ArticlePDF AvailableLiterature Review

Neuropathology of sporadic Parkinson disease before the appearance of parkinsonism: Preclinical Parkinson disease

Authors:
  • Universitat de Vic-Universitat Central de Catalunya and Autonomous University of Barcelona

Abstract and Figures

Parkinson disease (PD) is no longer considered a complex motor disorder characterized by parkinsonism but rather a systemic disease with variegated non-motor deficits and neurological symptoms, including impaired olfaction, sleep disorders, gastrointestinal and urinary abnormalities and cardiovascular dysfunction, in addition to other symptoms and signs such as pain, depression and mood disorders. Many of these alterations appear before or in parallel with motor deficits and then worsen with disease progression. Although there is a close relation between motor symptoms and the presence of Lewy bodies (LBs) and neurites filled with abnormal α-synuclein, other neurological alterations are independent of LBs, thereby indicating that different mechanisms probably converge in the degenerative process. This review presents cardinal observations at very early stages of PD and provides personal experience based on the study of a consecutive series of brains with PD-related pathology and without parkinsonism, mainly cases categorized as stages 2-3 of Braak. Alterations in the substantia nigra, striatum and frontal cortex in pPD are here revised in detail. Early modifications in the substantia nigra at pre-motor stages of PD (preclinical PD: pPD) include abnormal small aggregates of α-synuclein which is phosphorylated, nitrated and oxidized, and which exhibits abnormal solubility and truncation. This occurs in association with a plethora of altered molecular events including increased oxidative stress, altered oxidative stress responses, altered balance of L-ferritin and H-ferritin, reduced expression of neuronal globin α and β chains in neurons with α-synuclein deposits, increased expression of endoplasmic reticulum stress markers, increased p62 and ubiquitin immunoreactivity in relation to α-synuclein deposits, and altered distribution of LC3 and other autophagosome/lysosome markers. In spite of the relatively small decrease in the number of dopaminergic neurons in the substantia nigra, which does not reach thresholds causative of parkinsonism, levels of tyrosine hydroxylase and cannabinoid 1 receptor are reduced, whereas levels of adenosine receptor 2A are increased in the caudate in pPD. Moreover, biochemical alterations are also present in the cerebral cortex (at least in the frontal cortex) in pPD including increased oxidative stress and oxidative damage to proteins α-synuclein, β-synuclein, superoxide dismutase 2, aldolase A, enolase 1, and glyceraldehyde dehydrogenase, among others, indicating post-translational modifications of PD-related proteins, and suggesting altered function of pathways involved in glycolysis and energy metabolism in the cerebral cortex in pPD. Current evidence suggests convergence of several altered metabolic pathways leading to chronic neuronal dysfunction, mainly manifested as sub-optimal energy metabolism, altered synaptic function, oxidative and endoplasmic reticulum stress damage and corresponding altered responses, among others. By understanding that these alterations occur at very early stages of PD and that neuronal fatigue and exhaustion may precede, for years, cell death and neuronal loss, we may direct therapeutic strategies towards the prevention and delay of disease progression starting at pre-parkinsonian stages of PD.
Content may be subject to copyright.
BASIC NEUROSCIENCES, GENETICS AND IMMUNOLOGY - REVIEW ARTICLE
Neuropathology of sporadic Parkinson disease
before the appearance of parkinsonism:
preclinical Parkinson disease
Isidre Ferrer Anna Martinez Rosa Blanco
Ester Dalfo
´Margarita Carmona
Received: 26 July 2010 / Accepted: 30 August 2010 / Published online: 23 September 2010
ÓSpringer-Verlag 2010
Abstract Parkinson disease (PD) is no longer considered
a complex motor disorder characterized by parkinsonism
but rather a systemic disease with variegated non-motor
deficits and neurological symptoms, including impaired
olfaction, sleep disorders, gastrointestinal and urinary
abnormalities and cardiovascular dysfunction, in addition
to other symptoms and signs such as pain, depression and
mood disorders. Many of these alterations appear before or
in parallel with motor deficits and then worsen with disease
progression. Although there is a close relation between
motor symptoms and the presence of Lewy bodies (LBs)
and neurites filled with abnormal a-synuclein, other neu-
rological alterations are independent of LBs, thereby
indicating that different mechanisms probably converge in
the degenerative process. This review presents cardinal
observations at very early stages of PD and provides per-
sonal experience based on the study of a consecutive series
of brains with PD-related pathology and without parkin-
sonism, mainly cases categorized as stages 2–3 of Braak.
Alterations in the substantia nigra, striatum and frontal
cortex in pPD are here revised in detail. Early modifica-
tions in the substantia nigra at pre-motor stages of PD
(preclinical PD: pPD) include abnormal small aggregates
of a-synuclein which is phosphorylated, nitrated and oxi-
dized, and which exhibits abnormal solubility and trunca-
tion. This occurs in association with a plethora of altered
molecular events including increased oxidative stress,
altered oxidative stress responses, altered balance of
L-ferritin and H-ferritin, reduced expression of neuronal
globin aand bchains in neurons with a-synuclein deposits,
increased expression of endoplasmic reticulum stress
markers, increased p62 and ubiquitin immunoreactivity in
relation to a-synuclein deposits, and altered distribution of
LC3 and other autophagosome/lysosome markers. In spite
of the relatively small decrease in the number of dopami-
nergic neurons in the substantia nigra, which does not reach
thresholds causative of parkinsonism, levels of tyrosine
hydroxylase and cannabinoid 1 receptor are reduced,
whereas levels of adenosine receptor 2A are increased in
the caudate in pPD. Moreover, biochemical alterations are
also present in the cerebral cortex (at least in the frontal
cortex) in pPD including increased oxidative stress and
oxidative damage to proteins a-synuclein, b-synuclein,
superoxide dismutase 2, aldolase A, enolase 1, and glyc-
eraldehyde dehydrogenase, among others, indicating post-
translational modifications of PD-related proteins, and
suggesting altered function of pathways involved in gly-
colysis and energy metabolism in the cerebral cortex in
pPD. Current evidence suggests convergence of several
altered metabolic pathways leading to chronic neuronal
dysfunction, mainly manifested as sub-optimal energy
metabolism, altered synaptic function, oxidative and
endoplasmic reticulum stress damage and corresponding
altered responses, among others. By understanding that
these alterations occur at very early stages of PD and that
neuronal fatigue and exhaustion may precede, for years,
cell death and neuronal loss, we may direct therapeutic
strategies towards the prevention and delay of disease
progression starting at pre-parkinsonian stages of PD.
Keywords Preclinical Parkinson disease
Incidental Lewy body disease Oxidative stress
I. Ferrer (&)A. Martinez R. Blanco E. Dalfo
´
M. Carmona
Institut de Neuropatologia, Servei Anatomia Patolo
`gica,
CIBERNED, IDIBELL-Hospital Universitari de Bellvitge,
Universitat de Barcelona, carrer Feixa LLarga sn,
08907 Hospitalet de LLobregat, Spain
e-mail: 8082ifa@gmail.com
123
J Neural Transm (2011) 118:821–839
DOI 10.1007/s00702-010-0482-8
Endoplasmic reticulum stress Haemoglobin
Mitochondria Olfaction Cerebral cortex Striatum
Synuclein Glycolysis
Introduction
Parkinson disease is clinically characterized by a complex
motor disorder known as parkinsonism and is manifested
principally by resting tremor, slowness of initial move-
ment, rigidity and general postural instability. These
symptoms are mainly due to the loss of dopaminergic
neurons in the substantia nigra pars compacta, first the
lateral tier followed by the medial region, leading to
reduced dopaminergic input to the striatum, and accom-
panied by adaptive responses in the internal and external
globus pallidus, subthalamus, thalamus and substantia
nigra pars reticularis. Round, hyaline neuronal cytoplasmic
inclusions called Lewy bodies (LBs), and enlarged aberrant
neurites and threads are found in the parkinsonian sub-
stantia nigra (Forno 1996; Jellinger and Mizuno 2003). In
addition to the substantia nigra, other nuclei are involved
such as the locus coeruleus, reticular nuclei of the brain
stem, and dorsal motor nucleus of the vagus, as well as the
basal nucleus of Meynert, the amygdala and the CA2 area
of the hippocampus. LBs and aberrant neurites as well are
found in these locations (Forno 1996; Braak et al. 1999;
Dickson 2001; Goedert 2001; Jellinger and Mizuno 2003;
Braak and del Tredici 2008).
Cases with LB pathology in the brain stem without
parkinsonism are considered incidental LB disease because
they are unexpectedly discovered following appropriate
post-mortem neuropathological study (Jellinger 2004,
2009; Saito et al. 2004). Whether these cases constitute
pre-parkinsonian PD has been a matter of controversy for
some years, as nobody can ensure that these cases would
have progressed to parkinsonism if they had survived for
longer times. However, the study of consecutive cases in
large series and the recognition of several intermediate
degrees of involvement of the brain stem, limbic structures
and, eventually, the cerebral cortex make it clear that a
prediction of preclinical PD (pPD) as an anterior stage of
PD seems more than reasonable (Jellinger and Mizuno
2003; Braak and del Tredici 2008; Dickson et al. 2008).
LBs and neurites are composed of aggregates of normal,
misfolded and truncated proteins, and ubiquitin, which are
stored in the cytoplasm as non-degraded by-products of the
degenerative process (Schults 2006; Wakabayashi et al.
2007; Leverenz et al. 2007; Xia et al. 2008). The main
component of LBs and aberrant neurites is a-synuclein
which is abnormally phosphorylated, nitrated and oxidized,
has an abnormal crystallographic structure and abnormal
solubility, and is prone to the formation of aggregates and
insoluble fibrils (Spillantini et al. 1997; Wakabayashi et al.
1997; Baba et al. 1998; Hashimoto and Masliah 1999;
Duda et al. 2000; Giasson et al. 2000; Fujiwara et al. 2002;
Anderson et al. 2006).
Systematic study of cases with LB pathology has
prompted a staging classification of PD based on the
putative progression of LB pathology from the medulla
oblongata (and olfactory bulb) to the midbrain, dience-
phalic nuclei, and neocortex (Braak et al. 2002,2003,
2004). Stage 1 is characterized by LBs and neurites in the
dorsal IX/X motor nuclei and/or intermediate reticular
zone; there is also myenteric plexus involvement. Stage 2
affects the medulla oblongata and pontine tegmentum and
covers pathology of stage 1 plus lesions in the caudal raphe
nuclei, gigantocellular reticular nucleus, and coeruleus–
subcoeruleus complex; the olfactory bulb is also involved.
Stage 3 refers to pathology of stage 2 plus midbrain lesions,
particularly in the pars compacta of the substantia nigra.
Stage 4 includes basal prosencephalon and mesocortex
(cortical involvement confined to the transentorhinal region
and allocortex, and CA2 plexus) pathology in addition to
lesions in the midbrain, pons and medulla oblongata. Stage
5 extends to sensory association areas of the neocortex and
prefrontal neocortex. Stage 6 includes, in addition, lesions
in first order sensory association areas of the neocortex and
pre-motor areas; occasionally there are also mild changes
in primary sensory areas and the primary motor field.
This classification shows an acceptable correlation
between pathological findings and clinical data, mainly in a
subgroup of cases with early onset and prolonged duration
in which motor symptoms (parkinsonism) are clearly
dependent on the damage of dopaminergic neurons in the
substantia nigra pars compacta (Braak et al. 2002,2005;
Jellinger 2004; Wolters and Braak 2006). Thus, stages
1 and 2 are not accompanied by parkinsonism and they
fulfil the criteria of pPD. Cases at stage 3 may present with
parkinsonism depending on the amount of cell loss in the
substantia nigra. More strictly, pPD occurs in cases with
LB pathology limited to selected nuclei of the medulla
oblongata and pons, and in cases with a few LBs in the
substantia nigra but without substantial nigral neuron loss
not surpassing a determined threshold, usually considered
as 60% of the total population of dopaminergic neurons
of the pars compacta (Jellinger and Mizuno 2003;
DelleDonne et al. 2008).
Cumulative clinical evidence reveals that olfactory
dysfunction, dysautonomia, sleep fragmentation, rapid eye
movement behaviour disorder, mood and anxiety disorders,
and depression may precede parkinsonian symptoms in a
number of patients with PD clinically characterized by
parkinsonism (Chaudhuri et al. 2005; Postuma et al. 2006;
Iranzo et al. 2006; Siderowf and Stern 2008; Tolosa et al.
2007; Poewe 2007,2008; Ziemssen and Reichmann 2007;
822 I. Ferrer et al.
123
Herting et al. 2008; Ross et al. 2008; Natale et al. 2008).
