ArticlePDF Available

Loss of PINK1 enhances neurodegeneration in a mouse model of Parkinson's disease triggered by mitochondrial stress

Authors:

Abstract and Figures

Parkinson's disease (PD) shows a complex etiology, where both genetic and environmental factors contribute to initiation and advance of pathology. Mitochondrial dysfunction and mutation of genes implicated in mitochondria quality control are recognized contributors to etiopathology and progression of PD. Here we report the development and characterization of a genetic mouse model of PD with a combined etiology comprising: 1) induction of mitochondrial stress achieved through the expression of a mitochondrial matrix protein that accumulates in an unfolded state and 2) deletion of PINK1 gene. Using this model we address the role of PINK1 in mitochondrial quality control and disease progression. To induce mitochondrial stress specifically in catecholaminergic neurons we generated transgenic animals where the conditional expression of mitochondrial unfolded ornithine transcarbamylase (dOTC) is achieved under the tyrosine hydroxylase (Th) promoter. The mice were characterized in terms of survival, growth and motor behaviour. The characterization was followed by analysis of cell death induced in dopaminergic neurons and responsiveness to l-dopa. We demonstrate that accumulation of dOTC in dopaminergic neurons causes neurodegeneration and motor behaviour impairment that illustrates a parkinsonian phenotype. This associates with l-dopa responsiveness validating the model as a model of PD. The combined transgenic model where dOTC is overexpressed in PINK1 KO background presents increased neurodegeneration as compared to dOTC transgenic in wild-type background. Moreover, this combined model does not show responsiveness to l-dopa. Our in vivo data show that loss of PINK1 accelerates neurodegenerative phenotypes induced by mitochondrial stress triggered by the expression of an unfolded protein in this organelle.
Content may be subject to copyright.
Loss of PINK1 enhances neurodegeneration in a mouse model
of Parkinsons disease triggered by mitochondrial stress
q
Nicoleta Moisoi
b
,
*
, Valentina Fedele
a
, Jennifer Edwards
a
, L. Miguel Martins
a
,
**
a
Cell Death Regulation Laboratory, MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
b
Cell Physiology and Pharmacology Department, University of Leicester, Maurice Shock Building, University Road, Leicester LE1 9HN, UK
article info
Article history:
Received 14 July 2013
Received in revised form
12 September 2013
Accepted 7 October 2013
Keywords:
Mitochondria quality control
Neurodegeneration
Parkinsons disease
abstract
Parkinsons disease (PD) shows a complex etiology, where both genetic and environmental factors
contribute to initiation and advance of pathology. Mitochondrial dysfunction and mutation of genes
implicated in mitochondria quality control are recognized contributors to etiopathology and progression
of PD. Here we report the development and characterization of a genetic mouse model of PD with a
combined etiology comprising: 1) induction of mitochondrial stress achieved through the expression of a
mitochondrial matrix protein that accumulates in an unfolded state and 2) deletion of PINK1 gene. Using
this model we address the role of PINK1 in mitochondrial quality control and disease progression.
To induce mitochondrial stress specically in catecholaminergic neurons we generated transgenic
animals where the conditional expression of mitochondrial unfolded ornithine transcarbamylase (dOTC)is
achieved under the tyrosine hydroxylase (Th) promoter. The mice were characterized in terms of survival,
growth and motor behaviour. The characterization was followed by analysis of cell death induced in
dopaminergic neurons and responsiveness to
L
-dopa. We demonstrate that accumulation of dOTC in
dopaminergic neurons causes neurodegeneration and motor behaviour impairment that illustrates a
parkinsonian phenotype. This associates with
L
-dopa responsiveness validating the model as a model of
PD. The combined transgenic model where dOTC is overexpressed in PINK1 KO background presents
increased neurodegeneration as compared to dOTC transgenic in wild-type background. Moreover, this
combined model does not show responsiveness to
L
-dopa. Our in vivo data show that loss of PINK1 ac-
celerates neurodegenerative phenotypes induced by mitochondrial stress triggered by the expression of
an unfolded protein in this organelle.
Ó2013 The Authors. Published by Elsevier Ltd. All rights reserved.
1. Introduction
Parkinsons disease (PD) is the second most common neurode-
generative disorder, with a prevalence second only to that of Alz-
heimers disease. The primary hallmarks of the disease are
degeneration of multiple neuronal types including, most notably,
dopaminergic (DA) neurons in the Substantia Nigra of the midbrain
(Shulman et al., 2011), and formation of Lewy bodies, intra-
cytoplasmic inclusions that are mainly composed of brillar
a
-
synuclein. The, pathology of many non-dopaminergic neurons
including olfactory and brain stem neurons precedes that of DA
neurons (Braak et al., 2003). Patients with PD present characteristic
motor symptoms, such as resting tremor, slowness of movement,
rigidity, postural instability, and gait perturbation. PD patients also
present a combination of non-motor symptoms including psychi-
atric symptoms such as depression and anxiety, autonomic
dysfunction (involving cardiac and digestive systems), perturbed
sleep patterns, and musculoskeletal abnormalities (Simuni and
Sethi, 2008). PD has a complex multifactorial etiology where both
environmental and genetic factors appear to be important.
Mitochondrial dysfunction is at the core of several age related
neurodegenerative diseases including Parkinsons disease (Lezi and
Swerdlow, 2012). Over the last years mutations found in familial or
sporadic cases of neurodegenerative disorders provided insights
into the mechanisms underlying neurodegeneration. That is also
the case for PD where a number of proteins have been found
mutated and their function has been directly or indirectly corre-
lated with mitochondria, oxidative stress and accumulation of
q
This is an open-access article distributed under the terms of the Creative
Commons Attribution License, which permits unrestricted use, distribution, and
reproduction in any medium, provided the original author and source are credited.
*Corresponding author.
** Corresponding author. Tel.: þ44 116 252 5533; fax: þ4 4 116 252 5616.
E-mail addresses: nm105@le.ac.uk (N. Moisoi), martins.lmiguel@gmail.com
(L.M. Martins).
Contents lists available at ScienceDirect
Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm
0028-3908/$ esee front matter Ó2013 The Authors. Published by Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.neuropharm.2013.10.009
Neuropharmacology 77 (2014) 350e357
unfolded proteins (Kumar et al., 2012). The presence of protein
aggregates in neurodegeneration, including PD, has been well
documented. In PD, Lewy bodies are known to contain unfolded
proteins of mitochondrial origin, together with proteins that are
normally located in the cytosol or at neuronal synapses. Although
the accumulation of unfolded proteins in the mitochondria has
been debated (de Castro et al., 2010) there is now clear evidence
both from patient samples and animal models that in PD there is
accumulation of mitochondrial unfolded proteins correlated with
mitochondrial stress signalling (de Castro et al., 2012).
Cells have developed several defence mechanisms to cope with
mitochondrial damage (Tatsuta and Langer, 2008;Baker et al., 2011).
Molecular quality control represents the rst step in the mito-
chondrial defence mechanisms. This implicates the up-regulation of
Fig. 1. pZ/EG dOTC-FLAG expression construct. (A) The Z/EG construct for the expression of dOTC-FLAG consists of strong chicken
b
-actin promoter, directing the expression of a
loxP-anked (triangles)
b
geo (lacZ/neomycin-resistance) fusion gene and three SV40 polyadenylation sequences. Following that, there is the coding sequence of dOTC-FLAG, that
precedes an internal ribosomal entry site (IRES) and EGFP and a rabbit
b
globin polyadenylation sequence. In this conguration,
b
geo is expressed before Cre excision whereas dOTC-
FLAG and EGFP are expressed from a single mRNA after Cre excision. Indicated in the gure is the approximate position of oligonucleotide sequences used to detect the Cre-
mediated recombination by PCR analysis (P1, P2, P3). (B) PCR-analysis of Cre mediated recombination. The recombination in U2OS cells was achieved by overexpressing the Z/
EG dOTC-FLAG construct together with a plasmid expressing Cre recombinase under activation with tamoxifen. (C) Detection of EGFP in cells transfected with dOTC expression
plasmid following Cre-mediated recombination. Expression of dOTC at protein level following recombination is shown by immunostaining (red). (D) dOTC (green) colocalization
with mitochondria mtHsp70 (red) has been conrmed in U2OS cells transfected with dOTC in a vector for mammalian cells expression. (For interpretation of the references to
colour in this gure legend, the reader is referred to the web version of this article.).
