ArticlePDF Available

P268S in NOD2 associates with susceptibility to Parkinson’s disease in Chinese population

Authors:

Abstract and Figures

Background The cause of almost all cases of Parkinson’s disease (PD) remains unknown. Recent years have seen an explosion in the rate of discovery of genetic defects linked to PD. Different racial and geographical populations may have different distributions of genetic variants. Methods In the current study, we screened the following genetic variants, including some rare mutations and single nucleotide polymorphisms (SNPs), in a pedigree and cases-controls. To best of our knowledge, we first screened these variants known to be associated with neurodegeneration disease, E46K (rs104893875) in SNCA, A1442P in LRRK2, IVS9 in PARK2, A350V in SLC41A1, P268S (rs2066842), R702W (rs2066844), G908R (rs2066845), 1007fs (rs2066847) in NOD2 and G2385R (rs34778348) in LRRK2 from southern China population. Genotyping was performed by jointly using primers overlapping polymerase chain reaction (PCR) site-directed mutagenesis, restriction fragment length polymorphism (RFLP), and capillary electrophoresis (CE). Results We didn’t discover above 9 variants in the family members of the pedigree. Furthermore, of 237 patients with sporadic Parkinson’s disease and 190 controls, no heterozygosity or homozygosity were found from E46K, A1442P, A350V, R702W, G908R, or 1007fs but heterozygosity onto G2385R, IVS9, and P268S. No significant difference between cases and controls was found in both allele frequency (P = 0.572) and genotype frequency (P = 0.348) of IVS9. However, significant differences in genotype frequency (P = 0.009) of G2385R were consistent with prior observation. Eight patients with Parkinson’s disease (2 women and 6 men are over the age of 50 years at onset of PD) carried the P268S heterozygous variation in NOD2. There was no heterozygosity or homozygosity of P268S in the controls. Genotype frequency of P268S (P = 0.0450) had significant differences. Conclusions Our results suggested that the P268S variant in NOD2 might be a risk factor for susceptibility to sporadic Parkinson’s disease in Chinese populations. It also implied that the inflammatory response may play a role in PD.
Content may be subject to copyright.
R E S E A R C H Open Access
P268S in NOD2 associates with susceptibility to
Parkinsons disease in Chinese population
Qilin Ma
1
, Xingkai An
1
, Zhiming Li
2
, Huanjing Zhang
2
, Wenqing Huang
2
, Liangliang Cai
2
, Peng Hu
2
,
Qing Lin
1,2*
and Chi-Meng Tzeng
2*
Abstract
Background: The cause of almost all cases of Parkinsons disease (PD) remains unknown. Recent years have seen
an explosion in the rate of discovery of genetic defects linked to PD. Different racial and geographical populations
may have different distributions of genetic variants.
Methods: In the current study, we screened the following genetic variants, including some rare mutations and
single nucleotide polymorphisms (SNPs), in a pedigree and cases-controls. To best of our knowledge, we first
screened these variants known to be associated with neurodegeneration disease, E46K (rs104893875) in SNCA,
A1442P in LRRK2, IVS9 in PARK2, A350V in SLC41A1, P268S (rs2066842), R702W (rs2066844), G908R (rs2066845), 1007fs
(rs2066847) in NOD2 and G2385R (rs34778348) in LRRK2 from southern China population. Genotyping was
performed by jointly using primers overlapping polymerase chain reaction (PCR) site-directed mutagenesis,
restriction fragment length polymorphism (RFLP), and capillary electrophoresis (CE).
Results: We didnt discover above 9 variants in the family members of the pedigree. Furthermore, of 237 patients
with sporadic Parkinsons disease and 190 controls, no heterozygosity or homozygosity were found from E46K,
A1442P, A350V, R702W, G908R, or 1007fs but heterozygosity onto G2385R, IVS9, and P268S. No significant difference
between cases and controls was found in both allele frequency (P= 0.572) and genotype frequency (P= 0.348) of
IVS9. However, significant differences in genotype frequency (P= 0.009) of G2385R were consistent with prior
observation. Eight patients with Parkinsons disease (2 women and 6 men are over the age of 50 years at onset of
PD) carried the P268S heterozygous variation in NOD2. There was no heterozygosity or homozygosity of P268S in
the controls. Genotype frequency of P268S (P= 0.0450) had significant differences.
Conclusions: Our results suggested that the P268S variant in NOD2 might be a risk factor for susceptibility to
sporadic Parkinsons disease in Chinese populations. It also implied that the inflammatory response may play a role
in PD.
Keywords: Parkinsons disease, Variants, P268S
Background
Parkinsons disease (PD) is a complex neurodegenerative
disease caused by a variety of factors. The incidence in the
population over the age of 65 is about 1.8% [1]. The major
clinical features of PD include resting tremor, bradykinesia
and rigidity [2]. The pathological features of PD include pro-
gressive loss of dopaminergic neurons from the substantia
nigra pars compacta and the presence of intercellular Lewy
bodies in surviving neurons [3]. The cause of PD is unclear,
but it is generally considered to be associated with the im-
pairment in degradation of ubiquitin proteasome system
(UPS), mitochondrial dysfunction, and oxidative stress [4-6].
We hypothesize that genetic and environmental factors and
aging together lead to the development of PD. In this study,
we investigated five genes, including SNCA,LRRK2,PA RK2,
SLC41A1,andNOD2, all of which might be involved in the
cytological mechanisms of neurodegeneration disease aiming
to identify genetic variants associated with sporadic PD in
Chinese patients and understand the genetic etiology of PD.
* Correspondence: linqing2005602@aliyun.com;cmtzeng@xmu.edu.cn
Equal contributors
1
Department of Neurology, The First Affiliated Hospital of Xiamen University,
Xiamen, Fujian 361003, China
2
School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian
361005, China
© 2013 Ma et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.
Ma et al. Behavioral and Brain Functions 2013, 9:19
http://www.behavioralandbrainfunctions.com/content/9/1/19
The first gene to be identified as associated with PD was
SNCA (PARK1/4 ), which encodes α-synuclein. The major
mutations in SNCA include A53T, A30P, and E46K [7-9].
Although these mutations account for less than 1% of the
cases, patients carrying these mutations have obviously
clinical phenotypes [8]. The A53T and A30P mutations
have been found in Chinese patients with sporadic PD
[10]. However, the association of E46K with sporadic PD
has not been reported in Chinese patients.
LRRK2 (PARK8) encodes a large protein containing
five functional domains, involved in a number of
physiological functions, including substrate binding,
protein phosphorylation and protein interactions [11].
The most common mutation G2019 of LRRK2 in
European populations presents less than 0.1% in Asian
individuals [12]. The G2385R was at a significantly
higher frequency in Asian patients than in controls
[13]. One of the purposes of this study is to re-
validate this variant in southern China. In addition,
Yue Huang found A1442P in pedigrees of the Austra-
lian patients and considered it pathogenic, because it
is conserved across many species and a substitution of
Ala with Pro can change the secondary structure of
proteins [14]. However, A1442P has not been further
confirmed from a large number of cases and other
populations.
The most common cause of young-onset sporadic PD
is autosomal recessive PARK2 mutation [15]. The E3
ubiquitin ligase, parkin, which is encoded by PARK2, can
specifically degrade UPS through tagging ubiquitin on
protein [16]. Exon deletion, insertion, and point muta-
tions in PARK2 have been found in different ethnic
groups [17]. In 2009, Yih-Ru Wu screened 506 Taiwan
sporadic patients with age of onset below 50 years for
PARK2 gene mutation and identified a novel IVS9 inser-
tion (c.1084intron
+
) [18]. The c.1084intron
+
was due to
a G > A polymorphism at position 6 of a cryptic splice
acceptor site within IVS9.
