ArticlePDF Available

Elevated P75NTR expression causes death of engrailed-deficient midbrain dopaminergic neurons by Erk1/2 suppression

Authors:

Abstract and Figures

The homeodomain transcription factors Engrailed-1 and Engrailed-2 are required for the survival of mesencephalic dopaminergic (mesDA) neurons in a cell-autonomous and gene-dose-dependent manner. Homozygote mutant mice, deficient of both genes (En1-/-;En2-/-), die at birth and exhibit a loss of all mesDA neurons by mid-gestation. In heterozygote animals (En1+/-;En2-/-), which are viable and fertile, postnatal maintenance of the nigrostriatal dopaminergic system is afflicted, leading to a progressive degeneration specific to this subpopulation and Parkinson's disease-like molecular and behavioral deficits. In this work, we show that the dose of Engrailed is inversely correlated to the expression level of the pan-neurotrophin receptor gene P75NTR (Ngfr). Loss of mesDA neurons in the Engrailed-null mutant embryos is caused by elevated expression of this neurotrophin receptor: Unusually, in this case, the cell death signal of P75NTR is mediated by suppression of Erk1/2 (extracellular-signal-regulated kinase 1/2) activity. The reduction in expression of Engrailed, possibly related to the higher levels of P75NTR, also decreases mitochondrial stability. In particular, the dose of Engrailed determines the sensitivity to cell death induced by the classic Parkinson-model toxin MPTP and to inhibition of the anti-apoptotic members of the Bcl-2 family of proteins. Our study links the survival function of the Engrailed genes in developing mesDA neurons to the regulation of P75NTR and the sensitivity of these neurons to mitochondrial insult. The similarities to the disease etiology in combination with the nigral phenotype of En1+/-;En2-/- mice suggests that haplotype variations in the Engrailed genes and/or P75NTR that alter their expression levels could, in part, determine susceptibility to Parkinson's disease.
Differential activation of Erk1/2 in mesDA neurons. (A-D, G-K) Immunohistochemistry of E12 ventral midbrain cell culture stained against TH (green), total Erk1/2 protein (red) (A, B) and phosphorylated Erk1/2 (red) (C-D, G-K). (A-D) While Erk1/2 protein is present in mesDA neurons of both genotypes (A, B), it is only phosphorylated in En2-/- mesDA neurons (C) and not in the En1-/-;En2-/- (EnDM) counterparts (D). (E-I) Erk1/2 becomes activated in EnDM mesDA neurons after treatment with the survival-inducing neurotrophins, brain-derived neurotrophic factor (BDNF), neurotrophin (NT)4 and NT3, or after silencing of P75NTR (RNA interference (RNAi)) (G-J), but not when glial cell line-derived neurotrophic factor (GDNF) is applied (I). (E) Western blot of E12 ventral midbrain tissue confirms the immunohistochemical finding of differential phosphorylation between genotypes and shows that neither AKT, part of the phosphotidyl inositol-3 kinase pathway, nor other mitogen-activated protein kinases, such as JNK and P38, are differentially activated. (F) Quantification of phosphorylated Erk1/2 in western blot normalized against En2-/- tissue. (L) Number of TH-positive cells in EnDM and En2-/- ventral midbrain cultures after 72 hours, treated with the 400 nM Mek inhibitor U0126 in conjunction with BDNF, Penetratin-coupled P75NTR double-stranded RNA oligonucleotides and the P75NTR inhibiting antibody (Rex). Numbers are normalized against untreated cultures at 24 hours. The rescue effect is significantly reduced when the EnDM cultures are treated with the Erk1/2 inhibitor. Scale bars: 25 μm. Error bars indicate standard error. Ctl, control.
… 
Content may be subject to copyright.
BioMed Central
Page 1 of 13
(page number not for citation purposes)
Neural Development
Open Access
Research article
Elevated P75NTR expression causes death of engrailed-deficient
midbrain dopaminergic neurons by Erk1/2 suppression
Kambiz N Alavian1,2, Paola Sgadò1,3, Lavinia Alberi1,4,
Srinivasa Subramaniam1,5 and Horst H Simon*1
Address: 1Interdisciplinary Centre for Neuroscience, Department of Neuroanatomy, Ruprecht-Karls-Universität, 69120 Heidelberg, Germany,
2Harvard Medical School, Neuroregeneration Labs, MRC 1, McLean Hospital, Mill St, Belmont, MA 02478, USA, 3Paola Sgadò, Neurogenetics
Laboratory, Child Neurology Unit, Pediatric Hospital A Meyer, Piazza di Careggi, 50139 Florence, Italy, 4The Johns Hopkins Institute for Cell
Engineering, Department of Neurology, North Broadway Street, BRB 720, Baltimore, MD 2120, USA and 5Department of Neuroscience, Johns
Hopkins Medical School, N Wolfe Street, Baltimore, MD 21210, USA
Email: Kambiz N Alavian - kambiz.alavian@gmail.com; Paola Sgadò - paola.sgado@gmail.com; Lavinia Alberi - lalberi2@jhmi.edu;
Srinivasa Subramaniam - ssubram9@jhmi.edu; Horst H Simon* -hsimon@mac.com
* Corresponding author
Abstract
Background: The homeodomain transcription factors Engrailed-1 and Engrailed-2 are required for
the survival of mesencephalic dopaminergic (mesDA) neurons in a cell-autonomous and gene-dose-
dependent manner. Homozygote mutant mice, deficient of both genes (En1-/-;En2-/-), die at birth
and exhibit a loss of all mesDA neurons by mid-gestation. In heterozygote animals (En1+/-;En2-/-),
which are viable and fertile, postnatal maintenance of the nigrostriatal dopaminergic system is
afflicted, leading to a progressive degeneration specific to this subpopulation and Parkinson's
disease-like molecular and behavioral deficits.
Results: In this work, we show that the dose of Engrailed is inversely correlated to the expression
level of the pan-neurotrophin receptor gene P75NTR (Ngfr). Loss of mesDA neurons in the Engrailed-
null mutant embryos is caused by elevated expression of this neurotrophin receptor: Unusually, in
this case, the cell death signal of P75NTR is mediated by suppression of Erk1/2 (extracellular-signal-
regulated kinase 1/2) activity. The reduction in expression of Engrailed, possibly related to the
higher levels of P75NTR, also decreases mitochondrial stability. In particular, the dose of Engrailed
determines the sensitivity to cell death induced by the classic Parkinson-model toxin MPTP and to
inhibition of the anti-apoptotic members of the Bcl-2 family of proteins.
Conclusion: Our study links the survival function of the Engrailed genes in developing mesDA
neurons to the regulation of P75NTR and the sensitivity of these neurons to mitochondrial insult.
The similarities to the disease etiology in combination with the nigral phenotype of En1+/-;En2-/-
mice suggests that haplotype variations in the Engrailed genes and/or P75NTR that alter their
expression levels could, in part, determine susceptibility to Parkinson's disease.
Published: 16 March 2009
Neural Development 2009, 4:11 doi:10.1186/1749-8104-4-11
Received: 18 November 2008
Accepted: 16 March 2009
This article is available from: http://www.neuraldevelopment.com/content/4/1/11
© 2009 Alavian et al.; licensee BioMed Central Ltd.
This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 2 of 13
(page number not for citation purposes)
Background
Mesencephalic dopaminergic (mesDA) neurons are the
main source of dopamine in the mammalian central nerv-
ous system. They are located in three distinct nuclei, sub-
stantia nigra pars compacta, ventral tegmentum and
retrorubral field. Their main innervation targets are the
basal ganglia, where they play key roles in the control of
emotion, motivation and motor behavior, documented
by their connection to schizophrenia, addiction and, most
prominently, to Parkinson's disease (PD) [1,2]. The main
characteristic of PD is the slow progressive loss of
dopaminergic neurons in the substantia nigra pars com-
pacta, causing diminished release of dopamine in the cau-
date putamen and debilitating motor deficits. Although
the molecular causes for the selective vulnerability of this
neuronal population are hardly understood, multiple
lines of evidence suggest mitochondrial dysfunction as a
major contributing factor [3] and apoptosis as the execu-
tioner of cell death [4]. Several mutations associated with
familial forms of PD encode mitochondrial proteins [5]
and neurotoxins specific to the nigrostriatal system –
MPTP (1-methyl-4-phenyl-,1,3,6-tetrahydropyridine), 6-
hydroxydopamine and rotenone – cause mitochondrial
damage as inhibitors of complex-I of the electron trans-
port chain [6]. Mitochondrial insult can cause oxidative
stress by production of reactive oxygen species, leading to
increased permeability of the mitochondrial membrane,
release of pro-apoptotic molecules, including cyto-
chrome-C, into the cytoplasm and, subsequently, to acti-
vation of caspases and induction of apoptosis [7].
Neuronal cell death can be a result of neurotrophin defi-
ciency. Action of the neurotrophins, consisting of nerve
growth factor (NGF), brain-derived neurotrophic factor
(BDNF), neurotrophin (NT)4/5 and NT3, is mediated via
a set of specific tyrosine kinase (Trk) receptors (TrkA, B, C)
and a common receptor, P75NTR (Ngfr). While the Trk
receptors signal survival, P75NTR can relay a survival or cell
death signal, depending on the cellular context and the
molecular form of the ligand [8]. The pro-survival func-
tion of neurotrophins and their receptors has been mainly
attributed to the downstream effect of phosphotidyl inosi-
tol-3 kinase (PI3K) and the extracellular-signal-regulated
kinase 1/2 (Erk1/2) pathways [9], whereas cell death sig-
naling via P75NTR is mediated by phosphorylation of c-Jun
N-terminal kinase (JNK) and BH3-only members of the
Bcl-2 family [10].
The homeodomain transcription factors Engrailed-1 (En1)
and Engrailed-2 (En2) are required for the survival and
maintenance of mesDA neurons in a cell-autonomous
and gene-dose-dependent manner, demonstrated by in
vitro cell mixing experiments, RNA interference (RNAi)
and analysis of chimeric mice [11-13]. In Engrailed double
mutant mice (En1-/-;En2-/-; from here on EnDM), the
mesDA neurons are generated and begin to express their
neurotransmitter phenotype but then die by apoptosis
between embryonic day (E)12 and E14, the stage when
Engrailed expression starts in their wild-type counterparts
[11,12]. The intermediate genotypes between EnDM and
wild type show various degrees of cell loss in the mesDA
system. Most interestingly, mice heterozygous null for En1
and homozygous null for En2 (En1+/-;En2-/-; from here
on EnHT), which are viable and fertile, exhibit a slow pro-
gressive loss of nigral dopaminergic neurons within the
first two months after birth, resulting in diminished stor-
age and release of dopamine in the striatum and in PD-
like motor deficiencies [14].