N. Moisoi et al. / Neuropharmacology 77 (2014) 350e357 351
nuclear genes encoding for mitochondrial chaperones and proteases
to assist the removal of misfolded and non-assembled polypeptides.
This molecular quality control represents a form of mitochondrial
retrograde signalling known as the UPR
mt
(Haynes and Ron, 2010).
The second level of mitochondria quality control is achieved
through fusion and ssion processes facilitated by the highly dy-
namic nature of mitochondria (Twig et al., 2008a,b). Damaged
mitochondria can fuse with healthy organelles to restore the level of
healthy components necessary for proper mitochondrial function.
When the damage is excessive, the rst two levels of quality control
are overwhelmed mitochondria become depolarized and are tar-
geted for recycling through a specic form of autophagy, termed
mitophagy (Wang and Klionsky, 2011; Youle and Narendra, 2011).
Components of this quality control pathway such as PINK1 and
Parkin have already been found mutated in PD or associated with
increased susceptibility for PD like the mitochondrial protease
HTRA2 (reviewed in Rugarli and Langer, 2012).
The PINK1 gene encodes a highly conserved serineethreonine
kinase, mutations in which cause autosomal-recessive parkin-
sonism (Valente et al., 2004; Nuytemans et al., 2010). These mu-
tations compromise the kinase activity or interfere with protein
stability suggesting a loss-of-function mechanism in PD (Deas
et al., 2009; Kawajiri et al., 2011; Thomas and Cookson, 2009).
Further supporting this loss-of-function model, Drosophila pink1
mutants show reduced life span, and degeneration of ight mus-
cles and dopaminergic neurons (Clark et al., 2006; Park et al.,
2006).
Deletion of PINK1 in mice results in a subtle phenotype involving
decreased dopamine levels motor decits in aged mice (Gispert
et al., 2009) and impaired synaptic plasticity (Kitada et al., 2007).
A conditional KO of PINK1 displays phenotypes reminiscent of early
PD, including impaired gait, olfaction and serotonergic innervation
(Glasl et al., 2012). Moreover, mitochondrial respiration is impaired
in the PINK1 KO mice (Gautier et al., 2008; Gispert et al., 2009).
A model that has now been established to study mitochondrial
stress response and quality control is through overexpression of
unfolded mitochondrial ornithine transcarbamylase (OTC). OTC is a
mitochondrial enzyme involved in urea metabolism, normally only
expressed in liver and small intestine and absent from transformed
cells. Previous studies have established that accumulation of this
protein in the mitochondrial matrix in an unfolded structure
induced a mitochondrial stress response UPR
mt
, characterized by
transcriptional up-regulation of mitochondrial chaperones and
proteases (Zhao et al., 2002; de Castro et al., 2012).
Fig. 2. Expression of dOTC in Substantia Nigra. (A) The level of rat OTC mRNA in Substantia Nigra (SN) where the Cre recombination occurs under the tyrosine hydroxylase (Th)
promoter. We detected a signicantly higher mRNA level in the SN of the double transgenic mice. Statistical signicance was analysed with Student T-test (n¼3) between the
indicated groups. (B) EGFP uorescence image was taken from brain slices (perfused and xed in paraformaldehyde) with the same exposure for control and transgenic mice
expressing dOTC-EGFP. The imagescovered an area of the brain including most of the Substantia Nigra and Ventral Tegmental Area. (C) Expression of dOTC in dopaminergic neurons
conrmed using anti-GFP immunostaining (red) and by colocalization with tyrosine hydroxylase (green). (For interpretation of the references to colour in this gure legend, the
reader is referred to the web version of this article.).
N. Moisoi et al. / Neuropharmacology 77 (2014) 350e357352
Moreover we have recently reported the development of an
in vivo model for mitochondrial dysfunction as a consequence of
protein misfolding in Drosophila. In transgenic ies, the accumu-
lation of abnormally folded proteins resulted in mitochondrial
phenotypical alterations similar to those reported in Drosophila
models of PD (Clark et al., 2006; Park et al., 2006).
Here we present a mouse model that reproduces cardinal
features of the complex PD etiology. The model combines
mitochondrial stress achieved through the expression of dOTC in
the mitochondrial matrix together with impairment in the mito-
chondria quality control through genetic deletion of PINK1.
Using this model we show that the induction of mitochondrial
stress in dopaminergic neurons results in neurodegeneration
and motor impairment that is rescued by the administration of
L
-dopa. The process of neurodegeneration is accelerated by loss of
PINK1.
Fig. 3. Motor behaviour characterization. (A, B) Growth curves of double transgenic mice from two independent lines with single integration of the dOTC transgene do not show a sig-
nicantinuence of the transgeneexpressionon this parameter.(C, D) Motor abilityassessed byrearing activity in openeld systemis decreasedin both lines. Two-wayANOVA for age and
genotypeshow: panel C estatisticallysignicant differencesfor genotype (line 6-1C, p¼0.04, n¼3e5)and no statistical difference forage; panel D estatistically signicantdifferences for
genotype(line 8-3A, p¼0.03, n¼3e5) and no statisticaldifference forage; (E) Motor behaviouranalysis of tripletransgenic overexpressingdOTC single integrationfrom line 6-1C in PINK1
KObackground shows a more pronounceddecrease of motor ability.Two-wayANOVA shows signicantdifference with genotype,p¼0.05, n¼4e9, and not signicantdifference with age.
N. Moisoi et al. / Neuropharmacology 77 (2014) 350e357 353
Fig. 4. Neurodegeneration in the dopaminergic pathway. The neuronal cell death was assayed by immunohistochemistry of tyrosine hydroxylase positive neurons in brains of the
indicated genotypes. Overexpression of dOTC induces cell death in the Substantia Nigra with a signicant reduction in the dopaminergic neurons of 25% at one year of age (A, B)
(n¼3). In the PINK1 KO background this decrease is accentuated as shown by a more severe reduction in the number of Th positive neurons, to 40% (n¼3) (C) The level of
dopamine in Substantia Nigra was analysed by HPLC measurements in tissue of the indicated genotype. The dopamine content is reduced in the double transgenic in WT
N. Moisoi et al. / Neuropharmacology 77 (2014) 350e357354
2. Results
2.1. Generation of a system for the conditional expression of dOTC in
mice
In previous work we used dOTC to develop a model for mito-
chondrial stress in the fruit y, Drosophila melanogaster (de Castro
et al., 2012). In order to extend these studies to a mammalian sys-
tem, we made use of a system for the in vivo delivery of dOTC in
mouse. To achieve this, we generated a dOTC-FLAG expression
construct for in vivo studies based on the system described by Novak
and colleagues (Novak et al., 2000)(Fig.1A). This system enables the
expression of dOTC by Cre mediated recombination. Additionally,
cells where Cre-mediated recombination has taken place lose
expression of a lacZ reporter transgene with concomitant activation
of enhanced-GFP (EGFP) expression. We rst demonstrated the
efcient recombination of the transgene in U2OS cultured cells
transfected with the dOTC expression construct either in the pres-
ence or absence of a Cre recombinase expression vector. The
recombination at DNA level was conrmed by PCR (Fig. 1B), using
two sets of primers as indicated in Fig.1Aand B. Next, we transfected
cultured human cells with the transgene expression construct and
conrmed at protein level that co-expression of Cre recombinase
results in expression of dOTC with the EGFP reporter (Fig. 1C).
Additionally we have validated the localization of the dOTC to the
mitochondria by colocalization with mtHSP70 (Fig. 1D). We
demonstrated elsewhere that dOTC-FLAG overexpressed in vitro in
cell culture and in vivo in Drosophila accumulated in an unfolded
state in the mitochondria (de Castro et al., 2012) and its capability to
induce UPR
mt
in these systems (Zhao et al., 2002; de Castro et al.,
2012). Taken together this data validates our system for the
controlled delivery of dOTC in transgenic mouse models with the
purpose of inducing genetically a mitochondrial stress.
