ArticlePDF Available

Abstract and Figures

Variation in the genetic risk(s) of developing Parkinson's disease (PD) undoubtedly contributes to the subsequent phenotypic heterogeneity. Although patients with PD who undergo deep brain stimulation (DBS) are a skewed population, they represent a valuable resource for exploring the relationships between heterogeneous phenotypes and PD genetics. In this series, 94 patients who underwent DBS were screened for mutations in the most common genes associated with PD. The consequent genetic subgroups of patients were compared with respect to phenotype, levodopa (l-dopa), and DBS responsiveness. An unprecedented number (29%) of patients tested positive for at least 1 of the currently known PD genes. Patients with Parkin mutations presented at the youngest age but had many years of disease before needing DBS, whereas glucocerebrosidase (GBA) mutation carriers reached the threshold of needing DBS earlier, and developed earlier cognitive impairment after DBS. DBS cohorts include large numbers of gene positive PD patients and can be clinically instructive in the exploration of genotype-phenotype relationships.
Content may be subject to copyright.
Genotype and Phenotype in Parkinson’s Disease:
Lessons in Heterogeneity From Deep Brain Stimulation
Aikaterina Angeli, MD,
1
Niccolo E. Mencacci, MD,
2,3
Raquel Duran, MD,
2
Iciar Aviles-Olmos, MD,
1
Zinovia Kefalopoulou, MD, PhD,
1
Joseph Candelario, BSc,
1
Sarah Rusbridge, BSc,
4
Jennifer Foley, PhD,
4
Priyanka Pradhan, PhD,
4
Marjan Jahanshahi, PhD,
1
Ludvic Zrinzo, MD, PhD,
1
Marwan Hariz, MD, PhD,
1
Nicholas W. Wood, PhD,
FRCP,
2
John Hardy, PhD, FRS, FMedSci,
2
Patricia Limousin, MD, PhD,
1
and Tom Foltynie, MRCP, MD
1
*
1
Sobell Department of Motor Neuroscience, University College London (UCL) Institute of Neurology, Queen Square, London, United Kingdom
2
Reta Lila Weston Laboratories and Departments of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
3
Department of Neurology and Laboratory of Neuroscience, Dino Ferrari” Centre, Universit
a degli Studi di Milano-IRCCS Istituto
Auxologico Italiano, Milan, Italy
4
Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
ABSTRACT: Variation in the genetic risk(s) of devel-
oping Parkinson’s disease (PD) undoubtedly contributes to
the subsequent phenotypic heterogeneity. Although patients
with PD who undergo deep brain stimulation (DBS) are a
skewed population, they representavaluableresourcefor
exploring the relationships between heterogeneous pheno-
types and PD genetics. In this series, 94 patients who
underwent DBS were screened for mutations in the most
common genes associated with PD. The consequent
genetic subgroups of patients were compared with respect
to phenotype, levodopa (
L-dopa), and DBS responsiveness.
An unprecedented number (29%) of patients tested positive
for at least 1 of the currently known PD genes. Patients with
Parkin mutations presented at the youngest age but had
many years of disease before needing DBS, whereas gluco-
cerebrosidase (GBA) mutation carriers reached the thresh-
old of needing DBS earlier, and developed earlier cognitive
impairment after DBS. DBS cohorts include large numbers
of gene positive PD patients and can be clinically instructive
in the exploration of genotype-phenotype relationships.
Key Words: genetics; Parkinson’s disease; pheno-
type; deep brain stimulation; heterogeneity
Patients with Parkinson’s disease (PD) show marked
heterogeneity in their clinical features in relation to
motor phenotype, rate of progression, and development
of cognitive impairment.
1
Clinical studies of heterogene-
ity have been complemented by studies of the relation
between PD pathophysiological mechanisms and PD risk
factors.
2–4
Some aspects of PD heterogeneity almost cer-
tainly relate to the recognized genes
5,6
with superimposed
environmental, epistatic, as well as stochastic modifiers of
PD phenotype. Several studies have described the pheno-
type of patients with specific genetic mutations
7–9
;how-
ever, direct comparisons of PD phenotype between
different genetically defined groups are scarce.
10
Patients with PD who undergo deep brain stimulation
(DBS) tend to have a young age at onset and, thus, prob-
ably include an over-representation of genetic forms of
PD. In this study, an extensive genotyping of a series of
patients with PD who underwent DBS surgery was per-
formed, and subsequent comparisons were made of clini-
cal phenotypes among genetic subgroups both on and off
dopaminergic medication along with responses to DBS.
Patients and Methods
Clinical Assessments
All patients were followed at the National Hospital
for Neurology and Neurosurgery, Queen Square and
------------------------------------------------------------
*Correspondence to: Dr. Thomas Foltynie, Box 146, National Hospital
for Neurology & Neurosurgery, Queen Square, London, WC1N 3BG
United Kingdom; t.foltynie@ucl.ac.uk
Funding agencies: This work was supported by Parkinson’s UK (grant
number K-0901 awarded to T.F.), the National Institute for Health
Research (NIHR) UCL/UCLH Biomedical Research Centres funding
Scheme and The Wellcome Trust/Medical Research Council (MRC)
funded UK Parkinson’s disease consortium.
Relevant conflicts of interest/financial disclosures: Nothing to report.
Full financial disclosures and author roles may be found in the online ver-
sion of this article.
Received: 1 February 2013; Revised: 12 March 2013; Accepted: 17
April 2013
DOI: 10.1002/mds.25535
V
C
2013 The Authors. Movement Disorders published by Wiley on b-
ehalf of The Movement Disorder Society.
This is an open access article under the terms of the Creative Commons
Attribution License, which permits use, distribution and reproduction in
any medium, provided the original work is properly cited.
RESEARCH ARTICLE
Movement Disorders, Vol. 00, No. 00, 2013 1
underwent DBS between 2002 and 2011. Basic demo-
graphic details were recorded. The levodopa (
L-dopa)
equivalent dose was derived using standard
formulae.
11
The selection of surgical candidates and targets was
performed after assessments, including
L-dopa chal-
lenge, using the Unified Parkinson’s Disease Rating
Scale (UPDRS) part 3 (motor examination) in the
practically defined “OFF-state” and “ON-state” and
also using UPDRS parts 1 (nonmotor activities of daily
living), 2 (motor activities of daily life), and 4 (motor
complications). Additional assessments included brain
magnetic resonance imaging and neuropsychological
assessments (including the Mattis Dementia Rating
Scale [DRS-2]).
UPDRS part 3 subscale scores were derived to quan-
tify tremor (items 20 and 21), akinesia and rigidity
(items 22–26 and 31), and axial features (items 18,
19, and 27–30). Part 4 of the UPDRS was divided into
items 32 through 34 for dyskinesia and items 35
through 39 to reflect OFF periods.
The optimal DBS target was chosen according to the
frequency and severity of OFF-symptoms, dyskinesia,
tremor, speech-intelligibility, and cognition. DBS was
performed using a standard technique that has been
described previously.
12–15
Genotyping consent was
obtained from all patients.
