Transcript

www.elsevier.com/locate/brainres

Brain Research 1003 (2004) 86–97

Research report

Inhibition of mixed lineage kinase 3 attenuates MPP+-induced

neurotoxicity in SH-SY5Y cells

Joanne R. Mathiasena,*, Beth Ann W. McKennaa, Michael S. Saporitoa,Ghanashyam D. Ghadgeb, Raymond P. Roosb, Beverly P. Holskina, Zhi-Liang Wua,Stephen P. Truskoa, Thomas C. Connorsa, Anna C. Maroneya, Beth Ann Thomasa,

Jeffrey C. Thomasa, Donna Bozyczko-Coynea

aNeurobiology, Cephalon, Inc., 145 Brandywine Parkway, West Chester, PA 19380, USAbDepartment of Neurology, The University of Chicago, Chicago, IL 60637, USA

Accepted 3 November 2003

Abstract

The neuropathology of Parkinson’s Disease has been modeled in experimental animals following MPTP treatment and in dopaminergic

cells in culture treated with the MPTP neurotoxic metabolite, MPP+. MPTP through MPP+ activates the stress-activated c-Jun N-terminal

kinase (JNK) pathway in mice and SH-SY5Y neuroblastoma cells. Recently, it was demonstrated that CEP-1347/KT7515 attenuated MPTP-

induced nigrostriatal dopaminergic neuron degeneration in mice, as well as MPTP-induced JNK phosphorylation. Presumably, CEP-1347

acts through inhibition of at least one upstream kinase within the mixed lineage kinase (MLK) family since it has been shown to inhibit MLK

1, 2 and 3 in vitro. Activation of the MLK family leads to JNK activation. In this study, the potential role of MLK and the JNK pathway was

examined in MPP+-induced cell death of differentiated SH-SY5Y cells using CEP-1347 as a pharmacological probe and dominant negative

adenoviral constructs to MLKs. CEP-1347 inhibited MPP+-induced cell death and the morphological features of apoptosis. CEP-1347 also

prevented MPP+-induced JNK activation in SH-SY5Y cells. Endogenous MLK 3 expression was demonstrated in SH-SY5Y cells through

protein levels and RT-PCR. Adenoviral infection of SH-SY5Y cells with a dominant negative MLK 3 construct attenuated the MPP+-

mediated increase in activated JNK levels and inhibited neuronal death following MPP+ addition compared to cultures infected with a control

construct. Adenoviral dominant negative constructs of two other MLK family members (MLK 2 and DLK) did not protect against MPP+-

induced cell death. These studies show that inhibition of the MLK 3/JNK pathway attenuates MPP+-mediated SH-SY5Y cell death in culture

and supports the mechanism of action of CEP-1347 as an MLK family inhibitor.

D 2004 Elsevier B.V. All rights reserved.

Theme: Disorders of the nervous system

Topic: Degenerative disease: Parkinson’s

Keywords: Adenovirus; CEP-1347; c-Jun N-terminal kinase; Mixed lineage kinase; MPP+; SH-SY5Y

1. Introduction

Degeneration of nigro-striatal dopaminergic (DA) neu-

rons is a neuropathological hallmark of Parkinson’s Disease

(PD). Part of the underlying degenerative process is thought

to be due to a selective vulnerability of DA neurons to

mitochondrial dysfunction. This mitochondrial dysfunction

0006-8993/$ - see front matter D 2004 Elsevier B.V. All rights reserved.

doi:10.1016/j.brainres.2003.11.073

* Corresponding author. Tel.: +1-610-738-6634; fax: +1-610-344-

0065.

E-mail address: [email protected] (J.R. Mathiasen).

is widespread throughout many cell types of the PD patient

[54,56]. Thus, great interest lies in understanding the key

biochemical events that are triggered following blockade of

mitochondrial respiration and are causal to neuronal death.

Mechanistic studies of DA neuron death akin to PD are

routinely conducted using toxins that interfere with electron

transport at the site of mitochondrial complex I and/or

complex II. Classically, MPTP neurotoxicity has gained

broad acceptance as a model of PD since it is a potent

and selective nigro-striatal DA neurotoxin that produces

PD-like symptoms in humans, non-human primates and

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–97 87

mice [1,4,17,25,26,34]. Moreover, MPP+, the neurotoxic

byproduct of MPTP metabolic oxidation [43,50,62,65], is

selectively taken up into DA neurons and inhibits complex I

of mitochondrial electron transport.

Several intraneuronal signaling pathways have been im-

plicated in MPTP-induced neurotoxicity in vivo. For exam-

ple, mice overexpressing a mitochondrial membrane

signaling protein, Bcl-2, or mice that are deficient in another

signaling peptide, p53, are resistant to MPTP-induced neu-

rotoxicity [44,64,69]. MPTP also activates mitogen-activat-

ed protein kinase kinase 4 (MKK4) and a downstream

substrate, c-Jun N-terminal kinase (JNK) in both the striatum

and substantia nigra of mice [52]. Overexpression of all

members of the mixed lineage kinase (MLK) family leads to

an activation of the JNK pathway [5,9,13,21,27,49,58].

Downstream of the MLKs are the dual-specificity mitogen

activated protein kinase kinases (MKK4 and MKK7) which

phosphorylate JNKs on serine and threonine residues [14].

Gene transfer of JNK-interacting protein-1 (JIP) in SH-

SY5Y cells and mice has implicated the JNK pathway in

MPTP (MPP+)-induced DA cell death [67] and adenoviral

expression of dominant negative c-Jun in an axotomy-

induced rat model of DA cell death has demonstrated

inhibition of c-Jun activation and cell death [8]. Of note, in

a variety of neuronal cell types, signaling through these

pathways manifests in morphological and biochemical fea-

tures of apoptosis, including nuclear chromatin condensa-

tion, membrane blebbing, DNA laddering and activation of

caspase(s) [11,12,23,41,59].

Additionally, an inhibitor of the JNK pathway, the

indolocarbazole CEP-1347/KT7515 [36], attenuates MPTP-

mediated nigrostriatal DA neuron loss in mice [51]. Recent

studies exemplify that CEP-1347 likely inhibits JNK acti-

vation indirectly through the MLK family [37]. To establish

a direct role of MLK in DA neuron death elicited by MPP+,

studies were conducted in neuronally differentiated SH-

SY5Y cells, evaluating CEP-1347 protection against and

interception of a variety of events associated with apoptotic

death. MPP+-treated SH-SY5Y cells were chosen as a

model system to investigate signaling pathways causative

of cell death because these cells exhibit (1) DA neuron

characteristics including dopamine synthesis, (2) expression

of dopamine receptors, (3) specific uptake and sequestration

of dopamine consistent with expression of the dopamine

transporter [3,14,57] and (4) they can be differentiated into a

neuronal phenotype by incubation with retinoic acid [32].

Furthermore, MPP+ induces apoptotic cell death in SH-

SY5Y cells and activates JNK and the early transcription

factor nuclear factor nB [7,30,33,46,55]. The current studies

investigated the ability of CEP-1347 to (1) inhibit the

neurotoxic effects of MPP+ in retinoic acid (RA) differen-

tiated SH-SY5Y cells; and (2) inhibit MPP+-induced JNK

activation. Studies also determined whether SH-SY5Y cells

over-expressing dominant negative forms of MLK family

members would be protected from MPP+-induced JNK

activation and cell death.

2. Materials and methods

2.1. Cell culture

SH-SY5Y cells (J. Biedler, Memorial Sloan-Kettering

Cancer Center, Rye, NY, USA) were seeded at a density of

4� 104/cm2 in T150 flasks and propagated in Dulbecco’s

Modified Eagle’s Medium (DMEM; Gibco BRL, Gaithers-

burg, MD) containing 10% FBS and 2 mM L-glutamine

(Gibco BRL). Growing SH-SY5Y cells were maintained in

a humidified atmosphere at 37 jC in 10% CO2. Cells were

routinely subcultured once a week and used for assays at

passage 8–37. For assays, SH-SY5Y cells were plated at a

density of 1.2� 105/cm2 onto poly-ornithine/mouse laminin

(Sigma-Aldrich, St. Louis, MO/Becton Dickinson Bioscien-

ces, San Jose, CA) coated plates in Neurobasal medium

(Gibco BRL) with B27 supplement (Gibco BRL) and 2 mM

L-glutamine. Cells maintained in Neurobasal medium were

kept in a humidified atmosphere at 37 jC and 5% CO2. The

cells were allowed to attach for 1 h before the addition of 10

AM (final concentration) all trans-retinoic acid (RA, made in

ethanol as a 10 mM stock solution; Sigma-Aldrich) that was

used to induce neuronal differentiation.