2.2. In vivo expression of dOTC in dopaminergic neurons
We rst obtained transgenic mice containing the dOTC expres-
sion system. After Southern Blot analysis to conrm the successful
integration of the gene, two transgenic lines containing a single
integration (6-1C, 8-3A) were used for further characterization.
In order to induce mitochondrial stress in catecholaminergic
neurons we crossed dOTC mice with a line that expresses the Cre
recombinase under the control of the tyrosine hydroxylase pro-
moter (Th-Cre line) (Savitt et al., 2005).
We then demonstrated, that dOTC is expressed in the tissue of
interest. We measured the mRNA levels for the dOTC transgene in
the Substantia Nigra (SN) where the Cre recombination occurs
under the tyrosine hydroxylase promoter, detecting a signicantly
higher mRNA level in the double transgenic mice (Fig. 2A).
Additionally, we conrmed the correct expression of the trans-
gene by measuring the uorescence of the EGFP reporter that is co-
expressed with dOTC (Fig. 2B). In order to increase the GFP signal
and to verify the selective expression in dopaminergic neurons, we
used anti-GFP staining (red) and colocalization with tyrosine hy-
droxylase stain (green) (Fig. 2C).
2.3. PINK1 loss of function enhances the degenerative phenotype
induced by mitochondrial stress
To determine the consequences of enhanced stress caused by
the expression of a mitochondrial protein in a misfolded state, the
dOTC-expressing mice were characterized for both growth and
motor behaviour. We did not observe any growth defects in dOTC
expressing mice (Fig. 3A and B). However, in both lines there is a
signicant decrease in motor abilities as measured by reduced
rearing movements (Fig. 3C and D).
One of the single integration lines (6-1C) has been used to study
the effect of PINK1 loss of function on the phenotype induced by the
expression of dOTC (Fig. 3E). Due to the low probability to obtain all
the desired genotypes in the same colony, the mice were bred on
two different colonies: dOTC were crossed with Th-Cre onwild type
PINK1 background and on PINK1 KO background. Therefore the
comparisons were made between dOTCþ/Thþversus control/WT,
dOTCþ/Thþ/PINK1 KO versus control/PINK1 KO.
The motor ability analysis in the PINK1 KO background shows a
decrease in the rearing activity for the triple transgenic line as
compared to PINK1 KO. This appears to be accelerated as compared
to the decrease in rearing activity induced by dOTC in the WT
background. In the WT background the reduction in the rearing
activity shows at one year of age while in the PINK1 KO background
it diminishes from six months.
We then characterized the transgenic mice from one of the lines
(6-1C) for cell death and dopamine content in the Substantia Nigra.
The tyrosine hydroxylase (Th) positive cell counts show 25%
decrease in neuronal population in dOTCþ/Thþat one year of age
(Fig. 4A and B). Similarly with the motor dysfunction trait, loss of
PINK1 function accelerates this phenotype. Thus, in PINK1 KO
background overexpression of dOTC induces a cell loss of about 40%
in SN (Fig. 4A and B). We have then measured the level of dopamine
in the Substantia Nigra by HPLC. PINK1 KO mice are known to
present a reduction in dopamine levels that becomes signicant
from 18 months of age in spite of no dopaminergic cell loss being
detected (Gispert et al., 2009). In our experimental set-up the PINK1
KO strain versus WT presented a reduction in the dopamine levels
reported to the mass of tissue at 12 months of age, but this was not
signicant to 95% with a p>0.05 in the Student T-test. Due to the
breeding protocol in two separated colonies (WT respectively
PINK1 KO background) we have presented the data comparing the
dOTCþ/Thþtransgenics to littermates control within the individual
colonies. The dOTC overexpressed in WT background demonstrates
a signicant decrease in the dopamine levels versus control.
Interestingly in the PINK1 KO background overexpression of dOTC
does not induce a further reduction in the dopamine levels (Fig. 4C).
In order to assess whether the parkinsonian phenotype we have
achieved in this transgenic mice is dependent on
L
-dopa, we per-
formed a pharmacological rescue experiment analysing the motor
behaviour after
L
-dopa injection. Injection of
L
-dopa induced an
increase in the rearing movement in 50% of controls and in 60% of
double transgenics (Fig. 4D). The increase in motor ability is sig-
nicant suggesting that motor dysfunction caused by mitochon-
drial stress is rescued by
L
-dopa. Most strikingly the injection of
L
-
dopa in the PINK1 KO background does not improve the motor
behaviour in any of the mice assessed (Fig. 4E). This suggests that
the dopaminergic signalling is impaired in the transgenic mice
presenting both dOTC induced mitochondrial stress and PINK1 loss
of function.
3. Discussion
Sporadic PD appears to have a complex multifactorial etiology
with variable contributions from environmental factors and genetic
susceptibility.
background, versus control. Overexpression of dOTC in PINK1 KO background does not decrease the dopamine level as compared to PINK1 KO control. (D) Pharmacological rescue of
behavioural decit with L-dopa shows increased activity in a population of mice from double transgenics in both control and dOTC overexpressing mice. The data presented here are
from responsive mice (n¼4WT,n¼3PINK1 KO). (E) For triple transgenic mice with dOTC expressed in PINK1 KO background there is no response reected in rearing activity
changes following L-dopa treatment. (n¼8 for each WT and PINK1 KO). The analysis was performed with Student T-test between the indicated groups.
N. Moisoi et al. / Neuropharmacology 77 (2014) 350e357 355
It is now recognized that Parkinsons disease is a multisystem
neurodegenerative disorder that affects multiple areas of the brain
(Braak et al., 2003) with motor symptoms appearing when greater
than 50% of the dopamine neurons in the Substantia Nigra are lost.
Modelling prodromal symptomatology of PD appears possible using
transgenic mice with loss of function in PINK1,PARKIN,DJ-1 genes or
gain of function in LRKK2, and
a
-synuclein [reviewed in Smith et al.
(2012)], genes found mutated in PD. The early symptomatology from
these models offers the possibility of targeting dysfunctional path-
ways, prior to severe neuronal loss when drug treatments appear to
improve symptoms but not to target the disease progression.
Mutations in PINK1 serineethreonine kinase are the second
most common cause of autosomal recessive parkinsonism after
mutations in the PARKIN gene (Valente et al., 2004). About 30
pathogenic mutations have been associated with the disease
[reviewed in Nuytemans et al. (2010)]. Deletion of PINK1 in mice
does not result in an overt phenotype. The mice display only subtle
decits, which differ slightly between different loss-of-function
models, but converge to give a whole picture of a prodromal
model of PD. Thus the phenotype present only minor decrease in
total dopamine levels in very old mice (Gispert et al., 2009),
impaired synaptic plasticity in the striatum but no loss of dopa-
minergic neurons in Substantia Nigra (Kitada et al., 2007) respec-
tively a minor loss of dopamine cells in a more recent conditional
model of PINK1 loss-of-function (Glasl et al., 2012). Mitochondrial
respiration is impaired in the striatum of PINK1 KO and respiration
decits can be induced in the cortex by cellular stress (Gautier et al.,
2008). The mobilization of reserve pool synaptic vesicles at the
neuromuscular junction of pink1 decient ies is impaired during
rapid stimulation due to synaptic ATP depletion, indicating that
synaptic activity cannot be maintained under increased energy
demand in pink1 decient neurons (Morais, 2009).
There is strong published data from in vitro and Drosophila
models linking PINK1 to mitochondrial function and mitochondrial
quality control together with Parkin (reviewed in Pilsl and
Winklhofer, 2012). However, the role of the PINK1/Parkin pathway
in vivo needs to be studied further particularly due to the fact that
recent work from Larsson and colleagues could not demonstrate an
in vivo role for Parkin in mitochondrial quality control in a mouse
model of Parkinsonism based on dopaminergic neuron-specic loss
of the mitochondrial transcription factor A (TFAM) (Van Laar et al.,
2011; Sterky et al., 2011).
