dexras1, which in

turn binds to PAP7 [38, 39]. As this variety of lethal agents

(CoCl2, ErPC3, etc.) require the presence of TSPO to

induce cell death, we assumed that NO may be a common

mediator for all these lethal agents targeting the TSPO.

With the present study we showed that application of two

different NO donors, GSNO and SNP, induced apoptosis

both in U118MG human glioblastoma cells and in PC12 rat

pheochromocytoma cells. Because of the consistency of

the observed effects, we applied 1,000 lM of SNP to the

U118MG cell line for the most part of this study. The

concentrations and time periods of the NO donor applica-

tions used in the present study are as described previously

by others (see ‘‘Methods’’), and we found them to be

effective also in our pilot studies (e.g. Fig. 1). With this

approach we set out to determine potential interactions

between TSPO and NO in relation to cell death induced by

SNP. For this purpose, we applied TSPO knockdown and

their appropriate controls, and also assayed the effects of

the TSPO ligand PK 11195 on U118MG cells.

As the TSPO is able to activate the mitochondrial

apoptosis cascade, we decided to assay various aspects of

mitochondrial functions as well as indicators of cell death

after application of SNP. With the aid of the XTT kit, we

measured metabolic activity, which is also indicative to

cell proliferation and viability according to the manufac-

turer of the kit (Biological Industries). This showed that

SNP caused a reduction of 73 % in XTT signal. As the cell

death levels (46 % of the total population), measured with

Trypan blue and PI uptake, do not fully account for the

73 % reduction in XTT signal, we assume that SNP

application also affects metabolic processes in living cells,

in addition to cell death induction in our cell cultures. We

found here that SNP causes collapse of the DWm, which is

known to initiate cell death processes [35, 68–70]. TSPO

also is able to regulate the DWm [12, 13, 17, 18, 57]. With

the present study, we found that the TSPO specific ligand

PK 11195 was able to significantly reduce the cell death

levels effects of SNP on U118MG cells. The concentration

of PK 11195 (25 lM) used is the same concentration that

Fig. 7 Effects of SNP on mitochondrial functions is prevented by

TSPO knockdown in U118MG cells. a XTT assays show that in

siRNA cells SNP does not affect mitochondrial activity, as it does in

Scr cells. The y-axis indicates OD levels of XTT product, as an

indicator for relative mitochondrial activity (n = 8 for all groups).

b JC-1 fluorescence indicates that TSPO knockdown attenuates DWm

collapse, as it is normally induced by SNP in Scr cells. The y-axis

indicates red/green fluorescence ratio marking JC-1 polymerization

normalized to control, which reduction is indicative for the incidence

of collapse of the DWm (n = 6 for all groups). c NAO fluorescence

indicates that TSPO knockdown prevents cardiolipin oxidation

induced by SNP, as it normally occurs in Scr cells with SNP

exposure. The y-axis indicates normalized NAO signal for oxidized

cardiolipins (M1 in d), which is indicative to mitochondrial ROS

generation. Vehicle treated control groups for both siRNA and Scr

cells were set at ‘‘1’’ (n = 9 for all groups). d Representative FACS

results of NAO signal vehicle treated Scr cells (left hand figure); Scr

cells treated with SNP (1,000 lM) (right hand figure); M1 in each

histogram indicates the population of cells showing low intensity

NAO labeling, representing the % of cells displaying oxidized

cardiolipins. In (d) the x-axes indicate fluorescence intensity of NAO

signal, and the y-axes indicate the number of cells related to the

measured fluorescence intensity. ***p \ 0.001 versus Scr control;

**p \ 0.01 versus Scr control; *p \ 0.05 versus Scr control

b

658 Apoptosis (2012) 17:647–665

123

Page 13: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

showed protection against lethal effects of ErPC3 and

CoCl2 in these cells in previous studies [13, 17, 18]. In

contrast to the isoquinoline, PK 11195, which has high

affinity for the TSPO in all species studied so far, the

benzodiazepine Ro5 4864 shows only a low affinity for the

TSPO in the human brain and glioma (as well as kidney

and colon) [71, 72]. In particular, while [3H]PK 11195

binds with high affinity (Kd of about 2 nM) to human

cerebral cortex, kidney, and colon membranes, the specific

binding of [3H]Ro5 4864 is barely detectable (nonspecific

binding about 90 % of the total binding) [71]. Furthermore,

unlabeled PK 11195 is two orders of magnitude more

potent than unlabeled Ro5 4864 in displacing [3H]PK

11195 specific binding from human cerebral cortex and

kidney membranes [71]. Thus, we assume that the high

affinity of the isoquinoline PK 11195 for TSPO in human

cells is responsible for PK 11195’s ability to prevent the

lethal effects of the NO donor SNP in U118MG cells. In

another study, applying benzodiazepines, we found that the

TSPO ligand Ro5 4864 did not significantly modulate

effects of SNP in the human U118MG cells [61]. Treat-

ment with the benzodiazepine clonazepam, which is a CBR

specific ligand, also did not show any significant effect in

this regard [61]. Clonazepam is well known to not bind to

TSPO.