Postoperative assessments were performed 12
months after DBS surgery. Patients who refused to
have the stimulation switched off had their preopera-
tive “OFF-medication” scores imputed. A subset of 37
patients with >5 years of follow-up post-DBS under-
went repeat cognitive assessment and had the mean
annual change on the DRS-2 between baseline and
follow-up calculated.
Genotyping
Genetic testing for PD genes was performed in
accordance with the service supplied by the Depart-
ment of Neurogenetics, Queen Square. Multiplex
ligation-dependent probe amplification (MLPA) was
performed according to manufacturer’s instructions
using the P051 Salsa MLPA Parkinson probe set
(MRC Holland, Amsterdam, the Netherlands). This
set includes probes that detect exonic rearrangements
in PARK1 (synuclein alpha [SNCA]; exons 1–6),
PARK2 (Parkin: exons 1–12), PARK6 (phosphatase
and tensin homolog-induced putative kinase 1
[PINK1]; exons 1–8), PARK7 (DJ1; coding exons 3, 5,
6, and 7), PARK8 (leucine-rich repeat kinase 2
[LRRK2]; exons 1, 2, 10, 15, 27, 41, and 49), the
Ala30Pro mutation in PARK 1 (SNCA), and the
Gly2019Ser mutation in PARK8 (LRRK2). Patients
who were identified as positive for G2019S point
mutations had their results confirmed by Sanger
sequencing.
In addition, the Parkin (12 exons) and glucocerebro-
sidase (GBA) (11 exons) coding region and the flank-
ing intronic sequences were completely screened by
Sanger sequencing. The primers and polymerase chain
reaction (PCR) conditions we used are available on
request. PCR products were bidirectional sequenced
using the BigDye Terminator version 3.1 sequencing
chemistry, then loaded on the ABI3730xl genetic ana-
lyzer (Applied Biosystems, Foster City, CA).
Statistical Analysis
All statistical tests were performed using the Stata
statistical software package (version 8; StataCorp LC,
College Station, TX). The v
2
test was used for cate-
gorical data. Continuous data were checked for nor-
mality, and 1-way analysis of variance was used to
compare subgroups. Post hoc comparisons were per-
formed using Sidak’s method. Kruskal Wallis and non-
parametric post hoc comparisons also were used as
appropriate.
Results
Data are reported for 94 unselected patients with
PD who underwent DBS. The mean (6 standard devi-
ation [SD]) age of patients at the onset of symptoms
was 40.4 6 8.2 years (range, 7–58 years). Of these 94
patients, 27 (29%) had at least 1 mutation in a PD
gene. The results of genotyping for this cohort are pre-
sented in Table 1.
Genotypes
Parkin
All single mutation carriers are reported, but we
have only included Parkin homozygotes/compound
heterozygotes for phenotypic comparisons. Eight indi-
viduals had Parkin mutations, of which 5 patients had
2 mutations, and 3 patients had a single mutation (1
of whom also had a GBA T369M mutation).
GBA
Sixteen patients had at least 1 mutation in the GBA
gene, most common of which were the E326K
mutation.
9,16
LRRK2-G2019S
Five patients were identified with the G2019S muta-
tion. One patient with a G2019S mutation also car-
ried the E326K GBA mutation. The remaining 67
patients had no mutation detected.
Phenotypes
The mean age at onset for the individuals who had
2 Parkin mutations (24.0 years) was younger than the
age of those who had GBA mutations (P 5 0.0009)
PD HETEROGENEITY, GENOTYPES, AND DBS
2 Movement Disorders, Vol. 00, No. 00, 2013
and those without mutations (P 5 0.006) (Table
1).The mean duration of PD (6 SD) in patients at
DBS surgery was 15.0 6 6.6 years (range, 3.8–38.0
years). Patients with Parkin mutations had a longer
duration of disease at surgery than all other sub-
groups, whereas patients with GBA mutations had a
shorter duration of disease at surgery compared with
the Parkin mutation and mutation-negative subgroups
(P 5 0.001) (Table 2).
L-Dopa challenge revealed a
mean UPDRS part 3 improvement of 68% in the
group as a whole. UPDRS part 4 dyskinesia scores
were higher in the patients with Parkin mutations
TABLE 1. Description of abnormal genetic findings in a cohort of 94 patients with PD who underwent deep brain stimulation
surgery
Genetic test results
No. of
patients (sex) Description
Mean age 6 SD/age
at symptom onset, y Family history
a
Parkin double
mutation carriers
24 6 11.1
4 (3 M, 1 F) Homozygous [c.101_102delAG] 30 Nil
c.1289G>A, p.G430D and
c.823C>T; p.Arg275Trp
20 Sibling
c.337_376del and c.465–466del 36 Nil
Homozygous deletion of exon 3 and
4
27 Sibling
c.823C>T; p.Arg275Trp and Hetero-
zygous duplication of exon 6
7 Unknown
Parkin single
mutation carriers
5 (4M, 1F) c.1000C>T; p.Arg334Cys 41 Nil
c.337_376del, p.P113TfsX51 39 Nil
c.1310C>T; p.P437L and GBA
T369M
b
39 Nil
GBA confirmed
mutation
42.9 6 6.2
16 (9 M, 7 F) R463C/R463C 45 Parent
L444P/E326K 34 Nil
N370S 45 2 Second-degree
relatives
D409H 39 Nil
recNcil 40 Nil
R463C 45 Parent
N188S 49 Nil
R275Q 42 Parent
IVS2 1 1G>A 41 Nil
L444P 45 Nil
E326K/E326K 42 1 Third-degree relative
E326K 36 1 Second-degree relative
E326K 51 Parent
E326K 58 Nil
E326K and LRRK2 G2019S
b
35 Nil
T369M and parkin c.1310C>T;
p.P437L
b
39 Nil
LRRK2 43 6 8.7
5 (3 M, 2 F) G2019S 40 Nil
G2019S 36 Parent
G2019S 49 Nil
G2019S 55 Parent
G2019S and GBA-E326K
b
35 Nil
No mutation found
67 (46 M, 21 F) 40.8 6 7.2 Parent, n 5 9
Sibling, n 5 2
Grandparent, n 5 2
Half sibling, n 5 1
Cousin, n 5 4
Aunt, n 5 2
a
Family history data details the number of patients reporting a positive family history of PD, together with affected relative in each genetic subgroup.
b
Note that
the numbers add up to 96, because 2 individuals who carried 2 confirmed PD mutations are represented twice in the table.SD, standard deviation; M, males;
F, females; GBA, glucosidase beta acid; LRRK2, leucine-rich repeat kinase 2.
ANGELI ET AL.
Movement Disorders, Vol. 00, No. 00, 2013 3
despite their receipt of lower doses of L-dopa. No
other motor phenotypic differences were detectable
(Table 3).
There was a difference in the target choice for DBS
according to genotypic subgroups (v
2
statistic, 29.1;
P < 0.001), with an excess of patients who had Par-
kin and GBA mutations allocated to bilateral globus
pallidus internus DBS (GPi-DBS) rather than subtha-
lamic nucleus DBS (STN-DBS). The percentage
improvement in the UPDRS part 3 score “OFF-
medication” was less with bilateral GPi-DBS than
with STN-DBS for all groups (P 5 0.001) (Table 4).