The data presented here demonstrates for the rst time in an
in vivo model that PINK1 loss of function is capable to accelerate
neurodegenerative phenotypes induced by mitochondrial stress.
We made use of a model of mitochondrial stress induction through
the expression of a protein in the mitochondrial matrix that has
been previously characterized both in vitro and in vivo (Zhao et al.,
2002; de Castro et al., 2012). Our data indicates that the accumu-
lation of a misfolded protein in the dopaminergic neurons is
capable of inducing neurodegeneration and motor behaviour
impairment that illustrates a parkinsonian phenotype. This asso-
ciates with
L
-dopa responsiveness validating the model as a model
of Parkinsonism. We hypothesise further that increased expression
of dOTC would be able to accelerate the parkinsonian phenotype
that we have obtained here and would produce a model that can be
used in pharmacological studies for Parkinsons treatments.
Our results show that PINK1 loss of function accelerates the
motor decit and the dopaminergic degeneration induced by
overexpression of dOTC in the mitochondria. First, the motor
behaviour impairment although mild, as expected from a pro-
dromal model of PD, appears faster in the PINK1 KO background
than in the WT. PINK1 KO mice did not present a loss of dopami-
nergic neurons up to one year of age, similarly with the pheno-
types reported in the literature and reviewed here. However, the
PINK1 loss of function appears to accentuate the loss of dopami-
nergic neurons determined by the dOTC induced mitochondrial
stress. Interestingly, overexpression of dOTC in the PINK1 KO
background does not decrease the dopamine levels as compared to
the PINK1 KO control. These results suggest that loss of dopamine
production might occur in the same neuronal population or
through the same mechanistic pathway for both PINK1 loss of
function and dOTC expression.
Most strikingly, the loss of PINK1 compromised the rescue of the
motor decit induced by dOTC mitochondrial stress with
L
-dopa.
Previous published data mention that PINK1 loss of function results
in impaired dopaminergic signalling manifested in low dopamine
release from nigrostriatal terminals and consequent reduction in
activation of specic postsynaptic receptors. This is accompanied by
defects in corticostriatal electrophysiological properties (Kitada et al.,
2007). The fact that
L
-dopa injection does not appear to improve the
motor decit caused by mitochondrial stress in PINK1 KO back-
ground supports the hypothesis that the model presented here
demonstrates a decit in transmission of dopaminergic signalling.
In spite of the mild phenotypes that PINK1 KO mice show, this
model has established itself as a useful prodromal model of PD
(Smith et al., 2012). Here we make use of it to demonstrate that
PINK1 loss of function contributes to parkinsonian neuro-
degeneration. Moreover the combination of mitochondrial stress
and PINK1 loss of function demonstrates for the rst time that PD
has a multifactorial etiology where genetic and environmental
factors might synergize to increase neurodegeneration.
4. Materials and methods
4.1. Expression of dOTC
The deletion mutant dOTC (30-114) fused to a carboxy-terminal FLAG tag
sequence was clonedinto the conditional Z/EG expression vector (Novaket al., 200 0).
Recombination was assayed using a PCR strategy with primers P1 (5
0
-
TCTGCTAACCATGTTCATGCC-3
0
), P2 (5
0
-ATGTGCTGCAAGGCGATTAAG-3
0
) and P3 (5
0
-
TCTGACAGTCCGTTGACAATTG-3
0
). The recombination was tested in U2OS cells by
transfection (with Effectene) of the Z/EG dOTC-FLAG expression vector together with
the Cre expression vector pCAGGS_Cre-ER (a gift from Prof. Catrin Pritchard, Uni-
versity of Leicester). Expression of Cre was induced 24 h after transfection by addi-
tion of 4-hydroxy-tamoxifen(1 mM) for 48 h. The cells were than xed and extracted
with 3% PFA in microtubule stabilizing buffer (Moisoi et al., 2002) and processed for
immunouorescence with the antibody against OTC (Sigma). For colocalization of
dOTC with the mitochondria, dOTC was overexpressed in U2OS using a mammalian
expression vector, pcDNA3-dOTC, and the cells were xed and processed for
immunouorescence with the indicated antibodies (OTC respectively mtHSP70).
4.2. Animal husbandry
Animal husbandry and experimental procedures were performed in full
compliance with the United Kingdom Animal (Scientic Procedures) Act 1986.
The dOTC lines have been produced and characterized for the integration of the
gene by GENEOWAY, Lyon, France. The mice used in these experiments have been
backcrossed 5e6 times to a C57/B6 background.
The PINK1 KO mice have been obtained from LEXICON GENETICS and have been
described previously (Wood-Kaczmar et al., 2008). They were fully backcrossed on
C57/B6 background (more than ten times).
The Th-Cre line was from Jacksons laboratory and they are bred on a C57/B6
background.
In order to obtain the desired genotypes the mice were bred on two different
colonies: dOTC were crossed with Th-Cre on wild type PINK1 background and on
PINK1 KO background. For the experiments presented here we used males.
4.3. Mice genotyping
The genotyping of dOTC mice was performed within the EGFP locus with
the primers: oIMR0042(CTAGGCCACAGAATTGAAAGATCT), oIMR0043(GTAGGTG-
GAAATTCTAGCATCATCC), oIMR0872(AAGTTCATCTGCACCACCG), oIM1416(A-
GATGGTGCG). The top band at 324 bp is a positive control band and the bottom band
at 173 bp represents the EGFP locus genotype.
The ThCre mice were genotyped using the primers:
Th-Cre(þ) (AAATGTTGCTGGATAGTTTTTACTGC)
Th-Cre() (GGAAGGTGTCCAATTTACTGACCGTA)
N. Moisoi et al. / Neuropharmacology 77 (2014) 350e357356
This genotyping protocol provides a single band for the mutant locus at 300 bp.
The PINK1 KO genotype was performed using the primers:
EB0088-26 (CTGCCCTCAGGGTCTCTAATGC),
EB0088-27 (GGAAGGAGGCCATGGAAATTGT),
Neo3a (GCAGCGCATCGCCTTCTATC)
The top band at 296 bp genotypes the wt locus, the bottom band at 193 bp
genotypes the mutant locus.
4.4. Behavioural testing
Locomotor activity was assessed using a computer-controlled photocell-based
system (Linton Instruments, UK) as described previously (Moisoi et al., 2009). The
activity has been recorded every 10min over 1 h and the result is given as average of
the 6 measurements. The parameter reported is Rearing activitycalculated as
number of rearings in active time.
4.5. Pharmacological rescue
Transgenic mice and control littermates were injected intraperitoneal with sa-
line control (0.9% NaCl) or methyl levodopa hydrochloride 25 mg/kg with benser-
azide 6.5 mg/kg (Sigma) in saline solution. Behavioural testing was performed
40 min after the injection.
4.6. Histology, immunohistochemistry and immunouorescence of brain tissues
In order to conrm the protein overexpression the brains of 2 months old mice
were perfused and xed in 4% paraformaldehyde. Coronal sections were cut at 10
m
m
thickness. For GFP signal, images were taken at the same exposure for both control
and transgenic mice. In order to check the localization of the GFP signal in the
dopaminergic neurons, the sections were processed for immunouorescence using
an anti-GFP (Roche) and anti-tyrosine hydroxylase antibodies.
For tyrosine hydroxylase staining, and dopaminergic cell counts, brains were
harvested from one-year mice old and processed as described previously (Moisoi
et al., 2009). Dopaminergic neurons were visualized by immunostaining with
anti-tyrosine hydroxylase antibody using the DAKO duet system (DAKO K0492)
with 3,3, diaminobenzidine (DAB) as the chromagen according to the manufac-
turers instruction. The immunostained sections were counterstained with
hematoxilin.
4.7. Neuronal quantitation
Counts of dopaminergic neurons(tyrosine hydroxylase positive)were performed
in the area of Substantia Nigra and Ventral Tegmental Area. Whole brain coronal
sections were reconstituted from digital images acquired with a 10objective using
the Photomerge tool in Adobe Photoshop CS3. Counts of tyrosine hydroxylase cells
were performed using stereologic methods as described (Shin et al., 2011).