Apart from preventing cell death induced by SNP, PK

11195 also reduced metabolic activity, and collapse of the

DWm otherwise induced by SNP. Collapse of the DWm,

leading to cytochrome c release and the activation of cas-

pase-3 and caspase-9, is one of the initiating steps for the

mitochondrial apoptosis cascade [64, 68, 73]. Other studies

Table 2 Effects of the NO donor SNP on TSPO binding characteristics in U118MG cells

U118MG cells SNP treatment Vehicle treatment

Bmax 2146 ± 675 (fmol/mg protein) 2040 ± 424 (fmol/mg protein)

Kd 4.4 ± 2.0 nM 3.1 ± 1.2 nM

TSPO binding characteristics (Bmax and Kd) were determined with binding assays applying [3H]PK 11195 (n = 9 for all assays)

Control SNP + PK

β-Actin (42 KDa)

TSPO (18 KDa)

SNPPK

A

B

Fig. 8 Western blot of 18 kDa TSPO protein expression in U118MG

cells after exposure for 24 h to 1 mM of SNP (SNP), 25 lM of PK

11195 (PK), and the combination of 1 mM of SNP and 25 lM of PK

11195 (SNP ? PK). a A representative western blot of 18 kDa TSPO

protein expression. b-Actin was used as a loading control. b A bar

graph presenting the relative 18 kDa TSPO levels. No significant

differences were observed between the different types of exposure.

Control is application of vehicle

Fig. 9 Lack of TSPO and VDAC S-nitrosylation due to SNP (1 mM)

treatment of U118MG cells. Omission of ascorbate is applied as a

negative control (-), as described in ‘‘Methods’’. (?) Underneath

blots indicates the regular procedure with addition of ascorbate. As

positive control, cells are exposed to 1 mM of CysNO. a Represen-

tative western blot showing that SNP is not able to S-nitrosylate

18 kDa TSPO, while the positive control (CysNO) does S-nitrosylate

18 kDa TSPO. b Representative western blot showing that neither

SNP, nor the positive control CysNO, do affect normal 18 kDa TSPO

levels (starting material). c Representative western blot showing that

SNP is not able to S-nitrosylate VDAC, while the positive control

(CysNO) does S-nitrosylate VDAC. d Representative western blot

showing that neither SNP, nor the positive control CysNO, do affect

normal VDAC levels (starting material)

Apoptosis (2012) 17:647–665 659

123

Page 14: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

applying TSPO knockdown and TSPO ligands have shown

that the occurrence of these events of the mitochondrial

apoptosis cascade can be modulated by the TSPO [12, 13,

16–18, 74, 75]. For the present study, we also used TSPO

siRNA knockdown U118MG cells in comparison to their

sham controls (Scr), prepared previously in our laboratory

[18]. Comparable to the effects of PK 11195, our various

assays to test cell death, and cell death related processes,

showed that TSPO knockdown also counteracts cell

lethality induced by SNP. Thus both approaches for mod-

ulation of TSPO activity, i.e. application of the TSPO

ligand PK 11195 as well as TSPO knockdown by genetic

manipulation, implicate that TSPO is involved in the lethal

and metabolic effects of the NO donor SNP.

Interestingly, we found that applying 25 lM of PK

11195 to TSPO knockdown cells did not have any effect on

cell lethality, collapse of the DWm, metabolic activity, or

mitochondrial ROS generation, still occurring to TSPO

Fig. 10 Protein S-nitrosylation due to SNP (1 mM) treatment of

U118MG cells. Omission of ascorbate is applied as a negative control

(-), as described in ‘‘Methods’’. (?) Underneath blots indicates the

regular procedure with addition of ascorbate. As positive control,

cells are exposed to 1 mM of CysNO. a Coomassie stained gel

showing that CysNO (1 mM) exposure for 10 min (as positive

control) and SNP exposure for different time periods (5, 10 and

15 min) do S-nitrosylate proteins (Protein-SNO) of U118MG cells.

b Coomassie stained gel showing that SNP does not affect general

protein levels (starting material) in U118MG cells. Also the positive

control (CysNO) does not affect general protein levels (starting

material). In (a) and (b) the MW markers of the Precision Plus

ProteinTM Standards, KaleidoscopeTM (Bio-Rad) are presented on the

left hand sides of the gels. c Representative western blot showing that

SNP can cause S-nitrosylation of b-actin, displayed at the bottom ofthe figure. The bar chart presents the relative optical density of bands

labeled for S-nitrosylated b-actin showing that SNP, as well as the

positive control (CysNO), can cause S-nitrosylation of b-actin. The

y-axis indicates in arbitrary units the optical density of Actin-SNO

labeled bands. d Representative western blot indicating that SNP and

CysNO do not affect normal b-actin levels (Starting material).

**p \ 0.01 versus vehicle control. Vehicle = vehicle control. ‘‘-’’

Underneath blots indicates omission of ascorbate (negative control).

‘‘?’’ Underneath blots indicates inclusion of ascorbate (regular

procedure). TSPO-SNO, b-actin-SNO, and Protein-SNO are S-nitro-

sylated TSPO, b-actin, and total protein, respectively. 1 mM of SNP,

and 1 mM of CysNO as positive control, was applied for each assay

660 Apoptosis (2012) 17:647–665

123

Page 15: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

knockdown cells exposed to SNP. This makes sense as PK

11195 is a TSPO specific ligand and our siRNA U118MG

TSPO knockdown cells display reduced levels of TSPO

expression ([18]; present study). This point is not irrelevant

as PK 11195 is used at a concentration high above its

dissociation constant (Kd) for the TSPO (25 lM vs.

