ArticlePDF Available

Differential effects of PINK1 nonsense and missense mutations on mitochondrial function and morphology

Authors:

Abstract and Figures

Mutations of the PINK1 gene are a cause of autosomal recessive Parkinson's disease (PD). PINK1 encodes a mitochondrial kinase of unknown function which is widely expressed in both neuronal and non-neuronal cells. We have studied fibroblast cultures from four family members harbouring the homozygous p.Q456X mutation in PINK1, three of their wild-type relatives, one individual with the homozygous p.V170G mutation and five independent controls. Results showed bioenergetic abnormalities involving decreased activities of complexes I and IV along with increased activities of complexes II and III in the missense p.V170G mutant. There were increased basal levels of mitochondrial superoxide dismutase in these cells and an exaggerated increase of reduced glutathione in response to paraquat-induced free radical formation. Furthermore, swollen and enlarged mitochondria were observed in this sample. In the p.Q456X nonsense mutants, the respiratory chain enzymes were unaffected, but ATP levels were significantly decreased. These results confirm that mutations of PINK1 cause abnormal mitochondrial morphology, bioenergetic function and oxidative metabolism in human tissues but suggest that the biochemical consequences may vary between mutations.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
Differential effects of PINK1 nonsense and missense mutations on mitochondrial
function and morphology
A. Grünewald
a,b
, M.E. Gegg
c
, J.-W. Taanman
c
, R.H. King
c
, N. Kock
a,b
, C. Klein
a,b
, A.H.V. Schapira
c,
a
Department of Neurology, Lübeck University, Lübeck, Germany
b
Department of Human Genetics, Lübeck University, Lübeck, Germany
c
University Department of Clinical Neurosciences, Institute of Neurology, University College London, Rowland Hill Street, London NW3 2PF, UK
abstractarticle info
Article history:
Received 30 January 2009
Revised 21 May 2009
Accepted 22 May 2009
Available online 3 June 2009
Keywords:
PINK1
Parkinson's disease
Mitochondria
Respiratory chain
Oxidative stress
Complex I
Mutations of the PINK1 gene are a cause of autosomal recessive Parkinson's disease (PD). PINK1 encodes a
mitochondrial kinase of unknown function which is widely expressed in both neuronal and non-neuronal
cells. We have studied broblast cultures from four family members harbouring the homozygous p.Q456X
mutation in PINK1, three of their wild-type relatives, one individual with the homozygous p.V170G
mutation and ve independent controls. Results showed bioenergetic abnormalities involving decreased
activities of complexes I and IV along with increased activities of complexes II and III in the missense p.
V170G mutant. There were increased basal levels of mitochondrial superoxide dismutase in these cells and
an exaggerated increase of reduced glutathione in response to paraquat-induced free radical formation.
Furthermore, swollen and enlarged mitochondria were observed in this sample. In the p.Q456X nonsense
mutants, the respiratory chain enzymes were unaffected, but ATP levels were signicantly decreased. These
results conrm that mutations of PINK1 cause abnormal mitochondrial morphology, bioenergetic function
and oxidative metabolism in human tissues but suggest that the biochemical consequences may vary
between mutations.
© 2009 Elsevier Inc. All rights reserved.
Introduction
Parkinson's disease (PD) is characterised clinically by bradykinesia,
tremor, and rigidity. The aetiology of PD is varied (Warner and
Schapira, 2003), and an increasing proportion of patients, albeit still a
minority, have an identiable gene mutation (Klein and Schlossma-
cher, 2006). The biochemical mechanisms by which these single gene
mutations cause cell dysfunction and death provide valuable insight
into the pathogenesis of PD.
Recessive mutations in PTEN-induced putative kinase 1 (PINK1,
GenBank accession no. AB053323) are responsible for a familial form
of early-onset parkinsonism, previously mapped to chromosome 1p36
(the PARK6 locus). The protein product of PINK1 is ubiquitously
transcribed and encodes a kinase predominantly localised to mito-
chondria, but also expressed in the cytosol (Haque et al., 2008;
Valente et al., 2004).
Mitochondrial dysfunction has long been implicated in the
pathogenesis of PD. Evidence rst emerged when 1-methyl-4-
phenyl-1,2,3,4-tetrahydropyridine (MPTP), an environmental toxin,
was discovered to produce parkinsonian features in drug abusers
(Langston et al., 1983). This nding was further supported by the
detection of a complex I deciency in the substantia nigra of PD
patients (Schapira et al., 1989). Subsequently, complex I dysfunction
has also been observed in platelets (Benecke et al., 1993; Krige et al.,
1992; Parker et al., 1989) as well as in broblasts of patients (Hoepken
et al., 2007; Winkler-Stuck et al., 2004).
The complex I deciency is thought to result in increased free
radical production, and it contributes to the oxidative mediated
damage seen in the PD nigra (Schapira, 2008). This relationship is bi-
directional. The enhanced release of free radicals causes a decrease in
the activity of the respiratory chain enzymes, particularly of
complexes I and IV (Schapira, 2006). The mitochondrial deciency
can contribute to the dysfunction of the energy-dependent ubiquitin
proteasomal system (UPS) and to enhanced dopaminergic cell
damage and death (Hoglinger et al., 2003).
Preliminary data suggest that PINK1 may play a role in
protecting cells against stress conditions that affect the mitochon-
drial membrane potential (Valente et al., 2004). Recent mitochon-
drial studies on patient cells support these ndings but different
mutation types appear to induce different functional consequences.
Hoepken et al. observed elevated levels of oxidative stress and a
mild decrease in complex I activity in lymphoblasts from three
patients homozygous for the p.G309D PINK1 missense mutation
(Hoepken et al., 2007). In contrast, Piccoli et al. studied broblasts
Experimental Neurology 219 (2009) 266273
Corresponding author.
E-mail address: a.schapira@medsch.ucl.ac.uk (A.H.V. Schapira).
0014-4886/$ see front matter © 2009 Elsevier Inc. All rights reserved.
doi:10.1016/j.expneurol.2009.05.027
Contents lists available at ScienceDirect
Experimental Neurology
journal homepage: www.elsevier.com/locate/yexnr
from a PD patient with the p.W437X nonsense mutation and observed
unaltered levels of the antioxidant glutathione but a decrease in the
superoxide dismutase activity. The enzyme activities of complexes
IIV were unaffected, however ATP-ase activity was reduced (Piccoli
et al., 2008).
Similar differences emerged when the mitochondrial morphology
was investigated in mutant PINK1 cell models and patient broblasts.
Swollen and ruptured mitochondria were observed in Drosophila
PINK1 null mutants (Clark et al., 2006; Park et al., 2006; Poole et al.,
2008; Yang et al., 2006) and in human missense mutant broblasts (p.
Q126P and p.G309D PINK1) (Exner et al., 2007). In contrast, no
structural changes were seen in PD patient cells with the p.W437X
PINK1 mutation (Piccoli et al., 2008).
Here, we investigated the consequences of the PINK1 p.Q456X
nonsense and the p.V170G missense mutations on mitochondrial
function and morphology, and on free radical metabolism in broblast
cultures from patients with PD.
Materials and methods
PD patients with mutations in PINK1 and controls
Family W consists of three generations. Among the members there
were four patients with a homozygous c.1366CNT, p.Q456X nonsense
mutation (three female, mean age: 66.8 years [±4.8], mean age at
onset: 50.0 years [±9.3]). They had an average Hoehn and Yahr score
of 2.6 [± 0.5] and an average motor Unied Parkinson's Disease Rating
Scale (UPDRS) III score of 30.8 [± 4.4]. Three healthy family members
were also included (three female, mean age: 44.6 [± 10.0]) (for details
see Hedrich et al., 2006).
In addition, one patient (female, age: 70 years, age at onset:
31 years) harbouring a homozygous missense mutation c.509TNG, p.
V170G was studied. For this case, a Hoehn and Yahr score of 3.0 and an
UPDRS III score of 35.5 was determined (see also Moro et al., 2008). No
relatives of this individual were available.
Five independent age-matched healthy individuals of British origin
(C3, 4, 5, 7 and D5) served as additional controls (one female, mean
age: 64.8 years [±13.1]).
The investigated wild-type members of Family W and the
independent controls did not show signicant differences in any of
the performed experiments, therefore they were combined in the
following analyses. All control subjects were examined and found to
be free of any signs of PD.
Phenotypic features of all individuals, investigated in this study,
are summarised in Table 1.
Tissue culture
Primary broblasts cultures were established from skin biopsies.
Fibroblasts were cultured in high glucose (4.5 g/l) or 0.9 g/l galactose
Dulbecco's Modied Eagle's Medium (DMEM; Invitrogen) supple-
mented with 10% foetal bovine serum (BioSera) and 1 mM sodium
pyruvate (Sigma) at 37 °C, 5% CO
2
. The medium was changed weekly.