Whether these clinical symptoms are associated with LBs
in selected regions of the central, autonomous and
peripheral nervous systems is a matter of study.
a-Synuclein pathology affecting neurons and neurites
occurs in the olfactory bulb and related olfactory nuclei at
very early stages of cases with PD-related pathology
(Daniel and Hawkes 1992; Pearce et al. 1995; Del Tredici
et al. 2002; Braak et al. 2004; Bloch et al. 2006; Hubbard
et al. 2007).
The cardiovascular autonomic system is also affected in
pPD and PD, and alterations implicate both tyrosine
hydroxylase-positive (extrinsic) and negative (intrinsic)
nerves of the cardiac plexus (Iwanaga et al. 1999).
Together, these observations can be summarized as
follows: (a) variegated non-motor deficits and neurological
symptoms, covering impaired olfaction, sleep disorders,
gastrointestinal and urinary abnormalities and cardiovas-
cular dysfunction, in addition to other symptoms and signs
as pain, depression and mood disorders, may occur at early
stages of the disease. (b) A relationship between neuro-
logical symptoms and Lewy pathology exists, but it is not
clear whether the severity of morphological lesions corre-
lates with clinical symptoms. (c) Olfactory tests, poly-
somnographic studies and MIBG myocardial scintigraphy
in combination may be used to discover early signatures of
the disease (Stiasny-Kolster et al. 2005).
Lack of correlation between a-synuclein aggregates and
impaired function is dramatically represented in relation
with cognitive and mental functions. Retrospective clinical
and pathologic studies have shown that there is no rela-
tionship between LB stage and severity of cognitive
impairment in advanced stages of PD (Parkkinen et al.
2005; Weisman et al. 2007; Parkkinen et al. 2008; Jellinger
2008,2009).
These and other studies have led to the visualization of
PD as a systemic disease in which functional deficits are
not exclusively related with LB pathology in neurons and
neurites. This is dramatically manifested in relation with
cerebral cortex involvement in PD in which variegated
pathological molecular events converge to alter neuronal
function (Ferrer 2009a).
Neuropathological examination of pPD in a cohort
of consecutive cases in a general population
Over the last 10 years (2001–2010), 115 cases of LB-rela-
ted pathology were recovered in a consecutive series of
cases subjected to post-mortem study in a general hospital
(Institut de Neuropatologia, Hospital Universitari de
Bellvitge). These were classified as stages 1–2: 24%, stages
3–4: 42%, stages 5–6: 26%, and atypical: 8%. Interestingly,
mixed pathology was very common. AD-related pathology
was found in 81% of cases (40% stages I–II of Braak, 46%
corresponding to stages III–IV, and 14% to stages V and
VI), argyrophilic grain disease and other tauopathies in
24%, vascular pathology in 28%, and other pathological
conditions in 12%. Some cases had multiple pathologies
(mainly LB disease, AD and vascular pathology including
lacunae, multi-infarct encephalopathy, haemorrhages and
demyelination of the centrum semi-ovale reminiscent of
Binswanger encephalopathy). These figures are similar to
those already reported in other series (Jellinger 2004,2008,
2009,2010; Braak et al. 2006; Markesbery et al. 2009;
Beach et al. 2009; Dickson et al. 2010).
The distribution of PD-related pathology in our series
has a gradient with age; cases with stages 5 and 6 are older
as a group than cases at stage 4. However, a number of
cases at stages 1 and 2 were remarkably aged. More than
half were older than 75, indicating that, within the same
age interval, some patients had suffered from fully devel-
oped PD whereas others had remained at pre-motor stages.
These findings are inline with the observation that, at least
some cases of incidental LB disease could represent pre-
clinical PD, arrested PD, or a partial syndrome due to a
lesser burden of causative factors (Frigerio et al. 2009).
For the present purposes, parkinsonism (including slight
motor symptoms, revealed by relatives following an
unbiased post-mortem interview in cases that had not been
diagnosed during life) occurred in 68% of the cases.
Excluding atypical cases, pPD was seen in 28 cases of
stages 1–2, 10 cases of stage 3, and 3 cases of stage 4.
Morphological studies of the substantia nigra were carried
out in this group. In none of these cases did nerve cell loss
in the substantia nigra exceed 50% of the total number of
dopaminergic neurons as revealed by quantitative studies
using tyrosine hydroxylase immunohistochemistry. A sum-
mary of antibodies used for immunohistochemistry, and
single- and double-labelling immunofluorescence and
confocal microscopy, is given in Table 1.
Molecular studies in the frontal cortex were restricted to
20 pPD cases with no associated pathology or with
accompanying AD-related changes stages I–II, to exclude
cases in which cortical pathology could interfere with
changes interpreted to be exclusive of pPD. Control sam-
ples were from cases with no neuropathological changes or
with AD-related pathology stages I–II. Methods employed
were mono- and bi-dimensional gel electrophoresis and
western blotting, immunoprecipitation and functional
assays (Dalfo
´et al. 2004,2005; Dalfo
´and Ferrer 2008;
Muntane
´et al. 2008; Martı
´nez et al. 2010).
The present observations are mainly focused on the
substantia nigra pars compacta, caudate and cerebral cortex
(frontal cortex area 8) in pPD. The olfactory bulb and tract
also deserve a brief comment.
Neuropathology of sporadic Parkinson disease 823
123
The substantia nigra in pre-motor stages of PD
It is classically considered that the substantia nigra in pre-
motor stages of PD is not morphologically affected or
damaged to an extent not surpassing a threshold currently
accepted as involving less than 60% of the total of dopa-
minergic neurons of the substantia nigra pars compacta.
Using the staging nomenclature of PD-related pathology in
our series, pre-motor or incidental PD may correspond to
stages 1–3. Surprisingly, parkinsonian symptoms have
been reported in stage 2 of Braak (Jellinger 2009). This is
not our experience: parkinsonian symptoms and signs
appear at stage 3 in our series and stage 3 may be also
encountered in pPD cases in which decline of substantia
nigra dopaminergic neurons does not exceed 50% of the
total. Since vascular pathology is not rare in ours and in
other series, we cannot rule out the possibility of primary
vascular lesions in the striatum may account for motor
deficits in cases with PD-related pathology stage 2 in other
series.
a-Synuclein in the substantia nigra
An important issue is the relative amount of a-synuclein in
the substantia nigra pars compacta when compared with
other brain regions. Studies in normal human brain are
puzzling since it is not clear whether the substantia nigra
and striatum had the lowest (Rockenstein et al. 2001)or
the highest levels (Solano et al. 2000)ofa-synuclein.
Regarding PD, comparative studies of control and diseased
Table 1 Antibodies, species,
source and dilution used for
immunohistochemistry and
immunofluorescence in the
present study
Tissue sections were pre-treated
with formic acid for 3 min and
then with 10 mM sodium citrate
buffer pH 6 for 20 min at 95°C
Antibody Species Manufacturer Dilution
ATP-synthase aMouse BD Biosciences (Madrid, Spain) 1:1,000
Casein kinase II Rabbit Stressgen (bioNova, Madrid, Spain) 1:100
COX7A2 Mouse Abnova (Taipei, Taiwan) 1:50
eif2aRabbit Abcam (Cambridge, UK) 1:100
eif2a-P Rabbit Cell signalling (Izasa, Barcelona, Spain) 1:100
Ferritin Goat Affinity Bioreagents (bioNova, Madrid, Spain) 1:500
Haemoglobin (bchain) Mouse Abcam (Cambridge, UK) 1:200
8-Hydroxy-2
deoxyguanosine
Mouse Abcam (Cambridge, UK) 1:200
LAMP-1 Rabbit Sta. Cruz (Quimigen, Barcelona) 1:100
LAMP-2 Rabbit Sta. Cruz (Quimigen, Barcelona) 1:500
LC-3 Rabbit Cell signalling (Izasa, Barcelona, Spain) 1:100
Neuroketals Goat Chemicon (Millipore, Madrid, Spain) 1:200
Oxphos Mouse Mitosciences (Eugene, Oregon, USA) 1:1,000
Phospho-p38 Rabbit Cell signalling (Izasa, Barcelona, Spain) 1:100
p62 Rabbit BIOMOL (Quimigen, Madrid, Spain) 1:500
Peroxiredoxin-2 Rabbit Abcam (Cambridge, UK) 1:1,000
VDAC, porin Mouse Calbiochem (bioNova, Madrid, Spain) 1:300
SOD-1 (Cu/Zn) Mouse Novocastra (Newcastle, UK) 1:50
SOD-2 (Mn) Rabbit Stressgen (bioNova, Madrid, Spain) 1:100
a-Synuclein Mouse Novocastra (Newcastle, UK) 1:100
a-Synuclein Rabbit Chemicon (Millipore, Madrid, Spain), epitope
111–131
1:500
a-Synuclein Rabbit Abcam (Cambridge, UK), epitope 11–26 1:100
a-Synuclein Guinea
pig
Calbiochem (bioNova, Madrid, Spain), epitope
123–140
1:50
a-Synuclein Mouse ATGen (Montevideo, Uruguay), epitope 61–90 1:100
Nitrated a-synuclein Mouse Zymed (San Francisco, CA, USA) 1:300
Phospho-a-synuclein Mouse WAKO (Rafer, Madrid, Spain) 1:500
Transferrin Rabbit Affinity Bioreagents (bioNova, Madrid, Spain) 1:300
Ubiquitin Rabbit DAKO (Barcelona, Spain) 1:500
8-dOHG Mouse Abcam 1:50
Tyrosine hydroxylase Mouse Chemicon 1:1,000
CB-1 receptor Rabbit Frontier Science 1:200
824 I. Ferrer et al.
123
brains are not illuminating. a-Synuclein mRNA levels in
the PD substantia nigra have been reported to be unmodi-
fied, increased, or decreased when compared with age-
matched controls (Rockenstein et al. 2001; Wirdefeldt et al.
2001; Kingsbury et al. 2004; Chiba-Falek et al. 2006).
Cleaved (truncated) fragments of a-synuclein are prone to
aggregate in vitro and have been considered prime com-
ponents in LBs (Murray et al. 2003; Li et al. 2005).
Our recent studies have shown consistently lower levels
of a-synuclein protein expression in the human substantia
nigra and nucleus basalis of Meynert when compared with
other brain regions independent of age and pathology.
Phosphorylated a-synuclein at Ser129 is increased in the
same regions, thus correlating with the total amount of
a-synuclein. Additionally, truncated a-synuclein is natu-
rally observed in control and diseased brains, correlating
with the total amount of a-synuclein (Muntane
´et al., in
preparation).
a-Synuclein pathology and Lewy inclusions
in the substantia nigra in pPD
Phosphorylated a-synuclein at Ser129 is considered a key
step in the development of a-synuclein aggregates
(Anderson et al. 2006). Phosphorylated a-synuclein at
Ser129 co-localizes with a-synuclein in punctuate Lewy-
type aggregates in the substantia nigra at stage 3 of Braak
(Fig. 1a–c). It is difficult to ascertain the amount of phos-
phorylated a-synuclein of the total a-synuclein on the basis
of immunohistochemistry using antibodies that may have
different binding capacities to their corresponding sub-
strates. However, double-labelling studies suggest that
phosphorylated a-synuclein at Ser129 predominates in LB
lesions in pPD. Similar pattern was seen in affected nuclei
of the medulla oblongata and affected neurons in the sub-
stantia nigra in pPD. Casein kinase II, one of the enzymes
that phosphorylates a-synuclein at Ser129 (Ishii et al. 2007;
Waxman and Giasson 2008), is currently found in LB in
the substantia nigra in advanced stages of PD (Ryu et al.
2008). Yet casein kinase II is also observed in neurons with
early a-synuclein inclusions not sequestered by LBs in pPD
(Fig. 1d–f), indicating that increased casein kinase II
expression is associated with increased a-synuclein phos-
phorylation at early stages of PD-related pathology. Other
activated kinases, the functions of which have not been
clearly identified in PD, have also been reported in asso-
ciation with LBs. One of them, active p38 (p38-P), is
accumulated in cytoplasmic granules in the vicinity of
LBs or in association with irregular-shaped or diffuse
a-synuclein deposits in a percentage of substantia nigra
pars compacta neurons in PD (Ferrer et al. 2001) and in a
few neurons in pPD at stage 3 (Fig. 1g–i). In addition,
nitrated a-synuclein is also found in small granules in
dopaminergic neurons before the appearance of LBs in
pPD. Finally, oxidative damage of a-synuclein has also
been found using biochemical methods in the substantia
nigra in pPD (Dalfo
´and Ferrer 2008). These observations
indicate that modifications of a-synuclein, including oxi-
dation, nitration and phosphorylation at Ser129, are early
events in the substantia nigra at stage 3 of PD and occur in
neurons with punctuate a-synuclein aggregates.