±2.5 nM). An interesting question is why concentrations

of PK 11195, well above its Kd for TSPO, are needed to

induce TSPO specific effects. We assume that such high

concentrations of PK 11195 may be needed to compete

successfully with endogenous TSPO ligands potentially

having higher affinity for the TSPO than PK 11195. In a

generalized postulation, we suggest that TSPO specific

ligands, such as PK 11195, at concentrations up to 50 lM

can act as antagonists. Here we mean with antagonist the

capability to attenuate TSPO’s pro-apoptotic function. At

higher concentrations still, however, PK 11195 can induce

cell death, and we suggest that the latter is due to non-

TSPO specific effects. This postulation is presented by

various experimental studies by us targeting this question,

as well as by an extensive overview of experimental studies

by others [12–18]. We believe that endogenous activators

of the TSPO are able to induce TSPO’s pro-apoptotic

function, which appears to be blocked by TSPO knock-

down and synthetic TSPO ligands such as PK 11195.

Further studies are required to obtain better insights into

this possibility.

A priori, we assumed that NO, as generated for

example by SNP, acts directly on the TSPO. However, we

found that SNP did not affect TSPO’s expression or

binding characteristics, neither did SNP interfere with

[3H]PK 11195 binding to TSPO. Thus, SNP neither did

appear to damage the TSPO nor to interact directly via a

ligand binding mechanism with the TSPO. Nonetheless,

applying TSPO knockdown and the TSPO ligand PK

11195, our study shows that SNP application causes sev-

eral effects that are dependent on the presence of the

TSPO. Thus, we assumed that SNP may cause TSPO

S-nitrosylation. It is well known that S-nitrosylation can

affect protein function [31, 76–78]. In addition, co-local-

ization of NOS enzymes with target proteins, including

direct interactions, may be an important determinant of

S-nitrosylation [31]. As NOS may be closely associated

with the TSPO [38, 39], we assumed that NO generated by

NOS may affect TSPO function. Furthermore, human

TSPO possesses two cysteine residues, one at the N-ter-

minus and the other one at the C-terminus, therefore we

considered it likely that the TSPO can be nitrosylated [79,

80]. Such a nitrosylation of the TSPO would affect

TSPO’s interactions with molecules at both sides of the

outer mitochondrial membrane, i.e. it would affect TSPO

function. With the present study we found that the NO

donor SNP causes S-nitrosylation of various proteins,

including b-actin. Thus, our study suggests that S-nitro-

sylation of various proteins due to SNP exposure may

contribute to the induction of cell death by SNP. However,

S-nitrosylation of TSPO by SNP did not appear to occur.

Nonetheless, application of CysNO as a positive control

for detection of S-nitrosylation of TSPO showed that

TSPO in U118MG cells can be nitrosylated. In any case,

TSPO appears to be relatively resistant against S-nitrosy-

lation due to application of SNP. Thus, it appears that

S-nitrosylation of the TSPO is not required to allow TSPO

to initiate cell death processes. Noteworthy, the other cells

of our study, PC12 cells, are from rat, where TSPO does

not possess cysteine residues and thus cannot be S-nitro-

sylated [81]. Nevertheless, the effects of SNP on rat PC12

cells are not much different from what we observed in the

human U118MG cells (Fig. 1). This indicates that indeed

S-nitrosylation of the TSPO is not required for SNP to

exert its effects observed in the present study. It appears

that S-nitrosylation of proteins other than TSPO may lead

to activation of the TSPO, although at present it is not

clear how. Interestingly, Dexras1, which presents a link

between NOS and TSPO, can be nitrosylated [82]. Our

experiments, assaying protein S-nitrosylation, demon-

strated in our paradigm that protein S-nitrosylation takes

place within a short time after NO donor application, and

subsides after that. Thus, we assume that SNP in our

model affects the cell machinery soon after application,

for example by nitrosylating various proteins. This then

over an extended period of time leads to cell death

including apoptosis. Apart from inducing S-nitrosylation

of various proteins, the NO donor SNP can also be cyto-

toxic by other means, for example due to cyanide pro-

duction, as a consequence of SNP degradation [83]. This,

however, requires light [84]. Our experiments were per-

formed in the dark, excluding the possibility of cell death

induction due to degradation products of SNP. Interest-

ingly, in the light, our application of SNP did not induce

cell death (unpublished results). We assumed that degra-

dation of SNP in the light prevented NO production by

SNP and thereby pre-empted cell death induction due to

SNP application. In any case, our experiments performed

in the light do not support the possibility that cyanide

products due to SNP application present the major cause

of cell death detected under conditions of darkness.