To mimic conditions of oxidative stress, cells were treated with
0.5 mM paraquat (PQ; Aldrich) for 48 h. The medium containing PQ
was refreshed after 24 h.
Real-time PCR: mitochondrial DNA levels
Gene dosage analysis was performed in a quantitative SYBR green
PCR on the LightCycler according to manufacturer's protocol (Roche
Diagnostics). The mitochondrial DNA concentration was measured
using primers against the mitochondrial displacement loop (D-loop)
(for sequences see Bai and Wong, 2005). The expression of the nuclear
single copy gene thymidine kinase 2 (TK2) was quantied as an
external standard (forward primer: 5-TCC TGC AGA TGC CAC TTT GA-
3; reverse primer: 5-CCC CAA GTC TGA AGA AAA CG-3). PCR
conditions were as follows: 95 °C for 15 min, 95 °C for 15 s, 60 °C for
30 s, 72 °C for 30 s (40 cycles); measurement of uorescence in each
cycle at 72 °C. The mitochondrial DNA level was measured at least
twice per broblast sample.
Protein determination
Sample protein concentration was determined by use of the
bicinchoninic acid (BCA) protein assay kit (Pierce) according to the
manufacturer's protocol. Sample protein concentration was detected
in duplicate and calculated from the bovine serum albumin standard
calibration curve (0150 0 μg/ml).
Mitochondrial enzyme assays and citrate synthase activity measurement
Fibroblasts were trypsinised and washed in PBS supplemented
with 1 mM phenylmethanesulphonyluoride (PMSF), 1 μg/ml of
leupeptin and 1 μg/ml of pepstatin A. Cells were then homogenised in
isolation medium (250 mM sucrose, 10 mM Tris, pH 7.4, 1 mM
ethylenediaminetetraacetic acid (EDTA)), nuclei and cell debris were
removed by centrifugation at 1500 ×g, and mitochondria were
isolated by centrifugation at 11,800 ×g. The mitochondrial pellet was
then resuspended in isolation medium. Mitochondrial respiratory
chain complex and citrate synthase (CS) activities were measured by
Table 1
Genotypic and phenotypic characterisation of investigated individuals.
Mutational status Code Sex Age (yr) Age of onset (yr) Mutation (homozygous) H-Y stage UPDRS III motor score (off)
Nonsense mutants (Family W) IP2123 M 69 39 p.Q456X 3.0 34.0
R2122 F 67 61 p.Q456X 2.0 27.0
R2124 F 60 53 p.Q456X 2.5 27.0
R2126 F 71 47 p.Q456X 3.0 35.0
Mean+/STD 66.8+/4.8 50.0 +/9.3 2.6 +/0.5 30.8 +/4.4
Missense mutant IP1703 F 70 31 p.V170G 3.0 35.5
Controls (Family W) R2132 F 35 None ––
R2133 F 46 None ––
R2134 F 42 None ––
Mean+/STD 44.6+/10.0
Controls (independent) C3 F 60 None ––
C4 M 73 None ––
C5 M 45 None ––
C7 M 79 None ––
D5 M 67 None ––
Mean+/STD 64.8+/13.1
The index patients are denoted by IP.Relatives of IP2123 are marked by an R. Given are the patients' ages at the time of their rst examination. Sex: F female, M male; other: H-Y
Hoehn and Yahr, STD standard deviation, UPDRS Unied Parkinson Disease Rating Scale.
267A. Grünewald et al. / Experimental Neurology 219 (2009) 266273
spectrophotometric methods in these mitochondrial preparations as
reported (Bolanos et al., 1995). Data were expressed as a ratio with
citrate synthase activity. The activities of each complex were
determined in 24 assays per individual.
Malate dehydrogenase assay
Fibroblasts were harvested 72 h after feeding with fresh culture
media and resuspended in isolation media. Malate dehydrogenase
(MDH) activity was determined spectrophotometrically according to
Lai and Clark (1976). Samples were measured in duplicate and activity
expressed as μmol/min/mg protein.
Blue native polyacrylamide gel electrophoresis (PAGE)
Preparation of blue native gels was performed on isolated
mitochondria according to Schagger and Pfeiffer (2000). All gels
were transferred to Hybond-P membranes (GE Healthcare). The
membranes were incubated with primary antibodies raised against
subunits of the respiratory chain enzyme complexes (mouse anti-
complex I NDUFA9, mouse anti-complex II SDHA, mouse anti-complex
III UQCRC2, mouse anti-complex IV MTCO2, mouse anti-complex V
ATP5A1; Mitosciences). This was followed by incubation with a
horseradish peroxidase (HRP)-conjugated secondary mouse antibody
(Dako) and enhanced chemiluminescence (ECL) detection (Pierce).
Mitochondrial membrane potential
The mitochondrial membrane potential (ψm) was analysed by
means of 5,5,6,6-tetrachloro-1,1,3,3-tetraethylbenzimidazolylcar-
bocyanine iodide (JC-1, Invitrogen). Fibroblasts (5× 10
4
cells per 12-
well) were treated with 1 μg/ml of JC-1 for 15 min at 37 °C. The cells
were washed with phosphate-buffered saline (PBS), and mitochon-
drial JC-1 aggregates were measured with a uorescent plate reader
(excitation 530 nm, emission 590 nm). In a sister well, JC-1
uorescence was measured in the presence of the ionophore
valinomycin (100 nM), which destroys the mitochondrial membrane
potential, and was subtracted from the data. JC-1 uorescence was
expressed against protein.
Cellular ATP levels
ATP levels were determined with the ATP Bioluminescence
Assay Kit HS II (Roche). Prior to the measurement with a Jade
luminometer (Labtech International), the samples were corrected
for cell number. The experiment was performed with 1× 10
5
cells
per ml of PBS. Each measurement was performed at least three
times per sample.
ATP synthesis
Fibroblasts were trypsinised and washed three times with ice cold
PBS. Cells were resuspended at 2×10
5
cells/ml in incubation medium
(25 mM Tris, 150 mM KCl, 2 mM K
+
-EDTA, 10 mM K
2
HPO4, pH 7.4). An
aliquot of cells (2 × 10
4
) was mixed with an equal volume of
incubation buffer containing 1 mg/ml bovine serum albumin, 1 mM
ADP and substrates (complexes I, III, IV: glutamate+ malate (10 μM);
complexes II, III, IV: succinate (10 μM) + rotenone (40 μg/ml);
complex IV: ascorbate (2 mM)+ N,N,N,N-tetramethyl-p-phenylene-
diamine (TMPD; 50 μM)), permeabilised with digitonin (40 μg/ml),
and incubated at 37 °C for 20 min. The reaction was stopped with
perchloric acid, and samples neutralised with 3 M K
2
CO
3
dissolved in
0.5 M tri-ethanolamine. Debris was removed by centrifugation and
ATP measured with the ATP Bioluminesence Assay kit HSII (Roche).
Data were expressed as pmoles ATP synthesised/min/10
5
cells and
represent means of two analyses per sample.
Western blotting
Cell lysates were separated by 12% sodium dodecyl sulfate (SDS)-
PAGE and protein transferred to Hybond-P membranes. Blots were
incubated with rabbit polyclonal anti-superoxide dismutase 2 (SOD2;
sc-30080, Santa Cruz Biotechnology) or mouse monoclonal anti-porin
31HL (clone 89-173/016, Calbiochem). The blots were then incubated
with HRP-conjugated secondary antibodies and bands detected by ECL.
Equal loading was assessed using an antibody against succinate dehy-
drogenase subunit (SDHA) of the electron transport chain (Mitos-
ciences) or glyceraldehyde 3-phosphate dehydrogenase (GAPDH;
Abcam). Band density was measured using Alpha DigiDoc software.
For quantitative studies three separate western blots were analysed.
Cellular reduced glutathione (GSH) levels
The levels of GSH in broblasts were determined by trypsinising the
cells, resuspending in isolation medium and extracting into 15 mM ortho-
phosphoric acid. The cellular GSH concentration was then measured by
reverse-phase HPLC and electrochemical detection (Riederer et al., 1989).
Oxyblot
Protein carbonyl levels were measured in broblasts using the
Oxyblot Protein Oxidation Detection kit (Millipore). Briey, cells were
lysed with 0.25% (v/v) Triton X-100 in PBS supplemented with 50 mM
dithiotreitol and protease inhibitors. Following removal of insoluble
material, protein carbonyls were derivatised to 2,4-dinitrophenylhy-
drazone (DNP) by reaction with 2,4-dinitrophenylhydrazine. The
DNP-derivatised protein samples were then separated by SDS PAGE
followed by western blotting. Carbonyls were detected by incubation
with an antibody specic to DNP. Equal loading was determined by
reprobing the same blot with GAPDH antibody.