Truncated a-synuclein variants, detected with antibodies
raised against different C- and N-terminal epitopes of
a-synuclein, are components of punctate a-synuclein
aggregates and LBs (Fig. 2).
These observations indicate that altered expression of
proteins is not restricted to LBs, at least at early stages of
LB formation. Rather, altered expression of proteins is seen
in the vicinity but not within LBs in pPD. On the other
hand, the structure of LBs is not homogeneous but different
forms of a-synuclein are differentially incorporated into
LBs.
Mitochondria dysfunction
Deficiencies in complex I subunits of the respiratory chain
and impaired activity of the electron transport chain are
well-known abnormalities in the substantia nigra in spo-
radic PD with parkinsonism (Mizuno et al. 1989; Parker
et al. 1989; Schapira 2008; Onyango 2008).
Early studies showed no modifications in the activity of
mitochondrial complexes II, III and IV in pPD (Jenner
et al. 1992; Dexter et al. 1994). More especially, complex I
activity in pPD was normal or reduced to intermediate
levels among controls and cases with PD (Jenner et al.
1992; Dexter et al. 1994). However, studies in total
homogenates do not permit the detection of modifications
restricted to individual neurons. The use of immunofluo-
rescence with appropriate markers offers limited informa-
tion, as oxphos, porin, cytochrome Coxidase subunits and
ATP synthase immunoreactivities are preserved in neurons
of the substantia nigra pars compacta in stage 3 of Braak,
independent of the presence or absence of a-synuclein
inclusions (data not shown). Whether subunits of mito-
chondrial complex I are oxidatively damaged, functionally
impaired and misassembled in pPD substantia nigra as
reported in PD (Keeney et al. 2006) is not known.
Oxidative stress
Reduced glutathione levels are decreased in the substantia
nigra in pPD and they are considered one of the first
alterations in the substantia nigra in PD (Jenner et al. 1992;
Jenner 1993,1998; Dexter et al. 1994; Zeevalk et al. 2008).
Oxidative damage in the substantia nigra is already
present in pPD at stages 2 and 3 of Braak (Dalfo
´et al.
Neuropathology of sporadic Parkinson disease 825
123
Fig. 1 Double-labelling immunofluorescence and confocal micros-
copy of a-synuclein (green) and a-synuclein PSer129 (red)(ac),
casein kinase II (green) and a-synuclein (red)(df), and p38 kinase-P
(green) and a-synuclein (red)(gi) in the substantia nigra pars
compacta in pPD stage 3. a-Synuclein co-localizes with a-synuclein
PSer129 in several but not all punctiform, neuritic and cytoplasmic
aggregates. Casein kinase II and p38 kinase-P are expressed in the
cytoplasm of neurons with a-synuclein aggregates. p38-P also co-
localizes with synuclein in LBs. jlNegative controls without
primary antibodies
826 I. Ferrer et al.
123
2005). This is further documented by increased neuroketal
(NKT) immunofluorescence in neurons with a-synuclein
deposits and by the presence of NKTs in LBs (Fig. 3a–c).
Neuroketals are a class of compounds that result from the
oxidation of docosahexaenoic acid (DHA), a membrane
polyunsaturated fatty acid especially vulnerable to free
radical attack, which is present in large amounts in the
brain. Therefore, the presence of neuroketals indicates
oxidation of DHA in substantia nigra dopaminergic neu-
rons at early stages of PD.
Oxidative stress responses are also augmented in neurons
with a-synuclein pathology. Expression of peroxiredoxin 2,
superoxide dismutase 1 (SOD1) and SOD2 is increased in
neurons with a-synuclein inclusions (Fig. 3d–l). In addition
to cytoplasmic increase in SOD1 and SOD2, these proteins
can also be sequestered in LBs (Fig. 3j–l). It is interesting to
have in mind that SOD2 has been reported oxidatively
damaged in PD (Choi et al. 2005; Dalfo
´et al. 2005). Thus,
proteins produced to counteract oxidative stress are, in turn,
oxidatively damaged and sequestered into LBs.
In the line evidencing early oxidative damage in the
substantia nigra in pPD is the observation of lipoxidative
damage of a-synuclein in pPD (Dalfo
´and Ferrer 2008).
Therefore, available observations show that oxidative
Fig. 2 Triple-labelling immunofluorescence and confocal micros-
copy of a-synuclein inclusions in the substantia nigra in pPD. Triple
labelling with antibodies directed to the NAC region (AB9, green),
N-terminal (AB2, red) and C-terminal (AB8, blue)ofa-synuclein
show slight differences in the staining of a-synuclein inclusions and
LBs in the substantia in pPD. Several punctiform inclusions are
differentially stained with the different anti-a-synuclein antibodies
whereas co-localization of all three antibodies is found in LB (white
in the merge construction)
Neuropathology of sporadic Parkinson disease 827
123
Fig. 3 Double-labelling immunofluorescence and confocal micros-
copy of neuroketal (green) and a-synuclein (red)(ac), peroxiredoxin
(green) and a-synuclein (red)(df), superoxide dismutase 2 (green)
and a-synuclein (red)(gi), and superoxide dismutase 1 (green) and
a-synuclein (red)(jl) in the substantia nigra pars compacta in pPD
stage 3. Markers of oxidative damage and markers of oxidative stress
responses are expressed in neurons of the substantia nigra. In addition,
neuroketal, SOD1 and SOD2, but rarely peroxiredoxin accumulate in
LBs
828 I. Ferrer et al.
123
damage is an early event in PD and that oxidative damage
in the substantia nigra in stage 2 precedes the formation of
LBs in pPD substantia nigra.
Further biochemical studies have shown increased levels
of neuroketals in the substantia nigra in cases with PD-
related pathology stages 1 and 2 (Fig. 4), thus further
supporting that oxidative damage to specific lipids in the
substantia nigra occurs at very early stages of PD before
the appearance of regional a-synuclein aggregates. These
observations indicate early oxidative damage to lipids and
certain proteins. In addition, evidence of DNA damage in
the substantia nigra in pPD is shown in Fig. 4.
Iron
Iron plays an important role in the regulation of several
enzymatic activities and in redox modulation. Iron dysho-
meostasis has been observed in PD (Andersen 2004;
Salazar et al. 2006; Hirsch 2006; Barnham and Bush 2008;
Youdim 2008). Neuromelanin has a protective function
under physiological conditions contributing to iron
homeostasis (Double 2006). However, neuromelanin
lipoxidation, altered a-synuclein binding to neuromelanin
and increased levels of L-ferritin in neuromelanin have
harmful effects on dopaminergic cell fate (Halliday et al.
2005; Double and Halliday 2006; Zecca et al. 2006,2008;
Tribl et al. 2009). Iron is increased in the substantia nigra in
PD and it is stored in glial cells and macrophages at the
time that neuromelanin decreases in vulnerable neurons
(Gerlach et al. 2006; Fasano et al. 2006). In addition,
increased iron levels have been found in individual neurons
in PD (Oakley et al. 2007). As a result, increased iron
levels, and particularly increased levels of labile iron, may
increase iron-mediated oxidative damage and contribute to
redox imbalance in PD (Wang et al. 2008; Chinta and
Andersen 2008; Wypijewska et al. 2010).
No evidence of alteration in iron, copper, manganese or
zinc in the pPD substantia nigra was noted in early studies
(Jenner et al. 1992). However, recent observations have
shown that L-ferritin concentration in the substantia nigra
is lower in pPD (and PD) when compared with controls,
whereas H-ferritin in PD is higher than in pPD and con-
trols, thus indicating fine abnormalities in iron metabolism
in the substantia nigra at early stages of PD (Koziorowski
et al. 2007).
Neuronal globin aand bchain in the substantia
nigra in pPD
Recent studies have demonstrated the presence of globin a
and bin neurons in the mouse, rat and human brains, using
robust combined methods including transcriptome analysis
(cDNA microarrays and nanoCAGE) of laser-captured
micro-dissected neurons, quantitative transcriptase-poly-
merase chain reaction (RT-PCR), in situ hybridization and
immunohistochemistry with a panel of different specific
antibodies (Biagioli et al. 2009; Richter et al. 2009). Nerve
cells also express erythropoietin receptor, erythropoietin
and hypoxia-inducible factor Hif1a(Matsuda et al. 1994;
Digicaylioglu et al. 1995).
Globin aand bis present in dopaminergic neurons of the
substantia nigra, and its expression is reduced in vulnerable
neurons in parallel with abnormal a-synuclein deposition
(Fig. 5). In contrast, the expression of neuroglobin and
erythropoietin receptor is preserved in the same cells.
These preliminary data point to the possibility that
decreased globin aand bmay play a role in the patho-
genesis of PD.
Endoplasmic reticulum stress
Accumulation of misfolded proteins can trigger a reticu-
lum stress response which is manifested by the activation
of certain markers such as pancreatic endoplasmic retic-
ulum kinase (PERK) and eukaryotic initiation factor 2 a
(eIF2a). Phosphorylated PERK and phosphorylated eIF2a
expression is increased in dopaminergic neurons of the
substantia nigra in advanced stages of PD (Hoozemans
et al. 2007). Increased eIF2aand phosphorylated eIF2aare
also found in neurons of the substantia nigra pars com-
pacta, they partly co-localize with a-synuclein inclusions
in pPD (Fig. 6). Together, these observations support the
idea that endoplasmic reticulum stress plays a role in the
pathogenesis of early stages of PD (Wang and Takahashi
2007).
Protein degradation: the ubiquitin–proteasome system
and autophagy in iPD
LBs and neurites are manifestations of impaired protein
degradation in vulnerable cells. Since protein degradation
putatively occurs via cytosolic proteases, chaperone-
mediated autophagy (CMA), ubiquitin–proteasome system
(UPS) and micro-, macroautophagy, all these mechanisms
have been analysed in experimental models of PD. How-
ever, much less is known about what happens in PD.
Several in vitro studies have shown impaired UPS
function in PD (Olanow and McNaught 2006). In favour of
impaired UPS function is the cardinal early observation
that ubiquitin occurs in Lewy inclusions and that p62
immunoreactivity appears in association with abnormal
a-synuclein inclusions at early stages of LB (Kuusisto et al.
2003). However, the sequence of events is obscure. Mor-
phological evidence in PD neurons shows fine a-synuclein
deposition in the cytoplasm of selected neurons. Ubiquitin
and p62 decorates some of but not all these inclusions
Neuropathology of sporadic Parkinson disease 829
123
suggesting that abnormal synuclein deposition is a first step
followed by ubiquitination of a-synuclein and association
of p62 to polyubiquitinated proteins (Kuusisto et al. 2003;
Nakaso et al. 2004). More recent observations have pro-
vided evidence of altered expression of components of the
UPS in PD (Chu et al. 2009). Moreover, some of them
appear to be modified by oxidation thus resulting in lost of
function (Choi et al. 2004; Chung et al. 2004; see later).
Therefore, altered degradation by the UPS of abnormal
a-synuclein and several hundred proteins associated with
LBs does probably depend on multiple factors (Shamoto-
Nagai et al. 2007; Chu et al. 2009).
The role of autophagy, including CMA and macro-
autophagy in PD, is postulated mainly on the basis of
observations in cell models (Stefanis et al. 2001; Cuervo
et al. 2004; Engelender 2008; Martı
´nez-Vicente et al.
2008; Xilouri et al. 2008, Vogiatzi et al. 2008; Mak et al.
2010). Little is known, however, about the role of
autophagy in human PD. Several morphological studies
have shown that neuromelanin in the substantia nigra is
associated with the autophagosome/lysosome system
(Anglade et al. 1997; Tribl et al. 2006) thus providing a
possible subtle link between a-synuclein and lysosomes in
the substantia nigra under normal conditions and in PD.
Certain autophagy markers are altered in the substantia
nigra in PD (Pan et al. 2008). More straightforward in
the present context is the observation that the expression
and localization of proteins linked with autophagy and
Fig. 4 a Increased neuroketal expression in total homogenates of the
substantia nigra in pPD (iPD) compared with controls (CTL).