Two mutation-negative patients underwent bilateral
GPi-DBS and had worse OFF-medication/ON-stimu-
lation scores compared with their preoperative OFF-
medication assessment, but both experienced
improvement in dyskinesias.
There was no significant difference in the degree of
improvement achievable with STN-DBS between
mutation-negative patients and patients who had Par-
kin, GBA, or LRRK2 mutations. There was a worsen-
ing of postoperative OFF-medication/OFF-stimulation
scores compared with preoperative OFF-medication
scores in patients who underwent STN-DBS. Note
that medication doses are substantially reduced post-
operatively; therefore, the practically defined “OFF”
may be closer to the true “OFF” at this time point.
Longitudinal 5-year follow-up data with respect to
cognition were available for 35 individuals; all had
undergone STN-DBS, and 6 had GBA mutations. The
mean 6 SD decline in Mattis DRS-2 scores for patients
with GBA mutations was 4.4 6 7.3 points per year
compared with 0.5 6 0.9 points per year among
mutation-negative patients.
TABLE 2. Preoperative Unified Parkinson’s Disease Rating Scale scores and response to L-dopa according to genetic
subgroup
Mean 6 SD
UPDRS-II UPDRS-III UPDRS-IV
Genetic test results
Duration of
PD at DBS
assessment, y
LED, mg UPDRS-I Off meds On meds Off meds On meds
Percentage
improvement
in score
with
L-dopa
Dyskinesia
score
Off
meds
score
Parkin (compound
heterozygotes/
homozygotes;
N 5 5)
25.2 6 12.8 960 6 611 1.7 6 2.1 24.3 6 4.0 6 6 6.2 57.0 6 11.2 21.0 6 6.4 61.0 6 18.3 5.3 6 3.9 3.5 6 1.3
GBA (confirmed
mutation;
N 5 16)
11.2 6 5.0 1143 6 540 1.5 6 1.5 23.1 6 12.3 10.9 6 11.8 51.3 6 14.0 18.0 6 15.4 66.9 6 18.6 3.0 6 2.7 4.6 6 2.1
LRRK2 (N 5 5) 12.1 6 1.8 1317 6 803 1.2 6 1.1 26.8 6 7.9 4.5 6 4.4 65.4 6 14.9 10.8 6 5.1 84.9 6 5.5 4.2 6 3.1 5.2 6 1.3
No mutation
found
(N 5 67)
15.1 6 5.5 1259 6 559 2.0 6 1.6 22.9 6 7.9 8.0 6 7.2 47.4 6 14.7 15.6 6 11.3 68.5 6 19.3 3.0 6 2.2 4.5 6 1.3
SD, standard deviation; UPDRS-I through UPDRS-IV, Unified Parkinson’s Disease Rating Scale parts 1 (nonmotor activities of daily living), 2 (motor activities of
daily living), 3 (motor examination), and 4 (motor complications), respectively; DBS, deep brain stimulation; meds, medication; LED,
L-dopa equivalent dose;
GBA, glucosidase beta acid; LRRK2, leucine-rich repeat kinase 2.
TABLE 3. Preoperative Unified Parkinson’s Disease Rating Scale motor scores “off” and “on” L-dopa according to genetic
subgroup
a
UPDRS score: Mean 6 SD
Off
L-dopa On L-dopa
Genetic test results Tremor Bradykinesia/rigidity Axial Tremor Bradykinesia/rigidity Axial
Parkin (compound heterozygotes/
homozygotes; N 5 5)
6.4 6 3.8 36.4 6 7.4 14.2 6 4.2 1.4 6 1.7 13.0 6 4.5 6.6 6 2.4
GBA (confirmed mutation; N 5 16) 6.9 6 5.1 31.2 6 11.0 13.1 6 5.7 1.2 6 1.9 12.1 6 11.0 4.7 6 4.6
LRRK2 (N 5 5) 9.4 6 2.2 40.8 6 8.8 15.2 6 6.7 0 6 0.0 8.3 6 3.6 2.5 6 2.1
No mutation found (N 5 67) 7.3 6 5.3 29.4 6 10.3 10.8 6 4.8 1.7 6 3.4 10.3 6 7.4 3.6 6 2.9
a
There were no significant differences in the motor phenotype according to genetic subgroup. Tremor was seen in all subgroups, and the proportion of tremor
compared with akinesia/rigidity or axial features was similar in each group.UPDRS, Unified Parkinson’s Disease Rating Scale; SD, standard deviation; GBA,
glucosidase beta acid; LRRK2, leucine-rich repeat kinase 2.
PD HETEROGENEITY, GENOTYPES, AND DBS
4 Movement Disorders, Vol. 00, No. 00, 2013
Discussion
The results from this study confirm that patients
who undergo DBS surgery are a valuable resource for
identifying genetic forms of PD. The frequency of
mutation-positive patients with PD was much greater
in our cohort (27 of 94 patients; 29%) than in
population-representative cohorts of PD.
17
Although
the use of DBS cohorts facilitates the rapid identifica-
tion of mutation-positive patients for study, patients
who have major cognitive or psychiatric problems or
a relative lack of
L-dopa response are excluded; there-
fore, the relevance of such data to the broader popula-
tion of PD patients is partially limited. Patients in our
cohort underwent detailed screening for the most com-
mon PD genes; nevertheless, not every gene previously
linked to PD was sequenced. Therefore, some patients
with genetic forms of PD may have been misclassified
as mutation-negative.
Apart from confirming that patients with Parkin
mutations had more severe
L-dopa–induced dyskinesia,
no consistent phenotypic difference was identified
between mutation-positive and mutation-negative sub-
types at a single time point. This said, the small num-
bers of patients in each genetic subgroup, together with
variable disease duration between groups, limits the
power of this study to reach definite conclusions. In a
previous study that examined the phenotype of GBA
patients, bradykinesia as a presenting feature was the
only difference compared with GBA-negative patients.
9
Together with our data, this suggests that the currently
known genetic mutations all lead to a similar pattern/
variety of patterns of neurodegeneration.
However, longitudinal data are more revealing. Dis-
ease duration at DBS differs significantly between
genetic subgroups. Patients with Parkin mutations are
younger at the onset of disease and have earlier dyski-
nesia, but they have longer disease duration before
DBS, indicating a more indolent form of the disease.
The patients in our study who had Parkin mutations
were receiving lower doses of
L-dopa replacement than
all other groups, probably because of their increased
severity of dyskinesia. PD patients with GBA mutations
required DBS earlier in their disease course, because
they developed disabling motor symptoms at an earlier
stage despite conventional oral treatment, consistent
with other published studies suggesting that patients
with PD who have GBA mutations also have higher
rates of cognitive decline (irrespective of DBS).
18
Previous reports have documented the generally
good response to DBS of small numbers of patients
with Parkin,
19,20
Pink-1,
19,20
and LRRK-2 muta-
tions.