MitoTracker Green staining of broblasts
The mitochondrial network in live broblasts was assessed with
MitoTracker Green FM (Invitrogen). Cells were cultured on coverslips
for 72 h in media containing either glucose (4.5 g/l) or galactose
(0.9 g/l). Cells were then incubated with 5 μg/ml 4-6-diamidino-2-
phenylindole (DAPI) for 2 h in the respective media. The cells were
then loaded with 250 nM MitoTracker Green FM in their respective
culture media for 45 min at 37 °C.
Electron microscopy
Cells of one 80% conuent T-175 ask were harvested and the pellet
was treated as described by Kinget al. (2002). Electron microscopy was
performed on ultrathin sections of four p.Q456X mutants, the p.V170G
mutant and ve controls. Sections were contrasted with uranyl acetate
and lead citrate. Images were captured with a digital camera attached
to a Zeiss 902C electron microscope. Images were analysed blinded.
Statistical analysis
A MannWhitney-Utest (nonparametric) was applied to evaluate
the statistical signicance of differences measured in the different
data-sets. Values outside of the 99% conference interval of the control
group were dened as outliers in the assays.
Results
Mitochondrial respiratory chain enzyme complexes
To test for the effects of PINK1 mutations on mitochondrial
function, the activities of respiratory chain enzyme complexes were
268 A. Grünewald et al. / Experimental Neurology 219 (2009) 266273
investigated. No difference in complex I activity was found when
comparing four homozygous PINK1 nonsense mutants with six
controls. In the culture of the patient with a PINK1 missense mutation
the complex I activity was reduced, although not signicantly, to 81%
of the median control level (Fig. 1A).
For the combined activity measurement of complexes II+III there
was no difference between the nonsense mutants and the control
samples. However, in the missense mutant, the complex II+III
activity was markedly higher (231%) than the median control level
(Fig. 1B).
The activity of complex IV in the sample with the missense
mutation was decreased to 54% of the median control activity. The
median activity in the nonsense mutants was indistinguishable from
the activity in the controls (Fig. 1C).
To investigate if the detected changes in the respiratory chain
complex activities were a result of changes in expression levels, the
assembly of the respiratory chain enzyme complexes was investigated
by blue native PAGE. The western blot analysis of complexes I to V
displayed bands of expected molecular weight. No additional bands
indicative of complex subassemblies were detected (Fig. 1D). When
comparing the expression of the complexes, no overt changes were
observed.
Mitochondrial content and mtDNA levels
The activities of the citric acid cycle (CAC) enzymes CS (data not
shown) and MDH (median MDH
controls
: 7.38 μg/min/mg, n=6;
median MDH
p.Q456X
: 5.81 μg/min/mg, n=4; MDH
p.V170G
: 6.91 μg/
min/mg, n=1) were similar in the cell lysates of control, nonsense
and missense cultures, indicating that the mitochondrial number was
comparable in all cultures. This nding was further supported by
similar levels of mitochondrial DNA in all broblast cultures (median
[mtDNA]
controls
: 72.71, n= 7; median [mtDNA]
p.Q456X
:70.10,n=4;
[mtDNA]
p.V170G
: 64.52, n= 1).
Since complex II is a component of both the electron transport
chain and the CAC, the observation that both CS and MDH activities
Fig. 1. Activities and protein expression of respiratorychain enzyme complexes IIV. (A) Controls and patients with a nonsense mutation showed comparable complex I activities. The
activity in the missense mutation sample was at the lower limit of the control range. (B) In the p.V170G mutant elevated complex II + III activity was detected. The activity of the
missense mutant amounted to 231% of the median control activity. (C) The complex IV activity of the p.V170G mutant was 54% of the median control activity. The activities of
complexes IIV were normalised to CS activity. (D) Protein expression of the respiratory chain enzyme complexes. Western blot showing complex I to V for two controls, two
homozygous PINK1 p.Q456X mutants and the p.V170G mutant. Horizontal bars represent medians of the investigated groups of individuals. CIIV complex IIV activity, CS
citrate synthase activity, ⁎⁎ — value outside of the 99% condence interval of the control group.
269A. Grünewald et al. / Experimental Neurology 219 (2009) 266273
were unaffected, suggests that the increased activities of complexes
II+III (Fig. 1B) in the missense culture are a result of increased
respiratory chain activity rather than the CAC.
Mitochondrial membrane potential
The inuence of mutant PINK1 on ψm was investigated by means
of a cytouorimetric assay. Neither the nonsense nor the missense
mutation had a signicant effect on ψm (median ψm
controls
: 100%,
n=3; median ψm
p.Q456X
: 100% of the median control value, n=3;
ψm
p.V170G
: 79% of the median control value, n=1). PINK1 was
previously reported to function in the response to cellular stress
(Pridgeon et al., 2007; Wang et al., 2007). Therefore, the effect of
treatment with the free radical generator paraquat (0.5 mM) on ψm
was measured. Ψm was unaffected in control or mutant PINK1
broblasts following stress, when compared to basal conditions
(median ψm
controls
: 87% of the median basal control value, n=3;
median ψm
p.Q456X
: 106% of the median basal control value, n=3;
median ψm
p.V170G
: 74% of the median basal control value, n=1).
Cellular ATP content and synthesis
The total cellular ATP concentration was quantied in all availa-
ble patient broblast cultures and eight control cultures. The
experiment revealed a signicant decrease of 21% (pb0.05) in the
nonsense mutants (median level) when compared with the median
of the control individuals (Fig. 2A). The ATP concentration in cells
of the missense mutant patient did not differ from the average
control level.
Furthermore, we examined ATP synthesis in digitonin-permeabi-
lised broblasts via complexes I, III, IV (substrates: glutamate and
malate), complexes II, III, IV (succinate and rotenone) or complex IV
(ascorbate and TMPD) only. Measuring ATP production via complexes
I, III and IV the median activity in the missense mutant broblasts was
Fig. 2. Cellular ATP levels (A) and ATP synthesis (B). (A) Controls and the missense mutation sample showed comparable cellular ATP concentrations under basal conditions. In
patients with a nonsense mutation a signicantly reduced median ATP level was detected (pb0.05). Results are displayed relative to the median control level. (B) In the missense
mutant broblasts enhanced ATP synthesis was determined when measuring the activity by use of glutamate and malate (ATP generation via complexes I, III and IV; for details see
Materials and methods section), or succinate and rotenone (ATP generation via complexesII, III and IV) as substrates. Here, the PINK1 p.V170G sample was compared to four p.Q456X
cultures and ve controls. Horizontal bars represent medians of the investigated groups of individuals. Vertical lines indicate the 95% condence interval.⁎⁎ — value outside of the 99%
condence interval of the control group.
Fig. 3. Indicators of oxidative stress. (A) SOD2 protein expression under basal conditions (normalised to porin and the median value in the control group). The missense
mutant showed a markedly higher SOD2 expression level (284%) than seen in controls (100%). The median expression in the nonsense mutants was comparable with the
median control level (85%). (B) GSH levels under basal conditions and following paraquat treatment. For controls (100%), patients with the p.Q456X mutation (95%) and the
sample with the p.V170G mutation (95%) comparable median GSH levels were detected when measured under basal conditions. After treatment with 0.5 mM paraquat, all
samples showed a signicant increase in the cellular glutathione concentration. However, the missense mutant sample tended to show a greater increase (+134%) than the
other groups (controls: + 85%; p.Q456X mutants: + 60%). All data are expressed relative to the median of the control group under basal conditions. Horizontal bars represent
medians of the investigated groups of individuals. ⁎⁎ — value outside of the 99% condence interval of the control group.
270 A. Grünewald et al. / Experimental Neurology 219 (2009) 266273
71% higher than in controls and via complexes II, III and IV, the median
activity increased by 88%. However, no signicant differences in ATP
synthesis were observed when using ascorbate and TMPD as
substrates (Fig. 2B).
Oxidative stress in PINK1 mutant broblasts
SOD2 and GSH are important antioxidants, which participate in the
oxidant stress defence system. Whereas SOD2 can only be found in
mitochondria, GSH is additionally detectable in the cytosol. The
median cellular SOD2 level was determined relative to porin
expression by densitometric analysis of three western blots prepared
with cell lysates of all available mutant samples and six controls. SOD2
expression in the missense cells was markedly increased (284%). The
median SOD2 expression in the lysates of the nonsense mutation cells
was normal (85%) (Fig. 3A).
No signicant changes of the median GSH levels were found in cell
lysates of control, nonsense or missense PINK1 broblast cultures.
Fig. 4. Mitochondrial network in primary broblasts grown in glucose and galactose medium. MitoTracker Green FM was used to visualise the mitochondrial network in control,
nonsense mutant (p.Q456X) and missense (p.V170G) mutant broblasts. Cell nuclei are stained with DAPI.