We show here that Engrailed-deficiency in mesDA neurons
leads to elevated P75NTR expression that is causal for cell
death if the neurons are null for En1 and En2. The death
signal is mediated by the suppression of Erk1/2 activity.
Probably linked to the P75NTR elevation, the dose of the
Engrailed genes also determines the sensitivity to mito-
chondrial insult.
Materials and methods
Animals
The generation of the En2 null mutant and the En1tau-
LacZ 'knock-in' mice have been previously described
[15,16]. The En2 mutants with an original mixed genetic
background of 129 and Swiss Webster were crossed three
times into a C57/BL6 background. The line was bred as
En1+/tlZ;En2-/- at the central animal facility, University of
Heidelberg.
Quantitative RT-PCR
Quantitative RT-PCR reactions were performed according
to the manufacture in a 7000 Sequence Detection System
from Applied Biosystems (Foster City, CA, USA) using
pre-formulated 'assays on-demand' and calculating the
results with the comparative cycle time (CT) method. The
pre-formulated 'assays on demand' had the following
identification tag: Mm00446294_m1 for Ngfr (P75NTR);
as standard control Mm00507222_s1 ribosomal protein
S18; Mm00435617_m1 for phosphoglycerate kinase 1
(Pgk1); and Mm00446973_m1 for TATA box binding
protein (Tbp). The dissected ventral midbrains were
homogenized, the RNA isolated and reverse transcribed
using random hexamers to initiate transcription. Each of
the individual PCR reactions was done in triplicate and at
least two of three standard controls were run in parallel.
Each of the experimental sets consisted of RNA from dif-
ferent pools of mutant and littermate control as well as
RNA from Tet-On induced Engrailed expressing N2A cells
and control.
Cell culture
All primary cell cultures were performed using E12.5
mouse embryos. EnDM embryos were distinguished from
the other genotypes in the litter by their midbrain/hind-
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 3 of 13
(page number not for citation purposes)
brain morphology [11], which was occasionally verified
by PCR. Distinction between En2-/- and En1+/tlZ;En2-/-
embryos was achieved by incubation of the limb buds in
X-Gal solution at 37°C (40 mg/ml X-Gal (Sigma-Aldrich;
Munich, Germany) in 5 mM K3Fe(CN), 5 mM
K4Fe(CN)6x3H2O, 1 mM MgCl2 in phosphate-buffered
saline). For the cell culture, the neural tubes were dis-
sected and ventral midbrains were isolated. The tissue was
then dissociated using trypsine (Invitrogen; Karlsruhe,
Germany). The preparation of laminin (Sigma) coated
coverslips was described elsewhere [11]. The medium was
DMEM-F12 supplemented with 5% fetal calf serum,
0.25% bovine serum albumin (Sigma), 33 mM glucose,
50 U/ml penicillin, 50 U/ml streptomycin, and 1% Fung-
izone (Invitrogen). The cells were seeded at approximately
150,000 per cover slip and incubated at 37°C. After 36
hours, EnDM mutant cells were fixed in 4% paraformalde-
hyde and processed for immunostaining. The typical
numbers of tyrosine hydroxylase (TH)-positive cells per
cover slip were between 100 and 300 cells if wild-type or
heterozygote mutant tissue was dissociated. The numbers
of TH-positive neurons was always significantly lower if
the dissociated ventral midbrains were derived from E12
EnDM embryos. To obtain comparable numbers for
mutant and wild-type experiments, all cell counts were
normalized against each of the controls. Numbers pre-
sented in the results section (n) refer to the number of
experiments. Each of the experimental conditions is repre-
sented by at least three cover slips in each experiment. The
optimum concentration for the toxic substances was
determined by titration and checking for an intermediate
rate of survival (between 30% and 60%) in cultures of
mixed genotypes (En2-/- and En1+/-;En2-/-). For induc-
tion of cell death, serum was withdrawn from the medium
after 48 hours and the cultures were treated with the com-
pounds HA14-1, chelerythrine chloride (Axxora, San
Diego, CA)), prima-1, Apoptosis Activator-2, tumor
necrosis factor (TNF)α and 1-methyl-4-phenylpyridinium
(MPP+). Used concentrations, solvents, durations of treat-
ment and vendor sources are provided in Additional file
1[17-33]. The number 'n' corresponds to the number of
individual experiments conducted.
Design and transfection of siRNA oligos
The design of the 21-mer RNAi oligonucleotides was car-
ried out at Biomers.net (Ulm, Germany) and in accord-
ance with the protocol by Ebashir et al. [34]. Both RNA
duplexes targeted the coding sequence of P75NTR (A, sense
ACAGAACACAGUGUGUGAA(dTdT) and anti-sense
UUCACACACUGUGUUCUGU(dTdT); B, sense CAUUC-
CGACCGCUGAUGUUCU(dTdT) and anti-sense AACAU-
CAGCGGUCGGAAUGUG(dTdT)). For coupling with
Penetratin-One (QBiogene; Strasbourg, France), the sense
strand was modified with a thiol group on the 5' end. Tris-
2-carboxyethylphosphine (TCEP; 1 μl; Pierce; Bonn, Ger-
many) was added to 224 μl of the small interfering RNA
(siRNA; stock solution, 900 μM) and incubated for 15
minutes at room temperature. Then, 25 μl of Penetratin-1
was added, mixed and incubated for 5 minutes at 65°C
following by 1 hour at 37°C. Penetratin-1 solution was
reconstituted to 2 mg/ml (0.8 mM) in sterile water. A
stock solution of 20 mM TCEP in sterile RNAse/DNAse
free water was made. The aliquots were frozen at -80°C.
The Penetratin-coupled siRNA oligos were heated to 65°C
for 15 minutes and 3 μl of the mix was dissolved in com-
plete growth medium. As controls, we used Penetratin-1
coupled double-stranded RNA oligos directed against
Maged1 (Nrage) (UAACUUGAAUGUGGAAGAG(dTdT)
and CUCUUCCACAUUCAAGUUA(dTdT)) and the ran-
domly generated Scramble I Duplex (Dpharmacon; Hei-
delberg, Germany) sense CAGTCGCGTTTGCGACTGG
and antisense CCAGTCGCAAACGCGACTG. Both had no
effect on the survival rate of control or EnDM mesDA neu-
rons. Alternatively, uncoupled double-stranded RNA oli-
gos with the same sequences were transfected using
HiPerfect (Qiagen; Hilden, Germany) in accordance with
the manufacturer's protocol.
Immunohistochemistry and western blot analysis
The immunohistochemistry and western blot analysis was
done according to the protocol described elsewhere [35].
All the phospho-specific antibodies were purchased from
Cell Signaling Technology, the neurotrophin and TH anti-
bodies from Chemicon (Molsheim, France) and the anti-
bodies for neurotrophin receptors from Santa Cruz
Biotechnology (Heidelberg, Germany).
Statistical analysis
Values are expressed as mean ± standard error. Differences
between means were analyzed by using a paired, two-
tailed Student t-test. All shown p-values are rounded up at
the third or fourth digit.
Results
Elevated P75NTR expression in absence of Engrailed genes
A genome-wide expression analysis, using microarrays on
En1 inducible N2A cell line, identified the NGF receptor
P75NTR as downstream of the Engrailed transcription fac-
tors (data not shown). To confirm the microarray, we
examined by quantitative RT-PCR the levels of P75NTR in
the cell line as well as in ventral midbrain tissue in rela-
tionship to En1 expression. As a result of En1 induction in
N2A cells, the endogenous expression of P75NTR decreased
by 10-fold (9.9 ± 1.9%, p < 0.001, n = 8). Likewise, the
ventral midbrain tissue, derived from control littermates
(En2-/-) expressed almost 2.5-fold less P75NTR (42% ±
14.0%, p < 0.001, n = 6) than EnDM mutants (Figure 1A).
The western blot analysis confirmed the latter results; ven-
tral midbrain from En2-/- and EnHT embryos contained
74.4 ± 4.4% (p < 0.001, n = 3) and 60.2 ± 8.8% (p = 0.002,
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 4 of 13
(page number not for citation purposes)
n = 3) less P75NTR protein, respectively, than the same tis-
sue from EnDM littermates (Figure 1B,C).
Since P75NTR can mediate cell death in neurons [8], we
began to investigate whether its elevated expression is
causal for the death of mesDA neurons in EnDM embryos.
In order to functionally interfere with P75NTR, we applied
an activity-blocking antibody (Rex) [28] to primary ven-
tral midbrain cell cultures. This antibody increased the
survival rate from 7.5 ± 1.24% to 34.8 ± 4.6% (p < 0.001,
n = 6; Figure 1D). Furthermore, to lower P75NTR expres-
sion levels in the mutant neurons, we applied specific
Penetratin-coupled siRNA duplexes [36]; 72 hours after
transfection, the total P75NTR protein was reduced by 83.2
± 6.3% (p = 0.05, n = 3; western blot not shown) and the
survival rate increased from 7.5 ± 1.24% to 25.1 ± 2.1% (p
< 0.001 n = 16) (Figure 1D). These data suggested that ele-
vated expression of P75NTR is the direct cause of the induc-
tion of apoptosis in Engrailed-deficient mesDA neurons.
P75NTR mediates dual, opposing functions of cell survival
and death, controlled by the presence or absence of neu-
rotrophins. For the anti-apoptotic function, neuro-
trophins require their cognate Trk receptors as
heterodimerization partners for P75NTR [8]. In order to
assess a potential role of the Trk/P75NTR system during the
course of cell loss, we determined the expression of the
Trk-receptors in E12 mesDA neurons. TrkC and TrkB, but
not TrkA, were detectable by immunohistochemistry and
western blot at equal levels in wild type and EnDM mutants
(Figure 2A–G).
The up-regulation of P75NTR and the presence of Trk recep-
tors suggested that Engrailed deficiency introduces a neu-
rotrophin requirement to the E12 mesDA neurons that
cannot be satisfied at this age, since the neurotrophins
specific to TrkB and TrkC – that is, BDNF, NT4 and NT3 –
are not expressed in the E12 ventral midbrain as they are
in the adult (Figure 2H). To test this hypothesis, we
applied saturating concentrations of BDNF, NT4 and NT3
to ventral midbrain cultures. After 72 hours, 50.2 ± 2.9%
(p < 0.0001, n = 27), 42.3 ± 10.1% (p < 0.001, n = 9) and
26.0 ± 3.5% (p < 0.001, n = 9), respectively, of the other-
wise dying EnDM mesDA neurons were still present in the
cultures (Figure 2I). The addition of BDNF to the control
littermate cultures demonstrated that this was due to an
elevated survival rate and not attributable to a higher rate
of precursor cell proliferation (Figure 3L). As expected
from the lack of TrkA, application of its ligand, NGF, did
not change the survival rate significantly. To test the spe-
cificity of BNDF, NT3 and NT4, we applied glial cell line-
derived neurotrophic factor (GDNF), growth differentia-
tion factor (GDF)-15 and transforming growth factor
(TGF)-β to the mutant cultures, all known survival factors
for mesDA neurons [37-39]. Similar to NGF, none of
them prevented the death of the Engrailed-deficient
mesDA neurons (Figure 2I). Furthermore, the linear dose-
response trend-line of the survival effect of BDNF (Figure
Elevated P75NTR expression is causal for cell deathFigure 1
Elevated P75NTR expression is causal for cell death. (A) Quantitative RT-PCR of ventral midbrain tissue (VM) derived
from EnDM (En1-/-;En2-/-) and EnHT E12 embryos, and of En1-expressing N2A cells inducible by doxycycline (Dox). P75NTR
expression is inversely correlated with En1 expression levels in tissues and cell lines. (B, C) Western blot analysis of the ven-
tral midbrain tissue shows the same relationship between P75NTR protein levels and En1 expression. Each active En1 allele
decreases the P75NTR expression level (n = 3, p = 0.002). (D) Ventral midbrain cultures derived from EnDM and En2-/- embryos.