Chinese hamster ovary cells (CHO-K1; ATCC#CCL-61,

American Type Culture Collection, Manassas, VA) were

propagated as previously described [38].

2.2. MPP+ treatment

On 3–5 days in vitro subsequent to plating/differentia-

tion, SH-SY5Y cells were exchanged into fresh Neurobasal

medium with B27 supplement and 2 mM L-glutamine

without RA by serial dilution washing and treated for

indicated times with MPP+ (made as an 11� stock in

Neurobasal medium with supplements mentioned above;

Sigma-Aldrich, previously from RBI). Typically, CEP-1347

(stored as 4 mM in DMSO in amber glass vials), was diluted

to a 4� stock concentration in medium, then added to wells

establishing final 1� concentrations and incubated with

cells for 1 h prior to the addition of MPP+.

2.3. Lactate assay

Lactate measurement in culture medium from differenti-

ated SH-SY5Y cells was determined by a standard lactate

assay (Sigma-Aldrich #826-10) following 4, 24 or 48 h of 3

mM MPP+ treatment. Data are represented as fold increase

in lactate compared to untreated differentiated SH-SY5Y

cells.

2.4. DNA Characterization

For visualization of nuclear DNA, cell cultures were

fixed (10 min) with 4% paraformaldehyde and subsequently

incubated for 15–20 min with 1 Ag/ml bisBenzamide

(Sigma-Aldrich) in PBS. Photomicrographs were taken

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–9788

using a Nikon Eclipse TE300 microscope equipped with

both Hoffman and epifluorescence optics. For DNA ladder-

ing studies, cellular DNA was isolated [29] and resolved on

a 1.2% agarose gel containing 0.01% ethidium bromide. For

RT-PCR analyses, RNA was isolated from differentiated

SH-SY5Y cells and PC12 cells with RNAzol B according to

the manufacturer’s directions (TelTest, Friendswood, TX).

2.5. Real-time RT-PCR

Cells were lysed and RNA was isolated with a Qiagen

RNeasy Mini Kit (#74104, Qiagen, Valencia, CA) according

to the manufacturer’s instructions. Samples were run on a

1.2% agarose gel to check for RNA integrity. RNA was

quantified spectrophotometrically (OD 260). cDNA was

generated using oligo dT in Ambion’s Retroscript kit

(Ambion, Austin, TX). 1.5 Ag of total RNA was used per

20 Al reaction. Gene specific primers were identified by

using the Primer Express software (Applied Biosystems,

Foster City, CA). Real-time PCR was performed in an ABI

PrismR 7000 Sequence Detection System (Applied Biosys-

tems) using SYBRR Green PCR Master Mix (Applied

Biosystems). A 50-Al PCR reaction was run using manu-

facturer’s recommended cycling times. Real-time detection

of gene expression is displayed as a threshold cycle or CT

value. The CT value is the cycle at which the amplification

of the gene enters into the exponential phase. Quantification

of gene expression is possible with the assignment of a CT

value. Hence, relative mRNA expression for each sample

can be calculated by normalization to GAPDH CT values.

The relative quantitation value is expressed as D (delta) CT.

2.6. Cell lysis and immunoblot analysis

For measurement of activated, or phosphorylated JNK

(pJNK), differentiated SH-SY5Y cells (1.5� 106) were

lysed in 125-Al ice cold FRAK buffer (1% Triton X-100,

50 mMNaCl, 30 AM sodium pyrophosphate, 50 mM sodium

fluoride, 1 mM sodium vanadate, 10 mM Tris–HCl, pH 7.6)

containing protease inhibitors (1 mM phenylmethylsulfonyl-

fluoride (PMSF) and 20 Ag/ml aprotinin). Lysates were

passed through a 28-1/2 gauge needle and the Triton insol-

uble fraction was then removed from the lysate by centrifu-

gation at 4 jC (20 min, Effendorf microcentrifuge, 14,000

rpm). Supernatant was collected and an aliquot taken for

protein determination (BCA assay, Pierce, Rockford, IL).

Lysates were prepared for electrophoresis by adding Laem-

meli sample buffer and heating for 10 min at 95 jC. Samples

of equivalent total protein (20 Ag/lane) were run on 4–20%

Tris–Glycine gels (Invitrogen, Carlsbad, CA) and trans-

ferred (60 V/cm2) to 0.2 Am nitrocellulose (BioRad Labora-

tories, Hercules, CA). Membranes were blocked in 3% BSA/

Tris buffered saline (TBS) with 0.1% Triton X-100 followed

by overnight incubation at 4 jC with a polyclonal pJNK

antibody (New England Biolabs, Beverly, MA) diluted

1:1000 in block buffer. Following three 15-min washes in

0.5% Triton X-100/TBS membranes were incubated with a

horseradish peroxidase conjugated secondary antibody

(1:20,000, goat anti-rabbit; Southern Biotechnologies, Bir-

mingham, AL), washed and incubated for 1 min with

enhanced chemiluminescent (ECL) substrate (Amersham,

Buckinghamshire, UK) for imaging with BioMax Film

(Kodak, Rochester, NY). Total JNK levels across samples

were determined following stripping and reprobing mem-

branes with a monoclonal mouse antibody directed against

non-phosphorylated JNK proteins (JNK1/2; 1:1000; BD

PharMingen, Franklin Lakes, NJ). In experiments where

adenoviral infection was included in the protocol, immuno-

blots were stripped a second and/or third time and reprobed

with antibodies directed against MLK3 (Santa Cruz Biotech-

nology, Santa Cruz, CA; rabbit polyclonal sc-536; 1:1000)

and/or h-galactosidase (h-gal; Invitrogen, Carlsbad, CA;

rabbit polyclonal; 1:5000). Immunocytochemistry was per-

formed on AdCMVdnMLK3-infected cells to detect

dnMLK3 expression efficiency (anti-MLK3, Santa Cruz

Biotechnology; 1:300). h-gal histochemistry was performed

on AdCMVLacZ-infected cells to determine the expression

efficiency of LacZ (Invitrogenh-gal staining kit #K1465-01).

2.7. Cell viability assessment

Cell viability was determined based on measurement of

lactate dehydrogenase (LDH) release into the culture medi-

um. LDH assays were conducted in accordance with the

manufacturer’s instructions (Cytotoxicity Detection Kit

(LDH), Roche Diagnostics, Indianapolis, IN). Total LDH

release was obtained by completely lysing cells in desig-

nated wells and data are represented as percent total LDH

release.

2.8. Construction of recombinant replication-deficient

adenovirus

A HindIII to EcoRV fragment from vector pcDNA3EE

containing dnMLK3 cDNA (kinase dead/dominant negative)

[38] was inserted into respective sites of the vector pAdCMV

[18], downstream from the cytomegalovirus (CMV) promot-

er and upstream of the cellular heavy chain enhancer (4F2)

and the bovine growth hormone polyadenylation site.

pAdCMV contains 0–1 and 9–16 map units of the adeno-

virus 5 genome. pAdCMV containing mutant (dn) MLK3

was linearized with NheI and cotransfected, using the calci-

um phosphate precipitation method, with XbaI- and ClaI-

digested adenovirus 5 (sub360) DNA into HEK293 cells, a

trans-complementing cell line for E1 function. Viruses were

purified by CsCl isopycnic centrifugation, dialyzed against

HEPES-buffered saline (10 mM HEPES, 140 mM NaCl, 2

mM MgCl2, pH 7.5) containing 10% glycerol, and stored at

� 70 jC in small aliquots. AdCMVlacZ virus was a gener-

ous gift from Dr. Jerome Schaack (University of Colorado,

Denver, CO [53]). AdCMVdnMLK2 and AdCMVdnDLK

were prepared using the AdEasy Vector System (Quantum

Fig. 1. MPP+ concentration-dependent increases in release of LDH from

RA differentiated SH-SY5Y cell cultures measured 48 h post exposure (A).