In recent years, genome-wide association studies
(GWAS) have identified a number of new susceptibility
loci associated with PD in different ethnic groups. Of
these loci, the most striking locus is PARK16, which is lo-
cated in 1q23 and contains 5 genes (SLC45A3,NUCKS1,
RAB7L1,SLC41A1, and PM20D1) [19]. SLC41A1 en-
codes a 56 kDa Mg
2+
transporter consisting of 513 amino
acids, it has been proposed that PD might be associated
with lack of Ca
2+
and Mg
2+
in the brain [20]. In 2010,
Arianna Tucci sequenced PARK16 in 182 patients with
PD in the United Kingdom and found A350V in
SLC41A1, but failed to detect it in a large series of
ethnicity-matched controls (n = 483) [19]. To inspect
whether this coding mutations may or may not be the
disease-causing mutation, we aim to screen A350V in
Chinese Han populations.
Several obtained findings suggest that inflammation
also contributes to the pathogenesis of PD [21-23]. It
has been reported that the G174C variant in the IL-6
promoter may influence the risk for developing PD, par-
ticularly regarding early age of onset PD [24]. Recent
studies revealed that NF-κB-mediated inflammation
might also play an important role in the pathogenesis of
PD [25,26]. NOD (nucleotide-binding oligomerization
domain) proteins, i.e. NOD1 (a product of CARD4 gene)
and NOD2 (encoded by NOD2), are intracellular signal-
ing molecules that recognize bacterial components, me-
diate the activation of NF-κB and induce or enhance
apoptosis [27]. In 2007, Monika Bialecka et al. screened
308 Portland patients for 3 variants (R702W, G908R,
and 1007fs) in NOD2 which were associated with
Crohns disease (CD) in Europeans. They found that
NOD2 might be associated with susceptibility to PD
[28]. Although inflammatory response has long been
considered as one of the factors for PD development, it
has not yet been reported that R702W, G908R, 1007fs
and P268S in NOD2 is associated with Chinese PD
patients.
In this study, we screened 237 patients with sporadic PD
and 190 controls for the 9 variants, E46K (rs104893875),
G2385R (rs34778348), A1442P, IVS9, A350V, R702W
(rs2066844), G908R (rs2066845), 1007fs (rs2066847) and
P268S (rs2066842), in 5 genes which are possibly found to
be associated with Chinese PD patients, including SNC A,
LRRK2,PARK2,SLC41A1,andNOD2.Thesevariantsin-
cluded both rare mutations (e.g. E46K and A1442P) and
SNPs, so we also screened them in a pedigree with two PD
cases. In this way, we aimed to provide references for the
study of disease-causing or susceptibility gene for PD. In
addition, using the overlap extension polymerase chain reac-
tion (PCR)-based site-directed mutagenesis, we constructed
wild-type and homozygous mutant plasmids for these vari-
ants, which were used as controls to determine variants in
this study.
Material and methods
Study subjects
A total of 237 sporadic patients with PD (94 women and
143 men) and a pedigree with two PD cases were recruited
from Xiamen First Hospital, Fujian Province, China. The
average age of enrollment was 60.3 ± 11.3, ranging from 25
to 83 years. Their average age of onset was 56.4 ± 10.8 years,
rangingfrom23to80years.Ofthesepatients,51(21.5%)
had early-onset PD (EOPD), defined as the age at onset <50
years; 186 (78.5%) cases had late-onset PD (LOPD), with age
of onset 50 years. Patients were independently diagnosed
by two neurologists. Diagnostic criteria were taken from the
United Kingdom PD Brain Bank [29]. The severity of the
disease was determined using the Unified ParkinsonsDis-
ease Rating Scale (UPDRS). The control group consisted of
Ma et al. Behavioral and Brain Functions 2013, 9:19 Page 2 of 8
http://www.behavioralandbrainfunctions.com/content/9/1/19
190 individuals (75 women and 115 men), with an average
age of 51.7 ± 9.8 years (ranging from 23 to 80 years). The
cases and controls were matched with respect to age, gen-
der, and place of residence. This study was approved by the
Xiamen First Hospital Ethics Committee. Informed consent
was obtained from all participants.
Genomic DNA extraction
Five milliliters of peripheral blood with ethylene diamine
tetraacetic acid (EDTA) anticoagulant was collected from
each participant. Genomic DNA was extracted using the
MagCore Genomic DNA Whole Blood Kit and HF-16 ex-
tractor (Cat. No. MGB400-04, RBC Bioscience Taiwan)
according to the manufacturers instructions and stored all
samples at 20°C before use.
PCR amplification
Primers (Table 1) were designed using the software Pri-
mer Premier 5 according to the change of restriction
sites, caused by the mutation. Primers were synthesized
by Sangon Biotech (Shanghai, China).
Restriction endonuclease analysis
Target variants were detected using the PCR-RFLP
(restriction fragment length polymorphism) method.
The digestion reaction and restriction enzyme digestion
conditions were taken from the manuals included with
the restriction enzymes purchased from the New England
BioLab (United States). Then 20 μl of digestion products
were loaded on 2% agarose gel containing ethidium brom-
ide, electrophoresed under 100 V for 30 min in 1× TAE,
and imaged using the gel imager. Digestion results of
homozygous mutants, heterozygous mutants, and wild-
type are shown in Table 1.
DNA sequencing
Based on the results from PCR-RFLP and site-directed
mutagenesis, the target fragments were re-amplified
using PCR. PCR products were purified using a DNA gel
purification kit, sequenced using the dideoxy four-color
fluorescence method on an automatic sequencer (ABI,
U.S.), and compared using GenBank data to confirm the
mutation sites.
Capillary electrophoresis (CE)
In this study, we initially established the test method
using CE to detect gene polymorphisms for PD. Briefly,
based on the susceptibility gene polymorphisms of PD
identified from PCR-RFLP, restriction fragments were
detected using capillary electrophoresis (BIOptics
Table 1 Primer sequences, PCR conditions and restriction digestion predictions
Polymorphism Primer sequences (53) Annealing
temperature (°C)
Restriction
enzyme
Amplified products
length (bp)
Restriction products
length (bp)
E46K F: TGATGTGGGAACAAAGGGGA 58 BsaJI 747 46E allele: 287,460
R: GTGTTTCCTGAAATGCACTCTGA 46K allele: 747
A1442P F: GAGACTAAACTGCTGCTTGC 58 Hhal 801 1442A allele: 671,130
R: GTAATCTCGTATGGCAGGGA 1442P allele: 801
A350V F: TCAGTGGTCTTTGCGTCATT 58 MwoI 302 350A allele: 94,208
R: CTGTCCTTTTACTCTGCTCCC 350V allele: 302
P268S F: AGCCCATTGTCTGGTTAGGT 58 BamHI 309 268P allele: 309
R: ACAGTGTCCGCATCGTCAT 268S allele: 225,84
R702W F: AGATCACAGCAGCCTTCC 63 MspI 185 702R allele: 20, 35, 54, 76
R: CACGCTCTTGGCCTCACC 702W allele: 20, 35, 130
G908R F: CCCAGCTCCTCCCTCTTC 63 Hin6I 380 908G allele: 380
R: AAGTCTGTAATGTAAAGCCAC 908R allele: 242, 138
1007fs F:
GGCAGAAGCCCTCCTGCAGGGCC
58 ApaI 151 Wild type allele: 151
R: CCTCAAAATTCTGCCATTCC 1007f allele: 130, 21
IVS9* F:
ACTCCTGCGCTTGATTTAGGCAAT
58 Xhol 775 Wild type allele: 318,457
R:
TTGGAATTTAGCTGTTCCTTCGGG
IVS9 G A allele: 775
G2385R F:
AGACACTGCTCTCTATATTGCTAAG
58 AccI 261 2385G:261
R: CTGAAAAGATGGTGCTGAGAAG 2385R:77,184
*: IVS9: Intervening sequence 9, PARK2.