Fig. 5. Mitochondrial morphology in broblasts. Transverse sections of control and PINK1 mutant broblast pellets were examined using electron microscopy. (A) The micrograph
shows mitochondria of normal size and structure (highlighted by arrow heads) in a control. (B) In the p.V170G sample mitochondria fusion was detected. Abnormal mitochondria are
marked by arrows. Scale bar 2 μm.
271A. Grünewald et al. / Experimental Neurology 219 (2009) 266273
Following treatment with paraquat, the median GSH levels were
elevated in control and nonsense cultures by 85% and 60%,
respectively. The median GSH level was elevated to a greater extent
(134%) in the missense culture (Fig. 3B).
Finally, the Oxyblot Protein Oxidation Detection kit from Millipore
was used for the immunoblot detection of carbonyl groups introduced
into proteins by oxidative reactions. There were no differences
between the three groups (data not shown).
Mitochondrial network
The mitochondrial networks in live control, nonsense and
missense broblast cultures were stained with Mitotracker Green.
When grown in glucose, a typical reticular structure was seen in all
cultures. Culturing of the cells in galactose media for 72 h had no effect
on the mitochondrial network in control cells. However, a noticeable
minority of cells exhibited a more fragmented mitochondrial network
in both nonsense (22%) and missense cells (19%), when cultured in
galactose media (Fig. 4).
Electron microscopy studies
To study the mitochondrial morphology in the cells, pellets of
four controls, the four nonsense mutant samples and the missense
mutant sample were prepared for electron microscopy and
assessed blind. In the PINK1 p.V170G mutant broblasts a small
subset of mitochondria exhibited abnormal morphology when
compared with controls. The respective mitochondria in the missense
sample were swollen and noticeably enlarged with abnormal cristae
(Fig. 5).
Discussion
In this paper we have investigated the mitochondrial function in
broblasts containing homozygous nonsense (p.Q456X) or missense
(p.V170G) PINK1 mutations. The p.Q456X mutation, which is causing
nonsense-mediated mRNA decay (Grünewald et al., 2007), probably
leads to a loss of protein function in the four homozygous siblings
investigated here. However, this mutation had no effect on the
enzyme activities of the respiratory chain in these samples. This result
is in agreement with a previous report, which showed that another
PINK1 nonsense mutation (p.W347X) did not affect the activity of the
complexes of the electron transport chain when measured spectro-
photometrically. Although no decrease in complex IV activity was
detected spectrophotometrically in broblasts from this patient,
polarographic studies did indicate perturbed complex IV activity
(Piccoli et al., 2008).
In contrast, the missense mutant broblast culture exhibited
decreased activities of complexes I and IV, while the activity of
complexes II+ III was increased. These data lack statistical signi-
cance because only one p.V170G mutant broblast culture was
available for study and because of the wide range of control values,
a phenomenon that has been observed earlier (Williams et al., 2001).
Nevertheless, our results are in agreement with recently reported
ndings describing a link between reduced activity of complex IV and
loss of PINK1 activity (Gegg et al., 2009).
Despite the mitochondrial dysfunction observed in the missense
culture, mitochondrial membrane potential and steady-state ATP
levels were unaffected. In fact, the rate of ATP synthesis was increased
in the missense culture when electron donors feeding into the
respiratory chain at complexes I or II were used. This correlates with
the unexpected increase in the specic activity of complex II+III.
Since the activities of complexes I and IV appear decreased in these
cells, the respiratory chain activity via complexes II and/or III may be
increased to compensate, thereby maintaining the membrane poten-
tial, and subsequently ATP levels.
Steady-state ATP levels were signicantly decreased in the
nonsense cultures, although no loss of respiratory chain function or
ATP synthesis was detected in these cells. The lower ATP levels may be
areection of increased ATP consumption in the cell (e.g. main-
tenance of the mitochondrial membrane potential, increased activity
of Na
+
/K
+
transporters). Indeed, mitochondrial membrane potential
has been shown to be maintained in cells lacking PINK1 expression by
complex V hydrolysing ATP (Gandhi et al., 2009).
Recently, PINK1 deciency has been reported to cause mitochon-
drial accumulation of calcium in mammalian neurons, resulting in a
mitochondrial calcium overload which then stimulates the production
of reactive oxygen species (ROS) via NADPH oxidase (Gandhi et al.,
2009). Increased SOD2 expression levels in mitochondria of the
missense culture are most likely indicative of a cellular defence
mechanism against elevated ROS production. Similar effects have also
been detected earlier in broblasts harbouring PINK1 mutations
(Hoepken et al., 2007; Piccoli et al., 2008). Baseline GSH levels were
unaffected in both the missense and nonsense cultures and this
correlated with no detectable difference in the levels of oxidised
proteins.
GSH levels can be elevated in cells following exogenous oxidative
stress (Gegg et al., 2003; Moellering et al., 1999). Following paraquat
treatment, GSH levels were increased in control and nonsense cultures
to a similar extent, while the increase was much greater in the
missense culture. This result strengthens the idea that antioxidant
pathways have been activated in the missense culture to deal with the
oxidative stress induced by defective mitochondria.
The reason for altered mitochondrial function in the missense
culture is unclear. Studies have indicated that PINK1 function is
required to maintain the correct mitochondrial structure (Deng et al.,
2008; Exner et al., 2007; Poole et al., 2008; Yang et al., 2008). The
decreased activities of complexes I and IV in the missense broblasts
may be a direct result of decreased PINK1 activity, or a consequence of
the abnormal mitochondrial morphology seen in these cells. Culturing
both the nonsense and missense broblasts in galactose appears to
exacerbate PINK1-mediated changes in the mitochondrial network. A
similar phenomenon has also been reported in broblasts with
different PINK1 mutations (Exner et al., 2007). The use of galactose as
the major carbon source means that the cell predominantly relies on
oxidative phosphorylation for ATP synthesis (Robinson et al., 1992). It
is therefore expected that respiratory chain dysfunction as a result of
mutated PINK1 will be amplied under these conditions. Indeed, ATP
levels have been shown to be lower in PINK1 broblasts containing a
nonsense mutation only when they are cultured in galactose media
(Piccoli et al., 2008).
In summary, broblasts containing mutated PINK1 resulted in
mitochondrial dysfunction, with the missense PINK1 mutation in
particular having an effect on the respiratory chain. In support of
previous studies we have also found alterations in mitochondrial
morphology as a result of loss of PINK1 activity. The observation that
the missense mutation appears to be more pathogenic than the
nonsense mutation is intriguing. It should be noted that the PD patient
with the missense mutation had both an earlier age of onset and a
more severe clinical condition than the four patients with the
nonsense mutation. However, it remains to be determined in a larger
sample whether there may be a correlation between the severity of
clinical ndings and the degree of mitochondrial dysfunction seen in
the patients' cells. In addition, it is conceivable that the signicantly
longer duration of disease in the patient with the missense mutation
may have a possible impact on the mitochondrial integrity and
bioenergetic properties. Although this study on mitochondrial func-
tion comprises the largest number of broblast cultures from PD
patients and control individuals investigated to date, the results
received from the single p.V170G PINK1 missense mutant sample will
need further conrmation. To obtain a complete picture of the
situation on PINK1 missense and nonsense mutant backgrounds,
272 A. Grünewald et al. / Experimental Neurology 219 (2009) 266273
cellular samples from PD families with a large number of affected and
unaffected individuals will be required.
Acknowledgments
This study was supported by the Parkinson's Disease Society UK,
the Kattan Trust, the Fritz Thyssen Foundation, the DFG and the DAAD.
We are grateful to Prof. Simon Heales and Dr. Lee Stanyer (Institute of
Neurology, London) for allowing the use of the reverse phase HPLC to
measure GCH levels.
References
Bai, R.K., Wong, L.J., 2005. Simultaneous detection and quantication of mitochondrial
DNA deletion(s), depletion, and over-replication in patients with mitochondrial
disease. J. Mol. Diagn. 7, 613622.
Benecke, R., Strumper, P., Weiss, H., 1993. Electron transfer complexes I and IV of
platelets are abnormal in Parkinson's disease but normal in Parkinson-plus
syndromes. Brain 116 (Pt 6), 14511463.
Bolanos, J.P., Heales, S.J., Land, J.M., Clark, J.B., 1995. Effect of peroxynitrite on the
mitochondri al respiratory c hain: differential susc eptibility of neurones and
astrocytes in primary culture. J. Neurochem. 6 4, 19651972.
Clark, I.E., Dodson, M.W., Jiang, C., Cao, J.H., Huh, J.R., Seol, J.H., Yoo, S.J., Hay, B.A., Guo,
M., 2006. Drosophila pink1 is required for mitochondrial function and interacts
genetically with parkin. Nature 441, 11621166.