CEP-1347-dependent decreases in LDH release are significantly different

from 3 mM MPP+ at concentrations of 10–300 nM, indicating cell survival

promotion (B). Data is representative of three independent experiments

(*= significantly different from basal; **= significantly different from

MPP+ treated; p< 0.05 Student’s t-test).

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–97 89

Biotechnologies, also known as Qbiogene, Carlsbad, CA).

Briefly, MLK2(K125A) or DLK(K162A) cDNAwas cloned

into the pShuttle-CMV transfer vector at the 5VAcc65I and3VXbaI polylinker cloning sites and the positive clones

identified were sequence-verified. These pShuttle-CMV-

MLK2(K125A) or DLK(K162A) constructs were linearized

at the unique PmeI site, dephosphorylated and gel purified.

These constructs were co-transformed with supercoiled ad-

enovirus genome using highly competent BJ5183 E. coli

cells via electroporation. The transformed colonies were

grown on kanamycin plates, the smallest colonies picked

from these plates and amplified in LB/kanamycin medium.

Conventional alkaline lysis miniprep DNA protocols, utiliz-

ing phenol chloroform extractions and ethanol precipitations,

were performed. Following restriction digest and PCR con-

firmation, this DNAwas transformed into competent E. coli

DH5a cells. Positive clones were digested with PacI to

expose their ITRs and transfected into 293A cells using

Qiagen SuperFect reagent (Qiagen, Valencia, CA). Viruses

were purified and stored as described above.

2.9. Adenoviral infection

Differentiated SH-SY5Y cells were infected by removing

the culture medium and incubating cells at 37 jC for 2.5

h with high-titer virus diluted in a small volume of Neuro-

basal medium containing B27 supplement and L-glutamine

providing a multiplicity of infection (MOI) of 1000 based

on optical particle units (OPU; [40]). This MOI was

predetermined to infect f 90–100% of the SH-SY5Y cells.

The wells were gently rocked occasionally throughout the

infection period. Following infection warmed Neurobasal

medium containing B27 supplement and 2 mM L-glutamine

was added to the wells bringing the media volume up to

feeder levels (3–4 ml/6-well dish; 200 Al/96-well).For proof of dominant negative (dn) status of the viral

construct (AdCMVdnMLK3), CHO cells were treated in the

following manner. CHO cells were infected twice due to

their continual division. On day 0 cells were plated at 2� 105

cells/well in six-well culture dishes. On day 1 cells were

either uninfected, but subjected to the same conditions, or

infected with either control adenoviral construct, AdCMV-

LacZ (2000 MOI), or AdCMVdnMLK3 (2000 MOI). On

day 2 cells were transfected using Lipofectamine Plus (Gibco

#10964-013), as previously described [38]. All cells were

transfected with a total of 2 Ag of cDNA composed of the

following: 4% MLK3, 20% dnMLK3, and 50% dnMKK4.

The remaining percentage of cDNAwas composed of vector.

The ratio of MLK3 to dnMKK4 was predetermined to be in

the linear range with respect to MLK3 expression and

phosphorylation of the dnMKK4 substrate. A dominant

negative MKK4 substrate was used to prevent downstream

activation that would lead to cell death. The dominant

negative nature of these clones has been previously de-

scribed [38]. Four hours after the transfection, complete

growth medium was added. On day 3, all cells were re-

infected using the same protocol that was performed on day

1. On day 4 cells were lysed and prepared for phospho-

MKK4 ELISA [38] or immunoblot analysis.

2.10. Data computation

Data was computed using Microsoft Excel and statistics

were evaluated using either Student’s or Dunnett’s t-tests.

Significance (*) was based on p < 0.05. The graphics pack-

age used was Prism GraphPad.

3. Results

3.1. CEP-1347 inhibition of MPP+-induced loss in neuro-

nally differentiated SH-SY5Y cell survival, cellular mor-

phology and apoptosis

To establish cytotoxic effects of MPP+ in differentiated

SH-SY5Y cells, increases in the release of cellular lactate

dehydrogenase (LDH) were measured in the medium of

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–9790

cultures treated with increasing concentrations of MPP+ up to

a maximum of 10 mM. At 3 mM MPP+ significant increases

in LDH were observed at 48 h post treatment (Fig. 1A). This

was the lowest concentration of MPP+ that produced a

significant 30–50% increase in LDH release (relative to total

LDH) compared to untreated cultures. Although larger

increases in LDH release were observed at a higher concen-

tration of MPP+ (10 mM), the minimum concentration, 3

mM, which produced a significant increase in cytotoxicity

was used to measure neuroprotective effects of CEP-1347.

CEP-1347 provided significant protection from 3 mM

MPP+-induced LDH release at concentrations of 10, 30, 100

and 300 nM (Fig. 1B). Across multiple experiments 30–100

nM CEP-1347 achieved maximal efficacy against MPP+-

induced cell death ranging from f 40–60% rescue. After

48 h of treatment with 3 mM MPP+ SH-SY5Y cellular

morphology changes, going from adherent process bearing

neuronal-looking cells (Fig. 2A) to rounded, clustered cells

with decreased substrate adhesion and process extension

(Fig. 2C). Moreover, two features indicative of apoptosis

Fig. 2. MPP+-induced changes in cellular morphology and apoptosis in differentia

with Hoffman optics (A) and normal nuclear bis-benzimide (Hoechst) staining (B).

condensed nuclear chromatin. (D) Arrows represent apoptotic cells. Treatment with

(E) and chromatin changes (F). Insert panel D: DNA laddering following 3 mM

bar = 10 Am.

were observed in SH-SY5Y cells following 48 h of treatment

with 3 mM MPP+: (1) Condensed chromatin (Hoechst, Fig.

2B, D, and F) and (2) DNA laddering (Fig. 2D, insert). These

observations confirm reports that SH-SY5Y cells undergo

apoptosis in response to MPP+ treatment [30,33,46,55]. Of

significance, the morphologic changes observed in SH-

SY5Y cells following exposure to 3 mM MPP+ were mostly

prevented by pretreatment of cells with CEP-1347 (Fig. 2E).

Moreover, CEP-1347 (30 nM) prevented nuclear chromatin

condensation in a majority of cells when it was added 1

h prior to 3 mM MPP+ treatment (Fig. 2F), indicative of a

partial inhibition of MPP+-induced apoptosis.

3.2. CEP-1347 acts downstream of MPP+ effects on

mitochondrial demise in differentiated SH-SY5Y cells

MPP+ is a mitochondrial toxin that inhibits respiration at

complex I of the electron transport chain [43]. The resulting

increase in NADH due to this inhibition leads to pyruvate

being metabolized into lactate. To determine whether CEP-

ted SH-SY5Y cells. Untreated control cultures showing normal morphology

Cultures treated with 3 mM MPP+ show degenerating cells at 48 h (C) with

30 nM CEP-1347 1 h prior to 3 mMMPP+ exposure prevents morphologic

MPP+ exposure: lanes (1) MW markers; (2) 0 h; (3) 24 h; (4) 48 h. Scale

Fig. 3. Lack of CEP-1347 effects on cellular lactate production induced by

3 mM MPP+ in differentiated SH-SY5Y cells. Lactate levels in culture

medium were measured at 4, 24 and 48 h post exposure to 3 mM MPP+ in

quadruplicate. Groups as follows: (MPP) 3 mM MPP+; (MPP+/1347) 30

nM CEP-1347 (neuroprotective concentration) was tested with 3 mM

MPP+. Graph represents one of two experiments with the same result.

Fig. 4. JNK activation following 3 mM MPP+ and inhibition by CEP-

1347. Immunoblot analysis of differentiated SH-SY5Y cell lysate for

phosphorylated (activated) JNK and total JNK1/2 protein following 30

min of 3 mM MPP+ and as indicated below a 30-min pretreatment with a

concentration response of CEP-1347 (A). The p46 pJNK band is

quantitated in (B) and represented as a ratio of p46 optical density

relative to total JNK1/2 protein density as a gel loading control.