4.8. HPLC measurement of dopamine
For sample preparation, Substantia Nigra of one-year old males with the indi-
cated genotypes was dissected and quickly frozen in liquid nitrogen. The tissue was
weighed and homogenized in 200
m
l chilled 0.01 M perchloric acid using a motor-
ized, hand-held tissue homogenizer. The chilled homogenates were ltered through
a low-binding Durapore (0.22
m
m) PVDF membrane using Ultrafree-MC centrifugal
devices. The dopamine measurement was performed immediately after the sample
preparation. Supernatant uid was eluted at a ow rate of 50
m
l/min through a
150 1.0 mm C18 column (ALF-115, ANTEC). As recommended by the manufacturer
instructions the mobile phase contained: 50 mM phosphoric acid, 50 mM citric acid,
8 mM NaCl, 0.1 mM EDTA, 10% methanol, 350 mg/l OSA (pH 3.2). Analysis was
performed using an Alexys LC-EC system equipped with a DECADE II electro-
chemical detector (ANTEC). The level of dopamine was reported to the mass of tissue
and the values are presented as ratio to control.
4.9. Quantitative real-time RT-PCR
Quantitative RT-PCR was performed on an Mx4000 (Stratagene) real-time
cycler using the QuantiTect SYBR Green RT-PCR system (QIAGEN). Primers for rat
OTC were obtained from QIAGEN (QuantiTect Primer Assays). The relative tran-
script levels of the target genes were normalized against mouse GAPDH mRNA
levels. Quantication was performed using the comparative Ct method (Schmittgen
and Livak, 2008).
4.10. Statistical analysis
Data are presented as mean values, and error bars indicate SD. Inferential sta-
tisticalanalysis wasperformed usingthe Prism and StatMate software packages (www.
graphpad.com). The signicance level is indicated as * for p0.05 and NS for p>0.05.
References
Baker, M.J., et al., 2011. Quality control of mitochondrial proteostasis. Cold Spring
Harb. Perspect. Biol. http://dx.doi.org/10.1101/cshperspect.a007559.
Braak, H., et al., 2003. Staging of brain pathology related to sporadic Parkinsons
disease. Neurobiol. Aging 24, 197e211.
Clark, I.E., et al., 2006. Drosophila pink1 is required for mitochondrial function and
interacts genetically with parkin. Nature 441, 1162e1166.
de Castro, I.P., et al., 2010 Apr 19. Mitochondrial quality control and neurological
disease: an emerging connection. Expert Rev. Mol. Med. 12. http://dx.doi.org/
10.1017/S1462399410001456 e12.
de Castro, I.P., et al., 2012. Genetic analysis of mitochondrial protein misfolding in
Drosophila melanogaster. Cell Death Differ. 19, 1308e1316.
Deas, E., et al., 2009. PINK1 function in health and disease. EMBO Mol. Med. 1, 152e165.
Gautier, C.A., et al., 2008. Loss of PINK1 causes mitochondrial functional defects and
increased sensitivity to oxidative stress. PNAS 105, 11364e11369.
Gispert, S., et al., 2009. Parkinson phenotype in aged PINK1-decient mice is
accompanied by progressive mitochondrial dysfunction in absence of neuro-
degeneration. PLoS One 4 e5777.
Glasl, L., et al., 2012. Pink1-deciency in mice impairs gait olfaction and seroto-
nergic innervation of the olfactory bulb. Exp. Neurol. 235, 214e227.
Haynes, C.M., Ron, D., 2010. The mitochondrial UPR-protecting organelle protein
homeostasis. J. Cell Sci. 123, 3849e3855.
Kawajiri, S., et al., 2011. Genetic mutations and functions of PINK1. Trends Phar-
macol. Sci. 32, 573e580.
Kitada, T., et al., 2007. Impaired dopamine release and synaptic plasticity in the
striatum of PINK1-decient mice. PNAS 104, 11441e11 4 4 6.
Kumar, K.R., et al., 2012. Genetics of Parkinson disease and other movement dis-
orders. Curr. Opin. Neurol. 25, 466e474.
Lezi, E., Swerdlow, R.H., 2012. Mitochondria in neurodegeneration. Adv. Exp. Med.
Biol. 942, 269e286.
Moisoi, N., et al., 2002. Calmodulin-containing substructures of the centrosomal
matrix released by microtubule perturbation. J. Cell Sci. 115, 2367e2379.
Moisoi, N., et al., 2009. Mitochondrial dysfunction triggered by loss of HtrA2 results
in the activation of a brain-specic transcriptional stress response. Cell Death
Differ. 16, 449e464.
Morais, V.A., 2009. Parkinsons disease mutations in PINK1 result in decreased
complex 1 activity and decient synaptic function. EMBO Mol. Med. 1, 99e111.
Novak, A., et al., 2000. Z/EG, a double reporter mouse line that expresses enhanced
green uorescent protein upon Cre-mediated excision. Genesis 28, 147e155.
Nuytemans, K., et al., 2010. Genetic etiology of Parkinson disease associated with
mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation
update. Hum. Mutat. 31, 763e780.
Park, J., et al., 2006. Mitochondrial dysfunction in Drosophila PINK1 mutants is
complemented by parkin. Nature 4 41, 1157e1161.
Pilsl, A., Winklhofer, K.F., 2012. Parkin, PINK1 and mitochondrial integrity: emerging
concepts of mitochondrial dysfunction in Parkinsons disease. Acta Neuro-
pathol. 123, 173e188.
Rugarli, E.I., Langer, T., 2012. Mitochondrial quality control: a matter of life and
death for neurons. EMBO J. 31, 1336e1349.
Savitt, J.M., et al., 2005. Bcl-x is required for proper development of the mouse
substantia nigra. J. Neurosci. 25, 6721e6728.
Schmittgen, T.D., Livak, K.J., 2008. Analysing real time PCR data by the comparative
C(T) method. Nat. Protoc. 3, 1101e1108 .
Shin, J.H., et al., 2011. PARIS (ZNF746) repression of PGC-1a contributes to neuro-
degeneration in Parkinsons disease. Cell 144, 689e702.
Shulman, et al., 2011. Parkinsons disease: genetics and pathogenesis. Annu. Rev.
Pathol. 6, 193e222.
Simuni, Sethi, 2008 Dec. Non motor manifestations of Parkinsons disease. Ann.
Neurol. 64 (Suppl. 2), S65eS80.
Smith, G.A., et al., 2012. The search for the genetic mouse models of prodromal
Parkinsons disease. Exp. Neurol. 237, 267e273.
Sterky, F.H., et al., 2011. Impaired mitochondrial transport and Parkin-independent
degeneration of respiratory chain-decient dopamine neurons in vivo. PNAS
108, 12937e12942.
Tatsuta, T., Langer, T., 2008. Quality control of mitochondria: protection against
neurodegeneration and aging. EMBO J. 27, 306e314.
Thomas, K.J., Cookson, M.R., 2009. The role of PTEN-induced kinase 1 in mito-
chondrial dysfunction and dynamics. Int. J. Biochem. Cell Biol. 41, 2025e2035.
Twig, G., et al., 2008a. Fission and selective fusion govern mitochondrial segregation
and elimination by autophagy. EMBO J. 27, 433e446.
Twig, G., et al., 2008b. Mitochondrial fusion, ssion and autophagy as a quality
control axis: the bioenergetic view. Biochim. Biophys. Acta 1777, 1092e1097.
Valente, E.M., et al., 20 04. Hereditary early-onset Parkinsons disease caused by
mutations in PINK1. Science 304, 1158e116 0.
Van Laar, V.S., et al., 2011. Bioenergetics of neurons inhibit the translocation response of
Parkin following rapidmitochondrialdepolarization.Hum. Mol.Genet. 20,927e940.
Wang,K., Klionsky, D.J., 2011. Mitochondria removalby autophagy. Autophagy 7,297e300.
Wood-Kaczmar, A., et al., 2008 Jun 18. PINK1 is necessary for long-term survival and
mitochondrial function in human dopaminergic neurons. PLoS One 3 (6) e2455.