Accompanying diagram illustrtes significant differences P\0.001
(Student’s ttest). bIncreased oxidative damage to nucleic acids of
glial cells in the substantia nigra of pPD at stage 3. Double-labelling
immunofluorescence and confocal microscopy to 8-hydroxyguanine
(8-dOHG) (green) and a-synuclein (red) shows green staining in the
nuclei of several glial cels but not in neurons with a-synuclein
inclusions. Nuclei (blue) are stained with TO-PRO
830 I. Ferrer et al.
123
Fig. 5 Double-labelling immunofluorescence and confocal micros-
copy of ferritin (green) and a-synuclein (red)(af), a-synuclein
(green) and globin a-chain (red) (G-L) in the substantia nigra pars
compacta in pPD stage 3. Ferritin is decreased in neurons with
a-synuclein inclusions (compare acwith df). Similarly, globin
a-chain expression is reduced in neurons with a-synuclein inclusions
(compare giwith jl)
Neuropathology of sporadic Parkinson disease 831
123
lysosomes as microtubule-associated protein 1 light chain
3 alpha (LC3), lysosomal-associated protein 1 (LAMP1)
and LAMP2 (Klionsky et al. 2008) are altered in pPD
(Fig. 7). In the same line, recent findings suggest that
CMA activity is reduced in PD brain (Alvarez-Erviti et al.
2010).
Fig. 6 Double-labelling immunofluorescence and confocal micros-
copy of eif2a(green) and a-synuclein (red)(af), and eif2a(green)
and eif2a-P (red)(gl) in the substantia nigra pars compacta in pPD
stage 3. eif2aco-localizes with a-synuclein inclusions (af). The
majority of eif2ais phosphorylated (gl) thus suggesting activation of
the reticulum stress responses
832 I. Ferrer et al.
123
In summary, experiments in vitro strongly support the
idea of altered UPS and autophagosome/lysosome function
in models of altered a-synuclein turnover and clearance,
but precise information about the amount of a-synuclein
cleared by the UPS and the CMA systems is not known in
human disease.
Fig. 7 Double-labelling immunofluorescence and confocal micros-
copy of microtubule-associated protein 1 light chain 3 alpha (LC3)
(green) and a-synuclein (red)(ac), lysosomal associated protein 2
(LAMP2) (green) and a-synuclein (red)(df), LAMP1 (green)
and a-synuclein (red)(gh) in substantia nigra pars compacta in
pPD stage 3. Markers of autophagy are expressed in association with
a-synuclein aggregates. jlNegative controls processed without
primary antibodies
Neuropathology of sporadic Parkinson disease 833
123
The basal ganglia in pPD
Even in the absence of motor symptoms, loss of neurons in
the substantia nigra translates into loss of dopaminergic
innervation and consequent remodelling of the putamen.
This is clearly illustrated by the loss of tyrosine hydroxy-
lase (TH) immunoreactivity and loss of vesicular mono-
amine transporter 2 (VMAT2) in the putamen in pPD, with
values that are intermediate between those in normal
individuals and in parkinsonian stages of PD (DelleDonne
et al. 2008; Dickson et al. 2008) (Fig. 8). These morpho-
logical aspects are also sustained by observations using
serial metabolic imaging with [(18)F]-fluorodeoxyglucose
positron emission tomography (PET) in sporadic cases
(Tang et al. 2010). Multitracer PET scans are, therefore,
useful tools to unveil dopaminergic dysfunction in patients
with suspected pPD.
The balance of other molecules is also modified in the
striatum in pPD. Leu-encephalin levels are reduced in the
putamen and undetectable in the substantia nigra in PD, but
Leu-encephalin levels are only barely reduced in the
putamen in pPD (Fernandez et al. 1996).
Adenosine receptors 2A (A
2A
Rs) in the striatum are
practically restricted to GABAergic neurons projecting
from the caudate and putamen to the external globus pal-
lidus, which also expresses dopamine D
2
receptors (D
2
Rs)
(Fuxe et al. 2007). A
2A
Rs expression levels in the striatum
are increased in PD and correlate with motor symptoms
(Varani et al. 2010). Recently, we have seen increased
A
2A
R expression levels in the caudate in cases with pPD
stage 3 of Braak, thus indicating that this is an early
abnormality of the denervated striatum (Buira et al. 2010).
Finally, early and pre-symptomatic stages of PD are
associated with desensitization/downregulation of type 1
cannabinoid (CB1) receptors, whereas advanced stages are
characterized by up-regulatory responses of CB1 receptors
(Garcı
´a-Arencibia et al. 2009). Whether down-regulation
of CB1 receptors at early stages is pathogenic remains to be
elucidated; this may constitute an important putative target
for therapeutic intervention. Decreased CB1 receptor
expression is also observed in the caudate in pPD, showing
that this is a very early alteration in the course of PD
(Fig. 8).
The olfactory bulb and tract in pPD
In the present series, the morphological study of the
olfactory bulb and tract revealed the presence of small
numbers of neurons and neurites bearing a-synuclein
aggregates which are also recognized with antibodies
against nitrated a-synuclein and against phospho-specific
a-synuclein Ser129 antibodies (Fig. 9). However, it is hard
to assume that such a small number of structural anomalies
may account for the sophisticated olfactory deficits that
include not only loss of olfaction but also altered dis-
crimination of odours with increased perception in some
circumstances. It may be hypothesized that, as in other
regions, olfactory alterations in pPD and PD are the result
of more complicated settings resulting from several
molecular deficits.
The cerebral cortex in pPD
Altered behaviour, apathy and depression may occur in
patients with pPD (Poewe 2008; McKinlay et al. 2009;
Pedersen et al. 2009). The molecular substrates of such
alterations are scarcely known but pieces of knowledge are
rapidly growing (Ferrer 2009a).
Brain cortex and mitochondrial O
2
uptake and complex I
activity are significantly lower in PD, whereas mtNOS
activity, cytochrome content, expression of SOD2, mito-
chondrial mass, and oxidative damage are significantly
higher in the frontal cortex in PD. The decreases in tissue
and mitochondrial O
2
uptake and in complex I activity are
considered the consequences of mitochondrial oxidative
damage in the cerebral cortex in PD (Navarro et al. 2009).
Unfortunately, no similar data are available in pPD.
Recent observations have shown abnormal lipid com-
position in the frontal cortex in pPD which is manifested
Fig. 8 Expression levels of tyrosine hydroxylase (TH) and cannab-
inoid 1 receptor (CB-1 receptor) in the caudate nucleus in controls
and in pPD (iPD Braak 3). Reduced expression of TH (62 kDa) and
CB-1 receptor (52 kDa) is observed in diseased cases when compared
with controls (P\0.01, Student’s ttest)
834 I. Ferrer et al.
123
with significantly increased expression levels of the highly
peroxidizable DHA and increased peroxidability index
(Dalfo
´et al. 2005). More dramatically, several key proteins
are targets of oxidative damage in the frontal cortex, as
revealed by bi-dimensional gel electrophoresis, redox
proteomics and mass spectrometry, in pPD including
a-synuclein, b-synuclein and SOD2 (Dalfo
´et al. 2005;
Dalfo
´and Ferrer 2008). In addition, increased oxidative
damage of aldolase A, enolase 1 and glyceraldehyde
dehydrogenase (GAPDH), all of them involved in glycol-
ysis and energy metabolism, is found in the frontal cortex
in pPD (and PD as well) (Go
´mez and Ferrer 2009). Other
oxidatively damaged proteins are phosphoprotein enriched
in astrocytes 15, SH3 domain binding glutamic acid-rich
protein like, ubiquitin-conjugating enzyme E2N-like, pro-
hibitin, proteasome subunit Y and thioredoxin. Finally,
cortical synapses are abnormal in pPD (and PD), as tau
phosphorylation and a-synuclein phosphorylation are
increased in synaptic-enriched fractions of frontal cortex
homogenates (Muntane
´et al. 2008).
Concluding comments
Until recently, PD was considered to be a movement dis-
ease mainly characterized by the loss of dopaminergic
neurons and occurrence of LBs in the remaining cells in the
substantia nigra. This point of view has served to drive
major advances in the treatment of patients with PD,
including pharmacological and neurosurgical therapeutic
interventions geared to balancing dopamine deficits and
related neurotransmitters and modulators in the nigrostri-
atal system. However, PD is a systemic disease affecting
several distinct neuronal populations of the central and
peripheral nervous system, with the appearance of motor
symptoms in a substantial number of patients as the top of a
plethora of non-motor signs and symptoms affecting the
heart, gastrointestinal tract, olfaction, sleep and mental
functions. Moreover, neuropathological and clinical studies
have shown only a partial correlation between regional
a-synuclein inclusions and corresponding neurological
deficits at early stages of the disease, and no clear relation
between LBs, and motor and cognitive impairment.
Cumulative evidence in recent years indicates that mito-
chondrial dysfunction, oxidative stress and oxidative
damage to crucial proteins, post-translational modifications
of proteins, and endoplasmic reticulum stress converge in
the pathogenesis of PD. This is further supported by pro-
teomic studies showing dysregulation of proteins involved
in energy metabolism, oxidative stress, signal transduction,
electron transport and detoxification pathways in PD
(Srivastava et al. 2010). Furthermore, the use of redox
proteomics has revealed that protein altered by oxidative
damage may have an impact on protein function in spite of
apparently normal total levels of the protein (Martı
´nez
et al. 2010). Importantly, some of these changes occur at
very early stages of PD-related pathology in areas with no
accompanying Lewy-like inclusions, such as the substantia
Fig. 9 Images of the olfactory bulb at stages 2 and 3 of Braak. Scattered neurites and cell bodies are immunoreactive with anti-a-synuclein
antibodies. Paraffin sections slightly counterstained with haematoxylin
Neuropathology of sporadic Parkinson disease 835
123
nigra in stage 2, and the striatum and cerebral frontal cortex
in stages 2 and 3 of Braak. Furthermore, although still
patchy, several pieces of information suggest that failure in
energy metabolism may have an additive deleterious effect
on neuronal function. The term ‘exhausted neuron’ was
applied in the context of Alzheimer disease to describe the
particular situation of neurons working under suboptimal
conditions of reduced energy production, increased energy
demands and oxidative damage (Ferrer 2009b). This term
can also be applied to pPD and PD in which neurons suffer
from a convergence of intrinsic and external deficiencies
that may impair neuronal function and eventually result in
cell death. Recognition of this scenario helps us understand
day-to-day variations in neurological deficits, fluctuations,
apparent transient recoveries and subtle mining of neuronal
integrity in neurodegenerative disease. More importantly,
many of these alterations may be targets of therapeutic
intervention geared to delaying disease progression.
Acknowledgments Work carried out in the Institute of Neuropa-
thology was partially funded by grants from the Spanish Ministry of
Health, Instituto de Salud Carlos III PI05/1570, PI05/2214 and PI08/
0582, and supported by the European Commission: BrainNet Europe
II, LSHM-CT-2004-503039 and INDABIP FP6-2005-LIFESCI-
HEALTH-7 Molecular Diagnostics. Thanks to T. Yohannan for
editorial help.
References
Alvarez-Erviti L, Rodriguez-Oroz MC, Cooper JM, Caballero C,
Ferrer I, Obeso JA, Schapira AHV (2010) Chaperone-mediated
autophagy markers are abnormal in Parkinson’s disease brain.
Arch Neurol [Epub ahead of print]
Andersen JK (2004) Iron dysregulation and Parkinson’s disease.
J Alzheimers Dis 6:S47–S52
Anderson JP, Walker DE, Goldstein JM, Laat R, Banducci K,
Caccavello RJ, Barbour R, Huang J, Kling K, Lee M, Diep L,
Keim PS, Shen X, Chartaway T, Schlossmacher MG, Seubert P,
Schenk D, Sinha S, Gai WP, Chilcote TJ (2006) Phosphorylation
of Ser 129 is the dominant pathological modification of
synuclein in familial and sporadic Lewy body disease. J Biol
Chem 281:29739–29759
Anglade P, Vyas S, Javoy-Agid F, Herrero MT, Michel PP, Marquez
J, Mouatt-Prigent A, Ruberg M, Hirsch EC, Agid Y (1997)
Apoptosis and autophagy in nigral neurons of patients with
Parkinson’s disease. Histol Histopathol 12:25–31
Baba M, Nakajo S, Tu PH, Tomita T, Lee VM, Trojanowski JQ,
Iwatsubo T (1998) Aggregation of a-synuclein in Lewy bodies of
sporadic Parkinson’s disease and dementia with Lewy bodies.
Am J Pathol 152:879–884
Barnham KJ, Bush AI (2008) Metals in Alzheimer’s and Parkinson’s
diseases. Curr Opin Chem Biol 12:222–228
Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J,
Sasse J, Boyer S, Shirohi S, Brooks R, Eschbacher J, White CL
3rd, Akiyama H, Caviness J, Shill HA, Connor DJ, Sabbagh MN,
Walker DG, Arizona Parkinson’s Disease Consortium (2009)
Unified staging system for Lewy body disorders: correlation with
nigrostriatal degeneration, cognitive impairment and motor
dysfunction. Acta Neuropathol 117:613–634
Biagioli M, Pinto M, Cesselli D, Zaninello M, Lazarevic D,
Roncaglia P, Simone R, Vlachouli C, Plessy C, Bertin N,
Beltrami A, Kobayashi K, Gallo V, Santoro C, Ferrer I, Rivella
S, Beltrami CA, Carninci P, Raviola E, Gustincich S (2009)
Unexpected expression of alpha- and beta-globin in mesence-
phalic dopaminergic neurons and glial cells. Proc Natl Acad
Sci 106:15454–15459
Bloch A, Probst A, Bissing H, Adams H, Tolnay M (2006) Alpha-
synuclein pathology of the spinal and peripheral autonomic
nervous system in neurologically unimpaired elderly subjects.