Numbered duplicate wells in A are as follows: (1) and (2) basal untreated

cultures; (3) and (4) 3 mM MPP+; (5) and (6) MPP+ with 0.01 AM CEP-

1347; (7) and (8) MPP+ with 0.1 AM CEP-1347; (9) and (10) MPP+ with

0.3 AM CEP-1347; (11) and (12) MPP+ with 1 AM CEP-1347; (13) and

(14) MPP+ with 10 AM CEP-1347. * indicates significantly different from

basal ( p< 0.05); ** indicates significantly different from MPP+ ( p< 0.05).

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–97 91

1347 altered the toxic effects of MPP+ by interfering with

the initial events of mitochondrial complex I inhibition,

lactate levels were measured in SH-SY5Y cell culture

medium following MPP+ treatment in the presence or

absence of CEP-1347. As early as 4 h after 3 mM MPP+

addition, lactate production increased f 2-fold over basal

and this lactate induction was maintained to 48 h after 3 mM

MPP+ addition (Fig. 3). This increase in lactate production

was not prevented by addition of CEP-1347 to the cultures

(Fig. 3). These data indicate that CEP-1347 does not

interfere with MPP+ toxicity by blocking MPP+ inhibition

of complex I and verifies that CEP-1347 is acting down-

stream of this initial insult. Therefore, the neuroprotective

effects of CEP-1347 were not due to decreases in MPP+

interactions with the mitochondria.

3.3. CEP-1347 inhibits SAPK/JNK pathway activation

following MPP+ treatment in neuronally differentiated SH-

SY5Y cells

Evidence for activation of the SAPK/JNK pathway was

evaluated by immunoblot analysis for pJNK. In SH-SY5Y

cells the total JNK and pJNK antibodies detected two protein

bands of 46 and 54 kDa (Fig. 4). These molecular weight size

bands correspond to the reported sizes of cloned and

expressed JNK [22]. Initial time course experiments showed

JNK activation between 30 min and 4 h following MPP+

exposure to SH-SY5Y cells, with a maximal activation of

2.4-fold above untreated cultures at the 30-min time point.

Typically in MPP+-treated SH-SY5Y cells the p46 kDA band

Fig. 5. Inhibition of plasmid overexpressed wild-type MLK3-stimulated p-

dominant negative (dn) MKK4 by plasmid dominant negative (dn) MLK3

and adenoviral dnMLK3 in CHO cells. Data represent duplicate samples

from one experiment. *= significantly different from MLK3 alone.

**= significantly different from MLK3-stimulated p-dnMKK4 ( p< 0.05,

Student’s t-test).

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–9792

showed a more robust and reliable activation than did the p54

kDA band. CEP-1347 inhibited the pJNK signal seen 30 min

following treatment of cells with 3 mM MPP+ in a concen-

tration-dependent manner from 0.1 to 10 AM with complete

inhibition achieved at 0.1 AMand above (Fig. 4). Notably, the

MPP+-induced pJNK signal fell below basal levels following

treatment with CEP-1347.

3.4. Dominant negative MLK3 inhibits MPP+-induced

toxicity and pJNK in neuronally differentiated SH-SY5Y

cells

To verify the dominant negative action of overexpressed

AdCMVdnMLK3, a CHO cell system was infected and

transfected with the following constructs. CHO cells infected

Fig. 6. AdCMVdnMLK3 inhibition of MPP+-induced cell death in differentiated SH

into culture medium measured 48 h post exposure. AdCMVdnMLK3 (1000 MO

different from 3 mM MPP+ (A). *= significantly different from basal. **= signifi

Student’s t-test. (B) Upper immunoblot shows overexpression of MLK3. Lanes:

(1000 MOI, 48 h), (3) AdCMVdnMLK3-infected SH-SY5Y cells (1000 MOI, 48

immunoblot with same samples in A probed for hgal. Lanes: (1) uninfected SH-

differentiated SH-SY5Y cells immunostained for MLK3 showing very low level

infected differentiated SH-SY5Y cells immunostained with MLK3 antibody show

with AdCMVdnMLK3 were inhibited from wild-type plas-

mid transfected MLK3 phosphorylation of transfected

dnMKK4, MKK4 being a downstream target of MLK3

(Fig. 5). A dominant negative MKK4 substrate was used

to measure overexpressed MLK3 kinase activity so as to

prevent death of the cells through downstream activation

mechanisms. Due to the continual division of CHO cells,

they were subjected to infection on two separate days

bracketing the plasmid transfection day. The MOI was

increased to 2000 per day to provide 90–100% transfection

efficiency demonstrated in parallel sister cultures. Control

viral infection with AdCMVLacZ had no effect on the ability

of MLK3 to phosphorylate dnMKK4. Following confirma-

tion of the dominant negative status of the AdCMVdnMLK3

in CHO cells, overexpression of dnMLK3 in neuronally

-SY5Y cells. MPP+ (3 mM) increased release of LDH from SH-SY5Y cells

I, 48 h expression)-dependent decreases in LDH release are significantly

cantly different from 3 mM MPP+-treated. Significance based on p< 0.05,

(1) uninfected SH-SY5Y cells, (2) AdCMVLacZ-infected SH-SY5Y cells

h). Lower immunoblot, same blot reprobed for phospho-JNK. (C) Separate

SY5Y cells, (2) AdCMVLacZ-infected (1000 MOI, 48 h). (D) Uninfected

s of endogenous MLK3 detected. (E) AdCMVdnMLK3 (1000 MOI, 48 h)

ing 90–100% infection efficiency and expression. Scale bar = 10 Am.

Fig. 8. AdCMVdnMLK2 infection of differentiated SH-SY5Y cells.

AdCMVdnMLK2 infection did not inhibit 3 mM MPP+-induced cell death

in SH-SY5Y cells. MPP+ (3 mM) increased release of LDH from SH-SY5Y

cell culture medium measured 48 h post exposure in cells infected with

either AdCMVLacZ (1000 MOI, 48 h) or AdCMVdnMLK2 (250 MOI, 48

h). Immunoblot shows overexpression of MLK2 with an HA antibody

probe. Lanes: (1–5) Adenoviral infection of SH-SY5Y cells for 24 h; (6–

9) adenoviral infection for 48 h. Lane (1) AdCMVLacZ 1000 MOI; (2) and

(6) AdCMVdnMLK2 250 MOI; (3) and (7) AdCMVdnMLK2 1000 MOI;

(4) and (8) AdCMVdnMLK2 2000 MOI; (5) and (9) AdCMVdnMLK2

4000 MOI. *= significantly different from basal. Significance based on

p< 0.05, Student’s t-test.

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–97 93

differentiated SH-SY5Y cells (Fig. 6, compare A lane 1—

endogenous MLK3 with lane 3—overexpressed MLK3 and

C endogenous immunocytochemically detected MLK3 vs.

D overexpressed MLK3) partially suppressed MPP+-in-

duced cell death, comparable to the protective effect of

CEP-1347 on MPP+ toxicity (Fig. 1). MPP+-stimulated

pJNK levels were also partially prevented by expression

of AdCMVdnMLK3 in neuronally differentiated SH-SY5Y

cells (Fig. 7). This dnMLK3 overexpression level did not

decrease basal levels of pJNK (Fig. 6, compare A: lanes 1

and 2 vs. 3). The control construct, AdCMVLacZ, used at

an equivalent MOI (1000) produced similar overexpression

when blots were probed with the h-gal antibody instead of

anti-MLK3 (Fig. 6B) and in histochemical evaluation of the

cells (not shown).

3.5. Real-time RT-PCR expression analysis of SH-SY5Y

cells

The existence of endogenous MLK3 expression in SH-

SY5Y cells was implicit from immunoblot analysis of cell

lysate with anti-MLK3 (Fig. 6). There are, however, no

commercially available antibodies for human MLK1, 2 or

ZPK(DLK). Therefore, real-time RT-PCR was performed on

SH-SY5Y cell lysate with gene specific primers identified

by the Primer Express software provided with the ABI

PrismR 7000 Sequence Detection System. Gene specific

Fig. 7. AdCMVdnMLK3 inhibition of MPP+-induced pJNK response in

differentiated SH-SY5Y cells. Immunoblot analysis of SH-SY5Y cell lysate

for phosphorylated JNK following 30 min of 3 mM MPP+ (A, B).