Youle, R.J., Narendra, D.P., 2011. Mechanisms of mitophagy. Nat. Rev. Mol. Cell. Biol.
12, 9e14.
Zhao, Q., et al., 2002. A mitochondrial specic stress response in mammalian cells.
EMBO J. 21, 4411e4419.
N. Moisoi et al. / Neuropharmacology 77 (2014) 350e357 357
... , we utilized a cell-based model, which expresses a deletion mutant of ornithine transcarbamylase (ΔOTC)(Fig. 5A)(Moisoi et al., 2014). This mutanyields detergent-insoluble, intra-mitochondrial ΔOTC protein aggregates within the mitochondrial matrix that can be cleared by PINK1/Parkin-mediated mitophagy(Burman et al., 2017;Moisoi et al., 2014). ...
... 5A)(Moisoi et al., 2014). This mutanyields detergent-insoluble, intra-mitochondrial ΔOTC protein aggregates within the mitochondrial matrix that can be cleared by PINK1/Parkin-mediated mitophagy(Burman et al., 2017;Moisoi et al., 2014). In HeLa cells expressing doxycycline-inducible ΔOTC and YFP-Parkin, enhancing PINK1 activity with MTK458 treatment resulted in the robust clearance of ΔOTC as measured by either immuno uorescence or Western blotting (Figs. ...
Preprint
Full-text available
PINK1 loss-of-function mutations and exposure to mitochondrial toxins are causative for Parkinson’s disease (PD) and Parkinsonism, respectively. We demonstrate that pathological α-synuclein deposition, the hallmark pathology of idiopathic PD, induces mitochondrial dysfunction, and impairs mitophagy as evidenced by the accumulation of the PINK1 substrate pS65-Ubiquitin (pUb). We discovered MTK458, a brain penetrant small molecule that binds to PINK1 and stabilizes its active complex, resulting in increased rates of mitophagy. Treatment with MTK458 mediates clearance of accumulated pUb and α-synuclein pathology in α-synuclein pathology models in vitro and in vivo. Our findings from preclinical PD models suggest that pharmacological activation of PINK1 warrants further clinical evaluation as a therapeutic strategy for disease modification in PD.
... Defects in PINK1 also impair the oxidative stress-induced antioxidant heme oxygenase-1 expression [60]. Furthermore, a transgenic mouse model that has a combined etiology of PINK1 deletion and mitochondrial stress caused by the conditional expression of mitochondrial unfolded ornithine transcarbamylase with the TH promoter showed accelerated neurodegeneration as compared to the transgenic mice with wild-type background and also no responsiveness to L-dopa [61]. ...
Article
Full-text available
Mitochondrial dysregulation, such as mitochondrial complex I deficiency, increased oxidative stress, perturbation of mitochondrial dynamics and mitophagy, has long been implicated in the pathogenesis of PD. Initiating from the observation that mitochondrial toxins cause PD-like symptoms and mitochondrial DNA mutations are associated with increased risk of PD, many mutated genes linked to familial forms of PD, including PRKN, PINK1, DJ-1 and SNCA, have also been found to affect the mitochondrial features. Recent research has uncovered a much more complex involvement of mitochondria in PD. Disruption of mitochondrial quality control coupled with abnormal secretion of mitochondrial contents to dispose damaged organelles may play a role in the pathogenesis of PD. Furthermore, due to its bacterial ancestry, circulating mitochondrial DNAs can function as damage-associated molecular patterns eliciting inflammatory response. In this review, we summarize and discuss the connection between mitochondrial dysfunction and PD, highlighting the molecular triggers of the disease process, the intra- and extracellular roles of mitochondria in PD as well as the therapeutic potential of mitochondrial transplantation.
... PINK1 and Parkin mutations are responsible for more than 50% of autosomal recessive juvenile parkinsonism cases (Bonifati et al., 2002). Interesting, due to their physiologically compelled actions, mutations in PINK1 and Parkin pairs may either complement each other or interact with other somatic mutations in the genome to develop more severe PD phenotypes with varying disease outcome (Creed & Goldberg, 2020;Kitada et al., 2009;Moisoi et al., 2014). Impaired mitochondrial respiration is observed in the striatum but not in the cerebral cortex, suggesting specificity of this defect for dopaminergic circuitry (Yang & Lu, 2009). ...
Article
Full-text available
Parkinson's disease (PD) relates to defective mitochondrial quality control in the dopaminergic motor network. Genetic studies have revealed that PINK1 and Parkin mutations are indicative of a heightened propensity to PD onset, pinpointing mitophagy and inflammation as the culprit pathways involved in neuronal loss in the substantia nigra (SNpc). In a reciprocal manner, LRRK2 functions in the regulation of basal flux and inflammatory responses responsible for PINK1/Parkin-dependent mitophagy activation. Pharmacological intervention in these disease-modifying pathways may facilitate the development of novel PD therapeutics, despite the current lack of an established drug evaluation model. As such, we reviewed the feasibility of employing the versatile global Pink1 knockout (KO) rat model as a self-sufficient, spontaneous PD model for investigating both disease etiology and drug pharmacology. These rats retain clinical features encompassing basal mitophagic flux changes with PD progression. We demonstrate the versatility of this PD rat model based on the incorporation of additional experimental insults to recapitulate the proinflammatory responses observed in PD patients.
... Besides, genetic analyses associated mutations of critical genes, including PARKIN (PARK2) 3 , PINK1 (PARK6) 4 , DJ-1 (PARK7) 5 and LRRK2 (PARK8) 6 with PD. Interestingly, studies documented neurodegeneration and parkinsonian phenotype, accompanied by impaired mitochondrial function upon loss of PINK1 and PARKIN in different models [7][8][9][10][11] . Further investigation revealed that these genes are involved in the regulation of mitochondrial biogenesis 12,13 , protein turnover 14 and autophagic clearance 15 . ...
Article
Full-text available
Parkinson’s Disease (PD) is a chronic and progressive neurodegenerative disease manifesting itself with tremors, muscle stiffness, bradykinesia, dementia, and depression. Mutations of mitochondrial E3 ligase, PARKIN, have been associated with juvenile PD. Previous studies have characterized muscle atrophy and motor deficits upon loss of functional Parkin in fly and rodent models. However, the mechanisms behind pathophysiology of Parkin deficient muscle remains to be elusive. Here, results suggested that knock down of Parkin significantly increases proteolytic activities in skeletal muscle cell line, the C2C12 myotubes. However, the atrogene levels increase moderately in Parkin deficient cell line. To further investigate the role of Parkin in skeletal muscle atrophy, Parkin knock out (KO) and wild type mice were subjected to 48 h starvation. After 48 h fasting, a greater reduction in skeletal muscle weights was observed in Parkin KO mice as compared to age matched wild type control, suggesting elevated proteolytic activity in the absence of Parkin. Subsequent microarray analyses revealed further enhanced expression of FOXO and ubiquitin pathway in fasted Parkin KO mice. Furthermore, a greater reduction in the expression of cytoskeleton genes was observed in Parkin KO mice following 48 h fasting. Collectively, these results suggest that Parkin deficiency exacerbates fasting-induced skeletal muscle wasting, through upregulating genes involved in catabolic activities in skeletal muscle.
... In addition, aged (30 days old) pink-1 mutant flies exhibit degeneration of DA neurons in the PPL1 (Protocerebral posterior lateral) cluster. Similarly, pink-1 mutant caused locomotor defects and degenerated DA neurons in mice (Moisoi et al., 2014). These behavioural phenotypes of the pink1 mutant were accompanied by mitochondrial dysfunction. ...
