You are on page 1of 9

Clinical Neurology and Neurosurgery 175 (2018) 16–24

Contents lists available at ScienceDirect

Clinical Neurology and Neurosurgery


journal homepage: www.elsevier.com/locate/clineuro

H. pylori and Parkinson’s disease: Meta-analyses including clinical severity T


a,⁎,1 a,1 b,c,1 a
Efthimios Dardiotis , Zisis Tsouris , Alexios-Fotios A. Mentis , Vasileios Siokas ,
Amalia Michalopouloua, Maria Sokratousa, Metaxia Dastamania, Dimitrios P. Bogdanosd,e,
Georgia Deretzif, Jannis Kountourasg
a
Department of Neurology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
b
Public Health Laboratories, Hellenic Pasteur Institute, Athens, Greece
c
Department of Microbiology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
d
Department of Rheumatology and Clinical Immunology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
e
Cellular Immunotherapy & Molecular Immunodiagnostics, Biomedical Section, Centre for Research and Technology-Hellas (CERTH), Institute for Research and
Technology-Thessaly (IRETETH), Larissa, Greece
f
Department of Neurology, Papageorgiou General Hospital, Thessaloniki, Greece
g
Department of Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece

A R T I C LE I N FO A B S T R A C T

Keywords: The exact etiology of Parkinson’s disease (PD) remains unclear. Some evidence supports Helicobacter pylori in-
Helicobacter pylori fection as a trigger or driving event, but detection and eradication of H. pylori are not part of PD management.
Parkinson’s disease The aims of this case-control study and meta-analysis were to determine (i) the prevalence of H. pylori infection
Infection in PD patients, (ii) associations between H. pylori infection and clinical status, and (iii) differences in motor
Eradication
status in PD patients before and after H. pylori eradication. A literature search was performed using the PubMed
Meta-analysis
database. The prevalence of H. pylori infection in PD, its association with the unified Parkinson’s disease rating
UPDRS
scale (UPDRS), and the association of H. pylori eradication therapy with the UPDRS-III score were determined by
calculating the odds ratios (OR) and the standardized mean differences (SMD) with 95% confidence intervals
(CI). Fixed- and random-effects models were applied. Ten studies were included in the first meta-analysis (5043
PD patients, 23,449 HCs); H. pylori infection prevalence was higher in PD patients than in HCs [OR (95% CI):
1.47 (1.27, 1.70), Pz < 0.00001]. In seven studies reporting UPDRS scores (150 H. pylori infected, 228 non-
infected PD patients), there was a significant association between H. pylori infection and mean UPDRS scores
[SMD (95% CI): 0.33 (0.12, 0.54), Pz = 0.003]. Regarding H. pylori eradication, in five studies (90 PD patients),
there was a significant reduction in UPDRS-III scores after treatment [SMD (95% CI): 6.83 (2.29, 11.38),
Pz = 0.003]. In conclusion, the present meta-analysis revealed a higher prevalence of H. pylori infection in PD
patients suggesting that H. pylori may contribute to PD pathophysiology. In addition, the significantly lower
UPDRS scores in non-infected PD patients and in patients after H. pylori eradication therapy demonstrate that the
infection may deteriorate the clinical severity of the disease.

1. Introduction Helicobacter pylori (H. pylori) infection is one of the commonest


chronic infections. Its prevalence approaches 50% in middle-aged
Parkinson’s disease (PD) is the commonest movement disorder and adults even in developed countries, although there is some suggestion
second most common neurodegenerative disorder to Alzheimer’s dis- that its prevalence has decreased over the last century [8]. H. pylori
ease [1,2]. Neuropathological hallmarks are progressive dopaminergic mostly causes gastritis and peptic ulcer disease but is also linked to
neuron degeneration and loss in the midbrain substantia nigra com- extra-gastrointestinal disorders [9] including neurological diseases such
pacta, Lewy body formation, and abnormal α-synuclein accumulation as cerebrovascular diseases [10], dementia [11,12], multiple sclerosis
[3,4]. Genetic, neuronal aging, and environmental factors (such as [13,14], glaucoma (described as “ocular Alzheimer disease”) [15], and
constipation and low levels of physical activity) appear to be associated PD [16]. Eradication of H. pylori might therefore inhibit the develop-
with an increased risk of PD [2,5–7]. ment and delay the progression of many diseases.