Silencing of P75NTR by double-stranded RNA oligos and application of P75NTR-inhibiting antibody (Rex) increases the survival
rate of EnDM mesDA neurons compared to untreated control (Ctl) or after treatment with scrambled RNA oligos (n 6, p <
0.01). Error bars indicate standard error.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 5 of 13
(page number not for citation purposes)
2J) indicated a Kd value of approximately 2.5 nM, which
corresponds with the reported affinity of BDNF for the
TrkB/P75NTRcomplex [40].
The survival-mediating role of Erk1/2
The survival function of neurotrophins has been mainly
attributed to Erk1/2 and PI3K pathways [41]. As the next
step, we examined whether these pathways play a role in
arbitrating the effect of the neurotrophins on Engrailed-
deficient mesDA neurons and whether they intersect with
the molecular events regulated by Engrailed expression.
Therefore, we studied activation of the two pathways,
using antibodies against the phosphorylated forms of
Erk1/2 and AKT [41]. Immunohistochemistry showed
that while total Erk1/2 protein was present in both En2-/-
and EnDM mesDA neurons, it was activated only in the
wild-type-like cells and not in EnDM mesDA neurons (Fig-
ure 3A–D). Western blot analysis of ventral midbrain tis-
sue showed a similar effect; Erk1/2 activity in E12 EnDM
ventral midbrain was reduced by 66.8 ± 18.2% (p = 0.02,
Loss of Engrailed induces neurotrophin requirement in mesDA neuronsFigure 2
Loss of Engrailed induces neurotrophin requirement in mesDA neurons. (A-F) Double immunohistochemistry on
dissociated cells derived from En2-/- (A-C) and En1-/-;En2-/- (EnDM) (D-F) E12 ventral midbrain using antibodies against tyrosine
kinase (Trk)B (A, D), TrkC (B, E), P75NTR(C, F) and TH (green) counterstained with DAPI. TrkB, TrkC and P75NTR are
expressed by TH+ cells from both genotypes; however, the immunohistochemistry is not sensitive enough to detect differ-
ences in P75NTR expression between genotypes. (G, H) Western blot of ventral midbrain tissue derived from different
Engrailed genotypes. The two Trk receptors do not depend on Engrailed expression (G). Brain-derived neurotrophic factor
(BDNF), neurotrophin (NT)4 and NT3 are not expressed in E12 ventral midbrain tissue, but they are in the adult (H). (I)
Treatments (>10 ng/ml) for 72 hours with TrkB/C-specific neurotrophins – BDNF, NT4 and NT3 – greatly increases the sur-
vival rate of EnDM mesDA neurons (n 6; p < 0.001), whereas nerve growth factor (NGF), glial cell line-derived neurotrophic
factor (GDNF), transforming growth factor (TGF)-β and growth differentiation factor (GDF)-15 do not significantly alter sur-
vival rate. (J) Dose response curve: BDNF concentration plotted against survival rate showing saturation at approximately the
10 ng/ml. Scale bars: 25 μm. Error bars indicate standard error. Ctl, control.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 6 of 13
(page number not for citation purposes)
Differential activation of Erk1/2 in mesDA neuronsFigure 3
Differential activation of Erk1/2 in mesDA neurons. (A-D, G-K) Immunohistochemistry of E12 ventral midbrain cell
culture stained against TH (green), total Erk1/2 protein (red) (A, B) and phosphorylated Erk1/2 (red) (C-D, G-K). (A-D) While
Erk1/2 protein is present in mesDA neurons of both genotypes (A, B), it is only phosphorylated in En2-/- mesDA neurons (C)
and not in the En1-/-;En2-/- (EnDM) counterparts (D). (E-I) Erk1/2 becomes activated in EnDM mesDA neurons after treatment
with the survival-inducing neurotrophins, brain-derived neurotrophic factor (BDNF), neurotrophin (NT)4 and NT3, or after
silencing of P75NTR (RNA interference (RNAi)) (G-J), but not when glial cell line-derived neurotrophic factor (GDNF) is applied
(I). (E) Western blot of E12 ventral midbrain tissue confirms the immunohistochemical finding of differential phosphorylation
between genotypes and shows that neither AKT, part of the phosphotidyl inositol-3 kinase pathway, nor other mitogen-acti-
vated protein kinases, such as JNK and P38, are differentially activated. (F) Quantification of phosphorylated Erk1/2 in western
blot normalized against En2-/- tissue. (L) Number of TH-positive cells in EnDM and En2-/- ventral midbrain cultures after 72
hours, treated with the 400 nM Mek inhibitor U0126 in conjunction with BDNF, Penetratin-coupled P75NTR double-stranded
RNA oligonucleotides and the P75NTR inhibiting antibody (Rex). Numbers are normalized against untreated cultures at 24
hours. The rescue effect is significantly reduced when the EnDM cultures are treated with the Erk1/2 inhibitor. Scale bars: 25
μm. Error bars indicate standard error. Ctl, control.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 7 of 13
(page number not for citation purposes)
n = 3) in comparison to the same tissue derived from their
En2-/- littermates (Figure 3E,F). In contrast to Erk1/2, we
did not detect differential activation of AKT in the western
blot (Figure 3E). Additionally, we could not detect signs of
differential activation of other components of the
mitogen-activated protein kinase (MAPK) pathways, such
as JNK or P38 (Figure 3E).
The loss of Erk1/2 phosphorylation suggested that this
MAPK pathway was differentially activated in EnDM
mesDA neurons in response to a survival or death signal.
To investigate this hypothesis, we examined the state of
activation of Erk1/2 after application of the survival fac-
tors. The addition of BDNF, NT4 and NT3, and the knock-
down of P75NTR by siRNA oligonucleotides all caused
phosphorylation of Erk1/2 (Figure 3G–J); however,
MAPK was not activated after application of NGF, GDNF,
TGF-β or GDF-15 (for example, see Figure 3K). If the loss
of Erk1/2 activity is the primary cause of cell death, when
P75NTR is elevated in mesDA neurons, and this correlation
is not accidental, then inhibition of the Erk1/2 pathway
should interfere with the rescue by the neurotrophins and
by P75NTR inhibition or silencing. To test this, we concur-
rently treated the cultures with U0126 [42], an inhibitor
of the MAPK kinase upstream of Erk1/2, MEK1/2 [32], at
a concentration (400 nM) not toxic to the wild-type (En2-
/-) neurons. The inhibition of Erk1/2 by U0126 signifi-
cantly reduced the rescue effect of all three survival factors;
from 42.2 ± 3.1% to 11.7 ± 3.5% (p = 0.002) for BDNF,
from 34.7 ± 4.7% to 8.3 ± 2.7% (p < 0.0001) for Rex and
from 25.4 ± 2.7% to 10.8 ± 1.9% (p = 0.009) after P75NTR
silencing (control mutant 4.2 ± 2.2%, p = 0.01, n = 4 for
all experiments) (Figure 3L).
To elaborate further on the correlation between the
expression of P75NTR and the state of phosphorylation of
Erk1/2, we silenced the P75NTR expression in EnHT ventral
midbrain cultures by RNAi, using two methods with dif-
ferent transfection efficiencies (the lipophilic transfection
reagent HiPerfect, and Penetratin-coupled oligos). The
former reduced P75NTR expression levels, on average, by
63.3 ± 2.0% (p = 0.011, n = 3) and the latter by 82.2 ±
6.3% (p = 0.05, n = 3). This, in turn, caused 6.09 ± 1.40-
fold (p = 0.01, n = 3) and 8.64 ± 1.45-fold (p = 0.008, n =
3) increases, respectively, in the phosphorylation of Erk1/
2 (Figure 4A–D).
Sensitivity to mitochondrial dysfunction and Engrailed
expression level
P75NTR signaling often mediates cell death via induction
of the mitochondrial (intrinsic) pathway of apoptosis
[43,44]. To further assess whether the elevated level of
P75NTR expression in the Engrailed-deficient mesDA neu-
rons is causal for demise of the cells, we investigated the
dying neurons for signs of this pathway. We had previ-
ously reported that loss of Engrailed expression in mesDA
neurons causes activation of caspase-3 [11,14], an effector
caspase, triggered by the intrinsic or extrinsic pathways of
apoptosis [45,46]. The intrinsic death pathway involves
release of cytochrome C from the mitochondria, which
participates in formation of apoptosome, which is
required for activation of caspase-9. We detected small,
rounded TH-positive cells with active caspase-9, and
pyknotic, fragmented nuclei in the ventral midbrain of
Engrailed-deficient E13 embryos as well as in EnHT mice
during the postnatal stages of nigral cell loss (postnatal
day 20; Figure 5A–C) confirming our hypothesis that the
pathway downstream of P75NTR signaling must be the
cause of the demise of these neurons in the absence of the
Engrailed genes [43].
Erk1/2 activation is inversely correlated with P75NTR expres-sionFigure 4
Erk1/2 activation is inversely correlated with P75NTR
expression. (A, B) Immunohistochemistry on E12 dissoci-
ated ventral midbrain cultures derived from EnHT E12
embryos stained against TH (green) and P75NTR. P75NTR
expression is absent in cultures treated with Penetratin-cou-
pled double-stranded RNA oligos. (C) Western blot of ven-
tral midbrain cultures using two methods of RNA
transfection with different efficiencies: HiPerfect (HiP) and
Penetratin-coupled RNA oligos show increase in phosphor-
ylated Erk1/2 after silencing of P75NTR, but no changes in
phosphorylation of P38. (D) Quantification of P75NTR
expression and Erk1/2 phosphorylation after transfection
with P75NTR double-stranded RNA oligonucleotides, normal-
ized against untreated EnHT cultures, showing the inverse cor-
relation between the two parameters. Scale bars: 25 μm.