19,21,22
However, the long-term response of
patients with GBA mutations to DBS is less well
known. In the current series, the response to either
STN-DBS or GPi-DBS did not differ significantly
between the subgroup with GBA mutations (at 1 year)
and any other subgroup. However, our longitudinal
TABLE 4. Target selection and response to deep brain stimulation according to target and genetic subgroup
UPDRS-III score: Mean 6 SD UPDRS-IV
Post-op
Dyskinesia score:
Mean 6 SD
Genetic
test results
No. of
patients
DBS
target
Disease
duration at DBS:
Mean 6 SD, y
Pre-op:
Off meds
Off meds/
off stim
Off meds/
on stim
Percentage
improvement
in UPDRS-III
score off meds/
on stim vs
pre-op off
meds
Post-op:
On meds/
on stim
Mean 6 SD
reduction in
LED, mg Pre-op Post-op
Percentage
improvement
in post-op
vs pre-op
UPDRS-IV
score
Parkin
(compound
Hets/Homozy’s)
3 GPi 21.1 6 16.2 53.3 6 13.9 43.3 6 16.4 42.0 6 19.0 21% 27.3 6 17.6 2237 6 315 8.0 6 1.4 1.67 6 2.0 70%
2 STN 31.3 6 0.6 62.5 6 3.5 84.0 6 22.6 43.0 6 0.0 31% 23.5 6 6.4 20 6 594 2.5 6 3.5 2.0 6 1.4 20%
GBA
(confirmed
PD mutation)
2 GPi 15.5 6 5.3 64.5 6 21.9 66.5 6 19.1 50.0 6 19.8 22% 41.0 6 15.6 1005 6 77 8.5 6 0.7 0.5 6 0.7 94%
13 STN 10.9 6 4.9 50.5 6 12.4 56.1 6 18.8 28 6 11.4 40% 15.9 6 10.4 146 6 510 2.4 6 1.7 1.5 6 1.6 37%
1 VIM
(unilateral)
3.9 35 35 20 43% 8 445 0 0
LRRK2 5 STN 12.1 6 1.8 65.4 6 14.9 69.2 6 12.4 30.6 6 16.1 53% 14.0 6 8.1 586 6 495 4.2 6 3.1 1.4 6 2.6 50%
No mutation
found
2 GPi 21.7 6 0.5 40.5
6 13.4 78 6 7.1 51.0 6 7.1 228% 25.5 6 4.9 227 6 89 6.0 6 1.4 3.5 6 0.7 42%
65 STN 14.8 6 5.4 47.6 6 14.8 50.0 6 15.4 24.6 6 11.3 48% 15.0 6 9.0 468 6 494 3.1 6 2.2 3.0 6 2.2 26%
SD, standard deviation; UPDRS-III, Unified Parkinson’s Disease Rating Scale, part 3 (motor examination); meds, medication; stim, stimulation; LED, L-dopa
equivalent dose; UPDRS-IV, Unified Parkinsons Disease Rating Scale, part 4 (motor complications); DBS, deep brain stimulation; GPi, globus pallidus internus;
STN, subthalamic nucleus; GBA, glucosidase beta acid; VIM, ventral intermediate nucleus; LRRK2, leucine-rich repeat kinase 2.
ANGELI ET AL.
Movement Disorders, Vol. 00, No. 00, 2013 5
follow-up of the subgroup with GBA mutations sug-
gested a faster rate of cognitive decline after DBS. A
previous report of 3 patients with GBA mutations
who underwent STN-DBS also identified a more
aggressive process that led to both cognitive impair-
ment and axial impairments
23
; whereas, of 2 other
patients with GBA mutations who underwent STN-
DBS, 1 had early cognitive decline, and the second
had persistent benefit.
9
Whether there is an interaction
between GBA status and the risk of subsequent cogni-
tive decline after STN-DBS needs to be clarified.
More aggressive disease in patients who have GBA
mutations may have relevance from a therapeutic
standpoint in terms of supporting the earlier introduc-
tion of
L-dopa and possibly the use of bilateral GPi-
DBS rather than bilateral STN-DBS (because there are
likely to be additional negative cognitive consequences
of STN-DBS
24
). Further prospective data would be
needed to confirm this. Our data support the idea that
the heterogeneity of PD should be considered longitu-
dinally in terms of the rate of progression of motor
and nonmotor features rather than the detailed scru-
tiny of purely cross-sectional data.
References
1. Foltynie T, Brayne C, Barker RA. The heterogeneity of idiopathic
Parkinson’s disease. J Neurol 2002;249:138–145.
2. Volpicelli-Daley LA, Luk KC, Patel TP, et al. Exogenous alpha-
synuclein fibrils induce Lewy body pathology leading to synaptic
dysfunction and neuron death. Neuron 2011;72:57–71.
3. Schapira AH. Mitochondrial diseases. Lancet 2012;379:1825–1834.
4. Aviles-Olmos I, Limousin P, Lees A, Foltynie T. Parkinson’s dis-
ease, insulin resistance and novel agents of neuroprotection. Brain
2013;136:374–384.
5. Nalls MA, Plagnol V, Hernandez DG, et al. Imputation of
sequence variants for identification of genetic risks for Parkinson’s
disease: a meta-analysis of genome-wide association studies. Lancet
2011;377:641–649.
6. International Parkinson’s Disease Genetics Consortium. A two-
stage meta-analysis identifies several new loci for Parkinson’s dis-
ease [serial online]. PLoS Genet 2011;7:e1002142.
7. Marras C, Schule B, Munhoz RP, et al. Phenotype in parkinsonian
and nonparkinsonian LRRK2 G2019S mutation carriers. Neurol-
ogy 2011;77:325–333.
8. Kim HJ, Kim HJ, Lee JY, et al. Phenotype analysis in patients with
early onset Parkinson’s disease with and without parkin mutations.
J Neurol 2011;258:2260–2267.
9. Lesage S, Anheim M, Condroyer C, et al. Large-scale screening of
the Gaucher’s disease-related glucocerebrosidase gene in Europeans
with Parkinson’s disease. Hum Mol Genet 2011;20:202–210.
10. Gan-Or Z, Bar-Shira A, Mirelman A, et al. LRRK2 and GBA
mutations differentially affect the initial presentation of Parkinson
disease. Neurogenetics 2010;11:121–125.
11. Tomlinson CL, Stowe R, Patel S, et al. Systematic review of levo-
dopa dose equivalency reporting in Parkinson’s disease. Mov Dis-
ord 2010;25:2649–2653.
12. Foltynie T, Zrinzo L, Martinez-Torres I, et al. MRI-guided STN
DBS in Parkinson’s disease without microelectrode recording: effi-
cacy and safety. J Neurol Neurosurg Psychiatry 2011;82:358–363.
13. Petersen EA, Holl EM, Martinez-Torres I, et al. Minimizing brain
shift in stereotactic functional neurosurgery. Neurosurgery 2010;
67:213–221.
14. Zrinzo L, van Hulzen AL, Gorgulho AA, et al. Avoiding the ven-
tricle: a simple step to improve accuracy of anatomical targeting
during deep brain stimulation. J Neurosurg 2009;110:1283–1290.