Corresponding author.
E-mail address: edar@med.uth.gr (E. Dardiotis).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.clineuro.2018.09.039
Received 5 January 2018; Received in revised form 24 September 2018; Accepted 26 September 2018
Available online 01 October 2018
0303-8467/ © 2018 Elsevier B.V. All rights reserved.
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

Patients with PD have an excessive number of peptic ulcers [17]. mentioned original data were excluded from the meta-analysis.
Altschuler [18] hypothesized on causal link between H. pylori and PD,
while neuronal damage in PD has been recently proposed as a response 2.3.2. Mean UPDRS scores between H. pylori infected and non-infected PD
to chronic H. pylori infection [16]. Several studies have suggested an patients
increased prevalence of H. pylori infection in PD patients [19–22]. Studies included in the second part of the meta-analysis met the
Furthermore, a large population study suggested that there is an in- following criteria: a) case-control as well as observational studies in-
creased prescription of anti-Helicobacter drugs for chronic H. pylori in- cluding H. pylori infected PD patients (i.e., cases) and H. pylori non-
fection prior to the diagnosis of PD [23]. Likewise, other studies have infected PD patients (i.e., controls); b) all PD patients tested for H.
suggested that motor performance in PD patients is affected by the pylori infection; c) mean UPDRS-III or total UPDRS score (and SD)
presence of H. pylori infection [20,24]. provided separately for both groups of patients; d) English language; e)
As the duodenum is the main site for levodopa absorption, duodenal studies conducted in humans. Studies with incomplete or not reported
H. pylori infection [25] might affect levodopa bioavailability by indu- data were excluded from the meta-analysis.
cing inflammation, disrupting the duodenal mucosa, and releasing re-
active oxygen species that reduce levodopa absorption or inactivate the 2.3.3. Mean UPDRS scores before and after H. pylori eradication therapy
drug [21]. However, there is no consensus in the different published Studies included in the third part of the meta-analysis met the fol-
studies regarding the extent to, and the way in which, H. pylori eradi- lowing criteria: a) randomized controlled trials (RCTs) including motor
cation influences motor symptoms and affects levodopa absorption assessment before and after H. pylori eradication therapy; b) eradica-
[20,26–28]. tion therapy and duration mentioned in all studies; c) mean UPDRS-III
Despite this potential link between H. pylori infection and PD, score (and SD) provided before and after eradication; d) English lan-
screening for H. pylori is not specifically advocated in the management guage; e) studies conducted in humans; f) supportive criteria were also
of PD. A recent meta-analysis [29] examined the effects of H. pylori sought from studies preferring to use objective rather than subjective
infection on PD; however, it did not include all available studies and did outcome criteria (e.g., [30]). Studies with incomplete or not reported
not examine the relationship between H. pylori infection and the clinical data were excluded from the meta-analysis.
status in PD patients, or the effect of H. pylori eradication on disease
severity. Hence, the aim of this meta-analysis was to assess the pre- 2.4. Statistical analysis
valence of H. pylori infection in PD patients and to elucidate the re-
lationship between the infection and PD clinical status. We were also 2.4.1. H. pylori infection in PD patients and healthy controls
interested in examining the possible influence of H. pylori eradication The association between H. pylori infection and PD was determined
on PD motor severity as expressed by the UPDRS. by calculating the 95% confidence interval (95% CI) and the pooled
odds ratio (OR). The Z test was used to determine the statistical sig-
2. Methods nificance of the OR, and significance was set at p < 0.05.

2.1. Literature search strategy 2.4.2. Mean UPDRS scores between H. pylori infected and non-infected PD
patients
Eligible studies were selected by searching the PubMed database To examine the association between H. pylori infection and mean
using combinations of the following terms: “Helicobacter pylori” and UPDRS scores in PD patients, a second meta-analysis was performed.
“Parkinson’s disease”. The complete search algorithm is available in S1 The association was determined by calculating the standardized mean
File. The final literature search was performed on 13 November 2017. difference (SMD) [31], and the 95% CI was estimated by using the Z
The references in all the retrieved publications were also reviewed to test. Statistical significance was set at p < 0.05.
identify any studies missed in our initial search.
2.4.3. Mean UPDRS scores before and after H. pylori eradication therapy
2.2. Data extraction To examine the association between H. pylori eradication therapy
and mean UPDRS scores in PD patients, a third meta-analysis was
Eligible articles were first sorted by screening titles and abstracts performed. The association was determined by calculating the stan-
and assessing the full text of eligible studies. Data extracted directly dardized mean difference (SMD) [32], and the 95% CI was estimated by
from the articles were: study design, H. pylori infection detection using the Z test. Statistical significance was set at p < 0.05.
method, clinical assessment scale, H. pylori eradication therapy pro-
tocol, number of participants, number of infected patients and controls, 2.4.4. Tests of heterogeneity
sex ratio, and mean age. Data values were presented as in the original Cochran’s Q and I2 indices were calculated to estimate the statistical
articles. If those data were not provided, they were calculated using heterogeneity of the studies. In cases of substantial heterogeneity
related study data. The number of participants and the means and (PQ < 0.10 or/ and I2 > 75%), a random effects model was applied.
standard deviations (SD) of unified PD rating scale (UPDRS) scores Otherwise, the fixed effects model was used. Funnel plots were used to
were extracted from the RCTs. Two investigators (E.D. and Z.T.) in- assess for possible publication bias [33]. All statistical analyses were
dependently conducted data extraction and evaluated the methodolo- performed in Review Manager (RevMan) Version 5.3 software (The
gical quality with disagreements resolved by a third investigator (V.S.). Nordic Cochrane Centre, The Cochrane Collaboration, Copenhagen,
Denmark (http://tech.cochrane.org/revman)). PRISMA guidelines for
2.3. Inclusion criteria reporting reviews and meta-analyses were applied (S2 File) [34].