Numbered wells are as follows: (A1) basal untreated cultures infected with

AdCMVLacZ (1000 MOI, 24 h); (A2) 3 mM MPP+-treated culture infected

with AdCMVLacZ; (B1) basal untreated cultures infected with

AdCMVdnMLK3 (1000 MOI, 24 h); (B2) 3 mM MPP+-treated culture

infected with AdCMVdnMLK3. The p46 band is quantitated from duplicate

distinct samples normalized to total JNK1/2 protein density and graphically

represented as fold over control p46 activated JNK. * indicates significantly

different from AdCMVLacZ ( p< 0.05).

products for MLK1, 2, 3 and ZPK(DLK) were detected in

differentiated SH-SY5Y cells. Two to three separate cell

preparations with three independent measures were gener-

ated and normalized to GAPDH expression to determine the

relative expression of MLK RNA in differentiated SH-

SY5Y cells. MLK1 and MLK3 were equivalently expressed

(5.91F 0.32 and 5.86F 0.21 DCT, respectively) and MLK2

Fig. 9. AdCMVdnDLK infection of differentiated SH-SY5Y cells.

AdCMVdnDLK infection did not inhibit 3 mM MPP+-induced cell death

in SH-SY5Y cells. MPP+ (3 mM) increased release of LDH from SH-SY5Y

cell culture medium measured 48 h post exposure in cells infected with

either AdCMVLacZ (250 MOI, 48 h) or AdCMVdnDLK (500 MOI, 48 h).

Immunoblot shows overexpression of DLK with an HA antibody probe.

Lanes: (1–5) Adenoviral infection of SH-SY5Y cells for 24 h; (6–9)

adenoviral infection for 48 h. Lane (1) AdCMVLacZ 1000 MOI; (2) and (6)

AdCMVdnDLK 250 MOI; (3) and (7) AdCMVdnDLK 500 MOI; (4) and

(8) AdCMVdnDLK 1000 MOI; (5) and (9) AdCMVdnDLK 2000 MOI.

*= significantly different from basal. Significance based on p< 0.05,

Student’s t-test.

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–9794

and ZPK(DLK) were equivalently expressed 1 DCT level

higher than MLK1 and MLK3 (6.83F 1.17 and 6.89F0.31,

respectively). This represents a twofold lower expression of

MLK2 and ZPK(DLK) in differentiated SH-SY5Y cells

than MLK1 and MLK3, as each critical threshold value

represents a doubling of RNA present upon replication in

the PCR reaction. Statistically, differences in MLK2 vs.

MLK3 expression did not reach significance ( p = 0.07),

whereas significant differences were established for MLK1

vs. MLK2; MLK1 vs. ZPK(DLK) and MLK3 vs.

ZPK(DLK).

3.6. Dominant negative MLK2 and dnDLK do not inhibit

MPP+-induced toxicity in differentiated SH-SY5Y cells

AdCMVdnMLK2 and AdCMVdnDLK represent domi-

nant negative adenoviral constructs of MLK family mem-

bers that are related to MLK3. The kinase domains of

MLK3 and MLK2 have 75% sequence homology and

MLK3 and DLK share 42% sequence homology. The

leucine zipper regions share similar homology with MLK3

and MLK2 having 68% homology while MLK3 and DLK

share only 32% homology. It is theoretically possible that

dnMLK2 or dnDLK would cross react by heterodimerizing

with endogenous MLK3 in SH-SY5Y cells and provide

some protection against MPP+. This was not the case over a

wide range of overexpressed protein (125–1000 MOI). In

the higher MOI conditions (500–1000 MOI), dnMLK2 and

dnDLK were toxic, especially when combined with MPP+

(data not shown). At MOI levels that were not toxic to

basal untreated cells, AdCMVdnMLK2 (Fig. 8) and

AdCMVdnDLK (Fig. 9) did not protect differentiated SH-

SY5Y cells against MPP+-induced death. An adenoviral

construct containing dnMLK1 was not tested in these

studies.

4. Discussion

CEP-1347 has been identified as a direct inhibitor of the

mixed lineage kinase (MLK) family and has previously

been found to attenuate MPTP-mediated DA neuronal death

and activation of the JNK signaling pathway in vivo

[38,51,52]. The present studies expand on those findings

by demonstrating that CEP-1347 and a dominant-negative

MLK3 adenovirus construct inhibit MPP+-induced death

and JNK signaling in SH-SY5Y cells. These studies further

implicate the MLK/JNK signaling pathway in MPP+-in-

duced neuronal death in vitro and suggest that this pathway

may be active in degenerating DA neurons in PD. Further,

these studies demonstrate the potential value of CEP-1347

as a neuroprotective compound in PD.

MPP+-treated SH-SY5Y cells are a useful in vitro model

for studying neurodegenerative events that may occur in PD

[7,30,33,46,55]. In the present study, CEP-1347 partially

prevented MPP+-induced cell death in retinoic acid differ-

entiated SH-SY5Y cells at low nanomolar concentrations.

CEP-1347 demonstrates neuroprotective properties in a

variety of primary neurons in culture including dorsal root

ganglion, sympathetic and motor neurons, and PC12 cells

after trophic factor withdrawal, DNA damage, or oxidative

stress [6,36,37]. CEP-1347 also maintains survival of motor

neurons in several in vivo models of programmed cell death,

such as developmental cell death of postnatal rat motor

neurons or of chick lumbar motor neurons in ovo and adult

rat hypoglossal neurons subjected to axotomy [19].

Morphologically, differentiated SH-SY5Y cells treated

with 3 mM MPP+ lose their processes, cluster, and eventu-

ally lift off the plate. This morphological change was

inhibited by pretreatment with CEP-1347. Additionally,

apoptotic indicators were demonstrated to be present fol-

lowing MPP+ addition to RA differentiated SH-SY5Y

cultures. CEP-1347 visually decreased the number of cells

displaying condensed chromatin, indicating that CEP-1347

may be preventing programmed cell death initiated by

MPP+ treatment. Inhibition of apoptotic cell death may be

important because markers of apoptosis have been observed

in many neurodegenerative diseases including PD [28].

However, others have shown that evidence of apoptosis

was missing in samples of patients with PD [15]. While a

snapshot of postmortem tissue is difficult to reconcile with

an ongoing neurodegenerative disease process, coupled with

the unknown clearance of apoptotic cells, it is difficult to

ascribe PD to a strictly apoptotic process. In other studies

CEP-1347 suppressed CHO cell-transfected MLK3-driven

apoptotic death at concentrations that inhibited MLK3

kinase activity [38]. Transient transfection of naive and

neuronally differentiated PC12 cells with MLK family

members showed apoptotic responses including membrane

blebbing, pyknotic nuclei and positive Hoechst staining in

addition to cell death [68], thereby relating MLK family

activation with potential apoptotic cell death.

To begin to address the mechanism of CEP-1347 inhibi-

tion of the MPP+ insult it was important to evaluate whether

the actual mitochondrial insult still occurred in the presence

of CEP-1347. When MPP+ interferes with mitochondrial

function at the level of complex I, lactate levels increase as a

result of an increase in glycolysis and increases in NADH

[45]. To determine whether CEP-1347 acted downstream of

mitochondrial demise, lactate levels were measured follow-

ing MPP+ treatment in the presence or absence of CEP-

1347. Lactate levels were increased twofold over basal

levels at 4, 24 and 48 h post MPP+. This increase was

unaffected by pretreatment with a neuroprotective concen-

tration of CEP-1347. These data indicated that CEP-1347

effects on cellular death pathways were downstream of this

initial toxic event.