Error bars indicate standard error.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 8 of 13
(page number not for citation purposes)
P75NTR expression in tumor cells induces a dose-depend-
ent increase of the pro-apoptotic members (Bad, Bax and
Bak) and a decrease of the anti-apoptotic members (Bcl-2
and Bcl-XL) of the Bcl-2 family [47]. Since we were unable
to directly measure the relative amounts of these proteins
in mesDA neurons themselves, we followed up on this
possibility by application of HA14-1 and chelerythrine
chloride, inhibitors of Bcl-2 and Bcl-XL, respectively.
HA14-1 was discovered as an inhibitor of Bcl-2 using a
computer screening strategy based on its predicted struc-
ture [24]. Chelerythrine chloride was identified by a high-
throughput screening of natural compounds [48]. Both
are highly specific for their targets. If the levels of the two
anti-apoptotic members of the Bcl-2 family of proteins are
lowered, as suggested by these previous tumor cell-exper-
iments [47], the sensitivity to HA14-1 and chelerythrine
chloride should be higher when the level of Engrailed pro-
tein decreases. Twenty-four hours after treatment with
HA14-1 and chelerythrine chloride, the rate of survival of
mesDA neurons, derived from E12 En2-/- embryos, was,
on average, 75.3 ± 6.8% and 92.7 ± 2.7% (p < 0.001, n =
7 for both compounds) higher, respectively, than their
counterparts derived from EnHT littermates (Figure 6A,B),
suggesting that reduced Engrailed and elevated P75NTR
expression lowers the threshold at which the intrinsic
pathway of apoptosis is triggered.
These results could indicate that the reduction in Engrailed
expression increases the vulnerability of mesDA neurons
to mitochondrial dysfunction in a general manner, since
the three most commonly used reagents to model PD –
MPP+, 6-Hydroxydopamine (6-OHDA) and rotenone
[17,29,49,50] – also cause cell death by induction of this
pathway of apoptosis, evident from activation of caspase-
9 [51-53] (Figure 7A–C). To test this hypothesis directly,
we treated EnHT mesDA neurons with MPP+, the metabo-
lite of MPTP, an inhibitor of complex-I of the mitochon-
drial electron transport chain [49]. After 48 hours in
culture, the rate of survival of En2-/- mesDA neurons was,
on average, 124.5 ± 5.0% (p < 0.001, n = 12) higher than
for their En1 heterozygote (EnHT) littermates, suggesting
that MPTP and Engrailed may act upon the same molecu-
lar pathway upstream of caspase-9 (Figure 6A,B).
To determine whether this sensitivity to cell death is spe-
cifically related to instability of the mitochondria, we also
employed Apoptosis Activator-2 [19], Prima-1 [27], and
TNFα [31]. The first induces cell death by triggering apop-
tosome formation, the second by activation of p53 and
the third, as a ligand of the TNF receptors, induces the
extrinsic, receptor-mediated pathway of apoptosis. In all
three cases, the level of En1 expression did not have a sig-
nificant influence on the survival rates of these neurons (-
8.1 ± 6.0% (p = 0.67) for Apoptosis Activator-2, 4.6 ±
Mitochondrial apoptosis in Engrailed-deficient mesDA neuronsFigure 5
Mitochondrial apoptosis in Engrailed-deficient mesDA neurons. (A-C) TH and activated caspase-9 immunohistochem-
istry on ventral midbrain coronal sections of E13 En1-/-;En2-/- (EnDM) embryos (A), of En1+/-;En2-/- (EnHT) postnatal day 20
brain (C), and of EnDM E12 cell culture (B) counterstained with the nuclear marker DAPI. Dying TH-positive neurons (arrows)
exhibit small rounded cell bodies and signs of apoptosis, that is, activated caspase-9 and pyknotic nuclei (DAPI). (D) EnDM ven-
tral midbrain cultures treated for 72 hours with inhibitors for caspases-3, -8 and -9, a pan-caspase inhibitor, z-vad-fmk, the
BAX inhibitor Ku70, the P53 inhibitor Pfithrin-α, and the JNK inhibitor SP600125. None of the treatments significantly changed
the survival rate of EnDM mesDA neurons. The number of surviving TH-positive cells was normalized in each case against
untreated cultures 24 hours after dissociation. Scale bars: 25 μm. Error bars indicate standard error.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 9 of 13
(page number not for citation purposes)
6.2% (p = 0.82) for Prima-1, and 5.1 ± 3.1% (p = 0.48) for
TNFα; n = 7 for all three experiments; Figure 6A,B).
Gain of function in a mesDA cell line
To confirm the Engrailed dose-dependent sensitivity of
mesDA neurons to mitochondrial insult, we performed a
gain-of-function experiment using the inducible Tet-On
system to express En1 in the dopaminergic cell line MN9D
[54]. As in the primary cultures, En1 was protective against
cell death if induced by administration of MPP+ (42.4 ±
0.7%, p < 0.0001, n = 23), chelerythrine chloride (46.7 ±
1.3%, p < 0.0001, n = 10) and HA14-1 (32.4 ± 1.3%, p <
0.0001, n = 10). Accordingly, survival rate was not signif-
icantly altered if the other three reagents were employed (-
10.7 ± 4.7% (p = 0.26) for Apoptosis Activator-2, -8.3 ±
5.2% (p = 0.50) for Prima-1, and 1.0 ± 7.4% (p = 0.95) for
TNFα; n = 12 for all experiments; Figure 6C,D).
Functional inhibition of Bcl-2 and Bcl-XL and application
of MPP+ induces apoptosis by release of cytochrome C
from the mitochondrial inter-membrane space into the
cytosol [55,56]. The protective effect of En1 against HA14-
1, chelerythrine chloride and MPP+ may be attributable to
higher mitochondrial stability. To test this hypothesis, we
compared the cytosolic and mitochondrial protein frac-
tions of En1-expressing MN9D cells to non-expressing
cells. The proportion of cytochrome C in the mitochon-
dria was always significantly higher after induction of En1,
either in the presence of MPP+ or in untreated control cul-
tures (76.1 ± 7.3%, p = 0.03, n = 3, and 41.6 ± 7.0%, p =
0.005, n = 3, respectively), whereas the total amount of
cellular cytochrome C was unaltered (Figure 6F,H). In
contrast, the proportional levels in mitochondria and
cytosol of the apoptosis inducing factor, which causes cas-
pase-independent mitochondrial apoptosis [57], were
independent of the level of En1 expression, suggesting
that Engrailed participates in the regulation of mitochon-
drial stability via the cytochrome C/caspase-dependent
pathway of apoptosis rather than the caspase-independ-
ent pathway, represented by apoptosis inducing factor.
The sensitivity to induction of the intrinsic pathway of apoptosis correlates with En1 expressionFigure 6
The sensitivity to induction of the intrinsic pathway of apoptosis correlates with En1 expression. (A-D) Ventral
midbrain cultures 24 hours after application of apoptosis-inducing compounds. Charts of EnHT (En1+/-;En2-/-) and En2-/- E12
cultures depicting the number of surviving TH-positive cells (A, B) and cultures of En1-inducible MN9D cells depicting cell sur-
vival measured by cell proliferation assay (C, D). Surviving cells were normalized against untreated EnHT cultures (A) or
untreated non-induced MN9D cells (C), treated EnHT cultures (B) or treated non-induced MN9D cells (D). Higher Engrailed
expression reduced the cell death rate after MPP+, HA14-1 and chelerythrine chloride (CC) treatment, whereas the rate of cell
survival after application of the tumor necrosis factor alpha (TNFa), Prima-1 and Apoptosis Activator-2 (AA2) does not corre-
late with the level of En1 expression. Dox, doxycycline. (E) Western blot analysis of mitochondrial and cytoplasmic protein
fractions of MN9D cells 72 hours after En1 induction and 24 hours after MPP+ treatment. (F) Proportion of cytochrome C
(Cyt-C) in cytosol is lower in En1-expressing MN9D cells before and after MPP+ treatment. Scale bars: 25 μm. Error bars indi-
cate standard error. 6 n 27, *p 0.001, **p 0.0001. Cyt, cytosolic; Mit, mitochondrial; ANT, adenine nucleotide trans-
porter; Dox, doxycycline.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 10 of 13
(page number not for citation purposes)
Discussion
In this study, we provide evidence that cell death in
Engrailed-deficient mesDA neurons is a result of higher
P75NTR expression and the loss of Erk1/2 activity. Further-
more, we show that the dose of Engrailed is part of the
molecular mechanism that determines the sensitivity of
these neurons to mitochondrial insult.
P75NTR can cause apoptosis in various neuronal popula-
tions by mere high expression [10] or as a mediator of a
(pro-)neurotrophin death signal [8]. Alternatively, since
TrkB and C are expressed by the mesDA neurons, the
abnormal increase in the expression of P75NTR could
introduce a neurotrophin dependency, which does not
occur in the wild type at this age. The latter is more likely
in mesDA neurons deprived of the Engrailed genes, since
the death signal could be counteracted by addition of neu-
rotrophins (BDNF, NT3, NT4) [58]. Furthermore, the Kd
calculated from the dose response of BDNF, corresponds
to the known affinity of neurotrophins for P75NTR and Trk
receptors [40], demonstrating that the survival effect of
the neurotrophins is attributable to direct binding to the
receptors on the surface of mesDA neurons, as opposed to
a survival signal that originates from the surrounding
cells, which would be reflected in a different shape of the
dose response curve. A direct interaction of the neuro-
trophins with receptors on mesDA neurons is also consist-
ent with our previous findings that the Engrailed genes are
cell-autonomously required for the survival of these cells
[11]. The ineffectiveness of Ngf can be readily explained
by the lack of expression of TrkA. It is noteworthy that nei-
ther neurotrophins nor inhibition of P75NTR completely
(that is, 100%) rescue mesDA neurons deprived of
Engrailed genes, and it is thus possible that P75NTR is only
one of many factors contributing to the death of mesDA
neurons in the absence of the Engrailed genes. However,
since the in vitro timeframe of loss of EnDM mesDA neu-
rons is 72 hours, it is also possible that those cells, which
died anyway, had already been committed to cell death
before the knock-down of P75NTR by the RNA duplexes
was sufficiently high or the neurotrophin effect set in.
Neurotrophin-induced Trk/P75NTR interaction, or lack
thereof, can provoke intracellular activation of the MAPK
and PI3K pathways. Among MAPKs, JNK and p38 partici-
pate in stress responses and often trigger apoptosis, while
Erk1/2 signaling regulates cell proliferation, differentia-
tion and survival [59]. Alternatively, the pro-survival role
of neurotrophins can be mediated by PI3K signaling. This
is in contrast to our findings. The activity of JNK, p38 or
PI3K is independent of the Engrailed genes and the level of
P75NTR expression in mesDA neurons. The lack of Erk1/2
activity in EnDM mesDA neurons, which is reversed under
any of the rescue conditions, demonstrates that the level
of Erk1/2 phosphorylation in mesDA neurons is corre-
lated with Engrailed expression and inversely to the level
of P75NTR, suggesting that the P75NTR death signal is medi-
ated in these neurons by suppression of Erk1/2 activity.