2.3.1. H. pylori infection in PD patients vs. healthy controls 3. Results


Studies included in the first part of the meta-analysis met the fol-
lowing criteria: a) case-control as well as observational studies in- 3.1. Selection and characteristics of the included studies
cluding PD patients (i.e., cases) and healthy controls (i.e., controls); b)
both PD patients and healthy controls tested for H. pylori infection; c) The initial PubMed database search and examination of the refer-
studies reporting either cardinal number or percentage of subjects re- ences of the retrieved publications yielded 85 studies published be-
garding H. pylori infection for both groups; d) English language; e) tween 1 November 1996 and 13 November 2017 (after removal of
studies conducted in humans. Studies with incomplete or not duplicates). After reviewing titles and abstracts, articles not eligible for

17
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

Fig. 1. Flow chart presenting the selection process of eligible studies.

the meta-analysis were excluded, resulting in 28 potentially eligible two studies 13C-urea breath testing (UBT) was performed [22,46]. From
studies. A case-control study was written in Polish and was therefore the remaining protocols, one was a population-based study and H.
excluded from the meta-analysis [35]. Furthermore, the following stu- pylori infection status was determined by medical prescription records
dies were excluded: a) five studies for not using the UPDRS scale for [23], one used gastroscopy and histological examination [15], and one
motor assessment [30,36–39]; b) two studies, as they did not provide H. determined positivity by detecting H. pylori-specific DNA by RT-PCR
pylori seropositivity results [40,41]; and c) three studies due to possible [43]. The cases in all studies were diagnosed with PD, and all the
sample overlap [20,26,42]. In one protocol, the mean (and SD) UPDRS subjects used as controls were healthy. The case and control groups
scores of H. pylori positive and negative groups were calculated from were age and sex matched in five studies and only age matched in one
the pooled mean and SD of the two positive and two negative groups, study [47].
respectively (S3 File). Finally, seventeen studies were included in the
quantitative meta-analysis involving in total 5043 PD patients and
23,449 healthy controls in the first part, one-hundred fifty H. pylori- 3.1.2. Mean UPDRS score between H. pylori infected and non-infected PD
infected and 228 non-infected PD patients in the second part, and 90 PD patients
patients before and after H. pylori eradication in the third part. The Seven studies used in the second part of the meta-analysis were
selection algorithm is shown in Fig. 1. published between 2008 and 2017 [21,22,24,27,28,46,48]. Their main
characteristics are shown in Table 2. Only 3 out of 7 studies [24,46,48]
reported that H. pylori infection may worsen the mean UPDRS score,
3.1.1. H. pylori infection in PD patients and healthy controls while the rest of them [21,22,27,28] could not demonstrate a sig-
Ten studies used in the first part of the meta-analysis were published nificant association between H. pylori and mean UPDRS score. In all
between 1999 and 2017 and their main characteristics are presented in studies, case-control analysis was conducted between H. pylori infected
Table 1. All were case-control studies. Higher prevalence of H. pylori and non-infected PD patients. Regarding H. pylori detection, UBT was
infection was reported in 7 studies [15,19,22,23,30,43,44] while 3 used in five studies [21,22,24,46,48], while the antigen stool test was
studies [13,45,46] did not report a significant difference.With respect used in one study [27] and ELISA in another [28]. Section III of the
to H. pylori detection, serology by enzyme-linked immunosorbent assay UPDRS and total UPDRS score were used for motor assessment of the
(ELISA) was used in five studies [13,19,44,45,47], while in the other patients. Specifically, in six studies [21,22,27,28,46,48], section III of

18
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

Table 1
Characteristics of the studies included in the meta-analysis: H. pylori infection in PD patients and healthy controls.
Author Year Study design H. pylori detection PD patients Controls