CEP-1347 is a known inhibitor of the SAPK/JNK

signaling pathway and in vitro studies have demonstrated

inhibition at the level of the MLK family [38]. Increased

activity of JNK has been demonstrated following MPP+

treatment in undifferentiated SH-SY5Y cells [7,67]. In these

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–97 95

studies, undifferentiated cells were used in addition to

slightly higher concentrations of MPP+ (5 mM). In addition,

adenoviral gene transfer of the JNK binding domain of

JNK-interacting protein-1 (a scaffold protein and inhibitor

of JNK) inhibited MPP+-induced activation of JNK, c-Jun

and caspase and inhibited cell death in undifferentiated SH-

SY5Y cells [67]. The present study confirms these findings

by demonstrating that (1) MPP+ elevated pJNK in differen-

tiated SH-SY5Y cells, (2) that this could be partially

inhibited by adenoviral gene transfer of dominant negative

MLK3 and (3) completely blocked by an inhibitor of the

MLK family, CEP-1347/KT7515. It has been demonstrated

that all three JNK isoforms are represented in SH-SY5Y

cells [39]. What has not been determined is whether the

expression of these isoforms changes upon differentiation or

after MPP+ treatment. It is interesting that JNK activation

was completely inhibited by pretreatment with CEP-1347,

often with inhibition below basal conditions. CEP-1347

inhibition of JNK activation in differentiated SH-SY5Y

cells following MPP+ treatment was concentration depen-

dent with significant inhibition beginning at 10 nM and

maximal inhibition at 300–1000 nM. CEP-1347 does not

directly inhibit JNK in CHO cells as measured by a c-Jun

luciferase reporter construct [38] driven by c-Jun phosphor-

ylation, which is one of the immediate downstream targets

of JNK. This report [38] showed that only MLK3-induced

JNK activation could be inhibited by CEP-1347 and not

activation produced by MEKK1. More recently, there has

been described a lack of JNK activation in SH-SY5Y cells

following MPP+ [20]. These studies, however, were accom-

plished again in undifferentiated SH-SY5Y cells with much

lower concentrations of MPP+ (5 AM), which the authors

claim was insufficient to decrease mitochondrial function or

activate oxidative stress.

The MLK family has alternate downstream targets other

than JNK such as the transcription factor NF-nB and p38

[10,24,42,61]. Others have shown dominant negative ver-

sions of MLK3 prevent JNK activation induced by Rac and

Cdc42 but not JNK activation induced by MEKK1 [60].

Taken together, it seems that acute mitochondrial damage

initiates multiple intracellular signaling pathways leading to

cell death. Complete inhibition by CEP-1347 of the pJNK

induced by MPP+ mitochondrial damage, while only pre-

venting up to 70% of the cell death induced by MPP+,

suggests that another cell death pathway (other than through

JNK activation) is induced by MPP+ in these cells. CEP-

1347 can completely inhibit basal and MPP+-stimulated

JNK activation while at the same time only partially

prevents MPP+-induced cell death and condensed chroma-

tin. This study demonstrates that the JNK pathway disrup-

tion is critical but may not be sufficient to completely

protect from MPP+ toxicity. Others have shown that when

JNK is activated following UV light the resulting apoptosis

is JNK dependent [63]. They showed that the absence of

JNK1/2, through JNK1/2 null murine embryonic fibroblasts

prevented UV-induced apoptosis and DNA fragmentation

indicating that JNK is required for the normal apoptotic

response of fibroblasts to UV light. JNK null fibroblasts

were also resistant to the apoptotic effects of a genotoxin

(methyl methanesulfonate) and anisomycin, while the Fas-

death signaling pathway remained intact.

Utilization of an adenoviral dominant negative MLK3,

which infects >90% of differentiated SH-SY5Y cells and

allows considerable overexpression of this protein has shed

some light on the role of MLK3 in MPP+-induced cell

death. The dimerization of adenoviral overexpressed dom-

inant negative MLK3 with endogenous MLK3 [35] is

expected to prevent downstream phosphorylation of targets

such as MKK4 and/or MKK7. Leung and Lassam [35] have

shown that MLK3 homodimerization is crucial for down-

stream activation of the SAPK/JNK pathway. It is clear that

MLK3 partially contributes to the toxic effects of MPP+

through activation of JNK as demonstrated by our dominant

negative MLK3 data. Similar to CEP-1347, overexpression

of dnMLK3 prevented some, but not all, of the MPP+-

induced cell death. In contrast to CEP-1347, dnMLK3 did

not completely inhibit elevated levels of pJNK. CEP-1347 is

an inhibitor of the MLK family and would therefore be

expected to have effects beyond a single member such as

MLK3. Through a real-time RT PCR analysis of differen-

tiated SH-SY5Y cell lysate, expression of MLK1, MLK2,

MLK3 and ZPK(DLK) was measured. Expression of MLK1

and MLK3 had the higher relative expression seen among

these MLKs. It is theoretically possible for dnMLK2 or

dnDLK to heterodimerize with endogenous MLK3 to pre-

vent downstream activation of MKK4 or MKK7. Previous

studies using the same kinase dead (dominant negative)

mutations of MLK2 or DLK in plasmid transfection studies

with MLK3-activated JNK in PC12 cells [68] showed cross

reactivity. Others have shown cross reactivity with dominant

negative DLK prevention of MLK3-induced JNK activity in

293T cells [58]. Even though these dominant negative

adenoviral constructs overexpressed a considerable amount

of the dominant negative proteins in differentiated SH-

SY5Y cells, they were not protective following MPP+

treatment; in fact, they became toxic themselves at high

MOI concentrations. This toxicity was not seen with the

control construct, AdCMVLacZ or the positive test con-

struct, AdCMVdnMLK3, demonstrating that it was not

toxicity of a viral origin. CEP-1347 may be acting upon

other as yet undiscovered MLK family members or alternate

kinases.

This study demonstrates that the indolocarbazole CEP-

1347, likely through its inhibition of the MLK family,

completely inhibited the SAPK/JNK pathway and partially

prevented the neurotoxic effects of MPP+ in differentiated

SH-SY5Y cells. While CEP-1347 shows tendencies towards

an inverted U-shaped concentration response curve when

measuring cell survival, this is not demonstrated when

measuring JNK inhibition. At very high concentrations of

CEP-1347, other pharmacological rather than physiological

events could be occurring that are not addressed in this work.

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–9796

Adenoviral overexpression of dominant negative MLK3 also

partially protected differentiated SH-SY5Y cells from cell

death and pJNK activation indicating that MLK3 has a role

in MPP+-induced cell death. These results taken with previ-

ous studies indicate that CEP-1347 may be protective in

neurodegenerative diseases such as Parkinson’s Disease that

may involve the SAPK/JNK pathway leading to apoptosis.

References

[1] Y. Agid, Parkinson’s disease pathophysiology, Lancet 337 (1991)

1–3.

[3] J.L. Biedler, S. Roffler-Tarlov, M. Schachner, L.S. Freedman, Multi-

ple neurotransmitter synthesis by human neuroblastoma cell lines and

clones, Cancer Res. 38 (1978) 3751–3757.

[4] P.J. Blanchet, S. Konitsiotis, K. Hyland, L.A. Arnold, K.D. Pettigrew,

T.N. Chase, Chronic exposure to MPTP as a primate model of pro-

gressive parkinsonism: a pilot study with a free radical scavenger,

Exp. Neurol. 153 (1998) 214–222.

[5] L.J. Bloem, T.R. Pickard, S. Aton, M. Donoghue, R.C. Beavis, M.D.

Knierman, X. Wang, Tissue distribution and functional expression of

a cDNA encoding a novel mixed lineage kinase, J. Mol. Cell Cardiol.

33 (2001) 1739–1750.

[6] G.D. Borasio, S. Horstmann, J.M. Anneser, N.T. Neff, M.A.

Glicksman, CEP-1347/KT7515, a JNK pathway inhibitor, supports the

in vitro survival of chick embryonic neurons, NeuroReport 9 (1998)

1435–1439.

[7] D.S. Cassarino, E.M. Halvorsen, R.H. Swerdlow, N.N. Abramova,

W.D. Parker Jr., T.W. Sturgill, J.P. Bennett Jr., Interaction among

mitochondria, mitogen-activated protein kinases and nuclear factor-

kB in cellular models of Parkinson’s disease, J. Neurochem. 74 (2000)

1384–1392.

[8] S.J. Crocker, W.R. Lamba, P.D. Smith, S.M. Callaghan, R.S. Slack,

H. Anisman, D.S. Park, c-Jun mediates axotomy-induced dopamine

neuron death in vivo, PNAS 98 (2001) 13385–13390.

[9] A. Cuenda, D.S. Dorow, Differential activation of stress-activated

protein kinase kinases SKK4/MKK7 and SKK1/MKK4 by the

mixed-lineage kinase-2 and mitogen-activated protein kinase kinase

(MKK) kinase-1, Biochem. J. 333 (1998) 11–15.