Although the association of P75NTR and the Erk proteins
has been shown in PC12 cells [60], regulation of the activ-
ity of the Erk1/2 signaling pathway as a result of death sig-
naling by P75NTR is a novel mechanism, which signifies
both the role of sustained activity of Erk1/2 in the survival
and/or of P75NTR in the demise of mesDA neurons. Our
data also suggest that the survival effect of neurotrophins
may be the result of dis-inhibition of a death signal (high
expression of P75NTR) that is triggered in the absence of
the proper transcriptional regulation (by the Engrailed
genes) during the course of development or possibly
throughout life.
The pro-apoptotic role of P75NTR has been established in
the animal models of neurodegenerative disorders, other
than PD, including beta-amyloid peptide-dependent cell
death [61], or stress conditions, including ischemia [62].
Although function of the ligands and the co-receptors of
P75NTR, TrkA/B/C, have been investigated in mesDA neu-
rons [63-65], the role of P75NTR, itself, has not been
reported in this system. Our work provides the first evi-
dence for the pivotal role of P75NTR in mesDA neurons, as
a negative mediating factor for the lifelong survival func-
tion of the Engrailed genes in this neuronal population.
The sensitivity to neurotoxin-induced cell death by MPTP
and to inducers of mitochondrial instability, such as the
Bcl-2 and Bcl-XL inhibitors, is inversely correlated with the
dose of Engrailed expression in mesDA neurons. Mito-
chondrial dysfunction was first implicated in the patho-
Mitochondrial apoptosis after complex I inhibitionFigure 7
Mitochondrial apoptosis after complex I inhibition.
(A-C) Immunohistochemistry of E12 ventral midbrain cell
cultures. Caspase-9 is activated in wild-type mesDA neurons
(arrows), treated with MPP+, rotenone, or 6-OHDA.
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 11 of 13
(page number not for citation purposes)
genesis of PD after accidental administration of MPTP by
drug abusers and a consequent Parkinsonian syndrome.
Lowered complex I activity and reduced mitochondrial
stability [3] are believed to be key factors in the etiology
of PD. In postmortem brains of PD patients, reduced
activity of complex-I (NADH/ubiquinone oxidoreduct-
ase) of the mitochondrial electron transport chain [49]
and elevated levels of the pro-apoptotic members of the
Bcl-2 family have been observed in the substantia nigra
[66]. Then, three genes, DJ1, PINK1 and OMI/HTRA2,
mutations of which have been associated with familiar
forms of PD, were discovered to have a function in the
mitochondria [5]. Furthermore, the toxicity of MPTP is
conferred by inhibition of complex I of the electron trans-
port chain, triggering the release of cytochrome-C from
the mitochondrial intermembrane space into the cytosol,
which involves Bcl-2 family members [7]. Here, we dem-
onstrate that the sensitivity to neurotoxin-induced cell
death and to inducers of mitochondrial apoptosis, such as
the Bcl-2 and Bcl-XL inhibitors, is inversely correlated to
the dose of Engrailed expression in mesDA neurons. This,
the previous findings of other groups, and the PD-like
slow progressive loss of nigral dopaminergic neurons in
EnHT mice [14] suggest that the mode of action of the
Engrailed genes converges with MPTP toxicity and possibly
also with the disease mechanism on the mitochondria.
Intriguingly, a recent association study into PD indicated
a single nucleotide polymorphism in the intron of En1 as
a potential risk factor for sporadic forms of this disease
[67].
Abbreviations
BDNF: brain-derived neurotrophic factor; E: embryonic
day; En: Engrailed; EnDM: Engrailed double mutant mice
(En1-/-;En2-/-); EnHT: heterozygous null for En1 and
homozygous null for En2 (En1+/-;En2-/-); Erk: extracellu-
lar-signal-regulated kinase; GDF: growth differentiation
factor; GDNF: glial cell line-derived neurotrophic factor;
JNK: c-Jun N-terminal kinase; MAPK: mitogen-activated
protein kinase; mesDA: mesencephalic dopaminergic;
MPTP: 1-methyl-4-phenyl-,1,3,6-tetrahydropyridine;
NGF: nerve growth factor; NT: neurotrophin; PD: Parkin-
son's disease; PI3K: phosphotidyl inositol-3 kinase; RNAi:
RNA interference; siRNA: small interfering RNA; TGF:
transforming growth factor; Trk: tropomyosin-receptor-
kinase.
Competing interests
The authors declare that they have no competing interests.
Authors' contributions
KNA and HHS conceived of, designed and discussed the
studies, carried out the experiments, analyzed and inter-
preted the data and wrote the draft and the final version
of the manuscript. PS and LA made the stable MN9D cell
lines. SS helped with immunoassays. The manuscript was
approved by all authors.
Additional material
Acknowledgements
This work was supported by grants from the Federal Secretary for Educa-
tion and Research BMBF Biofuture 98, the German Research Council SI
752/3-1 and the Michael J Fox Foundation. We thank Martyn Goulding for
the En1/tauLacZ, Alex Joyner for the En2 mutant mice, Louis Reichardt for
the Rex antibody, and Jutta Fey for technical assistance.
References
1. Schultz W: Getting formal with dopamine and reward. Neuron
2002, 36:241-263.
2. Girault JA, Greengard P: The neurobiology of dopamine signal-
ing. Arch Neurol 2004, 61:641-644.
3. Abou-Sleiman PM, Muqit MM, Wood NW: Expanding insights of
mitochondrial dysfunction in Parkinson's disease. Nat Rev
Neurosci 2006, 7:207-219.
4. Tatton WG, Chalmers-Redman R, Brown D, Tatton N: Apoptosis
in Parkinson's disease: signals for neuronal degradation. Ann
Neurol 2003, 53(Suppl 3):S61-70. discussion S70-62.
5. Farrer MJ: Genetics of Parkinson disease: paradigm shifts and
future prospects. Nat Rev Genet 2006, 7:306-318.
6. Schober A: Classic toxin-induced animal models of Parkin-
son's disease: 6-OHDA and MPTP. Cell Tissue Res 2004,
318:215-224.
7. Peri er C, T ieu K, Guegan C, Casp ersen C, Jac kson-Lewis V, Carelli V,
Martinuzzi A, Hirano M, Przedborski S, Vila M: Complex I defi-
ciency primes Bax-dependent neuronal apoptosis through
mitochondrial oxidative damage. Proc Natl Acad Sci USA 2005,
102:19126-19131.
8. Lu B, Pang PT, Woo NH: The yin and yang of neurotrophin
action. Nat Rev Neurosci 2005, 6:603-614.
9. Krieglstein K, Strelau J, Schober A, Sullivan A, Unsicker K: TGF-beta
and the regulation of neuron survival and death. J Physiol Paris
2002, 96:25-30.
10. Bhakar AL, Howell JL, Paul CE, Salehi AH, Becker EB, Said F, Bonni A,
Barker PA: Apoptosis induced by p75NTR overexpression
requires Jun kinase-dependent phosphorylation of Bad. J Neu-
rosci 2003, 23:11373-11381.
11. Alberi L, Sgado P, Simon HH: Engrailed genes are cell-autono-
mously required to prevent apoptosis in mesencephalic
dopaminergic neurons. Development 2004, 131:3229-3236.
12. Simon HH, Saueressig H, Wurst W, Goulding MD, O'Leary DD: Fate
of midbrain dopaminergic neurons controlled by the
Engrailed genes. J Neurosci 2001, 21:3126-3134.
13. Simon HH, Bhatt L, Gherbassi D, Sgado P, Alberi L: Midbrain
dopaminergic neurons: determination of their developmen-
tal fate by transcription factors. Ann NY Acad Sci 2003,
991:36-47.
14. Sgado P, Alberi L, Gherbassi D, Galasso SL, Ramakers GM, Alavian
KN, Smidt MP, Dyck RH, Simon HH: Slow progressive degenera-
tion of nigral dopaminergic neurons in postnatal Engrailed
mutant mice. Proc Natl Acad Sci USA 2006, 103:15242-15247.
15. Joyner AL, Herrup K, Auerbach BA, Davis CA, Rossant J: Subtle cer-
ebellar phenotype in mice homozygous for a targeted dele-
tion of the En-2 homeobox. Science 1991, 251:1239-1243.
Additional file 1
List of compounds and concentrations. Compounds employed for the cell
culture experiments: Final concentrations, solvents, duration of applica-
tion, activity, source and literature.
Click here for file
[http://www.biomedcentral.com/content/supplementary/1749-
8104-4-11-S1.pdf]
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 12 of 13
(page number not for citation purposes)
16. Saueressig H, Burrill J, Goulding M: Engrailed-1 and netrin-1 reg-
ulate axon pathfinding by association interneurons that
project to motor neurons. Development 1999, 126:4201-4212.
17. Fink DW, Horn PT, Mirkin BL: Differential effects of 6-hydroxy-
dopamine-induced ablation of peripheral and central adren-
ergic axons on C-1300 murine neuroblastoma tumor growth
and catecholamine concentration in the A/J mouse. J Pharma-
col Exp Ther 1992, 262:1070-1075.
18. Mocanu MM, Baxter GF, Yellon DM: Caspase inhibition and limi-
tation of myocardial infarct size: protection against lethal
reperfusion injury. Br J Pharmacol 2000, 130:197-200.
19. Nguyen JT, Wells JA: Direct activation of the apoptosis machin-
ery as a mechanism to target cancer cells. Proc Natl Acad Sci
USA 2003, 100:7533-7538.
20. Sawada M, Sun W, Hayes P, Leskov K, Boothman DA, Matsuyama S:
Ku70 suppresses the apoptotic translocation of Bax to mito-
chondria. Nat Cell Biol 2003, 5:320-329.
21. Thornberry NA, Lazebnik Y: Caspases: enemies within. Science
1998, 281:1312-1316.
22. Fadeel B, Hassan Z, Hellstrom-Lindberg E, Henter JI, Orrenius S,
Zhivotovsky B: Cleavage of Bcl-2 is an early event in chemo-
therapy-induced apoptosis of human myeloid leukemia cells.
Leukemia 1999, 13:719-728.
23. Tanaka K, Honda M, Takabatake T: Anti-apoptotic effect of ator-
vastatin, a 3-hydroxy-3-methylglutaryl coenzyme a reduct-
ase inhibitor, on cardiac myocytes through protein kinase C
activation. Clin Exp Pharmacol Physiol 2004, 31:360-364.