Deng, H., Dodson, M.W., Huang, H., Guo, M., 2008. The Parkinson's disease genes pink1
and parkin promote mitochondrial ssion and/or inhibit fusion in Drosophila. Proc.
Natl. Acad. Sci. U. S. A. 105, 1450314508.
Exner, N., Treske,B., Paquet,D., Holmstrom,K., Schiesling, C., Gispert,S., Carballo-Carbajal,I.,
Berg, D., Hoepken, H.H., Gasser, T., Kruger, R., Winklhofer, K.F., Vogel, F., Reichert, A.S.,
Auburger, G., Kahle, P.J., Schmid, B., Haass, C., 2007. Loss-of-function of human PINK1
results in mitochondrial pathology and can be rescued by parkin. J. Neurosci. 27,
1241312418.
Gandhi, S., Wood-Kaczmar, A., Yao,Z., Plun-Favreau, H., Deas, E., Klupsch, K., Downward,
J., Latchman, D.S., Tabrizi, S.J., Wood, N.W., Duchen, M.R., Abramov, A.Y., 2009.
PINK1-associated Parkinson's disease is c aused by neuronal vulnerability to
calcium-induced cell death. Mol. Cell 33, 627638.
Gegg, M.E., Beltran, B., Salas-Pino, S., Bolanos, J.P., Clark, J.B., Moncada, S., Heales, S.J.,
2003. Differential effect of nitric oxide on glutathione metabolism and mitochon-
drial function in astrocytes and neurones: implications for neuroprotection/
neurodegeneration? J. Neurochem. 86, 228237.
Gegg, M.E., Cooper, J.M., Schapira, A.H., Taanman, J.W., 2009. Silencing of PINK1
expression affects mitochondrial DNA and oxidative phosphorylation in dopami-
nergic cells. PLoS ONE 4, e4756.
Grünewald, A., Breedveld, G.J., Lohmann-Hedrich, K., Rohe, C.F., König, I.R., Hagenah, J.,
Vanacore, N., Meco, G., Antonini, A., Goldwurm, S., Lesage, S., Durr, A., Binkofski, F.,
Siebner, H., Münchau, A., Brice, A., Oostra, B.A., Klein, C., Bonifati, V., 2007. Biological
effects of the PINK1 c.1366CNT mutation: implications in Parkinson disease
pathogenesis. Neurogenetics 8, 103109.
Haque, M.E., Thomas, K.J., D'Souza, C., Callaghan, S., Kitada, T., Slack, R.S., Fraser, P.,
Cookson, M.R., Tandon, A., Park, D.S., 2008. Cytoplasmic Pink1 activity protects
neurons from dopaminergic neurotoxin MPTP. Proc. Natl. Acad. Sci. U. S. A. 105,
1716 172 1.
Hedrich, K., Hagenah, J., Djarmati, A., Hiller, A., Lohnau, T., Lasek, K., Grunewald, A.,
Hilker, R., Steinlechner, S., Boston, H., Kock, N., Schneider-Gold, C., Kress, W.,
Siebner, H., Binkofski, F.,Lencer, R., Munchau, A., Klein, C., 20 06. Clinical spectrum of
homozygous and heterozygous PINK1 mutations in a large German family with
Parkinson disease: role of a single hit? Arch. Neurol. 63, 833838.
Hoepken, H.H., Gispert, S., Morales, B., Wingerter, O., Del Turco, D., Mulsch, A.,
Nussbaum, R.L., Muller, K., Drose, S., Brandt, U., Deller, T., Wirth, B., Kudin, A.P., Kunz,
W.S., Auburger, G., 2007. Mitochondrial dysfunction, peroxidation damage and
changes in glutathione metabolism in PARK6. Neurobiol. Dis. 25, 401411.
Hoglinger, G.U., Carrard, G., Michel, P.P., Medja, F., Lombes, A., Ruberg, M., Friguet, B.,
Hirsch, E.C., 2003. Dysfunction of mitochondrial complex I and the proteasome:
interactions between two biochemical decits in a cellular model of Parkinson's
disease. J. Neurochem. 86, 12971307.
King, R.H., Muddle, J.R., Nourallah, M., Wong, J., Workman, J.M., Thomas, P.K., 2002.
Peripheral nerve regeneration in galactosaemic rats. Neuropathol. Appl. Neurobiol.
28, 381389.
Klein, C., Schlossmacher, M., 2006. The genetics of Parkinson disease. Nat. Clin. Pract.
Neurol. 2, 136146.
Krige, D., Carroll, M.T., Cooper, J.M., Marsden, C.D., Schapira, A.H., 1992. Platelet
mitochondrial function in Parkinson's disease. The Royal Kings and Queens
Parkinson Disease Research Group. Ann. Neurol. 32, 782788.
Lai, J.C., Clark, J.B., 1976. Preparation and properties of mitochondria derived from
synaptosomes. Biochem. J. 154, 423432.
Langston, J.W., Ballard, P., Tetrud, J.W., Irwin, I., 1983. Chronic Parkinsonism in humans
due to a product of meperidine-analog synthesis. Science 219, 979980.
Moellering, D., Mc Andrew, J., Patel, R.P., Forman, H.J., Mulcahy, R.T.,Jo, H., Darley-Usmar,
V.M., 1999. The induction of GSH synthesis by nanomolar concentrations of NO in
endothelial cells: a role for gamma-glutamylcysteine synthetase and gamma-
glutamyl transpeptidase. FEBS Lett. 448, 292296.
Moro, E., Volkmann, J., Konig, I.R., Winkler, S., Hiller, A., Hassin-Baer, S., Herzog, J.,
Schnitzler, A., Lohmann, K., Pinsker, M.O., Voges, J., Djarmatic, A., Seibler, P., Lozano,
A.M., Rogaeva, E., Lang, A.E., Deuschl, G., Klein, C., 2008. Bilateral subthalamic
stimulation in Parkin and PINK1 parkinsonism. Neurology 70, 1186119 1.
Park, J., Lee, S.B., Lee, S., Kim, Y., Song, S., Kim, S., Bae, E., Kim, J., Shong, M., Kim, J.M.,
Chung, J., 2006 . Mitochondrial dysfunction in Drosophila PINK1 mu tants is
complemented by parkin. Nature 441, 11571161.
Parker Jr., W.D., Boyson, S.J., Parks, J.K., 1989. Abnormalities of the electron transport
chain in idiopathic Parkinson's disease. Ann. Neurol. 26, 719723.
Piccoli, C., Sardanelli, A., Scrima, R., Ripoli, M., Quarato, G., D'Aprile, A., Bellomo, F.,
Scacco, S., De Michele, G., Filla, A., Iuso, A., Boffoli, D., Capitanio, N., Papa, S., 2008.
Mitochondrial respiratory dysfunction in familiar parkinsonism associated with
PINK1 mutation. Neurochem. Res. 33, 25652574.
Poole, A.C., Thomas, R.E., Andrews, L.A., McBride, H.M., Whitworth, A.J., Pallanck, L.J.,
2008. The PINK1/Parkin pathway regulates mitochondrial morphology. Proc. Natl.
Acad. Sci. U. S. A. 105, 16381643.
Pridgeon, J.W., Olzmann, J.A., Chin, L.S., Li, L., 2007. PINK1 protects against oxidative
stress by phosphorylating mitochondrial chaperone TRAP1. PLoS Biol. 5, e172.
Riederer, P., Konradi, C., Hebenstreit, G., Youdim, M.B., 1989. Neurochemical perspec-
tives to the function of monoamine oxidase. Acta Neurol. Scand. Suppl. 126, 4145.
Robinson, B.H., Petrova-Benedict, R., Buncic, J.R., Wallace,D.C., 1992. Nonviabilityof cells
with oxidative defects in galactose medium: a screening test for affected patient
broblasts. Biochem. Med. Metab. Biol. 48, 122126.
Schagger, H., Pfeiffer, K., 2000. Supercomplexes in the respiratory chains of yeast and
mammalian mitochondria. Embo J. 19, 17771783.
Schapira, A.H., 2006. Mitochondrial disease. Lancet 368, 7082.
Schapira, A.H., 2008. Mitochondria in the aetiology and pathogenesis of Parkinson's
disease. Lancet Neurol. 7, 97109.
Schapira, A.H., Cooper, J.M., Dexter, D., Jenner, P., Clark, J.B., Marsden, C.D., 1989.
Mitochondrial complex I deciency in Parkinson's disease. Lancet 1, 1269.
Valente, E.M., Abou-Sleiman, P.M., Caputo, V., Muqit, M.M., Harvey, K., Gispert, S., Ali, Z.,
Del Turco, D., Bentivoglio, A.R., Healy, D.G., Albanese, A., Nussbaum, R., Gonzalez-
Maldonado, R., Deller, T., Salvi, S., Cortelli,P., Gilks, W.P., Latchman, D.S., Harvey, R.J.,
Dallapiccola, B., Auburger, G., Wood, N.W., 2004. Hereditary early-onset Parkinson's
disease caused by mutations in PINK1. Science 304, 11581160.