H. pylori Mean age Sex (F/M) H. pylori Mean age Sex (F/M)
(+/−) (+/−)

Charlett et al. 1999 Case-control ELISA 23/10 75 19/14 14/25 69 23/16


Dobbs et al. 2000 Case-control ELISA 50/55 74 50/55 83/127 70 103/107
Charlett et al. 2009 Cross-sectional ELISA 57/120 NM NM 77/196 NM NM
Nielsen et al. 2012 Population-based case- Prescription of H. pylori eradication 138/4,346 70.8 1,808/ 505/21,911 70.8 9,038/
control treatment 2,676 13,378
Nafisah et al. 2013 Cross-sectional Urea breath test 14/15 64.1 13/16 5/18 61.3 8/15
Fasano et al. 2013 Case-control Urea breath test 11/22 67.8 15/18 8/22 65.4 17/13
Blaecher et al. 2013 Case-control RT-PCR 17/60 62 26/34 42/256 52 136/120
Bu et al. 2015 Case-control ELISA 60/71 67 63/68 44/97 68 66/75
Tsolaki et al. 2015 Case-control Gastroscopy and histological 6/9 NM NM 14/31 NM NM
examination
Efthymiou et al. 2017 Case-Control ELISA 14/39 68.7 20/19 33/68 47.4 40/28

ELISA, enzyme-linked immunosorbent assay; PD, Parkinson’s disease; UPDRS, unified Parkinson's disease rating scale; RCT, randomized controlled trial; NM, not
mentioned; SD, standard deviation.

the UPDRS was evaluated, and in one study, the total UPDRS score was 3.2. Subgroup analysis
used to assess clinical status. All the studies provided mean UPDRS
scores with SD. Furthermore, six of the seven studies presented mean 3.2.1. H. pylori infection in PD patients and healthy controls
age and sex ratios for both H. pylori infected and non-infected patients The fixed-effects model was applied to the first group of studies due
[21,24,27,28,46,48], while one did not [22]. to low heterogeneity (I2 = 31%, PQ = 0.16). H. pylori infection was
significantly more prevalent among PD patients than among controls
[OR (95% CI): 1.47 (1.27, 1.70); Pz < 0.00001]. The results of the
3.1.3. Mean UPDRS score before and after H. pylori eradication therapy meta-analysis for the prevalence of H. pylori infection in PD patients
Five studies used in the third part of the meta-analysis were pub- are presented in Fig. 2A. The funnel plot presented in Fig. 2B without
lished between 2001 and 2017 [21,24,28,49,50]. Their main char- revealing any significant asymmetry.
acteristics are shown in Table 3. Four of the studies [18,24,26,28,49]
reported a significant association between H. pylori eradication and 3.2.2. Mean UPDRS scores between H. pylori infected and non-infected PD
decrease of the mean UPDRS-III score, while one study [21] did not. patients
Except, in the study of Pierantozzi et al. [50], in which patients were The fixed-effects model was applied to the second group of studies
given 250 mg levodopa per day, the average daily consumption of le- due to low heterogeneity (I2 = 19%, PQ = 0.28). There was significant
vodopa in the Helicobacter positive patients was about 500 mg per day, association between H. pylori infection and higher mean UPDRS scores
in all the other included studies. In all studies, UPDRS section III was [standardized mean difference, SMD (95% CI): 0.33 (0.12, 0.54),
used for motor assessment before and after H. pylori eradication in PD Pz = 0.003]. The results of the meta-analysis for the mean UPDRS score
patients. Regarding the H. pylori eradication therapy, a seven-day between H. pylori infected and non-infected PD patients are presented
treatment protocol with omeprazole, amoxicillin, and clarithromycin in Fig. 3A. The funnel plot presented in Fig. 3B without revealing any
was used in one study [50], while in two studies [28,49] a 14-day significant asymmetry.
protocol was applied. In the other two studies [21,24], seven-day
therapy with esomeprazole, amoxicillin, and clarithromycin was used.
3.2.3. Mean UPDRS scores before and after H. pylori eradication therapy
All the studies provided mean UPDRS scores with SD.
The random-effects model was applied to the third group of studies
due to high heterogeneity (I2 = 67%, PQ = 0.02). There was significant

Table 2
Characteristics of the studies included in the meta-analysis: Mean UPDRS scores in H. pylori infected (+) and non-infected (-) PD patients.
Author Year Study H. pylori Assessment scale H. pylori (+) patients H. pylori (−) patients
design detection
n UPDRS Mean age Sex n UPDRS Mean age Sex (F/
(mean ± SD) (F/M) (mean ± SD) M)

Lee et al. 2008 RCT & Case- Urea Breath UPDRS section III 35 22.9 ± 7.4 60 15/20 30 23.1 ± 7 60.2 14/16
control Test medication “on”
Nafisah et al. 2013 Case-control Urea Breath UPDRS III motor 14 19.43 ± 8.864 NM NM 15 19.2 ± 11.66 NM NM
Test examination
Fasano et al. 2013 RCT & Case- Urea Breath UPDRS III total score 11 40.61 ± 7.98 69.3 6/5 22 36.86 ± 13.53 67.2 9/13
control Test medication “off”
Narozanska 2014 Case-control Antigen Stool UPDRS part III in “off” 18 38.8 ± 19.7 59.8 10/8 19 37.6 ± 12.8 60.4 8/11
et al. Test
Hashim et al. 2014 RCT & Case- Urea Breath UPDRS total score 21 85.05 ± 25.48 65.1 11/10 55 63.98 ± 29.17 67.5 31/24
control Test
Tan et al. 2015 Case-control Urea Breath UPDRS section III in 33 33.97 ± 13.04 68.4 13/20 69 27.32 ± 10.05 63.8 28/41
Test “on”
Mridula et al. 2017 RCT & Case- ELISA UPDRS part III in “off” 18 54.1 ± 5.2 59 5/13 18 52.3 ± 11 61 12/6
control