[10] R.J. Davis, Signal transduction by the JNK group of MAP kinases,

Cell 103 (2000) 239–252.

[11] B. Dipasquale, A.M. Marini, R.J. Youle, Apoptosis and DNA degra-

dation induced by 1-methyl-4-phenylpyridinium in neurons, BBRC

181 (1991) 1442–1448.

[12] C.P. Fall, J.P. Bennett Jr., Characterization and time-course of MPP+-

induced apoptosis in human SH-SY5Y neuroblastoma cells, J. Neuro-

sci. Res. 55 (1999) 620–628.

[13] G. Fan, S.E. Merritt, M. Kortenjann, P.E. Shaw, L.B. Holzman, Dual

leucine zipper-bearing kinase (DLK) activates p46SAPK and

p38mapk but not ERK2, J. Biol. Chem. 271 (1996) 24788–24793.

[14] S.M. Farooqui, Induction of adenylate cyclase sensitive dopamine

D2-receptors in retinoic acid induced differentiated human neuroblas-

toma SHSY-5Y cells, Life Sci. 55 (1994) 1887–1893.

[15] I. Ferrer, R. Blanco, M. Carmona, B. Puig, M. Barrachina, C. Gomez,

S. Ambrosio, Active, phosphorylation-dependent mitogen-activated

protein kinase (MAPK/ERK), stress-activated protein kinase/c-Jun

N-terminal kinase (SAPK/JNK), and p38 kinase expression in Parkin-

son’s disease and Dementia with Lewy bodies, J. Neural. Transm. 108

(2001) 1383–1396.

[17] M. Gerlach, W. Gsell, J. Kornhuber, K. Jellinger, V. Krieger, F. Pan-

tucek, R. Vock, P. Riederer, A post mortem study on neurochemical

markers of dopaminergic, GABA-ergic and glutamatergic neurons in

basal ganglia-thalamocortical circuits in Parkinson syndrome, Brain

Res. 741 (1996) 142–152.

[18] G.D. Ghadge, R.P. Roos, U.J. Kang, R. Wollmann, P.S. Fishman,

A.M. Kalynych, E. Barr, J.M. Leiden, CNS gene delivery by retro-

grade transport of recombinant replication-defective adenoviruses,

Gene Ther. 2 (1995) 132–137.

[19] M.A. Glicksman, A.Y. Chiu, C.A. Dionne, M. Harty, M. Kaneko, C.

Murakata, R.W. Oppenheim, D. Prevette, D.R. Sengelaub, J.L.

Vaught, N.T. Neff, CEP-1347/KT7515 prevents motor neuronal

programmed cell death and injury-induced dedifferentiation in vivo,

J. Neurobiol. 35 (1998) 361–370.

[20] C. Gomez-Santos, I. Ferrer, J. Reiriz, F. Vinals, M. Barrachina, S.

Ambrosio, MPP+ increases alpha-synuclein expression and ERK/

MAP-kinase phosphorylation in human neuroblastoma SH-SY5Y

cells, Brain Res. 935 (2002) 32–39.

[21] I. Gotoh, M. Adachi, E. Nishida, Identification and characterization of

a novel MAP kinase kinase kinase, MLTK, J. Biol. Chem. 276 (2001)

4276–4286.

[22] S. Gupta, T. Barrett, A.J. Whitmarsh, J. Cavanagh, H.K. Sluss,

B. Derijard, R.J. Davis, Selective interaction of JNK protein

kinase isoforms with transcription factors, EMBO J. 15 (1996)

2760–2770.

[23] A. Hartley, J.M. Stone, C. Heron, J.M. Cooper, A.H. Schapira, Com-

plex I inhibitors induce dose-dependent apoptosis in PC12 cells; rel-

evance to Parkinson’s disease, J. Neurochem. 63 (1994) 1987–1990.

[24] S.P. Hehner, T.G. Hofmann, A. Ushmorov, O. Dienz, I.W.-L. Leung,

N. Lassam, C. Scheidereit, W. Droge, M.L. Schmitz, Mixed-lineage

kinase 3 delivers CD3/CD28-derived signals into the IkB kinase com-

plex, Mol. Cell. Biol. 20 (2000) 2556–2568.

[25] R.E. Heikkila, A. Hess, R.C. Duvoisin, Dopaminergic neurotoxicity

of 1-methyl-4 phenyl-1,2,3,6-tetrahydropyridine in mice, Science 224

(1984) 1451–1453.

[26] R.E. Heikkila, L. Manzino, F.S. Cabbat, R.C. Duvoisin, Protection

against the dopaminergic neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine by monoamine oxidase inhibitors, Nature 311

(1984) 467–469.

[27] S. Hirai, M. Izawa, S. Osada, G. Spyrou, S. Ohno, Activation of the

JNK pathway by distantly related protein kinases, MEKK and MUK,

Oncogene 12 (1996) 641–650.

[28] E.C. Hirsch, S. Hunot, B. Faucheux, Y. Agid, Y. Mizuno, H. Mochi-

zuki, W.G. Tatton, N. Tatton, W.C. Olanow, Dopaminergic neurons

degenerate by apoptosis in Parkinson’s disease, Mov. Disord. 14

(1999) 383–385.

[29] D. Hockenbery, G. Nunez, C. Milliman, R.D. Schreiber, S.J.

Korsmeyer, Bcl-2 is an inner mitochondrial membrane protein that

blocks programmed cell death, Nature 348 (1990) 334–336.

[30] Y. Itano, Y. Nomura, 1-Methyl-4-phenyl-pyridinium ion (MPP+)

causes DNA fragmentation and increases the Bcl-2 expression in

human neuroblastoma, SH-SY5Y cells, through different mecha-

nisms, Brain Res. 704 (1995) 240–245.

[32] D.R. Kaplan, K. Matsumoto, E. Lucarelli, C.J. Thiele, Induction of

TrkB by retinoic acid mediates biologic responsiveness to BDNF and

differentiation of human neuroblastoma cells, Neuron 11 (1993)

321–331.

[33] Y. Kitamura, T. Kosaka, J.-I. Kakimura, Y. Matsuoka, Y. Kohno, Y.

Nomura, T. Taniguchi, Protective effects of the antiparkinsonian drugs

talipexole and pramipexole against 1-methyl-4-phenylpyridinium-in-

duced apoptotic death in human neuroblastoma SH-SY5Y cells, Mol.

Pharmacol. 54 (1998) 1046–1054.

[34] J.W. Langston, The etiology of Parkinson’s disease with emphasis on

the MPTP story, Neurology 47 (1996) S153–S160.

[35] I.W. Leung, N. Lassam, Dimerization via tandem leucine zippers is

essential for the activation of the nitrogen-activated protein kinase

kinase kinase, MLK3, J. Biol. Chem. 273 (49) (1998) 32408–32415.

[36] A.C. Maroney, M.A. Glicksman, A.N. Basma, K.M. Walton, E.

Knight Jr., C.A. Murphy, B.A. Bartlett, T. Angeles, Y. Matsuda,

N.T. Neff, C.A. Dionne, Motoneuron apoptosis is blocked by CEP-

1347 (KT 7515), a novel inhibitor of the JNK signaling pathway,

J. Neurosci. 18 (1998) 104–111.

J.R. Mathiasen et al. / Brain Research 1003 (2004) 86–97 97

[37] A.C. Maroney, J.P. Finn, D. Bozyczko-Coyne, T.M. O’Kane, N.T.

Neff, A.M. Tolkovsky, D.S. Park, C.Y. Yan, C.M. Troy, L.A. Greene,

CEP-1347 (KT7515), an inhibitor of JNK activation, rescues sympa-

thetic neurons and neuronally differentiated PC12 cells from death

evoked by three distinct insults, J. Neurochem. 73 (1999) 1901–1912.

[38] A.C. Maroney, J.P. Finn, T.J. Connors, J.T. Durkin, T. Angeles, G.

Gessner, Z. Xu, S.L. Meyer, M.J. Savage, L.A. Greene, R.W. Scott,

J.L. Vaught, CEP-1347 (KT7515), a semisynthetic inhibitor of the

mixed lineage kinase family, J. Biol. Chem. 276 (2001) 25302–25308.