Wang, H.L., Chou, A.H., Yeh, T.H., Li, A.H., Chen, Y.L., Kuo, Y.L., Tsai, S.R., Yu, S.T., 2007.
PINK1 mutants associated with recessive Parkinson's disease are defective in
inhibiting mitochondrial release of cytochrome c. Neurobiol. Dis. 28, 216226.
Warner, T.T., Schapira, A.H., 2003. Genetic and environmental factors in the cause of
Parkinson's disease. Ann. Neurol. 53 (Suppl. 3), S16S23 discussion S2325.
Williams, S.L., Scholte, H.R., Gray, R.G., Leonard, J.V., Schapira, A.H., Taanman, J.W., 2001.
Immunological phenotyping of broblast cultures from patients with a mitochon-
drial respiratory chain decit. Lab. Invest. 81, 10691077.
Winkler-Stuck, K., Wiedemann, F.R., Wallesch, C.W., Kunz, W.S., 2004. Effect of
coenzyme Q10 on the mitochondrial function of skin broblasts from Parkinson
patients. J. Neurol. Sci. 220, 4148.
Yang, Y., Gehrke, S., Imai, Y., Huang, Z., Ouyang, Y., Wang, J.W., Yang, L., Beal, M.F., Vogel,
H., Lu, B., 2006. Mitochondrial pathology and muscle and dopaminergic neuron
degeneration caused by inactivation of Drosophila Pink1 is rescued by Parkin. Proc.
Natl. Acad. Sci. U. S. A. 103, 1079310798.
Yang, Y., Ouyang, Y., Yang, L., Beal, M.F., McQuibban, A., Vogel, H., Lu, B., 2008. Pink1
regulates mitochondrial dynamics through interaction with the ssion/fusion
machinery. Proc. Natl. Acad. Sci. U. S. A.105, 70707075.
273A. Grünewald et al. / Experimental Neurology 219 (2009) 266273
... It is reported that loss of PINK1 results in increased reactive oxygen species (ROS) levels, enhanced lipid peroxidation and glutathione metabolism, defected complex I activity, reduced uptake of calcium, impaired ATP production, an elevated cytochrome C release in vitro [29][30][31] . Impaired mitochondrial function and altered morphology (swollen and enlarged mitochondria) have also been described in animal models and patient tissues [32][33][34][35] . Consistent with previous studies, we observed abnormal mitochondria in pink1 deficient zebrafish embryos. ...
Preprint
Full-text available
PTEN-induced putative kinase 1 (PINK1) is a well-characterized regulator of mitochondrial quality control through mitophagy and its mutations are associated with recessive Parkinson’s disease. However, little is known about its functions in normal and malignant hematopoiesis in vertebrates. Here we aim to unravel the roles of PINK1 in definitive hematopoiesis and its underlying mechanisms using zebrafish (Danio rerio). In this study, we utilized CRISPR/Cas9 system to generate pink1 knockout zebrafish model and PINK1-deficient leukemia cell line. We found that pink1 deficiency activated autophagy in hematopoietic cells and promoted definitive hematopoiesis in zebrafish embryos, which can be alleviated by canonical autophagy inhibition. Further, the proteomic and metabolic analysis revealed an elevated expression of cell proliferation markers and enhanced respiration in pink1-deficient zebrafish embryos. On the other hand, PINK1 deficiency also induced autophagy and cell proliferation in human leukemic cells. Therefore, our findings demonstrated that PINK1 functions as a negative regulator of normal and malignant hematopoiesis through the autophagy-mediated pathway.
... Elongated or enlarged mitochondria in iPSC-derived neurons and fibroblasts from PD patients with Parkin or PINK1 mutations (Bogetofte et al., 2019;Chung et al., 2016;Grunewald et al., 2009;Imaizumi et al., 2012) Excessive mitochondrial fission and fragmentation in iPSC-derived dopaminergic neurons from PD patients (LRRK2 G2019S, VPS35 D620N and SNCA A53T mutations) (Hanss et al., 2021;Nakamura et al., 2011;Ryan et al., 2018) Altered cristae morphology, increased mitochondrial diameter, dysregulated levels of mitochondrial shaping proteins Drp1, OPA1 and MFN1 in iPSC-derived dopaminergic neurons with PD-related GBA1 mutations (Schondorf et al., 2018) Fibroblasts from PD patients carrying parkin mutations exhibited longer and highly branched mitochondria (Mortiboys et al., 2008) Fibroblasts from PD patients carrying DJ-1 mutations exhibited decrease mitochondrial network and increase fragmentation (Krebiehl et al., 2010) Decreased levels of the short form of the fusion factor OPA1 in the substantia nigra of sporadic PD patients (Zilocchi et al., 2018) Microtubules destabilization in fibroblast from PD patients (Cartelli et al., 2012) iPSC-derived neuronal cells from PD patients (LRRK2 mutations) highlitght altered mitochondrial mobility in the axon (Cooper et al., 2012;Schwab et al., 2017) iPSC-derived dopaminergic neurons from PD patient (Parkin mutation) display microtubules fragmentation which precedes mitochondrial transport defects (Cartelli et al., 2018) iPSC-derived neuronal models with aberrant α-synuclein expression and oligomerization exhibit reduction of Miro1 and KCL1 proteins which leads to disrupted mitochondrial anterograde axonal transport (Prots et al., 2018) Huntington's disease (HD) ...
Article
Mitochondrial health is based on a delicate balance of specific mitochondrial functions (e.g. metabolism, signaling, dynamics) that are impaired in neurodegenerative diseases. Rescuing mitochondrial function by selectively targeting mitochondrial stressors, such as reactive oxygen species, inflammation or proteotoxic insults (“bottom-up” approaches) thus is a widely investigated therapeutic strategy. While successful in preclinical studies, these approaches have largely failed to show clear clinical benefits. Promoting the capacity of mitochondria - and other cellular components - to restore a healthy cellular environment is a promising complementary or alternative approach. Herein, we provide a non-technical overview for neurologists and scientists interested in brain metabolism on neuroprotective strategies targeting mitochondria and focus on top-down interventions such as metabolic modulators, exercise, dietary restriction, brain stimulation and conditioning. We highlight general conceptual differences to bottom-up approaches and provide hypotheses on how these mechanistically comparatively poorly characterized top-down therapies may work, discussing notably mitochondrial stress responses and mitohormesis.
... PINK1 genes are associated with production of mitochondrial proteins; and mutations in these genes can cause autosomal recessive PD (Rogaeva et al., 2004). Genetic mutations in PINK1 generated oxidative stress, mitochondrial inadequacies and ultimately neuronal apoptosis (Grünewald et al., 2009, Ibánez et al., 2006, Kumazawa et al., 2008. LARK2 gene (producing protein with kinase domain) mutation is a most general originator of familial form of PD (Zimprich et al., 2004, Alessi andSammler, 2018). ...
Chapter
Parkinson's disease (PD) is a neurodegenerative disorder prevalent in the elderly population worldwide. It is characterized by low dopamine levels secondary to the loss of dopaminergic neurons in the pars compacta of the substantia nigra; and deposition of abnormal protein aggregates known as Lewy bodies. Genetic mutations in α-synuclein, DJ-1, PINK1, LRRK2, parkin and chronic exposure to herbicides and pesticides are suspected in PD pathology. Mitochondrial dysfunction and oxidative stress are interdependent feedback mechanism of vicious cycle of neurodegeneration. Prolonged intake of L-Dopa is accompanied by unwanted side effect like dyskinesia. Nutraceuticals derived from maccuna and ginseng has been observed to mitigate neuroinflammation and mitochondrial dysfunction via modulation of NF-κB, mitochondrial homeostasis and MAPK signaling pathways. Soy, peanuts and Ginkgo biloba have all shown neuroprotective effects via the regulation of the JNK pathway whereas both epigallocatechin 3-gallate (EGCG) and resveratrol have shown similar neuroprotective effects through AMPK activation, regulation of mitochondrial biogenesis and modulation of SIRIT1, Bax and protein kinase signaling pathways. L-sulforaphane and allicin ameliorated neuronal apoptosis through inhibition of 8-iso-prostagandins and caspases 3. Polyphenolic nutraceuticals decreased oxidative stress via modulation of Nrf/Keap1, Nrf/ARE, P13K/AKT and MEK/ERK signaling cascades. This chapter is aimed to examine the neuroprotective properties of nutraceuticals as disease modifying therapeutic options via modulation of molecular mechanism in PD.