Nonetheless, SNP was not able to S-nitrosylate TSPO in

our model. We consider the possibility that NO generated

by NOS may be more effective for S-nitrosylation of

TSPO than can be achieved with SNP. Interestingly, NOS

can be found in the vicinity of the TSPO [38, 39]. How-

ever, we have found that glutamate applied at toxic levels

([25 mM) to U118MG cells induces S-nitrosylation of

b-actin, but not of TSPO (unpublished results). Furthermore,

applying TSPO knockdown to these cells prevents the

Apoptosis (2012) 17:647–665 661

123

Page 16: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

toxic effects of glutamate (unpublished results). It is well

known that glutamate can activate NOS to generate NO

leading to S-nitrosylation of various proteins [85]. One of

the proteins that can be S-nitrosylated in this way is

Dexras1, which is closely associated with the TSPO [38,

39, 82]. Also by applying NH4Cl as a toxic agent we could

induce cell death and S-nitrosylation of various proteins,

but no nitrosylation of the TSPO (unpublished results). It

is known that NH4Cl can cause collapse of the DWm via

NOS activation [86]. Thus, SNP, glutamate, and NH4Cl

can cause cell death via S-nitrosylation of proteins, and

this induction of cell death can be prevented by deacti-

vation of the TSPO. Other proteins than Dexras1 that are

known to interact with the TSPO are: VDAC, ANT,

PRAX-1, PAP7, and pk 10 [4, 12, 13]. VDAC, however,

which is also closely associated with the TSPO, was not

S-nitrosylated by our application of SNP. To our knowl-

edge, S-nitrosylation of VDAC is not considered a major

function modifier for this protein. We are also not aware

of studies addressing S-nitrosylation of ANT, PRAX-1,

PAP7, and pk 10. The question remains by which mech-

anism the NO donor SNP (and other agents such as

glutamate, NH4Cl, CoCl2, and ErPC3) modulate mito-

chondrial function and cell death, i.e. the question remains

which agent(s) interact immediately with the TSPO to

activate its cell death function. Apart from potential

S-nitrosylation of proteins closely associated with TSPO,

it may be that steroids regulate TSPO functions, as various

steroids are known to affect TSPO expression in vivo [11,

12]. Recent data by us suggest that dexamethasone may

reduce TSPO expression in T98G cells, in correlation with

reduction in cell death levels otherwise induced by various

agents, such as SNP, CoCl2, and ErPC3 (unpublished

results). Note that SNP, CoCl2, and ErPC3 do require the

TSPO to induce cell death [17, 18, and the present study].

Thus, it cannot be excluded that locally produced steroids

may affect TSPO expression in relation to cell death

induction. Compounds we have also considered are por-

phyrins, which are putative endogenous TSPO ligands

[87], however, it appears that interactions between por-

phyrins and TSPO primarily serve to regulate porphyrin

levels rather than that porphyrins regulate TSPO’s cell

death functions [88–92].

Conclusion

In the present study we show that the activation of the

TSPO due to application of NO donors, in particular SNP,

leads to cell death including collapse of the DWm, ROS

generation at mitochondrial levels, cardiolipin oxidation,

DNA fragmentation, and rounding and blebbing of cells

(Fig. 11). The results of this study indicate conclusively

that the TSPO is involved in cell death caused by the NO

donor SNP, as well as SNP effects on mitochondrial

functions. Our results suggest that the cell death inducing

effects of the NO donor SNP is due to S-nitrosylation of

various proteins other than the TSPO (Fig. 11). Other

cytotoxic effect of SNP, however, cannot be excluded. A

question remaining is how in the end TSPO is activated by

SNP (or other lethal agents) to induce cell death; for

example: is an endogenous TSPO ligand released, or do

proteins typically in conjugation with the TSPO regulate its

function. It for example may be that S-nitrosylation of

Dexras1, a protein associated with TSPO, may contribute

to the effects of SNP operated by way of TSPO activation.

Other proteins closely associated with the TSPO are

VDAC, ANT, pk 10, PAP7, and PRAX-1 (see reviews, [4,

12]). Alternatively, regulation of TSPO gene expression,

including post-transcriptional processes, may be the driv-

ing force to induce its capability to initiate cell death

processes. In any case, the presence of the TSPO is spe-

cifically required to achieve at least part of the lethal and

metabolic effects of exposure to the NO donor SNP.

Fig. 11 The present study shows that the NO donor SNP is able to

S-nitrosylate proteins, inducing TSPO to activate the mitochondrial

apoptosis cascade, including mitochondrial ROS generation, cardio-

lipin oxidation, and DWm collapse. The DWm collapse may also

contribute to cell death other than apoptosis. While activation of the

TSPO apparently does not require TSPO’s S-nitrosylation, TSPO still

is required for cell death to occur in this experimental model. Which

exactly are the intermediate agents between general protein S-nitro-

sylation and activation of the TSPO in this experimental model is

unknown. Steroids and/or proteins closely associated with TSPO, and/

or regulation of gene expression of the TSPO can be suggested

662 Apoptosis (2012) 17:647–665

123

Page 17: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

Acknowledgments Moran Benhar and the co-workers of his labo-

ratory are thankfully acknowledged for their help with the S-nitro-

sylation assays. The expert assistance of Ilana Spanier and Esther

Messer is acknowledged. Research support was provided by: Nie-

dersachsen-Israel Project (MG, LV, JB), Technion-John Hopkins

Joint Research Program (S-HS, MG, LV), Johnson & Johnson (MG,

LV), L. Aronberg Research Fund in Neurology (MG, LV), Sch-

warzbach Medical Research Fund (MG, LV). Dr. Beatriz Caballero

thankfully acknowledges her post-doctoral fellowship (Programa

Cların) from FICYT (Gobierno del Principado de Asturias), Spain.

Dr. Leo Veenman acknowledges his support by a joint grant from the

Center for Absorption in Science of the Ministry of Immigrant

Absorption and the Committee for Planning and Budgeting of the

Council for Higher Education under the framework of the KAMEA

Program.