15. Zrinzo L. The role of imaging in the surgical treatment of move-
ment disorders. Neuroimaging Clin North Am 2010;20:125–140.
16. Pankratz N, Beecham GW, DeStefano AL, et al. Meta-analysis of
Parkinson’s disease: identification of a novel locus, RIT2. Ann
Neurol 2012;71:370–384.
17. Winder-Rhodes SE, Evans JR, Ban M, et al. Glucocerebrosidase
mutations influence the natural history of Parkinson’s disease in a
community-based incident cohort. Brain 2013;136:392–399.
18. Alcalay RN, Caccappolo E, Mejia-Santana H, et al. Cognitive per-
formance of GBA mutation carriers with early-onset PD: the
CORE-PD study. Neurology 2012;78:1434–1440.
19. Johansen KK, Jorgensen JV, White LR, Farrer MJ, Aasly JO. Par-
kinson-related genetics in patients treated with deep brain stimula-
tion. Acta Neurol Scand 2012;123:201–206.
20. Moro E, Volkmann J, Konig IR, et al. Bilateral subthalamic stimu-
lation in Parkin and PINK1 parkinsonism. Neurology 2008;70:
1186–1191.
21. Gomez-Esteban JC, Lezcano E, Zarranz JJ, et al. Outcome of bilat-
eral deep brain subthalamic stimulation in patients carrying the
R1441G mutation in the LRRK2 dardarin gene. Neurosurgery
2008;62:857–862.
22. Schupbach M, Lohmann E, Anheim M, et al. Subthalamic nucleus
stimulation is efficacious in patients with Parkinsonism and
LRRK2 mutations. Mov Disord 2007;22:119–122.
23. Weiss D, Brockmann K, Srulijes K, et al. Long-term follow-up of
subthalamic nucleus stimulation in glucocerebrosidase-associated
Parkinson’s disease. J Neurol 2012;259:1970–1972.
24. Witt K, Daniels C, Reiff J, et al. Neuropsychological and
psychiatric changes after deep brain stimulation for Parkinson’s
disease: a randomised, multicentre study. Lancet Neurol 2008;7:
605–614.
PD HETEROGENEITY, GENOTYPES, AND DBS
6 Movement Disorders, Vol. 00, No. 00, 2013
... In another cohort, in 9 PRKN mutation carriers (4 biallelic and 5 monoallelic) patients underwent surgery with either bilateral STN DBS or bilateral GPi DBS. STN DBS resulted in greater decrease in motor scores, while GPI DBS patients exhibited a significant improvement in dyskinesias [22]. Lohmann and co-authors (2008) assessed a cohort which included 14 PRKN mutation carriers which underwent bitaleral STN DBS. ...
Preprint
Full-text available
Introduction: Parkinson’s disease (PD) patients harboring recessive gene variants exhibit a distinct clinical phenotype with an early disease onset and relatively mild symptoms. Data concerning individualized therapy for autosomal recessive PD forms are still scarce. Methods: Demographic and treatment data of a cohort of PD carriers of recessive genes (9 homozygous or compound heterozygous PRKN carriers, 4 Heterozygous PRKN carriers and 3 biallelic PINK1 carriers) were evaluated. Results: The average Levodopa Equivalent daily dose (LEDD) was 806.8±453.5 (range 152-1810) in PRKN carriers and 765±96.6 (range 660-850) in PINK1 carriers. The majority responded to low/moderate doses of Levodopa. The response to Dopamine Agonists (DA) was often favorable both as initial and longitudinal therapy. 8/13 PRKN and 1/3 PINK1 carriers were treated with amantadine successfully, and this also applied to patients who could not tolerate Levodopa or DA. Conclusions: In the era of personalized treatment, the therapeutic approach in recessive PD gene carriers might differ as compared to idiopathic PD. Lower LEDD doses were efficient even in patients with a very long disease duration, while a few patients were doing well without any Levodopa treatment decades after disease initiation. DA or amantadine could be used as a first and main line treatment regimen if well tolerated. Literature data on therapeutic strategies in carriers of pathogenic mutations in recessive PD genes, including device-aided treatments will be further discussed.
... A much earlier clinical study at Queen Square Hospital in the United Kingdom revealed clear differences in global cognition between individuals with PD with GBA risk variants versus mutation-negative patients. 8 Over a 10year period, only 16 individuals with GBA risk variants and mutations were recruited. It is safe to assume that even in large urban academic centers performing over 50-100 new DBS electrode implantations a year, there are likely only a handful of individuals with a GBA mutation seeking advice regarding DBS in a given year. ...
... 3,4 Monogenic cases of sporadic PD have also been reported (for instance, those related to SNCA mutations). The proportion of cases with genetic causes varies between cohorts; for instance, among patients undergoing implantation of deep brain stimulators, up to 29% carry variants of the PRKN, LRRK2,o rGBA genes, 5 whereas up to 40% of Arab-Berber patients and 20% of patients of Ashkenazi Jewish descent are carriers of the G2019S variant of LRRK2. 6 To date, over 20 loci (PARK) have been associated with PD of Mendelian inheritance (Table 1), with SNCA, LRRK2, and VPS35 presenting autosomal dominant inheritance; more recently, cases have been associated with the genes EIF4G1, CHCHD2, and LPR10. ...
... Even though there was a disparity in the number of patients in groups between the mild (non-neuronopathic) and severe (neuronopathic) variants with DBS (24 risk variant, 23 mild variant, 11 severe variant), it is important to keep in mind that there are differences between variants in clinical phenotype and therapeutic outcome (Pal et al., 2022). Regardless of risk, the benefit on motor outcomes and fluctuations is becoming clearer, as beneficial effects up to a 40% reduction have been seen on the Unified Parkinson's Disease Rating Scale Part III (UPDRS III) and almost complete reduction (97%) in the Unified Parkinson's Disease Rating Scale Part IV (UPDRS IV) scores (Angeli et al., 2013;Mangone et al., 2020). Finally, longterm follow-ups are important, as it has been shown that GBA-DBS patients can have axial deterioration and postural instability, despite positive earlier improvements (Straccia et al., 2022). ...
Article
Full-text available
In this perspective article, we highlight the possible applicability of genetic testing in Parkinson’s disease and dystonia patients treated with deep brain stimulation (DBS). DBS, a neuromodulatory technique employing electrical stimulation, has historically targeted motor symptoms in advanced PD and dystonia, yet its precise mechanisms remain elusive. Genetic insights have emerged as potential determinants of DBS efficacy. Known PD genes such as GBA, SNCA, LRRK2, and PRKN are most studied, even though further studies are required to make firm conclusions. Variable outcomes depending on genotype is present in genetic dystonia, as DYT-TOR1A, NBIA/DYTPANK2, DYT-SCGE and X-linked dystonia-parkinsonism have demonstrated promising outcomes following GPi-DBS, while varying outcomes have been documented in DYT-THAP1. We present two clinical vignettes that illustrate the applicability of genetics in clinical practice, with one PD patient with compound GBA mutations and one GNAL dystonia patient. Integrating genetic testing into clinical practice is pivotal, particularly with advancements in next-generation sequencing. However, there is a clear need for further research, especially in rarer monogenic forms. Our perspective is that applying genetics in PD and dystonia is possible today, and despite challenges, it has the potential to refine patient selection and enhance treatment outcomes.