UPDRS, unified Parkinson's disease rating scale; RCT, randomized controlled trial; NM, not mentioned; SD, standard deviation.

19
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

Table 3
Characteristics of the studies included in the meta-analysis: Mean UPDRS scores before and after H. pylori eradication in PD patients.
Author Year Study H. pylori eradication therapy L-DOPA Assessment n Mean Age Sex Before eradication After eradication
design Average scale (F/M)
Daily dose
mg/day
UPDRS UPDRS
(mean ± SD) (mean ± SD)

Pierantozzi 2001 RCT Omeprazole, amoxicillin and 250.0 UPDRS-III 7 70.4 NM 34.1 ± 10.5 23.1 ± 8.8
et al. clarithromycin 7-day therapy
Lee et al. 2008 RCT & Case- Esomeprazole, amoxicillin and 544.5 UPDRS-III 34 NM NM 22.9 ± 7.4 22.3 ± 8
control clarithromycin 7-day therapy
Hashim et al. 2014 RCT & Case- Esomeprazole, amoxicillin and 500.0 UPDRS-III 21 65.1 11/10 50.62 ± 13.62 37 ± 15.56
control clarithromycin 7-day therapy
Mridula et al. 2017 RCT & Case- Omeprazole, amoxicillin and 470.4 UPDRS-III 18 59 5/13 22.3 ± 6.6 15.3 ± 3.8
control clarithromycin 14-day therapy
Liu et al. 2017 Cohort Omeprazole, amoxicillin and 599.5 UPDRS-III 10 63.2 5/5 25.9 ± 8.37 18.3 ± 8.38
study clarithromycin 14-day therapy

UPDRS, unified Parkinson's disease rating scale; RCT, randomized controlled trial; NM, not mentioned; SD, standard deviation.

association between H. pylori eradication and lower mean UPDRS-III 4. Discussion


scores [standardized mean difference, SMD (95% CI): 6.83 (2.29,
11.38), Pz = 0.003]. The results of the meta-analysis for the mean Here we present the results of a meta-analysis examining the asso-
UPDRS-III score before and after H. pylori eradication in PD patients are ciation between H. pylori infection status and PD. The first part of the
presented in Fig. 4A. The funnel plot presented in Fig. 4B without re- meta-analysis revealed a significantly higher prevalence of H. pylori
vealing any significant asymmetry. infection in PD patients than in healthy controls. Regarding the motor
status of PD patients, there was a significant association between H.
pylori infection and mean UPDRS scores. Furthermore, we found a
significantly lower mean UPDRS-III score after H. pylori eradication

Fig. 2. (A) Forest plot: meta-analysis of the prevalence of H. pylori infection in the PD patient group compared with the healthy control group. (B) Funnel plot to
detect publication bias in the studies reporting H. pylori infection in PD patients and healthy controls.

20
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

Fig. 3. (A) Forest plot: standardized mean difference of mean UPDRS scores between H. pylori infected (Hp+) and non-infected (Hp-) PD patients. (B) Funnel plot to
detect publication bias in the studies reporting mean UPDRS scores in H. pylori infected and non-infected PD patients.

Fig. 4. (A) Forest plot: standardized mean difference of mean UPDRS-III scores before and after H. pylori eradication therapy in PD patients. (B) Funnel plot to detect
publication bias in the studies reporting mean UPDRS-III scores before and after H. pylori eradication therapy in PD patients.