Conflict of interest The authors declare that they don’t have con-

flict of interest.

References

1. Braestrup C, Squires RF (1977) Specific benzodiazepine recep-

tors in rat brain characterized by high-affinity (3H) diazepam

binding. Proc Natl Acad Sci USA 74:3805–3809

2. Gavish M, Bachman I, Shoukrun R, Katz Y, Veenman L, Wei-

singer G et al (1999) Enigma of the peripheral benzodiazepine

receptor. Pharmacol Rev 51:629–650

3. Veenman L, Gavish M (2000) Peripheral-type benzodiazepine

receptors: their implication in brain disease. Drug Dev Res

50:355–3570

4. Veenman L, Gavish M (2006) The peripheral-type benzodiaze-

pine receptor and the cardiovascular system. Implications for

drug development. Pharmacol Ther 110:503–524

5. Papadopoulos V, Baraldi M, Guilarte TR, Knudsen TB, Lacapere

JJ, Lindemann P et al (2006) Translocator protein (18 kDa): new

nomenclature for the peripheral-type benzodiazepine receptor

based on its structure and molecular function. Trends Pharmacol

Sci 27:402–409

6. Cosenza-Nashat M, Zhao ML, Suh HS, Morgan J, Natividad R,

Morgello S et al (2009) Expression of the translocator protein of

18 kDa by microglia, macrophages and astrocytes based on

immunohistochemical localization in abnormal human brain.

Neuropathol Appl Neurobiol 35:306–328

7. Anholt RR, Pedersen PL, De Souza EB, Snyder SH (1986) The

peripheral-type benzodiazepine receptor. Localization to the

mitochondrial outer membrane. J Biol Chem 261:576–583

8. Olson JMM, Ciliax BJ, Mancini WR, Young AB (1988) Presence

of peripheral-type benzodiazepine binding sites on human

erythrocyte membranes. Eur J Pharmacol 152:47–53

9. O’Beirne G, Woods M, Williams D (1990) Two subcellular

locations for peripheral-type benzodiazepine acceptors in rat

liver. Eur J Biochem 188:131–138

10. McEnery MW, Snowman AM, Trifiletti RR, Snyder SH (1992)

Isolation of the mitochondrial benzodiazepine receptor: associa-

tion with the voltage-dependent anion channel and the adenine

nucleotide carrier. Proc Natl Acad Sci USA 89:3170–3174

11. Veenman L, Gavish M (2012) The role of 18 kDa mitochondrial

Translocator Protein (TSPO) in programmed cell death, and effects

of steroids on TSPO expression. Curr Mol Med 12:398–412

12. Veenman L, Papadopoulos V, Gavish M (2007) Channel-like

functions of the 18-kDa translocator protein (TSPO): regulation

of apoptosis and steroidogenesis as part of the host-defense

response. Curr Pharm Des 13:2385–2405

13. Veenman L, Shandalov Y, Gavish M (2008) VDAC activation by

the 18 kDa translocator protein (TSPO), implications for apop-

tosis. J Bioenerg Biomembr 40:199–205

14. Veenman L, Alten J, Linnemannstons K, Shandalov Y, Zeno S,

Lakomek M et al (2010) Potential involvement of F0F1-ATP

(synth)ase and reactive oxygen species in apoptosis induction by

the antineoplastic agent erucylphosphohomocholine in glioblas-

toma cell lines: a mechanism for induction of apoptosis via the

18 kDa mitochondrial translocator protein. Apoptosis 15:753–768

15. Veenman L, Gavish M, Kugler W (in press) Apoptosis induction

by erucylphosphohomocholine via the 18 kDa mitochondrial

translocator protein: implications for cancer treatment. Antican-

cer Agents Med Chem

16. Levin E, Premkumar A, Veenman L, Kugler W, Leschiner S,

Spanier I et al (2005) The peripheral-type benzodiazepine

receptor and tumorigenicity: isoquinoline binding protein (IBP)