Neuropathol Appl Neurobiol 32:284–295
Braak H, del Tredici K (2008) Cortico-basal ganglia-cortical circuitry
in Parkinson’s disease reconsidered. Exp Neurol 212:226–229
Braak H, Sandmann-Keil D, Gai W, Braak E (1999) Extensive axonal
Lewy neurites in Parkinson’s disease: a novel pathological
feature revealed by alpha-synuclein immunocytochemistry.
Neurosci Lett 265:67–69
Braak H, del Tredici K, Bratzke H, Hamm-Clement J, Sandemann-
Keil D, Rub U (2002) Staging of the intracerebral inclusion body
pathology associated with idiopathic Parkinson’s disease (pre-
clinical and clinical stages). J Neurol 249(suppl 3):1–5
Braak H, del Tredici K, Ru
¨b U, de Vos RAI, Jansen Steur ENH,
Braak E (2003) Staging of brain pathology related to sporadic
Parkinson’s disease. Neurobiol Aging 24:197–211
Braak H, Ghebremedhin E, Ru
¨b U, Bratzke H, del Tredici K (2004)
Stages in the development of Parkinson’s disease-related
pathology. Cell Tissue Res 318:121–134
Braak H, Ru
¨b U, Jansen Steur EN, del Tredici K, de Vos RA (2005)
Cognitive status correlates with neuropathologic state in Parkin-
son disease. Neurology 64:1404–1410
Braak H, Muller CM, Ru
¨b U, Ackermann H, Bratzke H, de Vos RA,
del Tredici K (2006) Pathology associated with sporadic
Parkinson’s disease: where does it end? J Neural Transm Suppl
70:89–97
Buira SP, Dentesano G, Albasanz JL, Moreno J, Juve
´sS,Martı
´nM,
Ferrer I, Barrachina M (2010) Increased striatal ying yang-1 levels
in Parkinson ‘s disease: a potential negative regulator of adenosine
A
2a
receptor expression levels. J Neurochem 112:1273–1285
Chaudhuri KR, Yates L, Martı
´nez-Martı
´n P (2005) The non-motor
symptom complex of Parkinson’s disease: a comprehensive
assessment is essential. Curr Neurol Neurosci Rep 5:275–283
Chiba-Falek O, Lopez GJ, Nussbaum RL (2006) Levels of alpha-
synuclein mRNA in sporadic Parkinson disease patients. Mov
Disord 21:1703–1708
Chinta SJ, Andersen JK (2008) Redox imbalance in Parkinson’s
disease. Biochem Biophys Acta 1780:1362–1367
Choi J, Levey AI, Weintraub ST, Rees HD, Gearing M, Chin LS, Li L
(2004) Oxidative modifications and down-regulation of ubiquitin
carboxyl-terminal hydrolase L1 associated with idiopathic
Parkinson’s and Alzheimer’s diseases. J Biol Chem 279:
13256–13264
Choi J, Rees HD, Weintraub ST, Levey AI, Chin LS, Li L (2005)
Oxidative modifications and aggregation of Cu, Zn-superoxide
dismutase associated with Alzheimer and Parkinson diseases.
J Biol Chem 280:11648–11655
Chu Y, Dodiya H, Aebischer P, Olanow CW, Kordower JH (2009)
Alterations in lysosomal and proteasomal markers in Parkinson’s
disease: relationship to alpha-synuclein inclusions. Neurobiol
Dis 35:385–398
Chung KK, Thomas B, Li X, Pletnikova O, Troncoso JC, Marsh L,
Dawson VL, Dawson TM (2004) S-Nitrosylation of parkin
regulates ubiquitination and compromises Parkin’s protective
function. Science 304:1328–1331
Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D (2004)
Impaired degradation of mutant alpha-synuclein by chaperone-
mediated autophagy. Science 305:1292–1295
836 I. Ferrer et al.
123
Dalfo
´E, Ferrer I (2008) Early a-synuclein lipoxidation in neocortex
in Lewy body diseases. Neurobiol Aging 29:408–417
Dalfo
´E, Barrachina M, Rosa JL, Ambrosio S, Ferrer I (2004)
Abnormal a-synuclein interactions with rab3a and rabphilin in
diffuse Lewy body disease. Neurobiol Dis 16:92–97
Dalfo
´E, Portero-Otı
´n M, Ayala V, Martı
´nez A, Pamplona R, Ferrer I
(2005) Evidence of oxidative stress in the neocortex in incidental
Lewy body disease. J Neuropathol Exp Neurol 64:1–13
Daniel SE, Hawkes CH (1992) Preliminary diagnosis of Parkinson’s
disease by olfactory bulb pathology. Lancet 340:186
Del Tredici K, Ru
¨b U, De Vos RA, Bohl JR, Braak H (2002) Where
does Parkinson disease pathology begin in the brain? J Neuro-
pathol Exp Neurol 61:413–426
DelleDonne A, Klos KJ, Fujishiro H, Ahmed Z, Parisi JE, Josephs
KA, Frigerio R, Burnett M, Wszolek ZK, Uiti RJ, Ahlskog JE,
Dickson DW (2008) Incidental Lewy body disease and preclin-
ical Parkinson disease. Arch Neurol 65:1074–1080
Dexter DT, Sian J, Rose S, Hindmarsh JG, Mann VM, Cooper JM,
Wells FR, Daniel SE, Lees AJ, Schapira AH (1994) Indices of
oxidative stress and mitochondrial function in individuals with
incidental Lewy body disease. Ann Neurol 35:38–44
Dickson DW (2001) Alpha-synuclein and the Lewy body disorders.
Curr Opin Neurol 14:423–432
Dickson DW, Fujishiro H, DelleDonne A, Menke J, Ahmed Z, Klos KJ,
Josephs KA, Frigerio R, Burnett M, Parisi JE, Ahlskog JE (2008)
Evidence that incidental Lewy body disease is pre-symptomatic
Parkinson’s disease. Acta Neuropathol 115:437–444
Dickson DW, Uchikado H, Fujishiro H, Tsuboi Y (2010) Evidence in
favor of Braak staging of Parkinson’s disease. Mov Disord
25(Suppl 1):S78–S82
Digicaylioglu M, Bichet S, Marti HH, Wenger RH, Rivas LA, Bauer
C, Gassmann M (1995) Localization of specific erythropoietin
binding sites in defined areas of the human brain. Proc Natl Acad
Sci USA 93:3717–3720
Double KL (2006) Functional effects of neuromelanin and synthetic
melanin in model systems. J Neuarl Transm 113:751–756
Double KL, Halliday GM (2006) New face of neuromelanin. J Neural
Transm Suppl 70:119–123
Duda JE, Giasson BI, Chen Q, Gur TL, Hurtig HI, Stern MB,
Gollomp SM, Ischiropoulos H, Lee VM, Trojanowski JQ (2000)
Widespread nitration of pathological inclusions in neurodegen-
erative synucleinopathies. Am J Pathol 157:1439–1445
ECh Wolters, Braak H (2006) Parkinson’s disease: pre-motor clinico-
pathological correlations. J Neural Transm Suppl 70:309–319
Engelender S (2008) Ubiquitination of alpha synuynucelin and
autophagy in Parkinson’s disease. Autophagy 4:372–374
Fasano M, Bergamasco B, Lopiano L (2006) Modifications of the
iron-neuromelanin system in Parkinson’s disease. J Neurochem
96:909–916
Fernandez A, de Ceballos ML, Rose S, Jenner P, Marsden CD (1996)
Alterations in peptide levels in Parkinson’s disease and inciden-
tal Lewy body disease. Brain 119:823–830
Ferrer I (2009a) Early involvement of the cerebral cortex in
Parkinson’s disease: convergence of multiple metabolic defects.
Progr Neurobiol 88:89–103
Ferrer I (2009b) Altered mitochondria, energy metabolism, voltage-
dependent anion channel, and lipid rafts converge to exhaust
neurons in Alzheimer’s disease. J Bioenerg Biomembr
41:425–431
Ferrer I, Blanco R, Carmona M, Puig B, Barrachina M, Go
´mez C,
Ambrosio S (2001) Active, phosphorylation-dependent mitogen-
activated protein kinase (MAPK/ERK), stress-activated protein
kinase/c-Jun N-terminal kinase (SAPK/JNK), and p38 kinase
expression in Parkinson’s disease and dementia with Lewy
bodies. J Neural Transm 108:1383–1396
Forno LS (1996) Neuropathology of Parkinson’s disease. J Neuropa-
thol Exp Neurol 55:259–272
Frigerio R, Fujishiro H, Maraganore DM, Klos KJ, DelleDonne A,
Heckman MG, Crook JE, Josephs KA, Parisi JE, Boeve BF,
Dickson DW, Ahlskog JE (2009) Comparison of risk factor
profiles in incidental Lewy body disease and Parkinson disease.
Arch Neurol 66:1114–1119
Fujiwara H, Hasegawa M, Dohmae N, Kawashima A, Masliah E,
Goldberg MS, Shen J, Takio K, Iwatsubo T (2002) a-synuclein is
phosphorylated in synucleinopathy lesions. Nat Cell Biol
4:160–164
Fuxe K, Marcellino D, Genedani S, Agnati L (2007) Adenosine
A(2A) receptors, dopamine D(2) receptors and their interactions
in Parkinson’s disease. Mov Disord 22:1990–2017
Garcı
´a-Arencibia M, Garcı
´a C, Ferna
´ndez-Ruiz J (2009) Cannabi-
noids and Parkinson’s disease. CNS Neurol Disord Drug Targets
8:432–439
Gerlach M, Double KL, Youdim MB, Riederer P (2006) Potential
sources of increased iron in the substantia nigra of parkinsonian
patients. J Neural Transm Suppl 70:133–142
Giasson BI, Duda JE, Murray IV, Chen Q, Souza JM, Hurtig HI,
Ischiropuolos H, Trojanowski JQ, Lee VMY (2000) Oxidative
damage linked to neurodegeneration by selective a-synuclein
nitration in synucleinopathy lesions. Science 290:985–989
Goedert M (2001) Parkinson’s disease and other a-synucleinopathies.
Clin Chem Lab Med 39:308–312
Go
´mez A, Ferrer I (2009) Increased oxidation of certain glycolysis
and energy metabolism enzymes in the frontal cortex in Lewy
body diseases. J Neurosci Res 87:1002–1013
Halliday GM, Ophof A, Broe M, Jensen PH, Kettle E, Fedorow H,
Cartwright MI, Griffiths FM, Shepherd CE, Double KL (2005)
Alpha-synuclein redistributes to neuromelanin lipid in the
substantia nigra early in Parkinson’s disease. Brain 128:2654–
2664
Hashimoto M, Masliah E (1999) a-synuclein in Lewy body disease
and Alzheimer’s disease. Brain Pathol 9:707–720
Herting B, Schulze S, Reichmann H, Haehner A, Hummel T (2008) A
longitudinal study of olfactory function in patients with
idiopathic Parkinson’s disease. J Neurol 255:367–370
Hirsch EC (2006) Altered regulation of iron transport and storage in
Parkinson’s disease. J Neural Transm Suppl 71:201–204
Hoozemans JJ, van Haastert ES, Eikelenboom P, de Vos RA,
Rozemuller JM, Scheper W (2007) Activation of the unfolded
protein response in Parkinson’s disease. Biochem Biophys Res
Commun 354:707–711
Hubbard PS, Esiri MM, Reading M, McShane R, Nagy Z (2007)
a-synuclein pathology in the olfactory pathways of dementia
patients. J Anat 211:117–124
Iranzo A, Molinuevo JL, Santamaria J, Serradell M, Marti MJ,
Vallderiola F, Tolosa E (2006) Rapid-eye-movement sleep
behaviour disorder as an early marker for a neurodegenerative
disorder: a descriptive study. Lancet Neurol 5:572–577
Ishii A, Nonaka T, Taniguchi S, Saito T, Arai T, Mann D, Iwatsubo T,
Hisanaga S, Goedert M, Hasegawa M (2007) Casein kinase 2 is
the major enzyme in brain that phosphorylates Ser129 of human
alpha-synuclein: implication for alpha-synucleinopathies. FEBS
Lett 581:4711–4717
Iwanaga K, Wakabayashi K, Yoshimoto M, Tomita I, Satoh H,
Takashima H, Satoh M, Tsujihara M, Takahashi H (1999) Lewy
body-type degeneration in cardiac plexus in Parkinson’s and
incidental Lewy body disease. Neurology 52:1269–1271
Jellinger KA (2004) Lewy body-related synucleinopathy in the aged
human brain. J Neural Transm 111:1219–1235
Jellinger KA (2008) A critical reappraisal of current staging of Lewy-
related pathology in human brain. Acta Neuropathol 116:1–16
Neuropathology of sporadic Parkinson disease 837
123
Jellinger KA (2009) A critical evaluation of current staging of alpha-
synuclein pathology in Lewy body disorders. Biochim Biophys
Acta 1792:730–740
Jellinger KA (2010) Prevalence and impact of cerebrovascular lesions
in Alzheimer and Lewy body diseases. Neurodegener Dis
7:112–115
Jellinger K, Mizuno Y (2003) Parkinson’s disease. In: Dickson D (ed)
Neurodegeneration: the molecular pathology of dementia and
movement disorders. ISN Neuropathol Press, Bassel, pp 159–187
Jenner P (1993) Altered mitochondrial function, iron metabolism and
glutathione levels in Parkinson’s disease. Acta Neurol Scand
Suppl 146:6–13
Jenner P (1998) Oxidative mechanisms in nigral cell death in
Parkinson’s disease. Mov Disord 13:24–34
Jenner P, Dexter DT, Sian J, Schapira AH, Marsden CD (1992)
Oxidative stress as a cause of nigral cell death in Parkinson’s
disease and incidental Lewy body disease. The Royal Kings and
Queens Parkinson’s Disease Research Group. Ann Neurol
32:S82–S87
Keeney PM, Xie J, Capaldi RA, Bennett JP (2006) Parkinson’s
disease brain mitochondrial complex I has oxidatively damaged
subunits and is functionally impaired and misassembled.