24. Wang JL, Liu D, Zhang ZJ, Shan S, Han X, Srinivasula SM, Croce CM,
Alnemri ES, Huang Z: Structure-based discovery of an organic
compound that binds Bcl-2 protein and induces apoptosis of
tumor cells. Proc Natl Acad Sci USA 2000, 97:7124-7129.
25. Langston JW, Irwin I, Langston EB, Forno LS: 1-Methyl-4-phenylpy-
ridinium ion (MPP+): identification of a metabolite of MPTP,
a toxin selective to the substantia nigra. Neurosci Lett 1984,
48:87-92.
26. Hoagland MS, Hoagland EM, Swanson HI: The p53 inhibitor pifit-
hrin-alpha is a potent agonist of the aryl hydrocarbon recep-
tor. J Pharmacol Exp Ther 2005, 314:603-610.
27. Bykov VJ, Issaeva N, Shilov A, Hultcrantz M, Pugacheva E, Chumakov
P, Bergman J, Wiman KG, Selivanova G: Restoration of the tumor
suppressor function to mutant p53 by a low-molecular-
weight compound. Nat Med 2002, 8:282-288.
28. Kruttgen A, Moller JC, Heymach JV Jr, Shooter EM: Neurotrophins
induce release of neurotrophins by the regulated secretory
pathway. Proc Natl Acad Sci USA 1998, 95:9614-9619.
29. Testa CM, Sherer TB, Greenamyre JT: Rotenone induces oxida-
tive stress and dopaminergic neuron damage in organotypic
substantia nigra cultures. Brain Res Mol Brain Res 2005,
134:109-118.
30. Wang W, Shi L, Xie Y, Ma C, Li W, Su X, Huang S, Chen R, Zhu Z,
Mao Z, Han Y, Li M: SP60 a new JNK inhibitor, protects
dopaminergic neurons in the MPTP model of Parkinson's
disease. Neurosci Res 0125, 48:195-202.
31. Adams JM: Ways of dying/multiple pathways to apoptosis.
Genes Dev 2003, 17:2481-2495.
32. Duncia JV, Santella JB 3rd, Higley CA, Pitts WJ, Wityak J, Frietze WE,
Rankin FW, Sun JH, Earl RA, Tabaka AC, Teleha CA, Blom KF, Favata
MF, Manos EJ, Daulerio AJ, Stradley DA, Horiuchi K, Copeland RA,
Scherle PA, Trzaskos JM, Magolda RL, Trainor GL, Wexler RR, Hobbs
FW, Olson RE: MEK inhibitors: the chemistry and biological
activity of U its analogs, and cyclization products. Bioorg Med
Chem Lett 0126, 8:2839-2844.
33. Martin DA, Siegel RM, Zheng L, Lenardo MJ: Membrane oligomer-
ization and cleavage activates the caspase-8 (FLICE/
MACHalpha1) death signal. J Biol Chem 1998, 273:4345-4349.
34. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T:
Duplexes of 21-nucleotide RNAs mediate RNA interference
in cultured mammalian cells. Nature 2001, 411:494-498.
35. Thuret S, Bhatt L, O'Leary DD, Simon HH: Identification and
developmental analysis of genes expressed by dopaminergic
neurons of the substantia nigra pars compacta. Mol Cell Neu-
rosci 2004, 25:394-405.
36. Davidson TJ, Harel S, Arboleda VA, Prunell GF, Shelanski ML, Greene
LA, Troy CM: Highly efficient small interfering RNA delivery
to primary mammalian neurons induces MicroRNA-like
effects before mRNA degradation. J Neurosci 2004,
24:10040-10046.
37. Strelau J, Sullivan A, Bottner M, Lingor P, Falkenstein E, Suter-Craz-
zolara C, Galter D, Jaszai J, Krieglstein K, Unsicker K: Growth/Dif-
ferentiation factor-15/Macrophage inhibitory cytokine-1 is a
novel trophic factor for midbrain dopaminergic neurons in
vivo. J Neurosci 2000, 20:8597-8603.
38. Blum M: A null mutation in TGF-alpha leads to a reduction in
midbrain dopaminergic neurons in the substantia nigra. Nat
Neurosci 1998, 1:374-377.
39. Tomac A, Lindqvist E, Lin LF, Ogren SO, Young D, Hoffer BJ, Olson
L: Protection and repair of the nigrostriatal dopaminergic
system by GDNF in vivo. Nature 1995, 373:335-339.
40. Barker PA: p75NTR is positively promiscuous: novel partners
and new insights. Neuron 2004, 42:529-533.
41. Hetman M, Xia Z: Signaling pathways mediating anti-apoptotic
action of neurotrophins. Acta Neurobiol Exp (Wars) 2000,
60:531-545.
42. Davies SP, Reddy H, Caivano M, Cohen P: Specificity and mecha-
nism of action of some commonly used protein kinase inhib-
itors. Biochem J 2000, 351:95-105.
43. Troy CM, Friedman JE, Friedman WJ: Mechanisms of p75-medi-
ated death of hippocampal neurons. Role of caspases. J Biol
Chem 2002, 277:34295-34302.
44. Wang X, Bauer JH, Li Y, Shao Z, Zetoune FS, Cattaneo E, Vincenz C:
Characterization of a p75(NTR) apoptotic signaling pathway
using a novel cellular model. J Biol Chem 2001, 276:33812-33820.
45. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES,
Wang X: Cytochrome c and dATP-dependent formation of
Apaf-1/caspase-9 complex initiates an apoptotic protease
cascade. Cell 1997, 91:479-489.
46. Stennicke HR, Jürgensmeier JM, Shin H, Deveraux Q, Wolf BB, Yang
X, Zhou Q, Ellerby HM, Ellerby LM, Bredesen D, Green DR, Reed JC,
Froelich CJ, Salvesen GS: Pro-caspase-3 is a major physiologic
target of caspase-8. J Biol Chem 1998, 273:27084-27090.
47. Tabassum A, Khwaja F, Djakiew D: The p75(NTR) tumor sup-
pressor induces caspase-mediated apoptosis in bladder
tumor cells. Int J Cancer 2003, 105:47-52.
48. Chan SL, Lee MC, Tan KO, Yang LK, Lee AS, Flotow H, Fu NY, Butler
MS, Soejarto DD, Buss AD, Yu VC: Identification of cheleryth-
rine as an inhibitor of BclXL function. J Biol Chem 2003,
278:20453-20456.
49. Dauer W, Przedborski S: Parkinson's disease/mechanisms and
models. Neuron 2003, 39:889-909.
50. Tanner CM, Ottman R, Goldman SM, Ellenberg J, Chan P, Mayeux R,
Langston JW: Parkinson disease in twins/an etiologic study.
JAMA 1999, 281:341-346.
51. Kaul S, Kanthasamy A, Kitazawa M, Anantharam V, Kanthasamy AG:
Caspase-3 dependent proteolytic activation of protein
kinase C delta mediates and regulates 1-methyl-4-phenylpy-
ridinium (MPP+)-induced apoptotic cell death in dopaminer-
gic cells: relevance to oxidative stress in dopaminergic
degeneration. Eur J Neurosci 2003, 18:1387-1401.
52. Liang Q, Liou AK, Ding Y, Cao G, Xiao X, Perez RG, Chen J: 6-
Hydroxydopamine induces dopaminergic cell degeneration
via a caspase-9-mediated apoptotic pathway that is attenu-
ated by caspase-9dn expression. J Neurosci Res 2004,
77:747-761.
53. Watabe M, Nakaki T: Rotenone induces apoptosis via activation
of bad in human dopaminergic SH-SY5Y cells. J Pharmacol Exp
Ther 2004, 311:948-953.
54. Choi HK, Won L, Roback JD, Wainer BH, Heller A: Specific mod-
ulation of dopamine expression in neuronal hybrid cells by
primary cells from different brain regions. Proc Natl Acad Sci
USA 1992, 89:8943-8947.
55. Deshmukh M, Johnson EM Jr: Evidence of a novel event during
neuronal death: development of competence-to-die in
response to cytoplasmic cytochrome c. Neuron 1998,
21:695-705.
56. Cai J, Yang J, Jones DP: Mitochondrial control of apoptosis: the
role of cytochrome c. Biochim Biophys Acta 1998, 1366:139-149.
57. Lorenzo HK, Susin SA, Penninger J, Kroemer G: Apoptosis induc-
ing factor (AIF): a phylogenetically old, caspase-independent
effector of cell death. Cell Death Differ 1999, 6:516-524.
Publish with BioMed Central and every
scientist can read your work free of charge
"BioMed Central will be the most significant development for
disseminating the results of biomedical research in our lifetime."
Sir Paul Nurse, Cancer Research UK
Your research papers will be:
available free of charge to the entire biomedical community
peer reviewed and published immediately upon acceptance
cited in PubMed and archived on PubMed Central
yours — you keep the copyright
Submit your manuscript here:
http://www.biomedcentral.com/info/publishing_adv.asp
BioMedcentral
Neural Development 2009, 4:11 http://www.neuraldevelopment.com/content/4/1/11
Page 13 of 13
(page number not for citation purposes)
58. Davey F, Davies AM: TrkB signalling inhibits p75-mediated
apoptosis induced by nerve growth factor in embryonic pro-
prioceptive neurons. Curr Biol 1998, 8:915-918.
59. Xia Z, Dickens M, Raingeaud J, Davis RJ, Greenberg ME: Opposing
effects of ERK and JNK-p38 MAP kinases on apoptosis. Sci-
ence 1995, 270:1326-1331.
60. Volente C, Angelastro JM, Greene LA: Association of protein
kinases ERK1 and ERK2 with p75 nerve growth factor recep-
tors. J Biol Chem 1993, 268:21410-21415.
61. Costantini C, Rossi F, Formaggio E, Bernardoni R, Cecconi D, Della-
Bianca V: Characterization of the signaling pathway down-
stream p75 neurotrophin receptor involved in beta-amyloid
peptide-dependent cell death. J Mol Neurosci 2005, 25:141-156.
62. Kokaia Z, Andsberg G, Martinez-Serrano A, Lindvall O: Focal cere-
bral ischemia in rats induces expression of P75 neurotrophin
receptor in resistant striatal cholinergic neurons. Neuro-
science 1998, 84:1113-1125.
63. von Bohlen und Halbach O, Minichiello L, Unsicker K: Haploin-suf-
ficiency for trkB and trkC receptors induces cell loss and
accumulation of alpha-synuclein in the substantia nigra.
Faseb J 2005, 19:1740-1742.
64. Porritt MJ, Batchelor PE, Howells DW: Inhibiting BDNF expres-
sion by antisense oligonucleotide infusion causes loss of
nigral dopaminergic neurons. Exp Neurol 2005, 192:226-234.