Article
Full-text available
Parkinson’s disease (PD) is the second most popular age-associated neurodegenerative disorder after Alzheimer’s disease. The degeneration of dopaminergic neurons, aggregation of α-synuclein (α-syn), and locomotor defects are the main characteristic features of PD. The main cause of a familial form of PD is associated with a mutation in genes such as SNCA , PINK1 , Parkin , DJ-1 , LRKK2 , and others. Recent advances have uncovered the different underlying mechanisms of PD but the treatment of PD is still unknown due to the unavailability of effective therapies and preventive medicines in the current scenario. The pathophysiology and genetics of PD have been strongly associated with mitochondria in disease etiology. Several studies have investigated a complex molecular mechanism governing the identification and clearance of dysfunctional mitochondria from the cell, a mitochondrial quality control mechanism called mitophagy. Reduced mitophagy and mitochondrial impairment are found in both sporadic and familial PD. Pharmacologically modulating mitophagy and accelerating the removal of defective mitochondria are of common interest in developing a therapy for PD. However, despite the extensive understanding of the mitochondrial quality control pathway and its underlying mechanism, the therapeutic potential of targeting mitophagy modulation and its role in PD remains to be explored. Thus, targeting mitophagy using chemical agents and naturally occurring phytochemicals could be an emerging therapeutic strategy in PD prevention and treatment. We discuss the current research on understanding the role of mitophagy modulators in PD using Drosophila melanogaster as a model. We further explore the contribution of Drosophila in the pathophysiology of PD, and discuss comprehensive genetic analysis in flies and pharmacological drug screening to develop potential therapeutic molecules for PD.
... In a familial form of PD, mutations have been identified in genes encoding mitochondrionassociated proteins such as mitochondrial phosphatase and tensin homologue (PTEN)induced kinase 1 (PINK1), Parkinson juvenile disease protein 2 (parkin), protein deglycase DJ-1 (Parkinson disease protein 7), and coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) [197]. PINK1 KO mice may use a prodromal model of PD, as the mice show olfactory and gain disturbances [198]. The Parkin KO mouse is a classic transgenic PD model, while there are few studies using DJ-1 KO rats [199]. ...
Article
Full-text available
Nearly half a century has passed since the discovery of cytoplasmic inheritance of human chloramphenicol resistance. The inheritance was then revealed to take place maternally by mitochondrial DNA (mtDNA). Later, a number of mutations in mtDNA were identified as a cause of severe inheritable metabolic diseases with neurological manifestation, and the impairment of mitochondrial functions has been probed in the pathogenesis of a wide range of illnesses including neurodegenerative diseases. Recently, a growing number of preclinical studies have revealed that animal behaviors are influenced by the impairment of mitochondrial functions and possibly by the loss of mitochondrial stress resilience. Indeed, as high as 54% of patients with one of the most common primary mitochondrial diseases, mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) syndrome, present psychiatric symptoms including cognitive impairment, mood disorder, anxiety, and psychosis. Mitochondria are multifunctional organelles which produce cellular energy and play a major role in other cellular functions including homeostasis, cellular signaling, and gene expression, among others. Mitochondrial functions are observed to be compromised and to become less resilient under continuous stress. Meanwhile, stress and inflammation have been linked to the activation of the tryptophan (Trp)-kynurenine (KYN) metabolic system, which observably contributes to the development of pathological conditions including neurological and psychiatric disorders. This review discusses the functions of mitochondria and the Trp-KYN system, the interaction of the Trp-KYN system with mitochondria, and the current understanding of the involvement of mitochondria and the Trp-KYN system in preclinical and clinical studies of major neurological and psychiatric diseases.
Article
Age‐related macular degeneration (AMD) is a devastating eye disease that causes permanent vision loss in the central part of the retina, known as the macula. Patients with such severe visual loss face a reduced quality of life and are at a 1.5 times greater risk of death compared to the general population. Currently, there is no cure for or effective treatment for dry AMD. There are several mechanisms thought to underlie the disease, for example, ageing‐associated chronic oxidative stress, mitochondrial damage, harmful protein aggregation and inflammation. As a way of gaining a better understanding of the molecular mechanisms behind AMD and thus developing new therapies, we have created a peroxisome proliferator‐activated receptor gamma coactivator 1‐alpha and nuclear factor erythroid 2‐related factor 2 ( PGC1α / NFE2L2 ) double‐knockout (dKO) mouse model that mimics many of the clinical features of dry AMD, including elevated levels of oxidative stress markers, damaged mitochondria, accumulating lysosomal lipofuscin and extracellular drusen‐like structures in retinal pigment epithelial cells (RPE). In addition, a human RPE cell‐based model was established to examine the impact of non‐functional intracellular clearance systems on inflammasome activation. In this study, we found that there was a disturbance in the autolysosomal machinery responsible for clearing mitochondria in the RPE cells of one‐year‐old PGC1α/NFE2L2‐deficient mice. The confocal immunohistochemical analysis revealed an increase in autophagosome marker microtubule‐associated proteins 1A/1B light chain 3B (LC3B) as well as multiple mitophagy markers such as PTE‐induced putative kinase 1 (PINK1) and E3 ubiquitin ligase (PARKIN), along with signs of damaged mitochondria. However, no increase in autolysosome formation was detected, nor was there a colocalization of the lysosomal marker LAMP2 or the mitochondrial marker, ATP synthase β. There was an upregulation of late autolysosomal fusion Ras‐related protein (Rab7) in the perinuclear space of RPE cells, together with autofluorescent aggregates. Additionally, we observed an increase in the numbers of Toll‐like receptors 3 and 9, while those of NOD‐like receptor 3 were decreased in PGC1α/NFE2L2 dKO retinal specimens compared to wild‐type animals. There was a trend towards increased complement component C5a and increased involvement of the serine protease enzyme, thrombin, in enhancing the terminal pathway producing C5a, independent of C3. The levels of primary acute phase C‐reactive protein and receptor for advanced glycation end products were also increased in the PGC1α/NFE2L2 dKO retina. Furthermore, selective proteasome inhibition with epoxomicin promoted both nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial‐mediated oxidative stress, leading to the release of mitochondrial DNA to the cytosol, resulting in potassium efflux‐dependent activation of the absent in melanoma 2 (AIM2) inflammasome and the subsequent secretion of interleukin‐1β in ARPE‐19 cells. In conclusion, the data suggest that there is at least a relative decrease in mitophagy, increases in the amounts of C5 and thrombin and decreased C3 levels in this dry AMD‐like model. Moreover, selective proteasome inhibition evoked mitochondrial damage and AIM2 inflammasome activation in ARPE‐19 cells.
Article
Parkinson's disease (PD) is a severe neurodegenerative disorder, partly attributed to mutations, environmental toxins, oxidative stress, abnormal protein aggregation, and mitochon-drial dysfunction. However, the precise pathogenesis of PD and its treatment strategy still require investigation. Fortunately, natural products have demonstrated potential as therapeutic agents for alleviating PD symptoms due to their neuroprotective properties. To identify promising lead compounds from herbal medicines' natural products for PD management and understand their modes of action, suitable animal models are necessary. Drosophila melanogaster (fruit fly) serves as an essential model for studying genetic and cellular pathways in complex biological processes. Diverse Drosophila PD models have been extensively utilized in PD research, particularly for discovering neuroprotective natural products. This review emphasizes the research progress of natural products in PD using the fruit fly PD model, offering valuable insights into utilizing inver-tebrate models for developing novel anti-PD drugs.
Article
Malignant brain aging corresponds to accelerated age-related declines in brain functions eventually derailing the self-sustaining forces that govern independent vitality. Malignant brain aging establishes the path toward dementing neurodegeneration, including Alzheimer’s disease (AD). The full spectrum of AD includes progressive dysfunction of neurons, oligodendrocytes, astrocytes, microglia, and the microvascular systems, and is mechanistically driven by insulin and insulin-like growth factor (IGF) deficiencies and resistances with accompanying deficits in energy balance, increased cellular stress, inflammation, and impaired perfusion, mimicking the core features of diabetes mellitus. The underlying pathophysiological derangements result in mitochondrial dysfunction, abnormal protein aggregation, increased oxidative and endoplasmic reticulum stress, aberrant autophagy, and abnormal post-translational modification of proteins, all of which are signature features of both AD and dysregulated insulin/IGF-1-mechanistic target of rapamycin (mTOR) signaling. This article connects the dots from benign to malignant aging to neurodegeneration by reviewing the salient pathologies associated with initially adaptive and later dysfunctional mTOR signaling in the brain. Effective therapeutic and preventive measures must be two-pronged and designed to 1) address complex and shifting impairments in mTOR signaling through the re-purpose of effective anti-diabetes therapeutics that target the brain, and 2) minimize the impact of extrinsic mediators of benign to malignant aging transitions, e.g., inflammatory states, obesity, systemic insulin resistance diseases, and repeated bouts of general anesthesia, by minimizing exposures or implementing neuroprotective measures.