J Neurosci 26:5256–5264
Kingsbury AE, Daniel SE, Sangha H, Eisen S, Lees AJ, Foster OJ
(2004) Alteration in alpha-synuclein mRNA expression in
Parkinson’s disease. Mov Disord 19:162–170
Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G,
Askew DS et al (2008) Guidelines for the use and interpretation
of assays for monitoring autophagy in higher eukaryotes.
Autophagy 4:151–175
Koziorowski D, Friedman A, Arosio P, Santambrogio P, Dziewulska
D (2007) ELISA reveals a difference in the structure of substantia
nigra ferritin in Parkinson’s disease and incidental Lewy body
compared to control. Parkinsonism Relat Disord 13:214–218
Kuusisto E, Parkkinen L, Alafuzoff I (2003) Morphogenesis of Lewy
bodies: dissimilar incorporation of alpha-synuclein, ubiquitin,
and p62. J Neuropathol Exp Neurol 62:1241–1253
Leverenz JB, Umar I, Wang Q, Montine TJ, McMillan PJ, Tsuang
DW, Jin J, Pan C, Shin J, Zhu D, Zhang J (2007) Proteomic
identification of novel proteins in cortical Lewy bodies. Brain
Pathol 17:139–145
Li W, West N, Colla E, Pletnikova O, Troncoso JC, Marsh L, Dawson
TM, Jakala P, Hartmann T, Price DL, Lee MK (2005)
Aggregation promoting C-terminal truncation of alpha-synuclein
is a normal cellular process and is enhanced by the familial
Parkinson’s disease-linked mutations. Proc Natl Acad Sci USA
102:2162–2167
Mak SK, McCormack AL, Manning-Bog AB, Cuervo AM, Di Monte
DA (2010) Lysosomal degradation of a-synuclein in vivo. J Biol
Chem 285:13621–13629
Markesbery WR, Jicha GA, Liu H, Schmitt FA (2009) Lewy body
pathology in normal elderly subjects. J Neuropathol Exp Neurol
68:816–822
Martı
´nez A, Portero-Otin M, Pamplona R, Ferrer I (2010) Protein
targets of oxidative damage in human neurodegenerative diseases
with abnormal protein aggregates. Brain Pathol 20:281–297
Martı
´nez-Vicente M, Talloczy Z, Kaushik S, Massey AC, Mazzulli J,
Mosharov EV, Hodara R, Fredenburg R, Wu DC, Follenzi A,
Dauer W, Przedborski S, Ischiropoulos H, Lansbury PT, Sulzer
D, Cuervo AM (2008) Dopamine-modified alpha-synuclein
blocks chaperone-mediated autophagy. J Clin Invest 118:777–
788
Matsuda S, Okano M, Yamagishi K, Nagao M, Ueda M, Sasaki R
(1994) A novel site of erythropoietin production. Oxygen-
dependent production in cultured rat astrocytes. J Biol Chem
269:19488–19493
McKinlay A, Grace RC, Dalrymple-Alford JC, Roger D (2009)
Cognitive characteristics associated with mild cognitive impair-
ment in Parkinson’s disease. Dement Geriatr Cogn Disord
28:121–129
Mizuno Y, Ohta S, Tanaka M, Takamiya S, Suzuki K, Sato T, Oya H,
Ozawa T, Kagawa Y (1989) Deficiencies in complex I subunits
of the respiratory chain in Parkinson’s disease. Biochem Biophys
Res Commun 163:1450–1455
Muntane
´G, Ferrer I, Martı
´nez-Vicente M a-Synuclein phosphoryla-
tion and lysosomal-mediated a-synuclein truncation are normal
events in the adult human brain (in preparation)
Muntane
´G, Dalfo
´E, Martinez A, Ferrer I (2008) Phosphorylation of
tau and alpha-synuclein in synaptic-enriched fractions of the
frontal cortex in Alzheimer’s disease, and in Parkinson’s disease
and related a-synucleinopathies. Neuroscience 152:913–923
Murray IV, Giasson BI, Quinn SM, Koppaka V, Axelsen PH,
Ischiropoulos H, Trojanowski JQ, Lee VM (2003) Role of alpha-
synuclein carboxy-terminus on fibril formation in vitro. Bio-
chemistry 42:8530–8540
Nakaso K, Yoshimoto Y, Nakano T, Takeshima T, Fukuhara Y, Yasui
K, Araga S, Yanagawa T, Ishii T, Nakashima K (2004)
Transcriptional activation of p62/A170/ZIP during the formation
of the aggregates: possible mechanisms and the role in Lewy
body formation in Parkinson’s disease. Brain Res 1012:42–51
Natale G, Pasquali L, Riuggieri S, Paparelli A, Fornai F (2008)
Parkinson’s disease and the gut: a well known clinical associ-
ation in need of an effective cure and explanation. Neurogas-
troenterol Motil 20:741–749
Navarro A, Boveris A, Ba
´ndez MJ, Sa
´nchez-Pino MJ, Go
´mez C,
Muntane
´G, Ferrer I (2009) Human brain cortex: mitochondrial
oxidative damage and adaptive response in Parkinson disease
and in dementia with Lewy bodies. Free Radic Biol Med
46:1574–1580
Oakley AE, Collingwood JF, Dobson J, Love G, Perrott HR,
Edwardson JA, Elstner M, Morris CM (2007) Individual
dopaminergic neurons show raised iron levels in Parkinson
disease. Neurology 68:1820–1825
Olanow CW, McNaught KS (2006) Ubiquitin-proteasome system and
Parkinson’s disease. Mov Disord 21:1806–1823
Onyango IG (2008) Mitochondrial dysfunction and oxidative stress in
Parkinson’s disease. Neurochem Res 33:589–597
Pan T, Kondo S, Le W, Jankovic J (2008) The role of autophagy-
lysosome pathway in neurodegeneration associated with Parkin-
son’s disease. Brain 131:1969–1978
Parker WD, Boyson SJ, Parks JK (1989) Abnormalities of the
electron transport chain in idiopathic Parkinson’s disease. Ann
Neurol 26:719–723
Parkkinen L, Kauppinen T, Pirttila
¨T, Autere JM, Alafuzoff I (2005)
a-Synuclein pathology does not predict extrapyramidal symp-
toms or dementia. Ann Neurol 57:82–91
Parkkinen L, Pirttila
¨T, Alafuzoff I (2008) Applicability of current
staging/categorization of a-synuclein pathology and their clinical
relevance. Acta Neuropathol 115:399–407
Pearce RK, Hawkes CH, Daniel SE (1995) The anterior olfactory
nucleus in Parkinson’s disease. Mov Disord 10:283–287
Pedersen KF, Alves G, Aarsland D, Larsen JP (2009) Occurrence and
risk factors for apathy in Parkinson disease: a 4-year prospective
longitudinal study. J Neurol Neurosurg Psychiatry 80:1279–
1782
Poewe W (2007) Dysautonomia and cognitive dysfunction in
Parkinson’s disease. Mov Disord Suppl 17:S374–S378
Poewe W (2008) Non-motor symptoms in Parkinson’s disease. Eur J
Neurol 15(Suppl 1):14–20
Postuma RB, Lang AE, Massicotte-Marquez J, Montplaisir J (2006)
Potential early markers of Parkinson disease in idiopathic REM
sleep behavioral disorder. Neurology 66:845–851
838 I. Ferrer et al.
123
Richter F, Meurers BH, Zhu C, Medvedeva VP (2009) Neurons
express haemoglobin a- and b-chains in rat and human brains.
J Comp Neurol 515:538–547
Rockenstein E, Hansen LA, Mallory M, Trojanowski JQ, Galasko D,
Masliah E (2001) Altered expression of the synuclein family
mRNA in Lewy body and Alzheimer’s disease. Brain Res
914:48–56
Ross GW, Petrovitch H, Abbott RD, Tanner CM, Popper J, Masaki K,
Launer L, White LR (2008) Association of olfactory dysfunction
with risk for future Parkinson’s disease. Ann Neurol 63:167–173
Ryu MY, Kim DW, Arima K, Mouradian MM, Kim SU, Lee G
(2008) Localization of CKII beta subunits in Lewy bodies of
Parkinson’s disease. J Neurol Sci 266:9–12
Saito Y, Ruberu NN, Sawabe M, Arai T, Kazama H, Hosoi T,
Yamanouchi H, Murayama S (2004) Lewy body-related a-syn-
ucleinopathy in aging. J Neuropathol Exp Neurol 63:742–749
Salazar J, Mena N, Nu
´n
˜ez MT (2006) Iron dyshomeostasis in
Parkinson’s disease. J Neural Transm Suppl 71:205–213
Schapira AH (2008) Mitochondria in the aetiology and pathogenesis
of Parkinson’s disease. Lancet Neurol 7:97–109
Schults CW (2006) Lewy bodies. Proc Nat Acad Sci USA
103:1661–1668
Shamoto-Nagai M, Maruyama W, Hashizume Y, Yoshida M, Osawa
T, Riederer P, Naoi M (2007) In parkinsonian substantia nigra,
a-synuclein is modified by acrolein, a lipid-peroxidation product,
and accumulates in the dopamine neurons with inhibition of the
proteasome activity. J Neural Transm 114:1559–1567
Siderowf A, Stern MB (2008) Premotor Parkinson’s disease: clinical
features, detection, and prospects for treatment. Ann Neurol
64(Suppl 2):S139–S147
Solano SM, Miller DW, Augood SJ, Young AB, Penney JB Jr (2000)
Expression of alpha-synuclein, parkin, and ubiquitin carboxy-
terminal hydrolase L1 mRNA in human brain: genes associated
with familial Parkinson’s disease. Ann Neurol 47:201–210
Spillantini MG, Schmidt M, Lee VM, Trojanowski JQ, Kaques R,
Goedert M (1997) a-Synuclein in Lewy bodies. Nature
388:839–840
Srivastava G, Singh K, Tiwari MN, Singh MP (2010) Proteomics in
Parkinson’s disease: current trends, translational snags and future
possibilities. Expert Rev Proteomics 7:127–139
Stefanis L, Larsen KE, Rideout HJ, Sulzer D, Greene LA (2001)
Expression of A53T mutant but not wild-type alpha-synuclein in
PC12 cells induces alterations of the ubiquitin-dependent
degradation system, loss of dopamine release, and autophagic
cell death. J Neurosci 21:9549–9560
Stiasny-Kolster K, Doerr Y, Mo
¨ller JC, Ho
¨ffken H, Behr TM, Oertel
WH, Mayer G (2005) Combination of idiopathic REM sleep
behaviour disorder and olfactory dysfunction as possible indi-
cator for alpha-synucleinopathy demonstrated by dopamine
transporter FP-CIT-SPECT. Brain 128:126–137
Tang CC, Poston KL, Dhawan V, Eidelberg D (2010) Abnormalities
in metabolic network activity precede the onset of motor
symptoms in Parkinson’s disease. J Neurosci 30:1049–1056
Tolosa E, Compta Y, Gaig C (2007) The premotor phase of
Parkinson’s disease. Parkinsonism Rel Disord 13:S2–S7
Tribl F, Marcus K, Meyer HE, Bringmann G, Gerlach M, Riederer P
(2006) Subcellular proteomics reveals neuromelanin granules to
be a lysosome-related organelle. J Neural Transm 113:741–749
Tribl F, Asan E, Arzberger T, Tatschner T, Langenfeld E, Meyer HE,
Bringmann G, Riederer P, Gerlach M, Marcus K (2009)
Identification of L-ferritin in neuromelanin granules of the
human substantia nigra: a targeted proteomics approach. Mol
Cell Proteomics 8:1832–1838
Varani K, Vincenzi F, Tosi A, Gessi S, Casetta I, Granieri G, Fazio P,
Leung E, MacLennan S, Granieri E, Borea PA (2010) A2A
adenosine receptor overexpression and functionality, as well as
TNF-alpha levels, correlate with motor symptoms in Parkinson’s
disease. FASEB J 24:587–598
Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L (2008) Wild type
a-synuclein is degraded by chaperone-mediated autophagy and
macroautophagy in neuronal cells. J Biol Chem 283:23542–
23556
Wakabayashi K, Matsumoto K, Takayama K, Yoshimoto M, Takahashi
H (1997) NACP, a presynaptic protein, immunoreactivity in Lewy
bodies in Parkinson’s disease. Neurosci Lett 249:180–182
Wakabayashi K, Tanji K, Mori F, Takahashi H (2007) The Lewy
body in Parkinson’s disease: molecules implicated in the
formation and degradation of a-synuclein aggregates. Neuropa-
thology 27:494–506
Wang HQ, Takahashi R (2007) Expanding insights on the involve-
ment of endoplasmic reticulum stress in Parkinson’s disease.