antisense knockdown in the C6 glioma cell line. Biochemistry

44:9924–9935

17. Kugler W, Veenman L, Shandalov Y, Leschiner S, Spanier I,

Lakomek M et al (2008) Ligands of the mitochondrial 18 kDa

translocator protein attenuate apoptosis of human glioblastoma cells

exposed to erucylphosphohomocholine. Cell Oncol 30:435–450

18. Zeno S, Zaaroor M, Leschiner S, Veenman L, Gavish M (2009)

CoCl2 induces apoptosis via the 18 kDa Translocator Protein

(TSPO) in U118MG human glioblastoma cells. Biochemistry

48:4652–4661

19. Carayon P, Portier M, Dussossoy D, Bord A, Petitpretre G, Canat

X et al (1996) Involvement of peripheral benzodiazepine recep-

tors in the protection of hematopoietic cells against oxygen rad-

ical damage. Blood 87:3170–3178

20. Stoebner PE, Carayon P, Casellas P, Portier M, Lavabre-Bertrand

T, Cuq P et al (2001) Transient protection by peripheral benzo-

diazepine receptors during the early events of ultraviolet light-

induced apoptosis. Cell Death Diff 8:747–753

21. Dawson VL, Dawson TM (1998) Nitric oxide in neurodegener-

ation. Prog Brain Res 118:215–229

22. Contestabile A, Bonanomi D, Burgaya F, Girault JA, Valtorta F

(2003) Localization of focal adhesion kinase isoforms in cells of

the central nervous system. Int J Dev Neurosci 21:83–93

23. Keynes RG, Garthwaite J (2004) Nitric oxide and its role in

ischaemic brain injury. Curr Mol Med 4:179–191

24. Duncan AJ, Heales SJ (2005) Nitric oxide and neurological dis-

orders. Mol Asp Med 26:67–96

25. Blokland A, Schreiber R, Prickaerts J (2006) Improving memory:

a role for phosphodiesterases. Curr Pharm Des 12:2511–2523

26. Calabrese LH, Dodick DW, Schwedt TJ, Singhal AB (2007)

Narrative review: reversible cerebral vasoconstriction syndromes.

Ann Int Med 146:34–44

27. Zhou L, Zhu DY (2009) Neuronal nitric oxide synthase: structure,

subcellular localization, regulation, and clinical implications.

Nitric Oxide 20:223–230

28. Saha RN, Pahan K (2006) Regulation of inducible nitric oxide

synthase gene in glial cells. Antioxid Redox Signal 8:929–947

29. Pannu R, Singh I (2006) Pharmacological strategies for the reg-

ulation of inducible nitric oxide synthase: neurodegenerative

versus neuroprotective mechanisms. Neurochem Int 49:170–182

30. Lancaster JR (2006) Nitroxidative, nitrosative, and nitrative

stress: kinetic predictions of reactive nitrogen species chemistry

under biological conditions. Chem Res Toxicol 19:1160–1174

31. Foster MW, Hess DT, Stamler JS (2009) Protein S-nitrosylation

in health and disease: a current perspective. Trends Mol Med

15:391–404

32. Bal-Price A, Brown GC (2000) Nitric oxide induced necrosis and

apoptosis in PC12 cells mediated by mitochondria. J Neurochem

75:1455–1464

Apoptosis (2012) 17:647–665 663

123

Page 18: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

33. Gegg ME, Beltran B, Salas-Pino S, Bolanos J, Clark JB, Moncada

S et al (2003) Differential effect of nitric oxide on glutathione

metabolism and mitochondrial function in astrocytes and neuro-

nes: implications for neuroprotection/neurodegeneration? J Neu-

rochem 86:228–237

34. Nakamura T, Lipton SA (2009) Cell death: protein misfolding

and neurodegenerative diseases. Apoptosis 14:455–468

35. Brown GC (2010) Nitric oxide and neuronal death. Nitric Oxide

23:153–165

36. Farinati F, Piciocchi M, Lavezzo E, Bortolami M, Cardin R

(2010) Oxidative stress and inducible nitric oxide synthase

induction in carcinogenesis. Dig Dis 28:579–584

37. Azad N, Iyer A, Vallyathan V, Wang L, Castranova V, Stehlik C

et al (2010) Role of oxidative/nitrosative stress-mediated Bcl-2

regulation in apoptosis and malignant transformation. Ann N Y

Acad Sci 1203:1–6

38. Fang M, Jaffrey SR, Sawa A, Ye K (2000) Dexras1: a G protein

specifically coupled to neuronal nitric oxide synthase via

CAPON. Neuron 28:183–193

39. Cheah JH, Kim SF, Hester LD, Clancy KW, Patterson SE III,

Papadopoulos V et al (2006) NMDA receptor-nitric oxide

transmission mediates neuronal iron homeostasis via the GTPase

Dexras1. Neuron 51:431–440

40. Dawson VL, Dawson TM, Bartley DA, Uhl GR, Snyder SH

(1993) Mechanisms of nitric oxide-mediated neurotoxicity in

primary brain cultures. J Neurosci 13:2651–2661

41. Boullerne AI, Nedelkoska L, Benjamins JA (1999) Synergism of

nitric oxide and iron in killing the transformed murine oligo-

dendrocyte cell line N20.1. J Neurochem 72:1050–1060

42. Liu QB, Liu LL, Lu YM, Tao RR, Huang JY, Han F et al (2010)

The induction of reactive oxygen species and loss of mitochon-

drial Omi/HtrA2 is associated with S-nitrosoglutathione-induced

apoptosis in human endothelial cells. Toxicol Appl Pharmacol

244:374–384

43. Romero C, Benedı J, Villar A, Martın-Aragon S (2010)

Involvement of Hsp70, a stress protein, in the resistance of long-

term culture of PC12 cells against sodium nitroprusside (SNP)-

induced cell death. Arch Toxicol 84:699–708

44. Forrester MT, Foster MW, Benhar M, Stamler JS (2009) Detec-

tion of protein S-nitrosylation with the biotin-switch technique.

Free Radic Biol Med 46:119–126

45. Notoya K, Jovanovic DV, Reboul P, Martel-Pelletier J, Mineau F,

Pelletier JP (2000) The induction of cell death in human osteo-

arthritis chondrocytes by nitric oxide is related to the production

of prostaglandin E2 via the induction of cyclooxygenase-2.