65. Pineda JR, Canals JM, Bosch M, Adell A, Mengod G, Artigas F, Ernfors
P, Alberch J: Brain-derived neurotrophic factor modulates
dopaminergic deficits in a transgenic mouse model of Hunt-
ington's disease. J Neurochem 2005, 93:1057-1068.
66. Tatton NA: Increased caspase 3 and Bax immunoreactivity
accompany nuclear GAPDH translocation and neuronal
apoptosis in Parkinson's disease. Exp Neurol 2000, 166:29-43.
67. Fuchs J, Mueller JC, Lichtner P, Schulte C, Munz M, Berg D, Wullner
U, Il lig T, S harma M , Gasser T: The transcription factor PITX3 is
associated with sporadic Parkinson's disease. Neurobiol Aging
2007.
... Notably, p75NTR can induce selective apoptosis of dopaminergic neurons and progression of PD [21]. In addition, p75NTR signaling inhibits the expression of transcription factors engrailed-1 and engrailed-2 which are necessary for the survival of dopaminergic neurons in the midbrain [66]. The reduction in expression of engraileds, possibly related to the higher levels of p75NTR, also decreases mitochondrial stability. ...
... The reduction in expression of engraileds, possibly related to the higher levels of p75NTR, also decreases mitochondrial stability. In particular, the dose of engrailed determines the sensitivity to cell death induced by the classic PD-model toxin MPTP and to inhibition of the anti-apoptotic members of the Bcl-2 family of proteins [66]. Furthermore, p75NTR expression is correlated with the severity of dopaminergic neuronal injury in 6-hydroxydopamine (6-OHDA)-induced striatal lesions in rats [22]. ...
Article
Full-text available
Parkinson’s disease (PD) is a chronic and progressive neurodegenerative disease of the brain. PD is characterized by motor and non-motor symptoms. The p75 neurotrophin receptor (p75NTR) is a functional receptor for different growth factors including pro-brain derived neurotrophic factor (pro-BDNF), neurotrophin 3 (NT-3), and neurotrophin 4 (NT-4). Consequently, this review aimed to illustrate the detrimental and beneficial role of p75NTR in PD. Diverse studies showed that p75NTR and its downstream signaling are intricate in the pathogenesis of PD. Nevertheless, pro-apoptotic and pro-survival pathways mediated by p75NTR in PD were not fully clarified. Of note, p75NTR plays a critical role in the regulation of dopaminergic neuronal survival and apoptosis in the CNS. Particularly, p75NTR can induce selective apoptosis of dopaminergic neurons and progression of PD. In addition, p75NTR signaling inhibits the expression of transcription factors which are essential for the survival of dopaminergic neurons. Also, p75NTR expression is connected with the severity of dopaminergic neuronal injury. These verdicts implicate p75NTR signaling in the pathogenesis of PD, though the underlying mechanistic pathways remain not elucidated. Collectively, the p75NTR signaling pathway induces a double-sword effect either detrimental or beneficial depending on the ligands and status of PD neuropathology. Therefore, p75NTR signaling seems to be protective via phosphoinositide 3-kinase (PI3K)/AKT and Bcl-2 and harmful via activation of JNK, caspase 3, nuclear factor kappa B (NF-κB), and RhoA pathways.
... Cdh13 can regulate neuronal migration and also has an effect on axonal outgrowth as demonstrated in the serotonergic system (Forero et al., 2017). The VTA-predominant gene Engrailed-2 (EN2) is a transcription factor known to promote survival of dopamine neurons by inducing survival gene expression and by protecting neurons from oxidative stress and blocking mitochondrial instability (Alavian et al., 2009;Alvarez-Fischer et al., 2011;Rekaik et al., 2015). The higher level of EN-2 in VTA compared to SNc neurons could in part explain the relative resilience of these neurons to PD. ...
... Regarding cell replacement therapies targeting PD (Alavian et al., 2009;Kriks et al., 2011;Ganat et al., 2012;Kefalopoulou et al., 2014;Kirkeby et al., 2017), there is still an urgent need to optimize the pluripotent stem cell preparations to specifically generate SNc rather than VTA neurons (Barker et al., 2017;Sonntag et al., 2018). Evaluation of the correct patterning and differentiation of pluripotent cells to midbrain dopamine neurons relies upon gene expression analysis using quantitative real time PCR (qPCR) or global transcriptome approaches such as RNA sequencing (Ganat et al., 2012;Barker et al., 2017;Nolbrant et al., 2017;Studer, 2017). ...
Article
Full-text available
Defining transcriptional profiles of substantia nigra pars compacta (SNc) and ventral tegmental area (VTA) dopamine neurons is critical to understanding their differential vulnerability in Parkinson’s Disease (PD). Here, we determine transcriptomes of human SNc and VTA dopamine neurons using LCM-seq on a large sample cohort. We apply a bootstrapping strategy as sample input to DESeq2 and identify 33 stably differentially expressed genes (DEGs) between these two subpopulations. We also compute a minimal sample size for identification of stable DEGs, which highlights why previous reported profiles from small sample sizes display extensive variability. Network analysis reveal gene interactions unique to each subpopulation and highlight differences in regulation of mitochondrial stability, apoptosis, neuronal survival, cytoskeleton regulation, extracellular matrix modulation as well as synapse integrity, which could explain the relative resilience of VTA dopamine neurons. Analysis of PD tissues showed that while identified stable DEGs can distinguish the subpopulations also in disease, the SNc markers SLIT1 and ATP2A3 were down-regulated and thus appears to be biomarkers of disease. In summary, our study identifies human SNc and VTA marker profiles, which will be instrumental for studies aiming to modulate dopamine neuron resilience and to validate cell identity of stem cell-derived dopamine neurons.
... In the present study, we evaluated the hypothesis that oxidative stress induces p75 NTR signaling in neuronal cells of the ventral mesencephalon. Previous reports have indicated that p75 NTR is expressed in dopaminergic neurons contained within this brain region (Wang et al. 2008;Wang et al. 2010;Alavian et al. 2009), yet how the receptor signals in mesencephalic cells and the impact of cellular injury on such signaling mechanisms are poorly understood. Here, we demonstrate that oxidative damage in mesencephalic cells stimulates regulated intramembrane proteolysis of p75 NTR . ...
... We have previously demonstrated that oxidative stress stimulates p75 NTR processing and thereby induces apoptosis of sympathetic neurons (Kraemer et al. 2014a), a cell population that, like dopaminergic neurons, are catecholinergic and susceptible to Lewy pathology associated with PD (Del Tredici et al. 2010;Orimo et al. 2008;Bloch et al. 2006). Moreover, expression of p75 NTR has been detected in injured dopaminergic neurons of the substantia nigra (Wang et al. 2008), and the receptor has been reported to contribute to neuronal death in cultures of mesencephalic neurons lacking expression of the pro-survival transcription factors Engrailed-1 and −2 (Alavian et al. 2009). Altogether, these findings support that p75 NTR signaling may underlie dopaminergic neurodegeneration. ...
Article
Full-text available
The p75 neurotrophin receptor (p75NTR) is a multifunctional protein that regulates cellular responses to pathological conditions in specific regions of the nervous system. Activation of p75NTR in certain neuronal populations induces proteolytic processing of the receptor, thereby generating p75NTR fragments that facilitate downstream signaling. Expression of p75NTR has been reported in neurons of the ventral midbrain, but p75NTR signaling mechanisms in such cells are poorly understood. Here, we used Lund Human Mesencephalic cells, a population of neuronal cells derived from the ventral mesencephalon, to evaluate the effects of oxidative stress on p75NTR signaling. Subjection of the cells to oxidative stress resulted in decreased cell-surface localization of p75NTR and intracellular accumulation of p75NTR fragments. Oxidative stress-induced p75NTR processing was reduced by pharmacological inhibition of metalloproteases or γ-secretase, but was unaltered by blockade of the ligand-binding domain of p75NTR. Furthermore, inhibition of c-Jun N-terminal Kinase (JNK) decreased p75NTR cleavage induced by oxidative damage. Altogether, these results support a mechanism of p75NTR activation in which oxidative stress stimulates JNK signaling, thereby facilitating p75NTR processing via a ligand-independent mechanism involving induction of metalloprotease and γ-secretase activity. These findings reveal a novel role for JNK in ligand-independent p75NTR signaling, and, considering the susceptibility of mesencephalic neurons to oxidative damage associated with Parkinson’s disease (PD), merit further investigation into the effects of p75NTR on PD-related neurodegeneration.
... MesDA neurons are located in three distinct nuclei, substantia nigra pars compacta (SNpc) or the A9 group, ventral tegmental area (VTA) or A10, and retrorubral field or A8. Dopaminergic neurons in the SNpc innervate dorsal striatum forming the nigrostriatal pathway and the VTA neurons give rise to the mesolimbic and mesocortical (mesocorticolimbic) systems by innervating the nucleus accumbens and olfactory tubercle, respectively [1][2][3][4][5][6] . These pathways perform several functions in the reward system, control of emotion, motivation, cognition, and motor behavior. ...
Article
Full-text available
Midbrain dopaminergic neurons are responsible for several functions in the reward system, control of emotion, motivation, cognition, and motor behavior. A set of well-characterized transcription factors involved in specification, neurogenesis, and neuronal differentiation determines the fate of these neurons and modulates their survival and maintenance postnatally. Identifying novel molecular connections with these factors might, therefore, lead to a better understanding of physiology as well as dysfunction and degeneration of these neurons in Parkinson's disease. To explore the links between developmental pathways and to identify novel linkages, we examined the correlations of phylogenetic profiles of these factors with those of the entire human proteome. Our analysis revealed two distinct evolutionary pathways comprising the early and late developmental factors and new linkages with these developmental cascades. These results suggest an evolutionary scenario for the development of the midbrain and dopaminergic neurons, with potential novel links to the canonical and non-canonical functions of the transcription factors.
Article
Engrailed‐1 (EN1) is a developmental gene that encodes En1, a highly conserved transcription factor involved in regionalization during early embryogenesis and in the later maintenance of normal neurons. After birth, EN1 still plays a role in the development and physiology of the body; for example, it exerts a protective effect on midbrain dopaminergic (mDA) neurons, and loss of EN1 causes mDA neurons in the ventral midbrain to gradually die approximately 6 weeks after birth, resulting in motor and nonmotor symptoms similar to those observed in Parkinson's disease. Notably, EN1 has been identified as a possible susceptibility gene for idiopathic Parkinson's disease in humans. EN1 is involved in the processes of wound‐healing scar production and tissue and organ fibrosis. Additionally, EN1 can lead to tumorigenesis and thus provides a target for the treatment of some tumors. In this review, we summarize the effects of EN1 on embryonic organ development, describe the consequences of the deletion or overexpression of the EN1 gene, and discuss the pathways in which EN1 is involved. We hope to clarify the role of EN1 as a developmental gene and present potential therapeutic targets for diseases involving the EN1 gene.