... PINK1 genes are associated with production of mitochondrial proteins; and mutations in these genes can cause autosomal recessive PD (Rogaeva et al., 2004). Genetic mutations in PINK1 generated oxidative stress, mitochondrial inadequacies and ultimately neuronal apoptosis (Grünewald et al., 2009, Ibánez et al., 2006, Kumazawa et al., 2008. LARK2 gene (producing protein with kinase domain) mutation is a most general originator of familial form of PD (Zimprich et al., 2004, Alessi andSammler, 2018). ...
... PINK1 genes are associated with production of mitochondrial proteins; and mutations in these genes can cause autosomal recessive PD (Rogaeva et al., 2004). Genetic mutations in PINK1 generated oxidative stress, mitochondrial inadequacies and ultimately neuronal apoptosis (Grünewald et al., 2009, Ibánez et al., 2006, Kumazawa et al., 2008. LARK2 gene (producing protein with kinase domain) mutation is a most general originator of familial form of PD (Zimprich et al., 2004, Alessi andSammler, 2018). ...
... PINK1 genes are associated with production of mitochondrial proteins; and mutations in these genes can cause autosomal recessive PD (Rogaeva et al., 2004). Genetic mutations in PINK1 generated oxidative stress, mitochondrial inadequacies and ultimately neuronal apoptosis (Grünewald et al., 2009, Ibánez et al., 2006, Kumazawa et al., 2008. LARK2 gene (producing protein with kinase domain) mutation is a most general originator of familial form of PD (Zimprich et al., 2004, Alessi andSammler, 2018). ...
... Mutations in these genes cause autosomal-recessive juvenile parkinsonism, associated with degeneration of midbrain dopaminergic neurons and motor impairments, among other symptoms and pathologies. Studies from a wide variety of model systems have shown various degrees of mitochondrial dysfunction associated with mutation of PINK1/PRKN homologues including disrupted fission/fusion [9][10][11][12][13][14][15][16][17]. Drosophila have proven to be a fruitful model for investigating the function of the conserved homologues Pink1 and parkin, with these mutants exhibiting robust mitochondrial disruption and neuromuscular phenotypes. ...
Article
Full-text available
Balanced mitochondrial fission and fusion play an important role in shaping and distributing mitochondria, as well as contributing to mitochondrial homeostasis and adaptation to stress. In particular, mitochondrial fission is required to facilitate degradation of damaged or dysfunctional units via mitophagy. Two Parkinson’s disease factors, PINK1 and Parkin, are considered key mediators of damage-induced mitophagy, and promoting mitochondrial fission is sufficient to suppress the pathological phenotypes in Drosophila Pink1/parkin mutants. We sought additional factors that impinge on mitochondrial dynamics and which may also suppress Pink1/parkin phenotypes. We found that the Drosophila phosphatidylinositol 4-kinase IIIβ homologue, Four wheel drive (Fwd), promotes mitochondrial fission downstream of the pro-fission factor Drp1. Previously described only as male sterile, we identified several new phenotypes in fwd mutants, including locomotor deficits and shortened lifespan, which are accompanied by mitochondrial dysfunction. Finally, we found that fwd overexpression can suppress locomotor deficits and mitochondrial disruption in Pink1/parkin mutants, consistent with its function in promoting mitochondrial fission. Together these results shed light on the complex mechanisms of mitochondrial fission and further underscore the potential of modulating mitochondrial fission/fusion dynamics in the context of neurodegeneration.
... Mitochondrial fractionation for measurement of Aconitase 2 activity. For measurement of ACO2 enzyme activity, mitochondrial fractions were prepared from fibroblasts as described before 49 . Fibroblasts were collected using trypsin-EDTA (0.25%) phenol red (Thermo Fisher Scientific, Braunschweig, Germany). ...
Article
Full-text available
ACO2 is a mitochondrial protein, which is critically involved in the function of the tricarboxylic acid cycle (TCA), the maintenance of iron homeostasis, oxidative stress defense and the integrity of mitochondrial DNA (mtDNA). Mutations in the ACO2 gene were identified in patients suffering from a broad range of symptoms, including optic nerve atrophy, cortical atrophy, cerebellar atrophy, hypotonia, seizures and intellectual disabilities. In the present study, we identified a heterozygous 51 bp deletion (c.1699_1749del51) in ACO2 in a family with autosomal dominant inherited isolated optic atrophy. A complementation assay using aco1-deficient yeast revealed a growth defect for the mutant ACO2 variant substantiating a pathogenic effect of the deletion. We used patient-derived fibroblasts to characterize cellular phenotypes and found a decrease of ACO2 protein levels, while ACO2 enzyme activity was not affected compared to two age- and gender-matched control lines. Several parameters of mitochondrial function, including mitochondrial morphology, mitochondrial membrane potential or mitochondrial superoxide production, were not changed under baseline conditions. However, basal respiration, maximal respiration, and spare respiratory capacity were reduced in mutant cells. Furthermore, we observed a reduction of mtDNA copy number and reduced mtDNA transcription levels in ACO2-mutant fibroblasts. Inducing oxidative stress led to an increased susceptibility for cell death in ACO2-mutant fibroblasts compared to controls. Our study reveals that a monoallelic mutation in ACO2 is sufficient to promote mitochondrial dysfunction and increased vulnerability to oxidative stress as main drivers of cell death related to optic nerve atrophy.
Chapter
Neurodegenerative disorders are rare hereditary disorders, which lead to the gradual deterioration of neural function in specific regions of the central nervous system. Neurodegenerative disorders are commonly depicted by the aggregation of extracellular and intracellular protein misfolds and neuronal degeneration in the selective areas of the nervous system. The misfolded proteins in neurodegenerative diseases possess unusual conformational effects, and the interaction between toxic proteins may escalate in different regions in the central nervous system, thereby causing degeneration of neurons. There are multiple pieces of evidence that specify substantial interaction between amyloidosis, tauopathies, synucleinopathies, and unusual protein aggregation of other disorders. The escalation of protein aggregates worsens over time due to abnormal functioning of intracellular mechanisms, leading to the inappropriate and unusual supplies of the essential factors and the mislaid interaction between the neurons. The molecular mechanism involves the dispersion and the transmission between the misfolded pathogenic proteins and the impact of the neuronal cells on the configuration and the effects of abnormal inclusions. In this regard, this chapter aims to highlight the mechanisms involved in neurodegenerative diseases, misfolding of proteins, the role of proteostasis, involvement of chaperones, and the association of ubiquitin-proteasome system and to emphasize the oxidative stress involvement and mitochondrial deficit in the progression of the diseases and the alternative splicing events in the neurodegenerative disorders.KeywordsAlzheimer’s diseaseParkinson’s diseaseProteostasisProtein aggregationProtein misfoldingMitochondrial dysfunctionUPSSplicing
Article
Full-text available
Significance Ceramide accumulates in Parkinson’s disease–related PINK1 deficiency to initiate ceramide-mediated mitophagy as an alternative pathway to overcome defective PINK1-related mitophagy and the concomitant increased requirements for mitochondrial clearance. Increased ceramide levels negatively correlate with β-oxidation and thus decrease efficiency of the electron transport chain, further increasing the need for mitochondrial clearance. Interfering with this vicious cycle can constitute a novel therapeutic strategy as suggested by our data showing that a reduction of ceramide levels or stimulation of β-oxidation improve the PINK1 -mutant phenotypes.
Article
Full-text available
Mutations in PINK1 cause autosomal recessive Parkinson's disease. PINK1 is a mitochondrial kinase of unknown function. We investigated calcium homeostasis and mitochondrial function in PINK1-deficient mammalian neurons. We demonstrate physiologically that PINK1 regulates calcium efflux from the mitochondria via the mitochondrial Na(+)/Ca(2+) exchanger. PINK1 deficiency causes mitochondrial accumulation of calcium, resulting in mitochondrial calcium overload. We show that calcium overload stimulates reactive oxygen species (ROS) production via NADPH oxidase. ROS production inhibits the glucose transporter, reducing substrate delivery and causing impaired respiration. We demonstrate that impaired respiration may be restored by provision of mitochondrial complex I and II substrates. Taken together, reduced mitochondrial calcium capacity and increased ROS lower the threshold of opening of the mitochondrial permeability transition pore (mPTP) such that physiological calcium stimuli become sufficient to induce mPTP opening in PINK1-deficient cells. Our findings propose a mechanism by which PINK1 dysfunction renders neurons vulnerable to cell death.