J Immunol 165:3402–3410

46. Suk K, Lee J, Hur J, Kim YS, Lee M, Cha S et al (2001) Activation-

induced cell death of rat astrocytes. Brain Res 900:342–347

47. Forrester MT, Thompson JW, Foster MW, Nogueira L, Moseley MA,

Stamler JS (2009) Proteomic analysis of S-nitrosylation and denitro-

sylation by resin-assisted capture. Nat Biotechnol 27:557–559

48. Decker T, Lohmann-Matthes ML (1988) A quick and simple

method for the quantitation of lactate dehydrogenase release in

measurements of cellular cytotoxicity and tumor necrosis factor

(TNF) activity. J Immunol Methods 115:61–69

49. Goodwin CJ, Holt SJ, Downes S, Marshall NJ (1995) Micro-

culture tetrazolium assays: a comparison between two new tet-

razolium salts, XTT and MTS. J Immunol Methods 179:95–103

50. Marshall NJ, Goodwin CJ, Holt SJ (1995) A critical assessment

of the use of microculture tetrazolium assays to measure cell

growth and function. Growth Regul 5:69–84

51. Wang S, Yu H, Wickliffe JK (2011) Limitation of the MTT and

XTT assays for measuring cell viability due to superoxide for-

mation induced by nano-scale TiO(2). Toxicol In Vitro

25:2147–2151

52. Bradford MM (1976) A rapid and sensitive method for the

quantitation of microgram quantities of protein utilizing the

principle of protein-dye binding. Anal Biochem 72:248–254

53. Fares F, Gavish M (1986) Characterization of peripheral benzo-

diazepine binding sites in human term placenta. Biochem Phar-

macol 35:227–230

54. Veenman L, Leschiner S, Spanier I, Weisinger G, Weizman A,

Gavish M (2002) PK 11195 attenuates kainic acid-induced sei-

zures and alterations in peripheral-type benzodiazepine receptor

(PBR) protein components in the rat brain. J Neurochem

80:917–927

55. Veenman L, Weizman A, Weisinger G, Gavish M (2010)

Expression and functions of the 18 kDa Mitochondrial Translo-

cator Protein (TSPO) in health and disease. In: Firer MA (ed)

Targeted drug delivery in cancer therapeutics. Research Signpost,

Kerala, pp 49–84

56. Shoukrun R, Veenman L, Shandalov Y, Leschiner S, Spanier I,

Karry R et al (2008) The 18-kDa translocator protein, formerly

known as the peripheral-type benzodiazepine receptor, confers

proapoptotic and antineoplastic effects in a human colorectal

cancer cell line. Pharmacogenetics Genom 18:977–988

57. Chelli B, Lena A, Vanacore R, Da Pozzo E, Costa B, Rossi L et al

(2004) Peripheral benzodiazepine receptor ligands: mitochondrial

transmembrane potential depolarization and apoptosis induction

in rat C6 glioma cells. Biochem Pharmacol 68:125–134

58. Knight SA, Dancis A (2006) Reduction of 2,3-bis-(2-methoxy-4-

nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide inner salt

(XTT) is dependent on CaFRE10 ferric reductase for Candida

albicans grown in unbuffered media. Microbiology 152:

2301–2308

59. Petrosillo G, Ruggiero FM, Paradies G (2003) Role of reactive

oxygen species and cardiolipin in the release of cytochrome c

from mitochondria. FASEB J 17:2202–2208

60. Bode J, Veenman L, Caballero B, Lakomek M, Kugler W, Gavish

M (in press) The 18 kDa Translocator Protein influences angio-

genesis, as well as aggressiveness, adhesion, migration, and

proliferation of glioblastoma cells. Pharmacogenet Genomics

61. Shargorodsky L (2010) The role of TSPO in mediating cell

death induced by nitric oxide. Thesis for the degree Master of

Science in Medical Science. Technion—Israel Institute of

Technology

62. Blachly-Dyson E, Zambronicz EB, Yu WH, Adams V, McCabe

ER, Adelman J et al (1993) Cloning and functional expression in

yeast of two human isoforms of the outer mitochondrial mem-

brane channel, the voltage-dependent anion channel. J Biol Chem

268:1835–1841

63. Choi J, Ifuku M, Noda M, Guilarte TR (2011) Translocator

protein (18 kDa)/peripheral benzodiazepine receptor specific

ligands induce microglia functions consistent with an activated

state. Glia 59:219–230

64. Erusalimsky JD, Moncada S (2007) Nitric oxide and mitochon-

drial signaling: from physiology to pathophysiology. Arterioscler

Thromb Vasc Biol 27:2524–2531

65. Bredt DS, Snyder SH (1989) Nitric oxide mediates glutamate-

linked enhancement of cGMP levels in the cerebellum. Proc Natl

Acad Sci USA 86:9030–9033

66. Garthwaite J, Garthwaite G, Palmer RM, Moncada S (1989)

NMDA receptor activation induces nitric oxide synthesis from

arginine in rat brain slices. Eur J Pharmacol 172:413–416

67. Chabrier PE, Demerle-Pallardy C, Auguet M (1999) Nitric oxide

synthases: targets for therapeutic strategies in neurological dis-

eases. Cell Mol Life Sci 55:1029–1035

68. Halestrap AP (2010) A pore way to die: the role of mitochondria

in reperfusion injury and cardioprotection. Biochem Soc Trans

38:841–860

664 Apoptosis (2012) 17:647–665

123

Page 19: The nitric oxide donor sodium nitroprusside requires the 18 kDa Translocator Protein to induce cell death