Chapter
In recent years, medical developments have resulted in an increase in human life expectancy. Some developed countries now have a larger population of individuals aged over 64 than those under 14. One consequence of the ageing population is a higher incidence of certain neurodegenerative disorders. In order to prevent these, we need to learn more about them. This book provides up-to-date information on the use of transgenic mouse models in the study of neurodegenerative disorders such as Alzheimer's and Huntington's disease. By reproducing some of the pathological aspects of the diseases, these studies could reveal the mechanism for their onset or development. Some of the transgenic mice can also be used as targets for testing new compounds with the potential to prevent or combat these disorders. The editors have extensive knowledge and experience in this field and the book is aimed at undergraduates, postgraduates and academics. The chapters cover disorders including: Alzheimer's disease, Parkinson's disease, Huntington's and other CAG diseases, amyotrophic lateral sclerosis (ALS), recessive ataxias, disease caused by prions, and ischemia.
Article
Full-text available
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson’s Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Article
The reticular thalamic nucleus (Rt) is a sheet of neurons that surrounds the dorsal thalamus laterally, along its dorso-ventral and rostro-caudal axes. It consists of inhibitory neurons releasing gamma-aminobutyric acid (GABA). This nucleus participates in the circuitry between the thalamus and the cerebral cortex, and its impairment is associated with neuro-psychiatric disorders. In this study, we investigated the Rt anatomy of Engrailed-2 knockout mice (En2-/- ), a mouse model of autism spectrum disorder (ASD), using parvalbumin as an immunohistochemical marker. We compared 4- and 6-week-old wild type (WT) and En2-/- mice using various morphometric parameters: cell area, shape factor, circularity and cell density. Significant differences were present in 6-week-old male mice with different genetic background (WT vs. En2-/- ): the Rt neurons of En2-/- mice showed a bigger cell area, shape factor and circularity when compared with WT. Age (4 weeks vs. 6 weeks) influenced the shape factor of WT females, the circularity and cell density of En2-/- males, and the shape factor and circularity of En2-/- females. Gender affected cell density in 4-week-old WT mice, shape factor and cellularity of 6-week-old WT mice, and cell area, shape factor and cell density of En2-/- at 6 weeks. Intrasubject (left-right) asymmetry of Rt was never observed. These results show for the first time that sex- and age-related changes occur in the Rt GABAergic neurons of the En2-/- ASD mouse model.
Article
Full-text available
Many forms of apoptosis, including that caused by the death receptor CD95/Fas/APO-1, depend on the activation of caspases, which are proteases that cleave specific intracellular proteins to cause orderly cellular disintegration. The requirements for activating these crucial enzymatic mediators of death are not well understood. Using molecular chimeras with either CD8 or Tac, we find that oligomerization at the cell membrane powerfully induces caspase-8 autoactivation and apoptosis. Death induction was abrogated by the z-VAD-fmk, z-IETD-fmk, or p35 enzyme inhibitors or by a mutation in the active site cysteine but was surprisingly unaffected by death inhibitor Bcl-2. Amino acid substitutions that prevent the proteolytic separation of the caspase from its membrane-associated domain completely blocked apoptosis. Thus, oligomerization at the membrane is sufficient for caspase-8 autoactivation, but apoptosis could involve a death signal conveyed by the proteolytic release of the enzyme into the cytoplasm.
Article
Full-text available
Context The cause of Parkinson disease (PD) is unknown. Genetic linkages have been identified in families with PD, but whether most PD is inherited has not been determined, Objective To assess genetic inheritance of PD by studying monozygotic (MZ) and dizygotic (DZ) twin pairs. Design Twin study comparing concordance rates of PD in MZ and DZ twin pairs. Setting and Participants A total of 19 842 white male twins enrolled in the National Academy of Sciences/National Research Council World War II Veteran Twins Registry were screened for PD and standard diagnostic criteria for PD were applied. Zygosity was determined by polymerase chain reaction or questionnaire. Main Outcome Measure Parkinson disease concordance in twin pairs, stratified by zygosity and age at diagnosis. Results Of 268 twins with suspected parkinsonism and 250 presumed unaffected twin brothers, 193 twins with PD were identified (concordance-adjusted prevalence, 8.67/1000). In 71 MZ and 90 DZ pairs with complete diagnoses, pairwise concordance was similar (0.129 overall, 0.155 MZ, 0.111 DZ; relative risk, 1.39; 95% confidence interval, 0.63-3.1). In 16 pairs with diagnosis at or before age 50 years in at least 1 twin, MZ concordance was 1.0 (4 pairs), and DZ was 0.167 (relative risk, 6.0; 95% confidence interval, 1.69-21.26). Conclusions The similarity in concordance overall indicates that genetic factors do not play a major role in causing typical PD, No genetic component is evident when the disease begins after age 50 years. However, genetic factors appear to be important when disease begins at or before age 50 years.
Article
Full-text available
The study of protein function in neurons has been hindered by the lack of highly efficient, nontoxic methods of inducing RNA interference in such cells. Here we show that application of synthetic small interfering RNA (siRNA) linked to the vector peptide Penetratin1 results in rapid, highly efficient uptake of siRNA by entire populations of cultured primary mammalian hippocampal and sympathetic neurons. This treatment leads to specific knock-down of targeted proteins within hours without the toxicity associated with transfection. In contrast to current methods, our technique permits study of protein function across entire populations with minimal disturbance of complex cellular networks. Using this technique, we found that protein knock-down (evident after 6 hr) precedes any decrease in targeted message (evident after 24 hr), suggesting an early, translational repression by perfectly targeted siRNAs.
Article
The specificities of 28 commercially available compounds reported to be relatively selective inhibitors of particular serine/threonine-specific protein kinases have been examined against a large panel of protein kinases. The compounds KT 5720, Rottlerin and quercetin were found to inhibit many protein kinases, sometimes much more potently than their presumed targets, and conclusions drawn from their use in cell-based experiments are likely to be erroneous. Ro 318220 and related bisindoylmaleimides, as well as H89, HA1077 and Y 27632, were more selective inhibitors, but still inhibited two or more protein kinases with similar potency. LY 294002 was found to inhibit casein kinase-2 with similar potency to phosphoinositide (phosphatidylinositol) 3-kinase. The compounds with the most impressive selectivity profiles were KN62, PD 98059, U0126, PD 184352, rapamycin, wortmannin, SB 203580 and SB 202190. U0126 and PD 184352, like PD 98059, were found to block the mitogen-activated protein kinase (MAPK) cascade in cell-based assays by preventing the activation of MAPK kinase (MKK1), and not by inhibiting MKK1 activity directly. Apart from rapamycin and PD 184352, even the most selective inhibitors affected at least one additional protein kinase. Our results demonstrate that the specificities of protein kinase inhibitors cannot be assessed simply by studying their effect on kinases that are closely related in primary structure. We propose guidelines for the use of protein kinase inhibitors in cell-based assays.
Article
Parkinson's disease (PD) results primarily from the death of dopaminergic neurons in the substantia nigra. Current PD medications treat symptoms; none halt or retard dopaminergic neuron degeneration. The main obstacle to developing neuroprotective therapies is a limited understanding of the key molecular events that provoke neurodegeneration. The discovery of PD genes has led to the hypothesis that misfolding of proteins and dysfunction of the ubiquitin-proteasome pathway are pivotal to PD pathogenesis. Previously implicated culprits in PD neurodegeneration, mitochondrial dysfunction and oxidative stress, may also act in part by causing the accumulation of misfolded proteins, in addition to producing other deleterious events in dopaminergic neurons. Neurotoxin-based models (particularly MPTP) have been important in elucidating the molecular cascade of cell death in dopaminergic neurons. PD models based on the manipulation of PD genes should prove valuable in elucidating important aspects of the disease, such as selective vulnerability of substantia nigra dopaminergic neurons to the degenerative process.
Article
Transforming growth factor-βs (TGF-βs) constitute a superfamily of multifunctional cytokines with important implications in morphogenesis, cell differentiation, and tissue remodeling. In the developing nervous system, TGF-β2 and -β3 occur in radial and astroglial cells as well as in many populations of postmitotic, differentiating neurons. TGF-β1 is restricted to the choroid plexus and meninges. In addition to functions related to glial cell maturation and performances, TGF-β2 and -β3 are important regulators of neuron survival. In contrast to neurotrophic factors, as for example, neurotrophins, TGF-βs are most likely not neurotrophic by themselves. However, they can dramatically increase the potency of select neurotrophins, fibroblast growth factor-2, ciliary neurotrophic factor, and glial cell line-derived neurotrophic factor (GDNF). In the case of GDNF, we have shown that GDNF fails to promote the survival of highly purified neuron populations in vitro unless it is supplemented with TGF-β. This also applies to the in vivo situation, where antibodies to all three TGF-β isoforms fully prevent the trophic effect of GDNF on axotomized, target-deprived neurons. In addition to the TGF-β isoforms -β2 and -β3, other members of the TGF-β superfamily are expressed in the nervous system having important roles in embryonic patterning, cell migration, and neuronal transmitter determination. We have cloned and expressed a novel TGF-β, named growth/differentiation factor-15 (GDF-15). GDF-15 is synthesized in the choroid plexus and released into the CSF, but also occurs in all regions investigated of the developing and adult brain. GDF-15 is a potent trophic factor for developing and 6-OHDA-lesioned midbrain dopaminergic neurons in vitro and in vivo, matching the potency of GDNF.
Article
Apoptosis plays an important role during neuronal development, and defects in apoptosis may underlie various neurodegenerative disorders. To characterize molecular mechanisms that regulate neuronal apoptosis, the contributions to cell death of mitogen-activated protein (MAP) kinase family members, including ERK (extracellular signal-regulated kinase), JNK (c-JUN NH2-terminal protein kinase), and p38, were examined after withdrawal of nerve growth factor (NGF) from rat PC-12 pheochromocytoma cells. NGF withdrawal led to sustained activation of the JNK and p38 enzymes and inhibition of ERKs. The effects of dominant-interfering or constitutively activated forms of various components of the JNK-p38 and ERK signaling pathways demonstrated that activation of JNK and p38 and concurrent inhibition of ERK are critical for induction of apoptosis in these cells. Therefore, the dynamic balance between growth factor-activated ERK and stress-activated JNK-p38 pathways may be important in determining whether a cell survives or undergoes apoptosis.
Article
MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) is a newly described neurotoxin which selectively destroys cells in the substantia nigra when administered systemically to primates. Because the substance creates a condition in humans which is virtually indistinguishable from Parkinson's disease, interest has focused on its mechanism of action. We now report that MPTP appears to be rapidly metabolized after systemic administration in toxic amounts, and identify 1-methyl-4-phenylpyridinium ion (MPP+) as a probable major metabolite.