Ma et al. Behavioral and Brain Functions 2013, 9:19 Page 3 of 8
http://www.behavioralandbrainfunctions.com/content/9/1/19
Qsep100 dna-CE, Taiwan), which used a molten molding
silica capillary column with an inner diameter of 75 μl
and column length of 150 mm and Mops-Tris buffer of
pH 7.55. Then 0.5 μl of sample was electrically injected
in 4kV × 15 s at 25°C, electrophoresed at a constant volt-
age of 8 Kv, and detected for the laser-induced ethidium
bromide emission wavelength at 590 nm. The gel elec-
trophoresis profiles were compared to determine the
genotypes.
Statistical analysis
SPSS 18.0 (Statistical Package for the Social Sciences,
version 18.0 for Windows) was used for statistical ana-
lysis. Allele and genotype frequencies were compared
between cases and controls using the Fishers exact test.
To avoid multiple easily false, false discovery rate was
used to correct p values. Odds ratios and 95% confi-
dence intervals were calculated. A two-sided Pvalue
0.05 was considered statistically significant. Power was
calculated by Power and Sample Size Calculations Ver-
sion 3.0, 2009.
Results
Pedigree
Figure 1 shows the pedigree (2 generations) of the family
that we studied. The age of PD onset for the affected in-
dividuals 3 and 4 in the kindred is 45 and 40, respect-
ively. We failed to discover any mutations of the 9
genetic variants, including rare mutations E46K and
A1442P originally found in the kindred. It may be
caused by low family members to detect the association.
Further studies using a larger Chinese pedigree with PD
patients are required to determine this.
Casecontrol study
We analyzed 9 variants, E46K, G2385R, A1442P, IVS9,
A350V, P268S, R702W, G908R, and 1007fs in 237 spor-
adic patients with PD and 190 controls using the PCR-
RFLP and CE. All the genotypic and allelic distributions
analyzed in this study were in accordance with the
HardyWeinberg equilibrium. No heterozygosity or
homozygosity of E46K, A1442P, A350V, R702W, G908R,
or 1007fs was found. Heterozygosity of G2385R, IVS9,
and P268S were found in the case groups and controls
(Figure 2). The genotype frequency, allele frequency,
odds ratios, and confidence interval of these variants are
shown in Table 2.
The distribution of G2385R heterozygosity in LRRK2
was significantly different between cases and controls
(10.1% vs. 2.1%, P= 0.009). The intron IVS9 heterozy-
gous variant in PARK2 was found in both the case group
(12.7%) and the control group (7.9%). Although this vari-
ant had a higher frequency in the case group than the
control group, the difference was not statistically signifi-
cant (P= 0.348).
Although only 8 out of 237 patients were found to
carry the P268S heterozygous variant in NOD2, all these
patients (2 women and 6 men) had age at onset of over
50 years. Since no controls were found to carry this vari-
ant, the difference between cases and controls was
slightly significant (P= 0.0450). These results suggest
that the variant P268S in NOD2 might be associated
with late-onset PD and is one of the risk factors for
sporadic PD in Chinese Han populations.
Discussion
Gene polymorphism is a risk factor of PD, and it shows
different distributions in different ethnic groups and
geographies. Although GWAS have identified a number
of susceptibility genes and relevant variants associated
with PD in different populations, few of them have been
analyzed in other populations. To best of out knowledge,
we first screened 8 variants that have been associated
with neurodegeneration, E46K, A1442P, A350V, IVS9,
P268S, R702W, G908R, and 1007fs in Chinese popula-
tions, and further verified the presence of the high-
frequency variant G2385R in southern China.
In casecontrol study, both PD patients and controls
carried the heterozygous G2385R mutation, and the
genotype difference was slightly significant. This is con-
sistent with the results of a previous study of 600
Chinese PD patients and 334 controls [30]. This variant
was initially reported by Xingkai An. in 2008 among PD
patients in Sichuan area [30]. However, we did not find
E46K, A1442P, or A350V variants in patients with PD
and in controls. All of them indicated following
phenomenon: the ancestry effect; familial PD results
from rare, highly penetrant pathogenic mutation (e.g.
12
34
Figure 1 Pedigree of Parkinsons disease (PD). Circle, woman;
square, man; filled square, affected.
Ma et al. Behavioral and Brain Functions 2013, 9:19 Page 4 of 8
http://www.behavioralandbrainfunctions.com/content/9/1/19
E46K, A1442P); multiple variants of low penetrant (e.g.
A350V) contribute to the risk of PD and are involved in
the etiology of PD.
Interestingly, none of homozygosity of these variants
was found in either the case or control groups [31,32]. It
was reported that heterozygosity of autosomal recessive
genes was very important in the initiation and develop-
ment of PD. For example, it has been found that patients
with PD who carry heterozygous PARK2 and PINK1 mu-
tations have an age of onset between that of wild-type
individuals and patients with homozygous mutations
[33,34]. Heterozygous mutation carriers indeed show
preclinical changes at the metabolic, structural, or func-
tional levels that are detectable by modern techniques of
neuroimaging and electrophysiology. Heterozygosity for
putative recessive mutations could lead to disease expression
by at least three mechanisms which are haploinsuffciency,
dominant-negative effect and novel gain-of-function [35].
Taken together, heterozygous mutations in putative
recessive genes seem to increase the susceptibility to
develop PD.
The new insertion mutation, c.1084intron
+
in PARK2,
introduces two new amino acids and a TAA stop codon,
resulting in reduced levels of parkin protein [36]. This
insertion mutation may be associated with the -6G > A
polymorphism of intron 9 in PARK2. One casecontrol
study found IVS9G > A to be a risk factor of PD in
Taiwan. A functional study of IVS9G > A by Guey-Jen
Lee-Chen showed that the shear efficiency of -6A was
slower than that of -6G at the protein level [37]. 12.7%
PD patients and 7.9% controls carried the heterozygous
IVS9 mutation, but the allele difference and genotype
difference were not statistically significant. This result is
consistent with the results of a previous study of 506
patients and 508 controls in Taiwan [18]. Further
investigation into the association between IVS9G > A
and PD need, through larger sample size from various
populations.
Inflammation is one of the suspected theories of PD.
The activation of NF-κB and microglial cells around
dopaminergic neurons have been analyzed in patients
with PD [38]. In 2007, Monika Bialecka reported that
three variants (R702W, G908R, and L1007fsinsC) of
NOD2 encoding NOD2, a protein that can activate the
apoptosis response, were significantly associated with
PD in a Polish population [28]. It has been demonstrated
that these three variants are not associated with CD in
Chinese Han, Korean, and Japanese populations [39-42].
Similarly, these three common SNPs (R702W, G908R,
and 1007fs) were not detected in our cohort. However, a
novel variant P268S was observed, which has also been
discovered to be associated with CD in Chinese popula-
tion [43]. Although linkage disequilibrium was observed
between P268S and the other three SNPs in NOD2, only
P268S of the four variants was slightly significant in our
study, which was consistent with recent study suggesting
that P268S instead of 1007fs, G908R, R702W was a risk
factor of Chinese Crohns disease population. It
ace
bdf
Figure 2 Capillary electrophoresis (CE) results of restriction fragments of 3 variants. a,c,eand b,d,fare the electropherograms and gel-
views of IVS9, P268S, and G2385R after restriction digestion, respectively. The green arrows indicate markers (20 bp, 1000 bp) and black arrows
indicate the restriction fragments. Restriction fragment sizes are shown in the Table 1: No mutation (wild-type/negative control), 2: Negative
results in the control group, 3: Positive results (heterozygote) in the case group, 4: Homozygous mutation (homozygote/positive control). Because
the signals are too strong for IVS9-4, P268S-1, P268S-2, G2385R-1, and G2385-2, 10 times markers are used to enhance the signal.
Ma et al. Behavioral and Brain Functions 2013, 9:19 Page 5 of 8
http://www.behavioralandbrainfunctions.com/content/9/1/19
indicated that P268S may be one of risk factors for PD
in Chinese individuals, or the P268S variant may be in
linkage disequilibrium with another causal rare variant
on NOD2 gene. The frequencies of minor allele 1007fs,
G908R, R702W and P268S variants are much lower in
Asians compare to Caucasians populations. In this study,
genotype frequency of P268S (P= 0.0450) had slightly
significant differences. The sample size has to be very
large or odds ratio has to be very high to have enough
power for detection effect of very rare variants.