... Thus, in patients carrying GBA pathogenic variants, this vicious cycle possibly leads to a rapid progression of nigrostriatal dopaminergic deficit due to α-synuclein accumulation and may explain the rapid development of LID. Also, PD patients carrying GBA pathogenic variants generally have a more rapid motor progression [5,33,34], which can be accounted for the same explanation. It is noteworthy that patients with other α-synucleinopathy spectrum disorders, such as idiopathic RBD and dementia with Lewy bodies, are also reported to have a higher proportion of GBA pathogenic variants carriers compared to control subjects [31]. ...
Article
Full-text available
Background With the benefit of using next-generation sequencing (NGS), our aim was to examine the prevalence of known monogenic causes in early-onset Parkinson’s disease (EOPD) patients in Thailand. The association between clinical features, such as levodopa-induced dyskinesia (LID), and genotypes were also explored. Method NGS studies were carried out for EOPD patients in the Tertiary-referral center for Parkinson’s disease and movement disorders. EOPD patients who had LID symptoms were enrolled in this study (n = 47). We defined EOPD as a patient with onset of PD at or below 50 years of age. LID was defined as hyperkinetic movements including chorea, ballism, dystonia, myoclonus, or any combination of these movements resulting from levodopa therapy, which could be peak-dose, off-period, or diphasic dyskinesias. Results Pathogenic variants were identified in 17% (8/47) of the Thai EOPD patients, of which 10.6% (5/47) were heterozygous GBA variants (c.1448T>C in 3 patients and c.115+1G>A in 2 patients), 4.3% (2/47) homozygous PINK1 variants (c.1474C>T) and 2.1% (1/47) a PRKN mutation (homozygous deletion of exon 7). The LID onset was earlier in patients with GBA mutations compared to those without (34.8±23.4 vs 106.2±59.5 months after starting levodopa, respectively, p = 0.001). LID onset within the first 30 months of the disease was also found to be independently associated with the GBA mutation (odds ratio [95% confidence interval] = 25.00 [2.12–295.06], p = 0.011). Conclusion Our study highlights the high prevalence of GBA pathogenic variants in Thai patients with EOPD and the independent association of these variants with the earlier onset of LID. This emphasizes the importance of genetic testing in this population.
... 38 The "severe" p.L483P variant in particular has been associated with worse disease progression in white populations, 39 and the findings in this DBS cohort (where p.L483P accounted for 90.0% of the pathogenic/likely pathogenic GBA1 variants, vs. 18.8-25.0% in previous reports 36,37 ) suggest that this may also be the case in Asians. 12 The rates of LRRK2 Asian risk variants in our cohort (p.R1628P, 10.3%; p.G2385, 7.6%) were comparable to overall (non-DBS) Asian PD populations (each variant being present in 5-10% of patients 12 ). ...
Preprint
Background: The availability of deep brain stimulation (DBS), a highly efficacious treatment for several movement disorders, remains low in developing countries, with scarce data available on utilization and outcomes. Objectives: We characterized the DBS cohort and outcomes at a Malaysian quaternary medical centre. Methods: A retrospective chart review was done on DBS-related surgery at the University of Malaya, including clinico-demographic, genetics, and outcomes data focusing on post-operative medication reduction and complications. Results: 149 Parkinson disease (PD) patients underwent DBS targeting the subthalamic nucleus. Six had globus pallidus internus DBS (primarily for dystonia). Only 16.1% of cases were government-funded. Of the 133 PD patients operated in the past decade (2013-2022), 25 (18.8%) had disease duration <5 years. At 6-12 months post-DBS, median levodopa-equivalent daily dosage (LEDD) reduction was 440.5 [418.9] mg/day, corresponding to a reduction of at least 50% and at least 30% in 42.2% and 69.8% of patients, respectively. LEDD reductions were larger in the early-onset and short-duration subgroups. Three patients (1.9% of 155) had symptomatic intracranial hemorrhage, resulting in stroke in two. Pathogenic monogenic or GBA1 variants were detected in 12/61 (19.7%) of patients tested, mostly comprising the GBA1 variant p.L483P (14.8%). Conclusion: This is the largest report on DBS from Southeast Asia. The procedures were effective, and complication rates on par with international norms. Our study found a high frequency of GBA1-PD; and included a substantial number of patients with short-duration PD, who had good outcomes. It also highlights the inequity of access to device-aided therapy.
Article
Background In the era of modern medicine, where high‐throughput sequencing techniques are readily available, it is desirable to elucidate the role of genetic background in patients with Parkinson's Disease (PD) undergoing Deep Brain Stimulation (DBS). Genetic stratification of PD patients undergoing DBS may assist in patient selection and prediction of clinical outcomes and complement existing selection procedures such as levodopa challenge testing. Objective To capture a broad spectrum of motor and non‐motor DBS outcomes in genetic PD patients with data from the recently updated literature. Methods A multi‐scale meta‐analysis with 380 genetic PD cases was conducted using the Cochrane Review Manager, JASP software and R. Results This meta‐analysis revealed that overall, patients with genetic PD are good candidates for DBS but the outcomes might differ depending on the presence of specific mutations. PRKN carriers benefited the most regarding motor function, daily dose medication and motor complications. However, GBA carriers appeared to be more prone to cognitive decline after subthalamic nucleus DBS accompanied by a low quality of life with variable severity depending on genetic variants and concomitant alterations in other genes. Apart from GBA , cognitive worsening was also observed in SNCA carriers. Pre‐operative levodopa responsiveness and a younger age of onset are associated with a favorable motor outcome. Conclusion A personalized approach with a variant‐based risk stratification within the emerging field of surgicogenomics is needed. Integration of polygenic risk scores in clinical‐decision making should be encouraged.
Article
Genetic subtyping of patients with Parkinson's disease (PD) may assist in predicting the cognitive and motor outcomes of subthalamic deep brain stimulation (STN‐DBS). Practical questions were recently raised with the emergence of new data regarding suboptimal cognitive outcomes after STN‐DBS in individuals with PD associated with pathogenic variants in glucocerebrosidase gene ( GBA1 ‐PD). However, a variety of gaps and controversies remain. (1) Does STN‐DBS truly accelerate cognitive deterioration in GBA1 ‐PD? If so, what is the clinical significance of this acceleration? (2) How should the overall risk‐to‐benefit ratio of STN‐DBS in GBA1 ‐PD be established? (3) If STN‐DBS has a negative effect on cognition in GBA1 ‐PD, how can this effect be minimized? (4) Should PD patients be genetically tested before STN‐DBS? (5) How should GBA1 ‐PD patients considering STN‐DBS be counseled? We aim to summarize the currently available relevant data and detail the gaps and controversies that exist pertaining to these questions. In the absence of evidence‐based data, all authors strongly agree that clinicians should not categorically deny DBS to PD patients based solely on genotype ( GBA1 status). We suggest that PD patients considering DBS may be offered genetic testing for GBA1 , where available and feasible, so the potential risks and benefits of STN‐DBS can be properly weighed by both the patient and clinician. © 2023 International Parkinson and Movement Disorder Society.