21
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

therapy. Additional H. pylori-mediated pathogenetic mechanisms in PD could be


Charlett et al. [19] reported that H. pylori seropositivity was two- caused by the production of toxins by H. pylori (intriguingly, toxic ac-
fold higher in patients with idiopathic parkinsonism than disease-free tions could even be raised by H. pylori-mediated glycosylation of human
controls [19]. It was reported that peptic ulcers were more common in cells’ cholesterol) a disrupted intestinal microbiome, or remarkably, by
PD patients [17,18]. Furthermore, several studies have revealed a high H. pylori-mediated alterations in the pharmacokinetic properties of
prevalence of H. pylori infection in PD patients [21,39,47,48] and these levodopa (all elegantly reviewed in [51]).
results were consistent with the results of the first part of our meta- This meta-analysis has certain limitations. First, we included studies
analysis. In addition, a recent meta-analysis [29] showed a statistically regardless of their H. pylori detection method. While both ELISA and
significant correlation between H. pylori infection and risk of PD [OR UBT reflect the infection status of patients, there are several differences
(95% CI): 1.59 (1.37,1.85)]. The same significant association was between these methods, especially that serology tends to overestimate
maintained in the subgroup analyses for study design, study location, the prevalence of infection. ELISA could remain positive months after
and studies adjusted and not adjusted for other covariates. This further potential H. pylori spontaneous elimination [8]. Second, H. pylori status
reinforces the notion that H. pylori infection plays an important role in could represent a marker of hygiene status, making the associations
the course or even in the pathogenesis of PD. more correlational than causal. Third, we observed significant hetero-
Nielsen et al. [23] showed that prescription of H. pylori eradication geneity between the scales used to assess the clinical status of PD pa-
therapy and proton pump inhibitors five or more years prior to the tients. Although UPDRS was used in most of the studies, different sec-
diagnosis of PD were associated with a 45% and 23% increase in PD tions of the scale were applied in different disease states. Moreover,
risk, respectively. They concluded that chronic H. pylori infection and/ when considering small-study effects, random-effects testing may
or gastritis contribute to PD pathogenesis or that they come before are overestimate summary effects [62]. Finally, several H. pylori eradica-
PD-related motor symptoms [23]. tion protocols with different treatment duration were applied in most of
Even though most studies suggest that the prevalence of H. pylori the studies.
infection is higher in PD than in disease-free status, considerable con-
troversy remains on whether the clinical status of H. pylori infected and 5. Conclusion
non-infected PD patients differs. Five relevant studies [21,24,28,49,50]
suggested that H. pylori-infected patients tended to have increased H. pylori infection may contribute to the pathogenesis and prognosis
motor fluctuations and worse mean UPDRS scores, which were found to of PD, given its higher prevalence in PD patients. Despite the difficulty
be controlled by L-Dopa. Also, H. pylori eradication has been shown to of therapeutic regimens due to high antibiotic resistance rates, suc-
ameliorate symptoms of PD [38]. H. pylori eradication has been sug- cessful H. pylori eradication therapy may positively influence the clin-
gested in PD patients treated with levodopa, as it may improve drug ical outcome of PD. However, further large-scale studies with unified
bioavailability and decrease motor fluctuations [21,24,37,46]. Thus, it clinical data and adequate statistical power for reproducibility (as
was previously reported [20,24,51] that there is an enhanced gut ab- emphasized in [62] for neurological studies) are required to accurately
sorption of levodopa in PD patients who had H. pylori eradication, as estimate the impact of the infection and elucidate the detailed me-
compared to PD patients with H. pylori infection who received placebo. chanisms by which H. pylori is involved in PD pathogenesis.