[39] K. Mielke, A. Damm, D.D. Yang, T. Herdegen, Selective expression

of JNK isoforms and stress-specific JNK activity in different neural

cell lines, Mol. Brain Res. 75 (2000) 128–137.

[40] N. Mittereder, K.L. March, B.C. Trapnell, Evaluation of the concen-

tration and bioactivity of adenovirus vectors for gene therapy, J. Virol.

70 (1996) 7498–7509.

[41] H. Mochizuki, N. Nakamura, K. Nishi, Y. Mizuno, Apoptosis is

induced by 1-methyl-4-phenylpyridinium ion (MPP+) in ventral

mesencephalic-striatal co-culture in rat, Neurosci. Lett. 170 (1994)

191–194.

[42] M. Mota, M. Reeder, J. Chernoff, C.E. Bazenet, Evidence for a role of

mixed lineage kinases in neuronal apoptosis, J. Neurosci. 21 (2001)

4949–4957.

[43] W.J. Nicklas, I. Vyas, R.E. Heikkla, Inhibition of NADH-linked ox-

idation in brain mitochondria by 1-methyl-4-phenylpyridine, a metab-

olite of the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine,

Life Sci. 36 (1985) 2503–2508.

[44] D. Offen, P.M. Beart, N.S. Cheung, C.J. Pascoe, A. Hocham, S.

Gorodin, E. Melamed, R. Bernard, O. Bernard, Transgenic mice

expressing human Bcl-2 in their neurons are resistant to 6-hydroxy-

dopamine and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neuro-

toxicity, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 5789–5794.

[45] S. Ofori, R.E. Heikkila, W.J. Nicklas, Attenuation by dopamine up-

take blockers of the inhibitory effects of 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine and some of its analogs on NADH-linked metab-

olism in mouse neostriatal slices, J. Pharmacol. Exper. Ther. 251

(1989) 258–266.

[46] C.-W. Park, H.-S. Lee, Y.-S. Kim, Mechanism of MPP+-induced

cytotoxicity in human neuroblastoma SH-SY5Y, J. Toxicol. Sci. 23

(Suppl. II) (1998) 184–188.

[49] H. Sakuma, A. Ikeda, S. Oka, Y. Kozutsumi, J.P. Zanetta, T. Kawa-

saki, Molecular cloning and functional expression of a cDNA encod-

ing a new member of mixed lineage protein kinase from human brain,

J. Biol. Chem. 272 (1997) 28622–28629.

[50] M.S. Saporito, R.E. Heikkila, S.K. Youngster, W.J. Nicklas, H.M.

Geller, Dopaminergic neurotoxicity of 1-methyl-4-phenylpyridinium

analogs in cultured mesencephalon: relationship to dopamine up-

take affinity and inhibition of mitochondrial respiration, J. Pharma-

col. Exp. Ther. 260 (1992) 1400–1409.

[51] M.S. Saporito, E.M. Brown, M.S. Miller, S. Carswell, CEP-1347/KT-

7515, An inhibitor of c-jun N-terminal kinase activation, attenuates

the 1-methyl-4-phenyl tetrahydropyridine-mediated loss of nigrostria-

tal dopaminergic neurons in vivo, J. Pharmacol. Exp. Ther. 288

(1999) 421–427.

[52] M.S. Saporito, B.A. Thomas, R.W. Scott, MPTP activates c-jun NH2-

terminal kinase (JNK) and its upstream regulatory kinase MKK4 in

nigrostriatal neurons in vivo, J. Neurochem. 75 (2000) 1200–1208.

[53] J. Schaack, S. Langer, X. Guo, Efficient selection of recombinant

adenovirus by vectors that express beta-galactosidase, J. Virol. 69

(1995) 3920–3923.

[54] A.H.V. Schapira, Evidence for mitochondrial dysfunction in Parkin-

son’s disease—a critical appraisal, Mov. Disord. 9 (1994) 125–138.

[55] J.P. Sheehan, P.E. Palmer, G.A. Helm, J.B. Tuttle, MPP+ induced

apoptotic cell death in SH-SY5Y neuroblastoma cells: an electron

microscope study, J. Neurosci. Res. 48 (1997) 226–237.

[56] R.H. Swerdlow, J.K. Parks, S.W. Miller, J.B. Tuttle, P.A. Trimmer,

J.P. Sheehan, J.P. Bennett Jr., R.E. Davis, W.D. Parker Jr., Origin and

functional consequences of the complex I defect in Parkinson’s dis-

ease, Ann. Neurol. 40 (1996) 663–671.

[57] T. Takahashi, Y. Deng, W. Maruyama, P. Dostert, M. Kawai, M. Naoi,

Uptake of a neurotoxin-candidate, (R)-1,2-dimethyl-6,7-dihydroxy-

1,2,3,4-tetrahydroisoquinoline into human dopaminergic neuroblasto-

ma SH-SY5Y cells by dopamine transport system, J. Neural Transm.

Gen. Sect. 98 (1994) 107–118.

[58] S. Tanaka, H. Hanafusa, Guanine-nucleotide exchange protein C3G

activates JNK1 by a ras-independent mechanism. JNK1 activation

inhibited by kinase negative forms of MLK3 and DLK mixed lineage

kinases, J. Biol. Chem. 273 (3) (1998) 1281–1284.

[59] N.A. Tatton, S.J. Kish, In situ detection of apoptotic nuclei in the

substantia nigra compacta of 1-methyl-4-phenyl-1,2,3,6-tetrahydro-

pyridine-treated mice using terminal deoxynucleotidyl transferase

labelling and acridine orange staining, Neuroscience 77 (1991)

1037–1048.

[60] H. Teramoto, O.A. Coso, H. Miyata, T. Igishi, T. Miki, J.S. Gutkind,

Signaling from the small GTP-binding proteins Rac1 and Cdc42 to

the c-Jun N-terminal kinase/stress-activated protein kinase pathway,

J. Biol. Chem. 271 (1996) 27225–27228.

[61] L.A. Tibbles, Y.L. Ing, F. Kiefer, J. Chan, N. Iscove, J.R. Woodgett,

N.J. Lassam, MLK-3 activates the SAPK/JNK and p38/RK pathways

via SEK1 and MKK3/6, EMBO J. 15 (1996) 7026–7035.

[62] K.F. Tipton, T.P. Singer, Advances in our understanding of the mech-

anisms of the neurotoxicity of MPTP and related compounds, J. Neu-

rochem. 61 (1993) 1191–1206.

[63] C. Tournier, P. Hess, D.D. Yang, J. Xu, T.K. Turner, A. Nimnual, D.

Bar-Sagi, S.N. Jones, R.A. Flavell, R.J. Davis, Requirement of JNK

for stress-induced activation of the cytochrome c-mediated death

pathway, Science 288 (2000) 870–874.

[64] P.A. Trimmer, T.S. Smith, A.B. Jung, J.P. Bennett Jr., Dopamine

neurons from transgenic mice with knockout of the p53 gene resist

MPTP neurotoxicity, Neurodegeneration 5 (1996) 233–239.

[65] I. Vyas, R.E. Heikkila, W.J. Nicklas, Studies on the neurotoxicity of

1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine: inhibition of NAD-

linked substrate oxidation by its metabolite, 1-methyl-4-phenylpyri-

dinium, J. Neurochem. 46 (1986) 1501–1507.

[67] X.G. Xia, T. Harding, M. Weller, A. Bieneman, J.B. Uney, J.B.

Schulz, Gene transfer of the JNK interacting protein-1 protects dopa-

minergic neurons in the MPTP model of Parkinson’s disease, PNAS

98 (2001) 10433–10438.

[68] Z. Xu, A.C. Maroney, P. Dobrzanski, N.V. Kukekov, L.A. Greene,

The MLK family mediates c-jun N-terminal kinase activation in neu-

ronal apoptosis, Mol. Cell. Biol. 21 (2001) 4713–4724.

[69] L. Yang, R.T. Matthews, J.B. Schulz, T. Klockgether, A.W. Liao, J.-C.

Martinou, J.B. Penney, B.T. Hyman, M.F. Beal, 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridne neurotoxicity is attenuated in mice overex-

pressing BCL-2, J. Neurosci. 18 (1998) 8145–8152.


Recommended