Antioxid Redox Signal 9:553–561
Wang R, Qing H, Liu XQ, Zheng XL, Deng YL (2008) Iron
contributes to the formation of catechol isoquinolines and
oxidative toxicity induced by overdose dopamine in dopaminer-
gic SH-SY5Y cells. Neurosci Bull 24:125–132
Waxman EA, Giasson BI (2008) Specificity and regulation of casein
kinase-mediated phosphorylation of alpha-synuclein. J Neuropa-
thol Exp Neurol 67:402–416
Weisman D, Cho M, Taylor C, Adame A, Thal LJ, Hansen LA (2007)
In dementia with Lewy bodies, Braak stage determines pheno-
type, not Lewy body distribution. Neurology 69:356–359
Wirdefeldt K, Bogdanovic N, Westerberg L, Payami H, Schalling M,
Murdoch G (2001) Expression of alpha-synuclein in the human
brain: relation to Lewy body disease. Brain Res Mol Brain Res
92:58–65
Wypijewska A, Galazka-Friedman J, Bauminger ER, Wszolek ZK,
Schweitzer KJ, Dickson DW, Jaklewicz A, Elbaum D, Friedman
A (2010) Iron and reactive oxygen species activity in parkin-
sonian substantia nigra. Parkinsonism Relat Disord 16:329–333
Xia Q, Liao L, Cheng D, Duong DM, Gearing M, Lah JJ, Levey AI,
Peng J (2008) Proteomic identification of novel proteins
associated with Lewy bodies. Front Biosci 13:3850–3856
Xilouri M, Vogiatzi T, Vekrellis K, Stefanis L (2008) alpha-synuclein
degradation by autophagic pathways: a potential key to Parkin-
son’s disease pathogenesis. Autophagy 4:917–919
Youdim MB (2008) Brain iron deficiency and excess; cognitive
impairment and neurodegeneration with involvement of striatum
and hippocampus. Neurotox Res 14:45–56
Zecca L, Lucca FA, Albertini A, Rizzio E, Fariello RG (2006) A
proposed dual role of neuromelanin in the pathogenesis of
Parkinson disease. Neurology 67(Suppl 2):S8–S11
Zecca L, Casella L, Albertini A, Bellei C, Zucca FA, Engelen M,
Zadlo A, Szewczyk G, Zareba M, Sarna T (2008) Neuromelanin
can protect against iron-mediated oxidative damage in system
modeling iron overload of brain aging and Parkinson’s disease.
J Neurochem 106:1866–1875
Zeevalk GD, Razmpour R, Bernard LP (2008) Glutathione and
Parkinson’s disease: is this the elephant in the room? Biomed
Pharmacother 62:236–249
Ziemssen T, Reichmann H (2007) Non-motor dysfunction in Parkin-
son’s disease. Parkinsonism Rel Disord 13:323–332
Neuropathology of sporadic Parkinson disease 839
123
... Further biochemical studies have shown increased levels of neuroketals in the SNc in post mortem tissue from Braak stages 1 and 2, supporting the notion that oxidative damage to specific lipids in the SNc occurs at very early stages of PD and prior to the appearance of LBP [83,84]. In line, recent observations have shown the concentration of L-ferritin in the SNc to be lower in iLBD (and PD) compared with controls, whereas H-ferritin in PD was found to be higher than in iLBD and controls. ...
... Tang et al. recently investigated autophagy-associated SNARE molecules in post mortem brain tissue from LBD cases and found a stagedependent decline of the v-SNARE SNAP29 -a member of the SNARE complex mediating autophagolysosome fusion -as early as in Braak stage 1 (Table 1) [86]. Additional experiments in cultured dopaminergic neurons ↓ TH-positive terminals in the striatum [73] Neurochemical ↓ Striatal TH [71,82] ↑ Oxidative damage in the SNc [83] ↑ Neuroketals [83,84] Changes to iron metabolism [85] Autophagy ↓ Autophagy-associated SNARE molecules (SNAP29) [86] Association of p62 with αSyn inclusions [87] Immunological ↑ TLR-2-positive microglia in the SNc [88] ↑ CD68-positive microglia/macrophages in the SNc [89] ↑ PAR-2-positive microglia in the anterior cingulate cortex (ACC) [90] ↑ MCM2-positiv cells in the hippocampus (HC) [88] Changes to expression of inflammatory and trophic molecules in the SNc and striatum [91] ↑ CD8-positive T-cells in the SNc [92] ↑ Angiogenesis marker a v β3 [89] Synaptic ↓ DA synaptic terminals [82,93,94] ↑ αSyn aggregates at presynaptic terminals [95] Gene Expression ...
Article
Full-text available
Background Parkinson’s disease (PD) is a progressive neurodegenerative disorder associated with a loss of dopaminergic (DA) neurons. Despite symptomatic therapies, there is currently no disease-modifying treatment to halt neuronal loss in PD. A major hurdle for developing and testing such curative therapies results from the fact that most DA neurons are already lost at the time of the clinical diagnosis, rendering them inaccessible to therapy. Understanding the early pathological changes that precede Lewy body pathology (LBP) and cell loss in PD will likely support the identification of novel diagnostic and therapeutic strategies and help to differentiate LBP-dependent and -independent alterations. Several previous studies identified such specific molecular and cellular changes that occur prior to the appearance of Lewy bodies (LBs) in DA neurons, but a concise map of such early disease events is currently missing. Methods Here, we conducted a literature review to identify and discuss the results of previous studies that investigated cases with incidental Lewy body disease (iLBD), a presumed pathological precursor of PD. Results Collectively, our review demonstrates numerous cellular and molecular neuropathological changes occurring prior to the appearance of LBs in DA neurons. Conclusions Our review provides the reader with a summary of early pathological events in PD that may support the identification of novel therapeutic and diagnostic targets and aid to the development of disease-modifying strategies in PD.
... ROS accumulation is the first step, followed by neuronal damage [10], and it may cause oxidative damage to DNA, protein, or lipids [11]. Several data collected from PD patients in their early stage reveal that oxidative stress is present from the beginning of the disease, which leads us to think that ROS may be the cause of dopaminergic neuronal damage and loss other than being the response to neurodegeneration [12]. Despite advances in understanding PD pathophysiology, effective treatments that can halt or reverse the progression of PD remain elusive. ...
Article
Full-text available
Parkinson’s disease (PD) is a progressive neurodegenerative condition marked by the gradual deterioration of dopaminergic neurons in the substantia nigra. Oxidative stress has been identified as a key player in the development of PD in recent studies. In the first part, we discuss the sources of oxidative stress in PD, including mitochondrial dysfunction, dopamine metabolism, and neuroinflammation. This paper delves into the possibility of mitigating oxidative stress as a potential treatment approach for PD. In addition, we examine the hurdles and potential of antioxidant therapy, including the challenge of delivering antioxidants to the brain and the requirement for biomarkers to track oxidative stress in PD patients. However, even if antioxidant therapy holds promise, further investigation is needed to determine its efficacy and safety in PD treatment.
... Importantly, evidence from earlystage PD patients revealed that elevated oxidative stress is a critical feature of the early disease stages, preceding major neuron loss. 26 This implicates uncontrolled ROS production as a potential cause of dopaminergic neurodegeneration rather than a secondary response to progressive neurodegeneration. 27 Previous research has found that FM has powerful anti-oxidative properties, as well as changes in neurotransmitter levels. ...
Article
Full-text available
The current study looked at the behavioral and antioxidant activity of Foxtail Millet (FM) against haloperidol-induced catalepsy in Parkinson’s Disease (PD) patients. It has been demonstrated that the antipsychotic drug haloperidol, which has a high capacity to block D2-type receptors, can cause motor impairments similar to those seen in people with PD. Catalepsy can develop when animals are placed in abnormal or unusual postures for an extended period of time. Foxtail millet significantly reduced lipid peroxidation (p 0.001) increased the antioxidant enzymes SOD (p 0.05) and GSH (p 0.05), and significantly improved motor deficits such as catalepsy, motor coordination, and locomotor activity in our study. These results show that foxtail millet can protect against the motor deficits (catalepsy) associated with PD and epilepsy.
... Parkin affects energy metabolism, as well as a cell-based investigation has shown this [85]. Its over-expression was discovered to inhibit glucose uptake, reduce glycolytic rate, and promote oxygen consumption [86]. The generation of MG as a result of triosephosphate accumulation due to diminished NAD + accessibility or impaired glyceraldehyde-3-phosphate dehydrogenase activity is one effect of excessive glycolysis, as mentioned above. ...
Article
Full-text available
Growing evidence suggest that there is a connection between Parkinson’s disease (PD) and insulin dysregulation in the brain, whilst the connection between PD and type 2 diabetes mellitus (T2DM) is still up for debate. Insulin is widely recognised to play a crucial role in neuronal survival and brain function; any changes in insulin metabolism and signalling in the central nervous system (CNS) can lead to the development of various brain disorders. There is accumulating evidence linking T2DM to PD and other neurodegenerative diseases. In fact, they have a lot in common patho-physiologically, including insulin dysregulation, oxidative stress resulting in mitochondrial dysfunction, microglial activation, and inflammation. As a result, initial research should focus on the role of insulin and its molecular mechanism in order to develop therapeutic outcomes. In this current review, we will look into the link between T2DM and PD, the function of insulin in the brain, and studies related to impact of insulin in causing T2DM and PD. Further, we have also highlighted the role of various insulin signalling pathway in both T2DM and PD. We have also suggested that T2DM-targeting pharmacological strategies as potential therapeutic approach for individuals with cognitive impairment, and we have demonstrated the effectiveness of T2DM-prescribed drugs through current PD treatment trials. In conclusion, this investigation would fill a research gap in T2DM-associated Parkinson’s disease (PD) with a potential therapy option. Graphical Abstract
Article
Full-text available
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) has been widely used due to its specific and reproducible neurotoxic effect on the nigrostriatal system, being considered a convenient model of dopaminergic neurodegeneration to study interventions therapeutics. The purple pitanga (Eugenia uniflora) is a polyphenol-rich fruit with antioxidant and antidepressant properties, among others. Therefore, this study investigated the effect of purple pitanga extract (PPE) on acute early oxidative stress induced by intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration in rats. Male Wistar rats were pre-treated orally with PPE (1000 mg/kg) or vehicle. After 24 h, MPTP (0.1 mg/10µL/nostril) or vehicle was administered bilaterally into the animal’s nostrils, and 6 h later, the olfactory bulb (OB), striatum (ST), and substantia nigra (SN) were collected to evaluate the oxidative stress parameters. Our findings revealed that OB and SN were the most affected areas after 6 h of MPTP infusion; an early increase in reactive oxygen species (ROS) levels was observed, while pretreatment with a single dose of PPE prevented this increment. No differences in thiobarbituric acid reactive species (TBARS) and 3-nitrotyrosine (3-NT) formation were observed, although 4-hydroxy-2-nonenal (4-HNE) levels increased, which is the most toxic form of lipid peroxidation, in the MPTP group. The PPE pretreatment could prevent this increase by increasing the NPSH levels previously decreased by MPTP. Furthermore, PPE prevents the Na+/K + ATPase strongly inhibited by MPTP, showing the neuroprotective capacity of the PPE by inhibiting the MPTP-generated oxidation. Thus, we demonstrated for the first time the antioxidant and neuroprotective effects of PPE against the early MPTP neurotoxicity.