NOD2 is a member of the Apaf-1 superfamily of apop-
tosis regulators that is expressed in monocytes and
involved in the activation of NF-κB, by bacterial compo-
nent muramyl dipeptide (MDP) [44]. Functional studies
revealed that both normal and P268S NOD2 induced
similar levels of NF-κB activation in response to MDP
[45] and the G908R, R702W, and L1007fsinsC variants
in the presence and absence of P268S are defective in
their ability to respond to bacterial lipopolysaccharides
and peptidoglycan, whereas P268S alone exhibited wild-
type activity [46]. Thus, P268S was confirmed as the
haplotype background of these three variants but has no
influence on functions of the CD-associated variants of
NOD2 [47]. Notably, above results from NF-κB activa-
tion assay using human embryonic kidney (HEK) 293
cell lines, but the function activity of NOD2 in
neurodegeneration field remain unclear. Additionally,
Crane et al. found that carriage of the P268S variant was
associated with greater disease activity and inversely as-
sociated with ulcerative colitis spondylarthritis [48]. We
regarded the P268S of NOD2 behaving as a common
factor of PD and CD. Crohns disease is a chronic in-
flammatory disorder of the gastrointestinal tract, which
is thought to result from the effect of environmental
factors in a genetically predisposed host [49]. A wealth
of new information has emerged to suggest that
inflammation-derived oxidative stress and cytokine
dependent toxicity may contribute to nigrostriatal path-
way degeneration and hasten progression of disease in
humans with idiopathic PD. Both them suggest that
inflammation may play a role connecting between PD
and CD.
Table 2 Genotype and allele frequencies of G2385R, P268S, and IVS9 in cases and controls
Genotype PD patients, n = 237 Control, n = 190 Odds ratio
(95% CI)*
p value** Power
§
No. % No. %
G2385R(G > A)
GG 213 89.9 186 97.9 1
GA 24 10.1 4 2.1 5.24 0.009 0.8973
(1.79-15.38)
AA 0 0 0 0 ——
G 450 94.9 376 98.9 1
A 24 5.1 4 1.1 5.01 0.090 0.8803
(1.72-14.58)
P268S(C > T)
CC 229 96.6 190 100 1
CT 8 3.4 0 0 0.045 NA
TT 0 0 0 0 ——
C 466 98.3 380 100 1
T 8 1.7 0 0 ——
IVS9(G > A)
GG 207 87.3 175 92.1 1
GA 30 12.7 15 7.9 1.7 0.348 0.2987
(0.88-3.24)
AA 0 0 0 0 ——
G 444 365 96.1 1
A 30 6.3 15 3.9 1.64 0.572 0.2865
(0.87-3.10)
*: Odds ratios (OR) and 95% confidence intervals (95% CI) were calculated by comparison to the common allele frequencies and genotype frequencies.
**: Fisher exact test and false discovery rate were used to calculate and correct the p values, respectively. Two sided Pvalue 0.05 was considered
statistically significant.
§
: Power was calculated by Power and Sample Size Calculations Version 3.0, 2009. NA: not available.
Ma et al. Behavioral and Brain Functions 2013, 9:19 Page 6 of 8
http://www.behavioralandbrainfunctions.com/content/9/1/19
Conclusions
We analyzed the genetic loci associated with PD in the
sporadic and family PD patients using PCR-RFLP and
found P268S might be a risk factor of sporadic PD in the
Chinese population. As it is the first study on NOD2
P268S in PD patients, the results should be treated with
caution, as one cannot completely exclude that they are
false positive. Further studies required to verify the asso-
ciation between P268S and susceptibility to sporadic
Parkinsons disease in large sample size, and explore the
NOD2 functions involved in neurodegenerative diseases.
Abbreviations
PD: Parkinsons disease; SNPs: Single nucleotide polymorphisms;
UPS: Ubiquitin proteasome system; PCR: Polymerase chain reaction;
RFLP: Restriction fragment length polymorphism; CE: Capillary
electrophoresis; GWAS: Genome-wide association studies; CD: Crohns
disease; NOD: Nucleotide-binding and oligomerization domain; EOPD: Early-
onset PD; LOPD: Late-onset PD; UPDRS: Unified parkinsons disease rating
scale; EDTA: Ethylene diamine tetraacetic acid; SPSS: Statistical package for
the social sciences; OR: Odds ratios; CI: Confidence intervals; MDP: Muramyl
dipeptide; NFAT: Nuclear factor of activated T-cells.
Competing interests
The authors declare that they have no competing interests.
Authorscontributions
QLM, XKA, and QL collected the blood samples from PD patients and
controls, extracted genomic DNA, and searched relevant literature. ZML
performed PCR, RFLP, and CE analysis and built positive and negative control
plasmids. HJZ and LLC designed the primers for PCR, cloning, and site-
directed mutagenesis and performed sequence alignment. WQH and PH
participated in statistical analysis and the production of tables. CMT designed
all the experiments and drafted the manuscript. All authors read and
approved the final manuscript.
Authorsinformation
QLM: MD, chief physician of the Department of Neurology of the First
Affiliated Hospital of Xiamen University; senior visiting scholar at Harvard
Medical School, Center of Neurological Diseases; Member of the World
Stroke Organization (WSO). XKA: Resident of the Department of Neurology of
Xiamen First Hospital; part of the team that first revealed the association of
G2385R and the LRRK2 gene with PD in Chinese Han populations. QL: MD,
associate-chief physician of the Department of Neurology of the he First
Affiliated Hospital of Xiamen University. ZML: Master student, School of
Pharmaceutical Sciences Xiamen University; participated in several projects
on screening the disease genes for PD of Chinese Han population. CMT: PhD
in Physical Biochemistry, Nuclear Science, National Tsing-Hua University,
Postdoctoral Fellow in Biochemistry, Stanford University School of Medicine,
School of Pharmaceutical Sciences; Executive Director Professor of Center for
Translational Medicine, Xiamen University.
Acknowledgements
We would like to thank all the patients and control subjects who
participated in this study. We would like to thank Professor Chi-Meng Tzeng
for his guidance and the members of our laboratory, Huiming Ye, Xiaonan
Lv, Yi Li, and Mingjie Lei for their help. This study was supported by Fujian
Province Natural Science Foundation Association of glucocerebrosidase gene
mutations with Parkinsons disease in Chinese Han patients (No. 2009D001),
Fujian Province Health Department Youth Research Projects Association of
glutamate transporter gene and patients with Parkinsons disease (No. 2009-2
-72) and Fujian Province Science and Technology Department Key Project of
The Screening of Susceptibility Gene in Parkinsons Disease and The Platform
Establishment of Molecular Diagnosis (No. 2012D062).
Received: 14 November 2012 Accepted: 1 May 2013
Published: 7 May 2013
References
1. Zhang ZX, Roman GC, Hong Z, Wu CB, Qu QM, Huang JB, Zhou B, Geng ZP,
Wu JX, Wen HB, et al:Parkinsons disease in China: prevalence in Beijing,
Xian, and Shanghai. Lancet 2005, 365(9459):595597.
2. Fahn S: Description of Parkinsons disease as a clinical syndrome. Ann N Y
Acad Sci 2003, 991:114.
3. Forno LS: Neuropathology of Parkinsons disease. J Neuropathol Exp Neurol
1996, 55(3):259272.
4. Dauer W, Przedborski S: Parkinsons disease: mechanisms and models.
Neuron 2003, 39(6):889909.
5. Dawson TM, Dawson VL: Molecular pathways of neurodegeneration in
Parkinsons disease. Science 2003, 302(5646):819822.
6. Cookson MR: The biochemistry of Parkinsons disease. Annu Rev Biochem
2005, 74:2952.
7. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B,
Root H, Rubenstein J, Boyer R, et al:Mutation in the alpha-synuclein gene
identified in families with Parkinsons disease. Science 1997,
276(5321):20452047.
8. Zarranz JJ, Alegre J, Gomez-Esteban JC, Lezcano E, Ros R, Ampuero I, Vidal L,
Hoenicka J, Rodriguez O, Atares B, et al:The new mutation, E46K, of alpha-
synuclein causes Parkinson and Lewy body dementia. Ann Neurol 2004,
55(2):164173.
9. Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S, Przuntek H,
Epplen JT, Schols L, Riess O: Ala30Pro mutation in the gene encoding
alpha-synuclein in Parkinsons disease. Nat Genet 1998, 18(2):106108.
10. Chan DK, Mellick G, Cai H, Wang XL, Ng PW, Pang CP, Woo J, Kay R: The
alpha-synuclein gene and Parkinson disease in a Chinese population.
Arch Neurol 2000, 57(4):501503.
11. Mata IF, Wedemeyer WJ, Farrer MJ, Taylor JP, Gallo KA: LRRK2 in Parkinsons
disease: protein domains and functional insights. Trends Neurosci 2006,
29(5):286293.
12. Tan EK, Shen H, Tan LC, Farrer M, Yew K, Chua E, Jamora RD, Puvan K,
Puong KY, Zhao Y, et al:The G2019S LRRK2 mutation is uncommon in an
Asian cohort of Parkinsons disease patients. Neurosci Lett 2005,
384(3):327329.
13. Funayama M, Li Y, Tomiyama H, Yoshino H, Imamichi Y, Yamamoto M,
Murata M, Toda T, Mizuno Y, Hattori N: Leucine-rich repeat kinase 2
G2385R variant is a risk factor for Parkinson disease in Asian population.
Neuroreport 2007, 18(3):273275.
14. Huang Y, Halliday GM, Vandebona H, Mellick GD, Mastaglia F, Stevens J,
Kwok J, Garlepp M, Silburn PA, Horne MK, et al:Prevalence and clinical
features of common LRRK2 mutations in Australians with Parkinsons
disease. Mov Disord 2007, 22(7):982989.
15. Lucking CB, Durr A, Bonifati V, Vaughan J, Michele G, Gasser T, Harhangi BS,
Meco G, Denefle P, Wood NW, et al:Association between early-onset
Parkinsons disease and mutations in the parkin gene. N Engl J Med 2000,
342(21):15601567.
16. Shimura H, Hattori N, Kubo S, Mizuno Y, Asakawa S, Minoshima S, Shimizu
N, Iwai K, Chiba T, Tanaka K, et al:Familial Parkinson disease gene
product, parkin, is a ubiquitin-protein ligase. Nat Genet 2000,
25(3):302305.
17. Mata IF, Lockhart PJ, Farrer MJ: Parkin genetics: one model for Parkinsons
disease. Hum Mol Genet 2004, 13:R127R133. Spec No 1.
18. Wu YR, Wu CH, Chao CY, Kuan CC, Zhang WL, Wang CK, Chang CY, Chang
YC, Lee-Chen GJ, Chen CM: Genetic analysis of Parkin in early onset
Parkinsons disease (PD): Novel intron 9 g > a single nucleotide
polymorphism and risk of Taiwanese PD. Am J Med Genet B Neuropsychiatr
Genet 2010, 153B(1):229234.
19. Tucci A, Nalls MA, Houlden H, Revesz T, Singleton AB, Wood NW, Hardy J,
Paisan-Ruiz C: Genetic variability at the PARK16 locus. Eur J Hum Genet
2010, 18(12):13561359.
20. Kolisek M, Launay P, Beck A, Sponder G, Serafini N, Brenkus M, Froschauer
EM, Martens H, Fleig A, Schweigel M: SLC41A1 is a novel mammalian Mg2
+ carrier. J Biol Chem 2008, 283(23):1623516247.
21. Wyss-Coray T, Mucke L: Inflammation in neurodegenerative diseasea
double-edged sword. Neuron 2002, 35(3):419432.
22. McGeer PL, McGeer EG: Inflammation and neurodegeneration in
Parkinsons disease. Parkinsonism Relat Disord 2004, 10(Suppl 1):S3S7.
23. Kim YS, Joh TH: Microglia, major player in the brain inflammation: their
roles in the pathogenesis of Parkinsons disease. Exp Mol Med 2006,
38(4):333347.
Ma et al. Behavioral and Brain Functions 2013, 9:19 Page 7 of 8
http://www.behavioralandbrainfunctions.com/content/9/1/19
24. Hakansson A, Westberg L, Nilsson S, Buervenich S, Carmine A, Holmberg B,
Sydow O, Olson L, Johnels B, Eriksson E, et al:Interaction of
polymorphisms in the genes encoding interleukin-6 and estrogen
receptor beta on the susceptibility to Parkinsons disease. Am J Med
Genet B Neuropsychiatr Genet 2005, 133B(1):8892.
25. Kaltschmidt B, Heinrich M, Kaltschmidt C: Stimulus-dependent activation
of NF-kappaB specifies apoptosis or neuroprotection in cerebellar
granule cells. Neuromolecular Med 2002, 2(3):299309.
26. Hirsch EC, Hunot S: Neuroinflammation in Parkinsons disease: a target for
neuroprotection? Lancet Neurol 2009, 8(4):382397.
27. Inohara N, Nunez G: NODs: intracellular proteins involved in inflammation
and apoptosis. Nat Rev Immunol 2003, 3(5):371382.
28. Bialecka M, Kurzawski M, Klodowska-Duda G, Opala G, Juzwiak S, Kurzawski
G, Tan EK, Drozdzik M: CARD15 variants in patients with sporadic
Parkinsons disease. Neurosci Res 2007, 57(3):473476.
29. Hughes AJ, Daniel SE, Kilford L, Lees AJ: Accuracy of clinical diagnosis of
idiopathic Parkinsons disease: a clinico-pathological study of 100 cases.
J Neurol Neurosurg Psychiatry 1992, 55(3):181184.
30. An XK, Peng R, Li T, Burgunder JM, Wu Y, Chen WJ, Zhang JH, Wang YC, Xu
YM, Gou YR, et al:LRRK2 Gly2385Arg variant is a risk factor of Parkinsons
disease among Han-Chinese from mainland China. Eur J Neurol 2008,
15(3):301305.
31. Abou-Sleiman PM, Muqit MM, McDonald NQ, Yang YX, Gandhi S, Healy DG,
Harvey K, Harvey RJ, Deas E, Bhatia K, et al:A heterozygous effect for
PINK1 mutations in Parkinsons disease? Ann Neurol 2006, 60(4):414419.
32. Klein C: Implications of genetics on the diagnosis and care of patients
with Parkinson disease. Arch Neurol 2006, 63(3):328334.
33. Hedrich K, Marder K, Harris J, Kann M, Lynch T, Meija-Santana H, Pramstaller
PP, Schwinger E, Bressman SB, Fahn S, et al:Evaluation of 50 probands
with early-onset Parkinsons disease for Parkin mutations. Neurology 2002,
58(8):12391246.
34. Sun M, Latourelle JC, Wooten GF, Lew MF, Klein C, Shill HA, Golbe LI, Mark
MH, Racette BA, Perlmutter JS, et al:Influence of heterozygosity for parkin
mutation on onset age in familial Parkinson disease: the GenePD study.
Arch Neurol 2006, 63(6):826832.
35. Klein C, Lohmann-Hedrich K, Rogaeva E, Schlossmacher MG, Lang AE:
Deciphering the role of heterozygous mutations in genes associated
with parkinsonism. Lancet Neurol 2007, 6(7):652662.
36. Matsuda N, Kitami T, Suzuki T, Mizuno Y, Hattori N, Tanaka K: Diverse effects
of pathogenic mutations of Parkin that catalyze multiple
monoubiquitylation in vitro. J Biol Chem 2006, 281(6):32043209.