Article
Full-text available
Mitochondrial dysfunction has been implicated in the pathogenesis of Parkinson's disease (PD). Impairment of the mitochondrial electron transport chain (ETC) and an increased frequency in deletions of mitochondrial DNA (mtDNA), which encodes some of the subunits of the ETC, have been reported in the substantia nigra of PD brains. The identification of mutations in the PINK1 gene, which cause an autosomal recessive form of PD, has supported mitochondrial involvement in PD. The PINK1 protein is a serine/threonine kinase localized in mitochondria and the cytosol. Its precise function is unknown, but it is involved in neuroprotection against a variety of stress signalling pathways. In this report we have investigated the effect of silencing PINK1 expression in human dopaminergic SH-SY5Y cells by siRNA on mtDNA synthesis and ETC function. Loss of PINK1 expression resulted in a decrease in mtDNA levels and mtDNA synthesis. We also report a concomitant loss of mitochondrial membrane potential and decreased mitochondrial ATP synthesis, with the activity of complex IV of the ETC most affected. This mitochondrial dysfunction resulted in increased markers of oxidative stress under basal conditions and increased cell death following treatment with the free radical generator paraquat. This report highlights a novel function of PINK1 in mitochondrial biogenesis and a role in maintaining mitochondrial ETC activity. Dysfunction of both has been implicated in sporadic forms of PD suggesting that these may be key pathways in the development of the disease.
Article
Full-text available
A method has been developed whereby a fraction of rat brain mitochondria (synaptic mitochondria) was isolated from synaptosomes. This brain mitochondrial fraction was compared with the fraction of "free" brain mitochondria (non-synaptic) isolated by the method of Clark & Nicklas (1970). (J. Biol. Chem. 245, 4724-4731). Both mitochondrial fractions are shown to be relatively pure, metabolically active and well coupled. 2. The oxidation of a number of substrates by synaptic and non-synaptic mitochondria was studied and compared. Of the substrates studied, pyruvate plus malate was oxidized most rapidly by both mitochondrial populations. However, the non-synaptic mitochondria oxidized glutamate plus malate almost twice as rapidly as the synaptic mitochondria. 3. The activities of certain tricarboxylic acid-cycle and related enzymes in synaptic and non-synaptic mitochondria were determined. Citrate synthase (EC 4.1.3.7), isocitrate dehydrogenase (EC 1.1.1.41) and malate dehydrogenase (EC 1.1.1.37) activities were similar in both fractions, but pyruvate dehydrogenase (EC 1.2.4.1) activity in non-synaptic mitochondria was higher than in synaptic mitochondria and glutamate dehydrogenase (EC 1.4.1.3) activity in non-synaptic mitochondria was lower than that in synaptic mitochondria. 4. Comparison of synaptic and non-synaptic mitochondria by rate-zonal separation confirmed the distinct identity of the two mitochondrial populations. The non-synaptic mitochondria had higher buoyant density and evidence was obtained to suggest that the synaptic mitochondria might be heterogeneous. 5. The results are also discussed in the light of the suggested connection between the heterogeneity of brain mitochondria and metabolic compartmentation.
Article
Background: Mitochondrial dysfunction has been implicated in the pathogenesis of Parkinson's disease (PD). Impairment of the mitochondrial electron transport chain (ETC) and an increased frequency in deletions of mitochondrial DNA (mtDNA), which encodes some of the subunits of the ETC, have been reported in the substantia nigra of PD brains. The identification of mutations in the PINK1 gene, which cause an autosomal recessive form of PD, has supported mitochondrial involvement in PD. The PINK1 protein is a serine/threonine kinase localized in mitochondria and the cytosol. Its precise function is unknown, but it is involved in neuroprotection against a variety of stress signalling pathways.Methodology/Principal Findings: In this report we have investigated the effect of silencing PINK1 expression in human dopaminergic SH-SY5Y cells by siRNA on mtDNA synthesis and ETC function. Loss of PINK1 expression resulted in a decrease in mtDNA levels and mtDNA synthesis. We also report a concomitant loss of mitochondrial membrane potential and decreased mitochondrial ATP synthesis, with the activity of complex IV of the ETC most affected. This mitochondrial dysfunction resulted in increased markers of oxidative stress under basal conditions and increased cell death following treatment with the free radical generator paraquat.Conclusions: This report highlights a novel function of PINK1 in mitochondrial biogenesis and a role in maintaining mitochondrial ETC activity. Dysfunction of both has been implicated in sporadic forms of PD suggesting that these may be key pathways in the development of the disease.
Article
Almost two decades after the identification of SNCA as the first causative gene in Parkinson's disease (PD) and the subsequent understanding that genetic factors play a substantial role in PD development, our knowledge of the genetic architecture underlying this disease has vastly improved. Approximately 5–10% of patients suffer from a monogenic form of PD where autosomal dominant mutations in SNCA, LRRK2, and VPS35 and autosomal recessive mutations in PINK1, DJ-1, and Parkin cause the disease with high penetrance. Furthermore, recent whole-exome sequencing have described autosomal recessive DNAJC6 mutations in predominately atypical, but also cases with typical PD. In addition, several other genes have been linked to atypical Parkinsonian phenotypes. However, the vast majority of PD is genetically complex, i.e. it is caused by the combined action of common genetic variants in concert with environmental factors. By the application of genome-wide association studies, 26 PD risk loci have been established to date. Similar to other genetically complex diseases, these show only moderate effects on PD risk. Increasing this etiologic complexity, many of the involved genetic and environmental risk factors likely interact in an intricate fashion. This article aims to provide a comprehensive overview of the current knowledge in PD genetics.
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
Mutations in PTEN-induced kinase 1 (pink1) or parkin cause autosomal-recessive and some sporadic forms of Parkinson's disease. pink1 acts upstream of parkin in a common genetic pathway to regulate mitochondrial integrity in Drosophila. Mitochondrial morphology is maintained by a dynamic balance between the opposing actions of mitochondrial fusion, controlled by Mitofusin (mfn) and Optic atrophy 1 (opa1), and mitochondrial fission, controlled by drp1. Here, we explore interactions between pink1/parkin and the mitochondrial fusion/fission machinery. Muscle-specific knockdown of the fly homologue of Mfn (Marf) or opa1, or overexpression of drp1, results in significant mitochondrial fragmentation. Mfn-knockdown flies also display altered cristae morphology. Interestingly, knockdown of Mfn or opa1 or overexpression of drp1, rescues the phenotypes of muscle degeneration, cell death, and mitochondrial abnormalities in pink1 or parkin mutants. In the male germline, we also observe genetic interactions between pink1 and the testes-specific mfn homologue fuzzy onion, and between pink1 and drp1. Our data suggest that the pink1/parkin pathway promotes mitochondrial fission and/or inhibits fusion by negatively regulating mfn and opa1 function, and/or positively regulating drp1. However, pink1 and parkin mutant flies show distinct mitochondrial phenotypes from drp1 mutant flies, and flies carrying a heterozygous mutation in drp1 enhance the pink1-null phenotype, resulting in lethality. These results suggest that pink1 and parkin are likely not core components of the drp1-mediated mitochondrial fission machinery. Modification of fusion and fission may represent a novel therapeutic strategy for Parkinson's disease. • mitofusin • drp1 • opa1 • parkin-pink1
Article
Diagnosis of respiratory chain defects in cultured skin fibroblasts is a difficult diagnostic procedure. We investigated the feasibility of using survival of skin fibroblasts in culture medium with galactose as the major carbon source as a method of quickly diagnosing cell lines that were compromised in oxidative metabolism. We found that cells from patients with most forms of cytochrome oxidase deficiency, cells with complex I deficiency, cells with multiple respiratory chain defects and cells with severe pyruvate dehydrogenase (PDH) complex deficiency failed to survive when subcultured into galactose (5 mM) medium. Cells from patients with Lebers hereditary optic neuropathy (LHON), Kearns-Sayre syndrome (KSS), myoclonus-epilepsy-lactic acidosis-stroke (MELAS), the hepatic form of cytochrome oxidase deficiency, and mild PDH complex deficiency survived well in galactose (5 mM)-containing medium. This could be used as a rapid screening test for skin fibroblasts with major oxidative defects.
Article
There is increasing evidence that defective function of the mitochondrial enzyme NADH CoQ reductase (complex I) is involved not only in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity, but also in idiopathic Parkinson's disease (PD). Complex I deficiency has been identified in PD substantia nigra and appears to be disease-specific and selective for the substantia nigra within the central nervous system. We describe a method for preparation of an enriched mitochondrial fraction from 60 mL blood. Using this technique, we analyzed respiratory chain function in 25 patients with PD and 15 matched control subjects. We confirm a previous report of a specific complex I deficiency in PD platelet mitochondria. Although there was a statistically significant decrease in complex I activity in the PD group compared with the control group (p = 0.005), the defect was mild (16%); it was not possible to distinguish PD from control values on an individual basis. This deficiency is not detectable in platelet whole-cell homogenates, presumably reflecting the relative insensitivity of this preparation and the limited decrease in complex I activity in PD. The presence of a mild complex I defect in platelets together with a more severe defect in substantia nigra suggests either that the pharmacological characteristics shared by these two tissues render them susceptible to a particular toxin or toxins, or that the defect is widely distributed and other biochemical events enhance the deficiency in substantia nigra.(ABSTRACT TRUNCATED AT 250 WORDS)