Article
Full-text available
Background GBA variants increase the risk of developing Parkinson disease (PD) and influence its outcome. Deep brain stimulation (DBS) is a recognised therapeutic option for advanced PD. Data on DBS long-term outcome in GBA carriers are scarce. Objective To elucidate the impact of GBA variants on long-term DBS outcome in a large Italian cohort. Methods We retrospectively recruited a multicentric Italian DBS-PD cohort and assessed: (1) GBA prevalence; (2) pre-DBS clinical features; and (3) outcomes of motor, cognitive and other non-motor features up to 5 years post-DBS. Results We included 365 patients with PD, of whom 73 (20%) carried GBA variants. 5-year follow-up data were available for 173 PD, including 32 mutated subjects. GBA-PD had an earlier onset and were younger at DBS than non-GBA-PD. They also had shorter disease duration, higher occurrence of dyskinesias and orthostatic hypotension symptoms. At post-DBS, both groups showed marked motor improvement, a significant reduction of fluctuations, dyskinesias and impulsive-compulsive disorders (ICD) and low occurrence of most complications. Only cognitive scores worsened significantly faster in GBA-PD after 3 years. Overt dementia was diagnosed in 11% non-GBA-PD and 25% GBA-PD at 5-year follow-up. Conclusions Evaluation of long-term impact of GBA variants in a large Italian DBS-PD cohort supported the role of DBS surgery as a valid therapeutic strategy in GBA-PD, with long-term benefit on motor performance and ICD. Despite the selective worsening of cognitive scores since 3 years post-DBS, the majority of GBA-PD had not developed dementia at 5-year follow-up.
Article
Full-text available
This article provides an overview of the various screening and selection tools which have been developed over the past 25 years to identify patients with Parkinson’s disease (PD) possibly eligible for device-aided therapies (DATs). For the available screening tools, we describe the target therapies (subtypes of DAT), development methods, validation data, and their use in clinical practice. In addition, the historical background and potential utility of these screening tools are discussed. The challenges in developing and validating these tools are also addressed, taking into account the differences in population, the local health care organization, and resource availability.
Article
Full-text available
Carriers of mutations in the glucocerebrosidase gene (GBA) are at increased risk of developing Parkinson's disease. The frequency of GBA mutations in unselected Parkinson's disease populations has not been established. Furthermore, no previous studies have investigated the influence of GBA mutations on the natural history of Parkinson's disease using prospective follow-up. We studied DNA from 262 cases who had been recruited at diagnosis into one of two independent community-based incidence studies of Parkinson's disease. In 121 cases, longitudinal data regarding progression of motor disability and cognitive function were derived from follow-up assessments conducted every 18 months for a median of 71 months. Sequencing of the GBA was performed after two-stage polymerase chain reaction amplification. The carrier frequency of genetic variants in GBA was determined. Baseline demographic and clinical variables were compared between cases who were either GBA mutation carriers, polymorphism carriers or wild-type homozygotes. Cox regression analysis was used to model progression to major motor (Hoehn and Yahr stage 3), and cognitive (dementia) end-points in cases followed longitudinally. We show that in a representative, unselected UK Parkinson's disease population, GBA mutations are present at a frequency of 3.5%. This is higher than the prevalence of other genetic mutations currently associated with Parkinson's disease and indicates that GBA mutations make an important contribution to Parkinson's disease encountered in the community setting. Baseline clinical characteristics did not differ significantly between cases with and without GBA sequence variants. However, the hazard ratio for progression both to dementia (5.7, P = 0.003) and Hoehn and Yahr stage 3 (4.2, P = 0.003) were significantly greater in GBA mutation carriers. We also show that carriers of polymorphisms in GBA which are not generally considered to increase Parkinson's disease risk are at significantly increased risk of progression to Hoehn and Yahr stage 3 (3.2, P = 0.004). Our results indicate that genetic variation in GBA has an important impact on the natural history of Parkinson's disease. To our knowledge, this is the first time a genetic locus has been shown to influence motor progression in Parkinson's disease. If confirmed in further studies, this may indicate that GBA mutation status could be used as a prognostic marker in Parkinson's disease. Elucidation of the molecular mechanisms that underlie this effect will further our understanding of the pathogenesis of the disease and may in turn suggest novel therapeutic strategies.
Article
Full-text available
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is a commonly employed therapeutic procedure for patients with Parkinson's disease uncontrolled by medical therapies. This series describes the outcomes of 79 consecutive patients that underwent bilateral STN DBS at the National Hospital for Neurology & Neurosurgery between November 2002 and November 2008 using an MRI-guided surgical technique without microelectrode recording (MER). Patients underwent immediate postoperative stereotactic MR imaging. The mean (SD) error in electrode placement was 1.3 (0.6) mm. There were no haemorrhagic complications. At a median follow up period of 12 months, there was a mean improvement in the off-medication motor part of the Unified Parkinson´s Disease Rating Scale (UPDRS III) of 27.7 points (SD 13.8) equivalent to a mean improvement of 52% (p<0.0001). In addition there were significant improvements in dyskinesia duration, disability and pain, with a mean reduction in on-medication dyskinesia severity (sum of dyskinesia duration, disability and pain from UPDRS IV) from 3.15 (SD 2.33) pre-operatively, to 1.56 (SD 1.92) post operatively (p=0.0001). Quality of life improved by a mean of 5.5 points (median 7.9 points, SD 17.3) on the Parkinson's disease Questionnaire 39 (PDQ39) summary index. This series confirms that image-guided STN DBS without microelectrode recording can lead to substantial improvements in motor disability of well-selected PD patients with accompanying improvements in quality of life and most importantly, with very low morbidity.
Article
Full-text available
Multiple avenues of research including epidemiology, molecular genetics and cell biology have identified links between Parkinson's disease and type 2 diabetes mellitus. Several recent discoveries have highlighted common cellular pathways that potentially relate neurodegenerative processes with abnormal mitochondrial function and abnormal glucose metabolism. This includes converging evidence identifying that peroxisome proliferator activated receptor gamma coactivator 1-α, a key regulator of enzymes involved in mitochondrial respiration and insulin resistance, is potentially pivotal in the pathogenesis of neurodegeneration in Parkinson's disease. This evidence supports further study of these pathways, most importantly to identify neuroprotective agents for Parkinson's disease, and/or establish more effective prevention or treatment for type 2 diabetes mellitus. In parallel with these advances, there are already randomized trials evaluating several established treatments for insulin resistance (pioglitazone and exenatide) as possible disease modifying drugs in Parkinson's disease, with only preliminary insights regarding their mechanisms of action in neurodegeneration, which may be effective in both disease processes through an action on mitochondrial function. Furthermore, parallels are also emerging between these same pathways and neurodegeneration associated with Alzheimer's disease and Huntington's disease. Our aim is to highlight this converging evidence and stimulate further hypothesis-testing studies specifically with reference to the potential development of novel neuroprotective agents in Parkinson's disease.