37. Wan-Ling Zhang C-HW, Tsu-Wei W, Lee-Chen G-J: Functional Analysis of
Parkin Gene Intron 9 g > a SNP. Bio Formosa 2010, 45(1):3138.
38. Hunot S, Brugg B, Ricard D, Michel PP, Muriel MP, Ruberg M, Faucheux BA,
Agid Y, Hirsch EC: Nuclear translocation of NF-kappaB is increased in
dopaminergic neurons of patients with parkinson disease. Proc Natl Acad
Sci USA 1997, 94(14):75317536.
39. Gao M, Cao Q, Luo LH, Wu ML, Hu WL, Hu WL, Si JM: [NOD2/CARD15 gene
polymorphisms and susceptibility to Crohns disease in Chinese Han
population]. Zhonghua Nei Ke Za Zhi 2005, 44(3):210212.
40. Croucher PJ, Mascheretti S, Hampe J, Huse K, Frenzel H, Stoll M, Lu T,
Nikolaus S, Yang SK, Krawczak M, et al:Haplotype structure and
association to Crohns disease of CARD15 mutations in two ethnically
divergent populations. Eur J Hum Genet 2003, 11(1):616.
41. Yamazaki K, Takazoe M, Tanaka T, Kazumori T, Nakamura Y: Absence of
mutation in the NOD2/CARD15 gene among 483 Japanese patients with
Crohns disease. J Hum Genet 2002, 47(9):469472.
42. Leong RW, Armuzzi A, Ahmad T, Wong ML, Tse P, Jewell DP, Sung JJ:
NOD2/CARD15 gene polymorphisms and Crohns disease in the Chinese
population. Aliment Pharmacol Ther 2003, 17(12):14651470.
43. Lv C, Yang X, Zhang Y, Zhao X, Chen Z, Long J, Zhong C, Zhi J, Yao G, Jiang
B, et al:Confirmation of three inflammatory bowel disease susceptibility
loci in a Chinese cohort. Int J Colorectal Dis 2012, 27(11):14651472.
44. Ogura Y, Inohara N, Benito A, Chen FF, Yamaoka S, Nunez G: Nod2, a
Nod1/Apaf-1 family member that is restricted to monocytes and
activates NF-kappaB. J Biol Chem 2001, 276(7):48124818.
45. Inohara N, Ogura Y, Fontalba A, Gutierrez O, Pons F, Crespo J, Fukase K,
Inamura S, Kusumoto S, Hashimoto M, et al:Host recognition of bacterial
muramyl dipeptide mediated through NOD2. Implications for Crohns
disease. J Biol Chem 2003, 278(8):55095512.
46. Bonen DK, Ogura Y, Nicolae DL, Inohara N, Saab L, Tanabe T, Chen FF,
Foster SJ, Duerr RH, Brant SR, et al:Crohns disease-associated NOD2
variants share a signaling defect in response to lipopolysaccharide and
peptidoglycan. Gastroenterology 2003, 124(1):140146.
47. Chamaillard M, Philpott D, Girardin SE, Zouali H, Lesage S, Chareyre F, Bui
TH, Giovannini M, Zaehringer U, Penard-Lacronique V, et al:Gene-
environment interaction modulated by allelic heterogeneity in
inflammatory diseases. Proc Natl Acad Sci USA 2003, 100(6):34553460.
48. Crane AM, Bradbury L, Van Heel DA, McGovern DP, Brophy S, Rubin L,
Siminovitch KA, Wordsworth BP, Calin A, Brown MA: Role of NOD2 variants
in spondylarthritis. Arthritis Rheum 2002, 46(6):16291633.
49. Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H,
Moran T, Karaliuskas R, Duerr RH, et al:A frameshift mutation in NOD2
associated with susceptibility to Crohns disease. Nature 2001,
411(6837):603606.
doi:10.1186/1744-9081-9-19
Cite this article as: Ma et al.:P268S in NOD2 associates with
susceptibility to Parkinsons disease in Chinese population. Behavioral
and Brain Functions 2013 9:19.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Ma et al. Behavioral and Brain Functions 2013, 9:19 Page 8 of 8
http://www.behavioralandbrainfunctions.com/content/9/1/19
... Nod-like receptors (NLRs) mainly recognize exogenous pathogen-associated molecular patterns (PAMP) such as bacteria. NLRs participate in the pathological mechanism of multiple CNS disorders, including AD (Faustin and Reed, 2013;Ma et al., 2013). Nucleotide-binding oligomerization domain 2 (NOD2) is a member of the NLRs and functions as a specific intracellular recognition receptor for MDP (Al Nabhani et al., 2017). ...
... Multiple studies indicate that NOD2 is a risk factor for neurodegenerative disorders like Parkinson's disease (PD) (Cheng et al., 2018;Ma et al., 2013) and multiple sclerosis (MS) (White et al., 2014). Previous research has revealed that NOD2 induces neuronal damage and cognitive dysfunction, thus implying a pathological role of NOD2 in AD as well (Chauhan et al., 2009;. ...
Article
Full-text available
The relationship between chronic bacterial colonization in the brain and Alzheimer's disease is attracting extensive attention. Recent studies indicated that the components of bacterial biofilm drive the amyloid-β production. Muramyl dipeptide, the minimal bioactive peptidoglycan motif common to all bacteria, contributes to the development of many central inflammatory and neurodegenerative disorders. However, the involvement of Muramyl dipeptide in amyloid-β production is not completely defined. In our present study, wild type mice received an intracerebroventricular injection of normal saline or Muramyl dipeptide. Data showed that the production of Aβ1-42 oligomers was significantly increased after Muramyl dipeptide injection in the wild type mice or incubation of the SH-SY5Y cells with Muramyl dipeptide. Moreover, the action of Muramyl dipeptide was dose- and time-dependent. The above results suggested a possibility that the Muramyl dipeptide-induced Aβ1-42 oligomer production might be related to the NOD2/p-p38 MAPK/BACE1 pathway. To confirm this, the SH-SY5Y cells were transfected with siRNA NOD2. Data showed that the transfected SH-SY5Y cells exhibited decreased expression of Aβ1-42 oligomer, NOD2, p-p38 MAPK, and BACE1 after treatment with Muramyl dipeptide. Finally, SH-SY5Y cells were pretreated with SB203580, an inhibitor of the p-38-MAPK pathway. The results indicated that these pretreated SH-SY5Y cells exhibited decreased expression of Aβ1-42 oligomer, p-p38 MAPK, and BACE1 after treatment with Muramyl dipeptide. In conclusion, these results suggested that Muramyl dipeptide was the trigger factor for Aβ1-42 oligomer production, which probably acts via the NOD2/p-p38 MAPK/BACE1 signaling pathway.
... This gene is located on chromosome 16 and encodes the NOD2 protein (23). Four single nucleotide polymorphisms (SNPs; R702W, G908R, L1007fs, and P268S) of the NOD2/CARD15 gene are highly expressed in patients with Crohn's disease and PD (22,24,25). However, Appenzeller et al. suggested that the three SNPs (R702W, G908R, and L1007fs) are not associated with PD (26). ...
Article
Full-text available
Parkinson’s disease (PD) and inflammatory bowel disease (IBD) are the two chronic inflammatory diseases that are increasingly affecting millions of people worldwide, posing a major challenge to public health. PD and IBD show similarities in epidemiology, genetics, immune response, and gut microbiota. Here, we review the pathophysiology of these two diseases, including genetic factors, immune system imbalance, changes in gut microbial composition, and the effects of microbial metabolites (especially short-chain fatty acids). We elaborate on the gut–brain axis, focusing on role of gut microbiota in the pathogenesis of PD and IBD. In addition, we discuss several therapeutic strategies, including drug therapy, fecal microbiota transplantation, and probiotic supplementation, and their potential benefits in regulating intestinal microecology and relieving disease symptoms. Our analysis will provide a new understanding and scientific basis for the development of more effective therapeutic strategies for these diseases.