69. Ordys BB, Launay S, Deighton RF, McCulloch J, Whittle IR

(2010) The role of mitochondria in glioma pathophysiology. Mol

Neurobiol 42:64–75

70. Shoshan-Barmatz V, Ben-Hail D (2011) VDAC, a multi-func-

tional mitochondrial protein as a pharmacological target. Mito-

chondrion 12:24–34

71. Awad M, Gavish M (1991) Peripheral-type benzodiazepine

receptors in human cerebral cortex, kidney, and colon. Life Sci

49:1155–1161

72. Junck L, Olson JM, Ciliax BJ, Koeppe RA, Watkins GL, Jewett

DM et al (1989) PET imaging of human gliomas with ligands for

the peripheral benzodiazepine binding site. Ann Neurol 26:

752–758

73. Green DR, Reed JC (1998) Mitochondria and apoptosis. Science

281:1309–1312

74. Veenman L, Levin E, Weisinger G, Leschiner S, Spanier I,

Snyder SH et al (2004) Peripheral-type benzodiazepine receptor

density and in vitro tumorigenicity of glioma cell lines. Biochem

Pharmacol 68:689–698

75. Ott M, Gogvadze V, Orrenius S, Zhivotovsky B (2007) Mito-

chondria, oxidative stress and cell death. Apoptosis 12:913–922

76. Lipton SA, Choi YB, Pan ZH, Lei SZ, Chen HSV, Sucher NJ et al

(1993) A redox-based mechanism for the neuroprotective and

neurodestructive effects of nitric oxide and related nitroso-com-

pounds. Nature 364:626–632

77. Stamler JS (1994) Redox signaling: nitrosylation and related

target interactions of nitric oxide. Cell 78:931–936

78. Haendeler J, Hoffmann J, Tischler V, Berk BC, Zeiher AM,

Dimmeler S (2002) Redox regulatory and anti-apoptotic func-

tions of thioredoxin depend on S-nitrosylation at cysteine 69. Nat

Cell Biol 4:743–749

79. Babbage A (2009) translocator protein (18 kDa) [Homo sapiens]

CAB55884.1 GenBank. NCBI http://www.ncbi.nlm.nih.gov/

protein/CAB55884.1

80. Batarseh A, Papadopoulos V (2010) Regulation of translocator

protein 18 kDa (TSPO) expression in health and disease states.

Mol Cell Endocrinol 327:1–12

81. Sprengel R, Werner P, Seeburg PH, Mukhin AG, Santi MR,

Grayson DR et al (1989) Molecular cloning and expression of

cDNA encoding a peripheral-type benzodiazepine receptor.

J Biol Chem 264:20415–20421

82. Jaffrey SR, Fang M, Snyder SH (2002) Nitrosopeptide mapping:

a novel methodology reveals S-nitrosylation of Dexras1 on a

single cysteine residue. Chem Biol 9:1329–1335

83. Vesey CJ, Cole PV, Simpson PJ (1976) Cyanide and thiocyanate

concentrations following sodium nitroprusside infusion in man.

Br J Anaesth 48:651–660

84. Arnold WP, Longnecker DE, Epstein RM (1984) Photodegrada-

tion of sodium nitroprusside: biologic activity and cyanide

release. Anesthesiology 61:254–260

85. Hara MR, Snyder SH (2007) Cell signaling and neuronal death.

Annu Rev Pharmacol Toxicol 47:117–141

86. Norenberg MD, Jayakumar AR, Rama Rao KV, Panickar KS

(2007) New concepts in the mechanism of ammonia-induced

astrocyte swelling. Metab Brain Dis 22:219–234

87. Verma A, Nye JS, Snyder SH (1987) Porphyrins are endogenous

ligands for the mitochondrial (peripheral-type) benzodiazepine

receptor. Proc Natl Acad Sci USA 84:2256–2260

88. Bisland SK, Goebel EA, Hassanali NS, Johnson C, Wilson B

(2007) Increased expression of mitochondrial benzodiazepine

receptors following low-level light treatment facilitates enhanced

protoporphyrin IX production in glioma-derived cells in vitro.

Lasers Surg Med 39:678–784

89. Rampon C, Bouzaffour M, Ostuni MA, Dufourcq P, Girard C,

Freyssinet JM et al (2009) Translocator protein (18 kDa) is

involved in primitive erythropoiesis in zebrafish. FASEB J

23:4181–4192

90. Balsemao-Pires E, Jaillais Y, Olson BJ, Andrade LR, Umen JG,

Chory J et al (2011) The Arabidopsis translocator protein (AtT-

SPO) is regulated at multiple levels in response to salt stress and

perturbations in tetrapyrrole metabolism. BMC Plant Biol

111:108

91. Vanhee C, Zapotoczny G, Masquelier D, Ghislain M, Batoko H

(2011) The Arabidopsis multistress regulator TSPO is a heme

binding membrane protein and a potential scavenger of porphy-

rins via an autophagy-dependent degradation mechanism. Plant

Cell 23:785–805

92. Zeno S, Veenman L, Katz Y, Bode J, Gavish M, Zaroor M (2012)

The 18 kDa mitochondrial Translocator Protein (TSPO) prevents

accumulation of protoporphyrin IX. A TSPO knockdown study.

Curr Mol Med 12:494–501

Apoptosis (2012) 17:647–665 665

123


Recommended