The results of our meta-analysis were consistent with most of the in-
cluded studies [21,24,27,28,46,48–50], showing an association be- Acknowledgements
tween H. pylori infection and the clinical status (i.e., mean UPDRS
score) of PD patients and a significant clinical improvement (i.e., lower A.-F.A.M. is supported by an educational scholarship fromthe
mean UPDRS-III score) after H. pylori eradication therapy. Onassis Public Benefit Foundation, who played no role in the design of
Mechanistically, there is evidence that inteleukin-1β (IL-1β), IL-6, the study, collection and/or interpretation of data, or writing of the
tumor necrosis factor-α (TNF-α), other proinflammatory cytokines, and article.
C-reactive protein play important roles in neurodegenerative disorders
including PD [52]. H. pylori induces the release of large amounts of IL- Appendix A. Supplementary data
1β, −6, −8, −10, −13 and TNF-α, eicosanoids, and acute phase
proteins and activates monocytes, which may result in disruption of the Supplementary material related to this article can be found, in the
blood brain barrier (BBB), microglial activation, and deleterious effects online version, at doi:https://doi.org/10.1016/j.clineuro.2018.09.039.
on the nigrostriatal dopaminergic system [53]. In this respect, neu-
roinflammation seems to contribute to neurodegeneration with acti- References
vation of microglia [54]. Alternatively, noxious chemicals produced by
H. pylori can be transmitted through the vagal axonal afferent pathway [1] O.B. Tysnes, A. Storstein, Epidemiology of Parkinson’s disease, J. Neural Transm.
and affect brainstem neurons, as indicated by animal and human stu- (Vienna) 124 (8) (2017) 901–905.
[2] V. Bellou, L. Belbasis, I. Tzoulaki, E. Evangelou, J.P. Ioannidis, Environmental risk
dies [53], or H. pylori might access the brain via the oral-nasal-olfactory factors and Parkinson’s disease: an umbrella review of meta-analyses, Parkinsonism
pathway through a disrupted BBB [55]. Conversely, α-synuclein has Relat. Disord. 23 (2016) 1–9.
been recently reported to reach the gastric wall via preganglionic vagal [3] J.M. Lee, P. Derkinderen, J.H. Kordower, R. Freeman, D.G. Munoz, T. Kremer,
W. Zago, S.J. Hutten, C.H. Adler, G.E. Serrano, T.G. Beach, The search for a per-
route, suggesting brain-to-stomach interactions with implications in PD ipheral biopsy Indicator of alpha-synuclein pathology for Parkinson disease, J.
[56]. Neuropathol. Exp. Neurol. 76 (1) (2017) 2–15.
In addition, H. pylori infection is capable of inducing apoptosis via [4] G. Xiromerisiou, E. Dardiotis, V. Tsimourtou, P.M. Kountra, K.N. Paterakis,
E.Z. Kapsalaki, K.N. Fountas, G.M. Hadjigeorgiou, Genetic basis of Parkinson dis-
multiple apoptotic pathways including the Fas-FasL pathway, the mi-
ease, Neurosurg. Focus 28 (1) (2010) E7.
tochondrial pathway, or through inducible nitric oxide to cause neu- [5] C.M. Lill, J.T. Roehr, M.B. McQueen, F.K. Kavvoura, S. Bagade, et al.,
rodegeneration [57]. Furthermore, auto-antibodies against dopami- Comprehensive research synopsis and systematic meta-analyses in Parkinson’s
disease genetics: the PDGene database, PLoS Genet. 8 (3) (2012) e1002548.
nergic neurons found in the cerebrospinal fluid or the blood of PD
[6] R. Kruger, M. Sharma, O. Riess, T. Gasser, C. Van Broeckhoven, J. Theuns, J. Aasly,
patients [58] might represent molecular mimicry from H. pylori infec- G. Annesi, A.R. Bentivoglio, A. Brice, A. Djarmati, A. Elbaz, M. Farrer, C. Ferrarese,
tion [59]. A potentially important mechanism in other neurological J.M. Gibson, G.M. Hadjigeorgiou, N. Hattori, J.P. Ioannidis, B. Jasinska-Myga,
diseases such as multiple sclerosis [60,61], molecular mimicry (in this C. Klein, J.C. Lambert, S. Lesage, J.J. Lin, T. Lynch, G.D. Mellick, F. de Nigris,
G. Opala, A. Prigione, A. Quattrone, O.A. Ross, W. Satake, P.A. Silburn, E.K. Tan,
case between H. pylori and host antigens) might trigger auto-antibody T. Toda, H. Tomiyama, K. Wirdefeldt, Z. Wszolek, G. Xiromerisiou,
production, leading to chronic inflammation that contributes to the D.M. Maraganore, A large-scale genetic association study to evaluate the con-
pathology of both gastric and systemic diseases including PD [36]. tribution of Omi/HtrA2 (PARK13) to Parkinson’s disease, Neurobiol. Aging 32 (3)