... In terms of neurodegenerative disorders, Parkinson's and Alzheimer's disease are also treated long-term, which is associated with high costs and a lot of attention. Several publications suggest that NOD2 gene variants may correlate with Parkinson's susceptibility, as exemplified by the polymorphism of c.2 857A> G p.K953E, P268S in the Chinese Han population [67]. Nonetheless, Appenzeller et al. discovered 9 SNPs of the NOD2 gene covering common variations throughout the whole sequence and announced that the NOD2 genetic mutation might not show the high susceptibility of Parkinson's disease in German patients [68]. ...
Article
Full-text available
Nucleotide-binding oligomerization domain containing 2 (NOD2), located in the cell cytoplasm, is a pattern recognition receptor belonging to the innate immune receptor family. It mediates the innate immune response by identifying conserved sequences in bacterial peptide glycans and plays an essential role in maintaining immune system homeostasis. Gene mutations of NOD2 lead to the development of autoimmune diseases such as Crohn’s disease and Blau syndrome. Recently, NOD2 has been shown to be associated with the pathogenesis of diabetes, cardiac-cerebral diseases, and cancers. However, the function of NOD2 in these non-communicable diseases (CNCDs) is not well summarized in reviews. Our report mainly discusses the primary function and molecular mechanism of NOD2 as well as its potential clinical significance in CNCDs.
... The risk of inflammatory bowel disease (IBD) increases by some identified genetic factors that cause sporadic PD. For instance, NOD2 is recognized to be a powerful indicator of IBD and to interact with LRRK2 [114][115][116]. Gut dysbiosis has been shown to induce gastrointestinal inflammation and increase proinflammatory cytokines, including TNF, IFN-γ, IL-6 and IL-1β [116]. ...
Article
Full-text available
Parkinson’s disease is a progressive neurodegenerative disorder with motor, physical and behavioral symptoms that can have a profound impact on the patient’s quality of life. Most cases are idiopathic, and the exact mechanism of the disease’s cause is unknown. The current hypothesis focuses on the gut-brain axis and states that gut microbiota dysbiosis can trigger inflammation and advances the development of Parkinson’s disease. This systematic review presents the current knowledge of gut microbiota analysis and inflammation based on selected studies on Parkinson’s patients and experimental animal models. Changes in gut microbiota correlate with Parkinson’s disease, but only a few studies have considered inflammatory modulators as important triggers of the disease. Nevertheless, it is evident that proinflammatory cytokines and chemokines are induced in the gut, the circulation, and the brain before the development of the disease’s neurological symptoms and exacerbate the disease. Increased levels of tumor necrosis factor, interleukin-1β, interleukin-6, interleukin-17A and interferon-γ can correlate with altered gut microbiota. Instead, treatment of gut dysbiosis is accompanied by reduced levels of inflammatory mediators in specific tissues, such as the colon, brain and serum and/or cerebrospinal fluid. Deciphering the role of the immune responses and the mechanisms of the PD-associated gut microbiota will assist the interpretation of the pathogenesis of Parkinson’s and will elucidate appropriate therapeutic strategies.
... The risk ratio for PD (1.4) is similar in Crohn's disease and ulcerative colitis [57]. Crohn's disease and PD share two genetic risk factors for inflammation: LRRK2 risk alleles [58] and a NOD2 mutation in a protein encoded by CARD15 [59,60]. LRRK2 deficiency leads to aberrant activation of macrophages and increased susceptibility to chemically-induced colitis in mice [61]. ...
Article
Full-text available
Depression is associated with constipation within and outside Parkinson’s disease (PD). Since inefficient cognitive-processing (bradyphrenia) features in PD and an enterokinetic agent improved cognitive performance in healthy individuals, bradyphrenia may be associated with constipation. We aim to define the archetypical bowel function of PD, and its association with cognition, mood, and motor features within and outside PD. We assessed colonic transit time (oral radio-opaque markers over 6 days), bowel function and psychometric questionnaires and measures of PD facets, including bradyphrenia, in 58 participants with diagnosed PD, and 71 without (controls). The best abdominal X-ray (day 7) predictors of PD status were total retained marker count and transverse colon segmental delay. However, Rome functional constipation status complemented segmental delay better, giving good specificity (85%) but low sensitivity (56%). Transverse colon marker count appeared to be age-associated only in PD. In PD, those correctly classified by bowel dysfunction had higher depression scores (p = 0.02) and longer cognitive-processing times than the misclassified (p = 0.05). Controls misclassified as PD by bowel dysfunction had higher depression and anxiety scores than the correctly classified (p = 0.002 and 0.003, respectively), but not slower cognitive processing. Measures of motor features were independent of sub-classification by bowel function in PD and in controls. In conclusion, constipation in PD has distinct localized pathophysiology, and is associated with bradyphrenia.
... Furthermore, NOD2 polymorphisms are proved to be associated with PD risk [27]. Ma et al. find that NOD2 P268S polymorphism might play a role in sporadic PD susceptibility based on Chinese population and meanwhile prove that inflammatory response involve in PD [28]. ...
Article
Full-text available
We, the Editors and Publisher of the journal Artificial Cells, Nanomedicine, and Biotechnology, have retracted the following article: Meiling Xu, Shuangyan Zhang, Hong Zhao, Chi Ma & Yujun Pan (2019) Association between nucleotide-binding oligomerization domain protein 2 (NOD2) gene polymorphisms and Parkinson’s disease (PD) susceptibility. Artificial Cells, Nanomedicine, and Biotechnology, 47:1, 2134–2138, DOI: https://doi.org/10.1080/21691401.2019.1617730 It has come to our attention that the full authorship list and affiliations for this manuscript were changed after the article was submitted. In addition, the location of the study site and the ethics committee were also changed. We have contacted the authors for an explanation, but we have not received a response within the requested timeframe. As determining authorship and the location of where the research was conducted, is core to the integrity of published work, we are therefore retracting the article. The authors listed in this publication have been sent notification. We have been informed in our decision-making by our policy on publishing ethics and integrity and the COPE guidelines on retractions. The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as ‘Retracted’.
Article
Full-text available
Emerging evidence implicates intestinal involvement in the onset and/or progression on the selective degeneration of dopaminergic neurons characterizing Parkinson’s disease (PD). On the one hand, there are studies supporting the Braak hypothesis that holds that pathologic α-synuclein, a hallmark of PD, is secreted by enteric nerves into intestinal tissue and finds its way to the central nervous system (CNS) via retrograde movement in the vagus nerve. On the other hand, there is data showing that cells bearing leucine-rich repeat kinase 2 (LRRK2), a signaling molecule with genetic variants associated with both PD and with inflammatory bowel disease, can be activated in intestinal tissue and contribute locally to intestinal inflammation, or peripherally to PD pathogenesis via cell trafficking to the CNS. Importantly, these gut-centered factors affecting PD development are not necessarily independent of one another: they may interact and enhance their respective pathologic functions. In this review, we discuss this possibility by analysis of studies conducted in recent years focusing on the ability of LRRK2 to shape immunologic responses and the role of α-synuclein in influencing this ability.
Article
Introduction Variants in the leucine-rich repeat kinase 2 gene (LRRK2) are risk factors for Parkinson's disease (PD), but their prevalence varies geographically, reflecting the locations of founder events and dispersion of founders' descendants. Methods A comprehensive literature review was conducted to identify studies providing prevalence estimates for any of ten variants in LRRK2 (G2019S, R1441C, R1441G, R1441H, I2020T, N1437H, Y1699C, S1761R, G2385R, R1628P) among individuals with PD globally. We calculated crude country-specific variant prevalence estimates and, when possible, adjusted estimates for ethno-racial composition. For clinic-based studies, probands were used over other familial cases, whereas for population-based studies, all PD cases were used. Results The analysis included 161 articles from 52 countries yielding 581 prevalence estimates across the ten variants. G2019S was the most common variant, exceeding 1.0% in 26 of 51 countries with estimates. The other variants were far less common. G2385R and R1628P were observed almost exclusively in East Asian countries, where they were found in ∼5–10% of cases. All prevalence estimates adjusted for ethno-racial composition were lower than their unadjusted counterparts, although data permitting this adjustment was only available for six countries. Conclusions Except for G2019S, the LRRK2 variants covered in this review were uncommon in most countries studied. However, there were countries with higher prevalence for some variants, reflecting the uneven geographic distribution of LRRK2 variants. The fact that ethno-racial group‒adjusted estimates were lower than crude estimates suggests that estimates derived largely from clinic-based studies may overstate the true prevalence of some LRRK2 variants in PD.