Article
Full-text available
Protein misfolding has a key role in several neurological disorders including Parkinson's disease. Although a clear mechanism for such proteinopathic diseases is well established when aggregated proteins accumulate in the cytosol, cell nucleus, endoplasmic reticulum and extracellular space, little is known about the role of protein aggregation in the mitochondria. Here we show that mutations in both human and fly PINK1 result in higher levels of misfolded components of respiratory complexes and increase in markers of the mitochondrial unfolded protein response. Through the development of a genetic model of mitochondrial protein misfolding employing Drosophila melanogaster, we show that the in vivo accumulation of an unfolded protein in mitochondria results in the activation of AMP-activated protein kinase-dependent autophagy and phenocopies of pink1 and parkin mutants. Parkin expression acts to clear mitochondria with enhanced levels of misfolded proteins by promoting their autophagic degradation in vivo, and refractory to Sigma P (ref(2)P), the Drosophila orthologue of mammalian p62, is a critical downstream effector of this quality control pathway. We show that in flies, a pathway involving pink1, parkin and ref(2)P has a role in the maintenance of a viable pool of cellular mitochondria by promoting organellar quality control.
Article
Two different methods of presenting quantitative gene expression exist: absolute and relative quantification. Absolute quantification calculates the copy number of the gene usually by relating the PCR signal to a standard curve. Relative gene expression presents the data of the gene of interest relative to some calibrator or internal control gene. A widely used method to present relative gene expression is the comparative CT method also referred to as the 2−ΔΔCT method. This protocol provides an overview of the comparative CT method for quantitative gene expression studies. Also presented here are various examples to present quantitative gene expression data using this method.
Article
Parkinson's disease is the second most common neurodegenerative disorder and is characterized by the degeneration of dopaminergic neurons in the substantia nigra. Mitochondrial dysfunction has been implicated as an important trigger for Parkinson's disease-like pathogenesis because exposure to environmental mitochondrial toxins leads to Parkinson's disease-like pathology
Article
Historically, mitochondrial reactive oxygen species (mROS) were thought to exclusively cause cellular damage and lack a physiological function. Accumulation of ROS and oxidative damage have been linked to multiple pathologies, including neurodegenerative diseases, diabetes, cancer, and premature aging. Thus, mROS were originally envisioned as a necessary evil of oxidative metabolism, a product of an imperfect system. Yet few biological systems possess such flagrant imperfections, thanks to the persistent optimization of evolution, and it appears that oxidative metabolism is no different. More and more evidence suggests that mROS are critical for healthy cell function. In this Review, we discuss this evidence following some background on the generation and regulation of mROS.
Article
Parkinson's disease is characterized and diagnosed by bradykinetic motor symptoms caused by the loss of dopamine neurons in the substantia nigra. The pathological and non-motor behavioral changes that occur prior to degeneration are less well characterized, although changes in gait, olfaction and cognition have been recognized in familial Parkinson's disease subjects. Gene mutations associated familial Parkinson's disease give rise to mitochondrial changes, altered energy homeostasis and intracellular trafficking deficits, and these can be modeled in transgenic mice. Here we discuss the recent finding of prodromal behavioral disturbances in a PINK1 deficient mouse that manifest prior to dopaminergic cell death and correlate to 5-HT fiber losses and mitochondrial morphological changes. We discuss the representation of the PINK1 deficient mouse and other genetic models to accurately recapitulate early Parkinson's disease. Prodromal symptoms and underlying pathology modeled in mice and cell lines from human subjects may have wide implications for earlier diagnosis. Current and emerging therapies need to be tailored to target both early cognitive and late stage motor symptoms.
Article
We will review the recent advances in the genetics of Parkinson disease and other movement disorders such as dystonia, essential tremor and restless legs syndrome (RLS). Mutations in VPS35 were identified as a novel cause of autosomal dominant Parkinson disease using exome sequencing. Next generation sequencing (NGS) was also used to identify PRRT2 mutations as a cause of paroxysmal kinesigenic dyskinesia (DYT10). Using a different technique, that is linkage analysis, mutations in EIF4G1 were implicated as a cause of Parkinson disease and mutations in SLC20A2 as a cause of familial idiopathic basal ganglia calcification. Furthermore, genome-wide association studies (GWAS) and meta-analyses have confirmed known risk genes and identified new risk loci in Parkinson disease, RLS and essential tremor. New models to study genetic forms of Parkinson disease, such as stem cell-derived neurons, have helped to elucidate disease-relevant molecular pathways, such as the molecular link between Gaucher disease and Parkinson disease. New genes have been implicated in Parkinson disease and other movement disorders through the use of NGS. The identification of risk variants has been facilitated by GWAS and meta-analyses. Furthermore, new models are being developed to study the molecular mechanisms involved in the pathogenesis of these diseases.
Article
Neuronal survival critically depends on the integrity and functionality of mitochondria. A hierarchical system of cellular surveillance mechanisms protects mitochondria against stress, monitors mitochondrial damage and ensures the selective removal of dysfunctional mitochondrial proteins or organelles. Mitochondrial proteases emerge as central regulators that coordinate different quality control (QC) pathways within an interconnected network of mechanisms. A failure of this system causes neuronal loss in a steadily increasing number of neurodegenerative disorders, which include Parkinson's disease, spinocerebellar ataxia, spastic paraplegia and peripheral neuropathies. Here, we will discuss the role of the mitochondrial QC network for neuronal survival and neurodegeneration.
Article
Parkinson's Disease (PD) is the most common neurodegenerative movement disorder. Autosomal-recessive mutations in the mitochondrial protein kinase PINK1 (PTEN-induced kinase 1) account for 1-2% of the hereditary early-onset cases. To study the mechanisms underlying disease development, we generated Pink1-deficient mice. In analogy to other genetic loss-of-function mouse models, Pink1(-/-) mice did not show morphological alterations in the dopaminergic system. As a consequence, no gross motor dysfunctions were observed indicating that these mice do not develop the cardinal symptoms of PD. Nonetheless, symptoms which develop mainly before bradykinesia, rigidity and resting tremor were clearly evident in Pink1-deficient mice. These symptoms were gait alterations and olfactory dysfunctions. Remarkably in the glomerular layer of the olfactory bulb the density of serotonergic fibers was significantly reduced. Concerning mitochondrial morphology, neurons in Pink1(-/-) mice had less fragmented mitochondria. In contrast, upon acute knock-down of Pink1 increased mitochondrial fragmentation was observed in neuronal cultures. This fragmentation was, however, evened out within days. Taken together, we demonstrate that Pink1-deficient mice exhibit behavioral symptoms of early phases of PD and present systematic experimental evidence for compensation of Pink1-deficiency at the cellular level. Thus, Pink1-deficient mice represent a model for the early phases of PD in which compensation may still impede the onset of neurodegeneration. Consequently, these mice are a valuable tool for studying Pink1-related PD development, as well as for searching for reliable PD biomarkers.
Article
Many neurodegenerative diseases demonstrate abnormal mitochondrial morphology and biochemical dysfunction. Alterations are often systemic rather than brain-limited. Mitochondrial dysfunction may arise as a consequence of abnormal mitochondrial DNA, mutated nuclear proteins that interact directly or indirectly with mitochondria, or through unknown causes. In most cases it is unclear where mitochondria sit in relation to the overall disease cascades that ultimately causes neuronal dysfunction and death, and there is still controversy regarding the question of whether mitochondrial dysfunction is a necessary step in neurodegeneration. In this chapter we highlight and catalogue mitochondrial perturbations in some of the major neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). We consider data that suggest mitochondria may be critically involved in neurodegenerative disease neurodegeneration cascades.