Preprint
Full-text available
Although Parkinson's disease (PD) coincides with altered immune functioning, there are few reproducible associations between blood DNA methylation (DNAm) and PD case-control status. Integrative analyses of genotype and blood DNAm can address this gap and can help us characterize the biological function of PD genetic risk loci. First, we tested for associations between a PD genetic risk score (GRS) and DNAm. Our GRS included 36 independent genome-wide significant variants from the largest GWAS of PD to date. Our discovery sample was TERRE, consisting of French agricultural workers (71 PD cases and 147 controls). The GRS associated with DNAm at 85 CpG sites, with 19 associations replicated in an independent sample (DIG-PD). The majority of CpG sites (73) are within a 1.5 Mb window on chromosome 17, and 36 CpG sites annotate to MAPT and KANSL1 , neighboring genes that affect neurodegeneration. All associations were invariant to non-genetic factors, including exposure to commercial-grade pesticides, and omitting chromosome 17 variants from the GRS had little effect on association. Second, we compared our findings to the relationship between individual PD risk loci and blood DNAm using blood mQTL from a large independent meta-analysis (GoDMC). We found 79 CpG sites that colocalized with PD loci, and via summary Mendelian randomization analysis, we show 25/79 CpG sites where DNAm causally affects PD risk. The nine largest causal effects are within chromosome 17, including an effect within MAPT. Thus, all integrative analyses prioritized DNAm on chromosome 17, drawing from multiple independent data sets, meriting further study of this region.
Article
Unlabelled: Objective: The purpose of this review was to assess the current evidence regarding the associated physiological and cognitive effects of aspartame (APM) consumption and Parkinson's Disease (PD). Methods: A total of 32 studies demonstrating effects of APM on monoamine deficiencies, oxidative stress, and cognitive changes were reviewed. Results: Multiple studies demonstrated decreased brain dopamine, decreased brain norepinephrine, increased oxidative stress, increased lipid peroxidation, and decreased memory function in rodents after APM use. In addition, PD animal models have been found to be more sensitive to the effects of APM. Discussion: Overall, studies of APM use over time yielded more consistent results; however, no study has examined long-term effects on APM in human PD patients. Based on the current evidence, long-term human based observational research is needed to further investigate the potential effect of APM on PD.
Article
Full-text available
The mesencephalic dopaminergic (mDA) cell system is composed of two major groups of projecting cells in the substantia nigra (SN) (A9 neurons) and the ventral tegmental area (VTA) (A10 cells). A9 neurons form the nigrostriatal pathway and are involved in regulating voluntary movements and postural reflexes. Their selective degeneration leads to Parkinson's disease. Here, we report that gene expression analysis of A9 dopaminergic neurons (DA) identifies transcripts for α- and β-chains of hemoglobin (Hb). Globin immunoreactivity decorates the majority of A9 DA, a subpopulation of cortical and hippocampal astrocytes and mature oligodendrocytes. This pattern of expression was confirmed in different mouse strains and in rat and human. We show that Hb is expressed in the SN of human postmortem brain. By microarray analysis of dopaminergic cell lines overexpressing α- and β-globin chains, changes in genes involved in O2 homeostasis and oxidative phopshorylation were observed, linking Hb expression to mitochondrial function. Our data suggest that the most famed oxygen-carrying globin is not exclusively restricted to the blood, but it may play a role in the normal physiology of the brain and neurodegenerative diseases.
Article
Although still considered a paradigmatic movement disorder, Parkinson's disease (PD) is associated with a broad spectrum of non-motor symptoms. These include disorders of mood and affect with apathy, anhedonia and depression, cognitive dysfunction and hallucinosis, as well as complex behavioural disorders. Sensory dysfunction with hyposmia or pain is almost universal, as are disturbances of sleep-wake cycle regulation. Autonomic dysfunction including orthostatic hypotension, urogenital dysfunction and constipation is also present to some degree in a majority of patients. Whilst overall non-motor symptoms become increasingly prevalent with advancing disease, many of them can also antedate the first occurrence of motor signs - most notably depression, hyposmia or rapid eye movement sleep behaviour disorder (RBD). Although exact clinicopathological correlations for most of these non-motor features are still poorly understood, the occurrence of constipation, RBD or hyposmia prior to the onset of clinically overt motor dysfunction would appear consistent with the ascending hypothesis of PD pathology proposed by Braak and colleagues. Screening these early non-motor features might, therefore, be one approach towards early 'preclinical' diagnosis of PD. This review article provides an overview of the clinical spectrum of non-motor symptoms in PD together with a brief review of treatment options.
Article
α-Synuclein is mutated in some hereditary cases of Parkinson’s disease and the protein precipitates in Lewy bodies, the pathological hallmark of both Parkinson’s disease and Lewy body disease. Transgenic mice overexpressing human wild-type α-synuclein develop α-synuclein-immunoreactive inclusions in brain regions typically affected with Lewy body disease. We used in situ hybridization to characterize α-synuclein expression and examine mRNA levels in patients affected with Lewy body disease and controls. Substantia nigra was avoided because of the extensive neuronal loss and cingulate gyrus was chosen as it is one of the diagnostic regions in Lewy body disease where Lewy bodies most frequently are demonstrated. β-tubulin was used to control for neuronal degeneration. The α-synuclein probe showed intense labeling of pyramidal cells in lamina III and V in both patients and controls. We found no difference in α-synuclein mRNA levels and β-tubulin mRNA was not significantly altered (P=0.06) in patient brains. There was no difference in the ratio of α-synuclein and β-tubulin mRNA levels between patients and controls. Further, we found no relationship between α-synuclein mRNA levels and Lewy bodies. Great variability in α-synuclein mRNA levels among patients indicates that Lewy body disease may be a heterogeneous disorder with regard to α-synuclein involvement.
Article
We reported previously that phosphorylation by casein kinase II (CKII) regulates the interaction between alpha-synuclein and its binding partner synphilin-1, and that both CKII alpha and beta subunits co-localize with alpha-synuclein in cytoplasmic inclusions in transfected cells. In this study, we extended these observations to the brains of patients with Parkinson's disease (PD) and examined whether CKII subunits are present in Lewy bodies. Immunohistochemical studies on PD brains harboring Lewy bodies revealed a positive stain for CKII beta but not for CKII alpha. In addition, CKII beta subunits co-localized with alpha-synuclein in most Lewy bodies. These findings suggest that CKII beta subunits may play a role in the formation of intracytoplasmic inclusions in human alpha-synucleinopathies either through phosphorylation events or through a separate mechanism linked to the beta subunit itself.
Article
We examine the evidence for free radical involvement and oxidative stress in the pathological process underlying Parkinson's disease, from postmortem brain tissue. The concept of free radical involvement is supported by enhanced basal lipid peroxidation in substantia nigra in patients with Parkinson's disease, demonstrated by increased levels of malondialdehyde and lipid hydroperoxides. The activity of many of the protective mechanisms against oxidative stress does not seem to be significantly altered in the nigra in Parkinson's disease. Thus, activities of catalase and glutathione peroxidase are more or less unchanged, as are concentrations of vitamin C and vitamin E. The activity of mitochondrial superoxide dismutase and the levels of the antioxidant ion zinc are, however, increased, which may reflect oxidative stress in substantia nigra. Levels of reduced glutathione are decreased in nigra in Parkinson's disease; this decrease does not occur in other brain areas or in other neurodegenerative illnesses affecting this brain region (i.e., multiple system atrophy, progressive supranuclear palsy). Altered glutathione metabolism may prevent inactivation of hydrogen peroxide and enhance formation of toxic hydroxyl radicals. In brain material from patients with incidental Lewy body disease (presymptomatic Parkinson's disease), there is no evidence for alterations in iron metabolism and no significant change in mitochondrial complex I function. The levels of reduced glutathione in substantia nigra, however, are reduced to the same extent as in advanced Parkinson's disease. These data suggest that changes in glutathione function are an early component of the pathological process of Parkinson's disease. The data presented suggest (1) there is oxidative stress in the substantia nigra at the time of death in advanced Parkinson's disease that manifests in terms of increased lipid peroxidation, superoxide dismutase activity, and zinc levels; (2) there is a major impairment of the glutathione pathway in Parkinson's disease; and (3) alterations in reduced glutathione levels may occur very early in the illness.
Article
In order to clarify the significance of Lewy body (LB)-related α-synucleinopathy in aging, we investigated the incidence of LBs in 1241 consecutive autopsy cases (663 male and 578 female) from a community-based general geriatric hospital between 1995 and 2002. LB pathology was evaluated histologically in sections stained with hematoxylin and eosin and immunocytochemically with anti-ubiquitin and anti-α-synuclein antibodies. Cases without LBs were classified into LB Stage 0 (987 cases). Cases with LBs were categorized as follows. LB Stage I: incidental LBs without neuronal degeneration (149 cases); LB Stage II: LB-related neuronal degeneration without documentation of attributable clinical symptoms (47 cases); LB Stage III: Parkinson disease (PD) without dementia (10 cases); LB Stage IV: dementia with Lewy bodies (DLB) and PD with dementia (PDD), presenting with a Lewy score fulfilling the criteria for transitional (limbic) form (25 cases) on the 1996 Consensus Guidelines for DLB; and LB stage V: DLB and PDD, with a Lewy body score fulfilling the criteria for neocortical form (23 cases). The average age at death of cases with LBs was older than that of cases without LBs. The average LB stage increased with age. There were no gender differences in the grade of LB pathology. The G284A polymorphism in the paraoxonase 1 gene was associated with men in LB stage equal to or above II, and suggests a gender-specific risk factor. LB Stage V had higher Braak Stage for neurofibrillary tangle (NFT) and senile plaque (SP) than the background and also had a higher frequency of apolipoprotein E ε4. Cases of higher SP stage with dementia tended to have higher LB stage. Our findings suggest that LBs cause cognitive decline synergistically with Alzheimer-type senile changes, especially with senile plaques. Further study of these correlations should be carried out.
Article
The etiology and pathogenesis of Parkinson’s disease have remained a mystery since the first description of the disease, but the neuropathology of the disorder, as we now know it, is fairly simple and straightforward. The most important lesion is in the substantia nigra pars compacta, where nerve cells degenerate together with their nigro-striatal fiber tract, causing a severe depletion of dopamine in the basal ganglia.
Article
Cannabinoid-based medicines have been proposed as clinically promising therapies in Parkinson's disease (PD), given the prominent modulatory function played by the cannabinoid signaling system in the basal ganglia. Supporting this pharmacological potential, the cannabinoid signaling system experiences a biphasic pattern of changes during the progression of PD. Thus, early and presymptomatic stages, characterized by neuronal malfunctioning but little evidence of neuronal death, are associated with desensitization/downregulation of CB1 receptors. It was proposed that these losses may be part of the pathogenesis itself, since they can aggravate different cytotoxic insults which are controlled in part by cannabinoid signals, mainly excitotoxicity but also oxidative stress and glial activation. By contrast, intermediate and, in particular, advanced stages of parkinsonism characterized by a profound nigral degeneration and occurrence of major parkinsonian symptoms (e.g. bradykinesia), are associated with upregulatory responses of CB1 receptors, possibly CB2 receptors too, and the endocannabinoid ligands for both receptor types. This would explain the motor inhibition typical of this disease and the potential proposed for CB1 receptor antagonists in attenuating the bradykinesia typical of PD. In addition, certain cannabinoid agonists have been proposed to serve as neuroprotective molecules in PD, given their well-demonstrated capability to reduce excitotoxicity, calcium influx, glial activation and, in particular, oxidative injury that cooperatively contribute to the degeneration of nigral neurons. However, the potential of cannabinoid-based medicines in PD have been still scarcely studied at the clinical level despite the existence of solid and promising preclinical evidence. Considering the relevance of these preclinical data, the need for finding treatments for motor symptoms that may be alternative to classic dopaminergic replacement therapy, and the lack of efficient neuroprotective strategies in PD, we believe it is of major interest to develop further studies that allow the promising expectations generated for these molecules to progress from the present preclinical evidence towards a real clinical application.