Article
Gut bacteria influence brain functions and metabolism. We investigated whether this influence can be mediated by direct sensing of bacterial cell wall components by brain neurons. In mice, we found that bacterial peptidoglycan plays a major role in mediating gut-brain communication via the Nod2 receptor. Peptidoglycan-derived muropeptides reach the brain and alter the activity of a subset of brain neurons that express Nod2. Activation of Nod2 in hypothalamic inhibitory neurons is essential for proper appetite and body temperature control, primarily in females. This study identifies a microbe-sensing mechanism that regulates feeding behavior and host metabolism.
Article
Parkinson’s disease (PD) is the most common neurodegenerative movement disorder in the elderly. As the pathogenesis of PD is still not fully understood, medications with the capacity of halting the disease progression are currently unavailable. The discovery of genes that are causative for, or increase susceptibility to, PD is pivotal for the development of novel therapeutic approaches, as they are critical for the onset of PD and the molecular pathways underlying its pathogenesis. By reviewing relevant data, we discuss causative genes, and those associated with PD susceptibility and quantitative traits. Through Gene Ontology database and STRING analysis, we emphasize the roles of inorganic cation transmembrane transport pathways and hypothalamic pituitary thyroid axis, in addition to the established roles of inflammation/oxidative stress and mitochondrial dysfunction in the pathogenesis of PD. It is hoped these insights 1) untangle the clinical complex presentations of PD, 2) reveal the interwoven molecular network leading to PD, and 3) identify critical molecular targets to facilitate novel PD drug discovery, with a view to providing improved consultation and personalized medicine for patients with PD in the future.
Article
Full-text available
Apaf-1 and Nod1 are members of a protein family, each of which contains a caspase recruitment domain (CARD) linked to a nucleotide-binding domain, which regulate apoptosis and/or NF-κB activation. Nod2, a third member of the family, was identified. Nod2 is composed of two N-terminal CARDs, a nucleotide-binding domain, and multiple C-terminal leucine-rich repeats. Although Nod1 and Apaf-1 were broadly expressed in tissues, the expression of Nod2 was highly restricted to monocytes. Nod2 induced nuclear factor κB (NF-κB) activation, which required IKKγ and was inhibited by dominant negative mutants of IκBα, IKKα, IKKβ, and IKKγ. Nod2 interacted with the serine-threonine kinase RICK via a homophilic CARD-CARD interaction. Furthermore, NF-κB activity induced by Nod2 correlated with its ability to interact with RICK and was specifically inhibited by a truncated mutant form of RICK containing its CARD. The identification of Nod2 defines a subfamily of Apaf-1-like proteins that function through RICK to activate a NF-κB signaling pathway.
Article
Few detailed clinico-pathological correlations of Parkinson's disease have been published. The pathological findings in 100 patients diagnosed prospectively by a group of consultant neurologists as having idiopathic Parkinson's disease are reported. Seventy six had nigral Lewy bodies, and in all of these Lewy bodies were also found in the cerebral cortex. In 24 cases without Lewy bodies, diagnoses included progressive supranuclear palsy, multiple system atrophy, Alzheimer's disease, Alzheimer-type pathology, and basal ganglia vascular disease. The retrospective application of recommended diagnostic criteria improved the diagnostic accuracy to 82%. These observations call into question current concepts of Parkinson's disease as a single distinct morbid entity.
Article
Parkinson's disease (PD) results primarily from the death of dopaminergic neurons in the substantia nigra. Current PD medications treat symptoms; none halt or retard dopaminergic neuron degeneration. The main obstacle to developing neuroprotective therapies is a limited understanding of the key molecular events that provoke neurodegeneration. The discovery of PD genes has led to the hypothesis that misfolding of proteins and dysfunction of the ubiquitin-proteasome pathway are pivotal to PD pathogenesis. Previously implicated culprits in PD neurodegeneration, mitochondrial dysfunction and oxidative stress, may also act in part by causing the accumulation of misfolded proteins, in addition to producing other deleterious events in dopaminergic neurons. Neurotoxin-based models (particularly MPTP) have been important in elucidating the molecular cascade of cell death in dopaminergic neurons. PD models based on the manipulation of PD genes should prove valuable in elucidating important aspects of the disease, such as selective vulnerability of substantia nigra dopaminergic neurons to the degenerative process.
Article
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
Results:Mutations were found in 23 families (12.6% of those screened). Among the mutation-positive families, 10 (43%) contained compound heterozygotes; 3 (13%), homozygotes; and 10 (43%), heterozygotes. The onset age in patients with parkin gene mutations ranged from 20 to 76 years. Patients with 1 parkin mutation had an 11.7-yearageatonsetthandidpatientswithnone(P=.04), andpatientswith2ormoreparkinmutationshada13.2- yeardecreaseinageatonsetcomparedwithpatientswith 1 mutation (P=.04). Conclusions: These data indicate that parkin muta- tions are not rare in multiply affected sibships, and that heterozygous mutation carrier status in PARK2 signifi- cantly influences age at onset of PD.
Article
The multifunctional cytokine interleukin-6 (IL-6) is involved in inflammatory processes in the central nervous system and increased levels of IL-6 have been found in patients with Parkinson's disease (PD). It is known that estrogen inhibits the production of IL-6, via action on estrogen receptors, thereby pointing to an important influence of estrogen on IL-6. In a previous study, we reported an association between a G/A single nucleotide polymorphism (SNP) at position 1730 in the gene coding for estrogen receptor beta (ERbeta) and age of onset of PD. To investigate the influence of a G/C SNP at position 174 in the promoter of the IL-6 gene, and the possible interaction of this SNP and the ERbeta G-1730A SNP on the risk for PD, the G-174C SNP was genotyped, by pyrosequencing, in 258 patients with PD and 308 controls. A significantly elevated frequency of the GG genotype of the IL-6 SNP was found in the patient group and this was most obvious among patients with an early age of onset (≤50 years) of PD. When the GG genotypes of the IL-6 and ERbeta SNPs were combined, the combination was much more robustly associated with PD, and especially with PD with an early age of onset, than respective GG genotype when analyzed separately. Our results indicate that the G-174C SNP in the IL-6 promoter may influence the risk for developing PD, particularly regarding early age of onset PD, and that the effect is modified by interaction of the G-1730A SNP in the ERbeta gene. © 2004 Wiley-Liss, Inc.
Article
Both epidemiological and genetic studies support a role of neuroinflammation in the pathophysiology of Parkinson's disease (PD). Furthermore, post mortem studies confirm the involvement of innate as well as adaptive immunity in the affected brain regions in patients with PD. Indeed, activated microglial cells and T lymphocytes have been detected in the substantia nigra of patients concomitantly with an increased expression of pro-inflammatory mediators. Preclinical investigations conducted in various animal models of PD indicate that inflammatory processes are instrumental in neuronal cell death even though they are unlikely to be a primary cause for neuronal loss. Neuroinflammatory processes in PD are rather involved in self-perpetuating deleterious events that lead to protracted neuronal degeneration. In line with this, recent data indicate that glucocorticoid receptors are important in curtailing microglial reactivity, and deregulation in their activity in PD could lead to sustained inflammation-mediated degeneration. Altogether, neuroinflammatory processes might represent a target for neuroprotection in PD.