Article
Full-text available
A previous genome-wide association (GWA) meta-analysis of 12,386 PD cases and 21,026 controls conducted by the International Parkinson's Disease Genomics Consortium (IPDGC) discovered or confirmed 11 Parkinson's disease (PD) loci. This first analysis of the two-stage IPDGC study focused on the set of loci that passed genome-wide significance in the first stage GWA scan. However, the second stage genotyping array, the ImmunoChip, included a larger set of 1,920 SNPs selected on the basis of the GWA analysis. Here, we analyzed this set of 1,920 SNPs, and we identified five additional PD risk loci (combined p<5×10(-10), PARK16/1q32, STX1B/16p11, FGF20/8p22, STBD1/4q21, and GPNMB/7p15). Two of these five loci have been suggested by previous association studies (PARK16/1q32, FGF20/8p22), and this study provides further support for these findings. Using a dataset of post-mortem brain samples assayed for gene expression (n = 399) and methylation (n = 292), we identified methylation and expression changes associated with PD risk variants in PARK16/1q32, GPNMB/7p15, and STX1B/16p11 loci, hence suggesting potential molecular mechanisms and candidate genes at these risk loci.
Article
Mitochondria have a crucial role in cellular bioenergetics and apoptosis, and thus are important to support cell function and in determination of cell death pathways. Inherited mitochondrial diseases can be caused by mutations of mitochondrial DNA or of nuclear genes that encode mitochondrial proteins. Although many mitochondrial disorders are multisystemic, some are tissue specific--eg, optic neuropathy, sensorineural deafness, and type 2 diabetes mellitus. In the past few years, several disorders have been associated with mutations of nuclear genes responsible for mitochondrial DNA maintenance and function, and the potential contribution of mitochondrial abnormalities to progressive neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease has been recognised. The process of mitochondrial fission-fusion has become a focus of attention in human disease. Importantly, the mitochondrion is now a target for therapeutic interventions that encompass small molecules, transcriptional regulation, and genetic manipulation, offering opportunities to treat a diverse range of diseases.
Article
Genome-wide association (GWAS) methods have identified genes contributing to Parkinson's disease (PD); we sought to identify additional genes associated with PD susceptibility. A 2-stage design was used. First, individual level genotypic data from 5 recent PD GWAS (Discovery Sample: 4,238 PD cases and 4,239 controls) were combined. Following imputation, a logistic regression model was employed in each dataset to test for association with PD susceptibility and results from each dataset were meta-analyzed. Second, 768 single-nucleotide polymorphisms (SNPs) were genotyped in an independent Replication Sample (3,738 cases and 2,111 controls). Genome-wide significance was reached for SNPs in SNCA (rs356165; G: odds ratio [OR]=1.37; p=9.3×10(-21)), MAPT (rs242559; C: OR=0.78; p=1.5×10(-10)), GAK/DGKQ (rs11248051; T: OR=1.35; p=8.2×10(-9)/rs11248060; T: OR=1.35; p=2.0×10(-9)), and the human leukocyte antigen (HLA) region (rs3129882; A: OR=0.83; p=1.2×10(-8)), which were previously reported. The Replication Sample confirmed the associations with SNCA, MAPT, and the HLA region and also with GBA (E326K; OR=1.71; p=5×10(-8) Combined Sample) (N370; OR=3.08; p=7×10(-5) Replication sample). A novel PD susceptibility locus, RIT2, on chromosome 18 (rs12456492; p=5×10(-5) Discovery Sample; p=1.52×10(-7) Replication sample; p=2×10(-10) Combined Sample) was replicated. Conditional analyses within each of the replicated regions identified distinct SNP associations within GBA and SNCA, suggesting that there may be multiple risk alleles within these genes. We identified a novel PD susceptibility locus, RIT2, replicated several previously identified loci, and identified more than 1 risk allele within SNCA and GBA.
Article
To assess the cognitive phenotype of glucocerebrosidase (GBA) mutation carriers with early-onset Parkinson disease (PD). We administered a neuropsychological battery and the University of Pennsylvania Smell Identification Test (UPSIT) to participants in the CORE-PD study who were tested for mutations in PARKIN, LRRK2, and GBA. Participants included 33 GBA mutation carriers and 60 noncarriers of any genetic mutation. Primary analyses were performed on 26 GBA heterozygous mutation carriers without additional mutations and 39 age- and PD duration-matched noncarriers. Five cognitive domains, psychomotor speed, attention, memory, visuospatial function, and executive function, were created from transformed z scores of individual neuropsychological tests. Clinical diagnoses (normal, mild cognitive impairment [MCI], dementia) were assigned blind to genotype based on neuropsychological performance and functional impairment as assessed by the Clinical Dementia Rating (CDR) score. The association between GBA mutation status and neuropsychological performance, CDR, and clinical diagnoses was assessed. Demographics, UPSIT, and Unified Parkinson's Disease Rating Scale-III performance did not differ between GBA carriers and noncarriers. GBA mutation carriers performed more poorly than noncarriers on the Mini-Mental State Examination (p = 0.035), and on the memory (p = 0.017) and visuospatial (p = 0.028) domains. The most prominent differences were observed in nonverbal memory performance (p < 0.001). Carriers were more likely to receive scores of 0.5 or higher on the CDR (p < 0.001), and a clinical diagnosis of either MCI or dementia (p = 0.004). GBA mutation status may be an independent risk factor for cognitive impairment in patients with PD.
Article
Parkinson's disease (PD) and dementia with Lewy bodies are common disorders of the aging population and characterized by the progressive accumulation of α-synuclein (α-syn) in the central nervous system. Aggregation of α-syn into oligomers with a ring-like appearance has been proposed to play a role in toxicity. However, the molecular mechanisms and the potential sequence of events involved in the formation of pore-like structures are unclear. We utilized computer modeling and cell-based studies to investigate the process of oligomerization of wild-type and A53T mutant α-syn in membranes. The studies suggest that α-syn penetrates the membrane rapidly, changing its conformation from α-helical towards a coiled structure. This penetration facilitates the incorporation of additional α-syn monomers in the complex, and the subsequent displacement of phospholipids and the formation of oligomers in the membrane. This process occurred more rapidly, and with a more favorable energy of interaction, for mutant A53T compared with wild-type α-syn. After 4 ns of simulation of the protein-membrane model, α-syn had penetrated through two-thirds of the membrane. By 9 ns, the penetration of the annular α-syn oligomers can result in the formation of pore-like structures that fully perforate the lipid bilayer. Experimental incubation of recombinant α-syn in synthetic membranes resulted in the formation of similar pore-like complexes. Moreover, mutant (A53T) α-syn had a greater tendency to accumulate in neuronal membrane fractions in cell cultures, resulting in greater neuronal permeability, as demonstrated with the calcein efflux assay. These studies provide a sequential molecular explanation for the process of α-syn oligomerization in the membrane, and support the role of formation of pore-like structures in the pathogenesis of the neurodegenerative process in PD.