22
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

(2011) 548 e9-18. D. Letley, L. Harbin, A.B. Price, M.A. Ibrahim, N.L. Oxlade, J. Bowthorpe,
[7] L. Wang, J.O. Aasly, G. Annesi, S. Bardien, M. Bozi, A. Brice, J. Carr, S.J. Chung, D. Leckstroem, C. Smee, J.M. Plant, D.W. Peterson, Differential effect of
C. Clarke, D. Crosiers, A. Deutschlander, G. Eckstein, M.J. Farrer, S. Goldwurm, Helicobacter pylori eradication on time-trends in brady/hypokinesia and rigidity in
G. Garraux, G.M. Hadjigeorgiou, A.A. Hicks, N. Hattori, C. Klein, B. Jeon, Y.J. Kim, idiopathic parkinsonism, Helicobacter 15 (4) (2010) 279–294.
S. Lesage, J.J. Lin, T. Lynch, P. Lichtner, A.E. Lang, V. Mok, B. Jasinska-Myga, [31] E. Dardiotis, S. Arseniou, M. Sokratous, Z. Tsouris, V. Siokas, A.A. Mentis,
G.D. Mellick, K.E. Morrison, G. Opala, L. Pihlstrom, P.P. Pramstaller, S.S. Park, A. Michalopoulou, A. Andravizou, M. Dastamani, K. Paterakis, D. Bogdanos,
A. Quattrone, E. Rogaeva, O.A. Ross, L. Stefanis, J.D. Stockton, P.A. Silburn, A. Brotis, Vitamin B12, folate, and homocysteine levels and multiple sclerosis: a
J. Theuns, E.K. Tan, H. Tomiyama, M. Toft, C. Van Broeckhoven, R.J. Uitti, meta-analysis, Mult. Scler. Relat. Disord. 17 (2017) 190–197.
K. Wirdefeldt, Z. Wszolek, G. Xiromerisiou, K.C. Yueh, Y. Zhao, T. Gasser, [32] E. Dardiotis, A. Nousia, V. Siokas, Z. Tsouris, A. Andravizou, A.A. Mentis, D. Florou,
D.M. Maraganore, R. Kruger, M. Sharma, Large-scale assessment of polyglutamine L. Messinis, G. Nasios, Efficacy of computer-based cognitive training in neu-
repeat expansions in Parkinson disease, Neurology 85 (15) (2015) 1283–1292. ropsychological performance of patients with multiple sclerosis: a systematic re-
[8] A. Mentis, P. Lehours, F. Megraud, Epidemiology and Diagnosis of Helicobacter view and meta-analysis, Mult. Scler. Relat. Disord. 20 (2018) 58–66.
pylori infection, Helicobacter 20 (Suppl. 1) (2015) 1–7. [33] V. Siokas, E. Dardiotis, E.E. Tsironi, G. Tsivgoulis, D. Rikos, M. Sokratous,
[9] J. Kountouras, M. Boziki, S.A. Polyzos, P. Katsinelos, E. Gavalas, C. Zeglinas, S. Koutsias, K. Paterakis, G. Deretzi, G.M. Hadjigeorgiou, The role of TOR1A
D. Tzivras, I. Romiopoulos, N. Giorgakis, K. Anastasiadou, E. Vardaka, polymorphisms in dystonia: a systematic review and meta-analysis, PLoS One 12 (1)
C. Kountouras, E. Kazakos, G. Xiromerisiou, E. Dardiotis, G. Deretzi, Impact of re- (2017) e0169934.
active oxygen species generation on Helicobacter pylori-related extragastric dis- [34] D. Moher, A. Liberati, J. Tetzlaff, D.G. Altman, P. Group, Preferred reporting items
eases: a hypothesis, Free Radic. Res. 51 (1) (2017) 73–79. for systematic reviews and meta-analyses: the PRISMA statement, PLoS Med. 6 (7)
[10] J. Kountouras, S.A. Polyzos, P. Katsinelos, C. Zeglinas, F. Artemaki, D. Tzivras, (2009) e1000097.
E. Vardaka, E. Gavalas, I. Romiopoulos, C. Simeonidou, N. Grigoriadis, [35] D. Wlodarek, W. Pakszys, J. Bujko, Helicobacter pylori infection and its influence
C. Kountouras, E. Dardiotis, G. Deretzi, Helicobacter pylori eradication to prevent on Parkinson disease, Pol. Merkur. Lekarski 15 (89) (2003) 428–431.
cardio-cerebrovascular disease: Are current data useful for clinical practice? Int. J. [36] G. Suwarnalata, A.H. Tan, H. Isa, R. Gudimella, A. Anwar, M.F. Loke, S. Mahadeva,
Cardiol. 233 (2017) 92. S.Y. Lim, J. Vadivelu, Augmentation of autoantibodies by Helicobacter pylori in
[11] W.S. Huang, T.Y. Yang, W.C. Shen, C.L. Lin, M.C. Lin, C.H. Kao, Association be- Parkinson’s disease patients may Be linked to greater severity, PLoS One 11 (4)
tween Helicobacter pylori infection and dementia, J. Clin. Neurosci. 21 (8) (2014) (2016) e0153725.
1355–1358. [37] K.E. Rahne, C. Tagesson, D. Nyholm, Motor fluctuations and Helicobacter pylori in
[12] J. Kountouras, M. Boziki, E. Gavalas, C. Zavos, G. Deretzi, S. Chatzigeorgiou, Parkinson’s disease, J. Neurol. 260 (12) (2013) 2974–2980.
P. Katsinelos, N. Grigoriadis, E. Giartza-Taxidou, I. Venizelos, Five-year survival [38] I.T. Bjarnason, A. Charlett, R.J. Dobbs, S.M. Dobbs, M.A. Ibrahim, R.W. Kerwin,
after Helicobacter pylori eradication in Alzheimer disease patients, Cogn. Behav. R.F. Mahler, N.L. Oxlade, D.W. Peterson, J.M. Plant, A.B. Price, C. Weller, Role of
Neurol. 23 (3) (2010) 199–204. chronic infection and inflammation in the gastrointestinal tract in the etiology and
[13] G. Efthymiou, E. Dardiotis, C. Liaskos, E. Marou, V. Tsimourtou, E.I. Rigopoulou, pathogenesis of idiopathic parkinsonism. Part 2: response of facets of clinical
T. Scheper, A. Daponte, W. Meyer, L.I. Sakkas, G. Hadjigeorgiou, D.P. Bogdanos, idiopathic parkinsonism to Helicobacter pylori eradication. A randomized, double-
Immune responses against Helicobacter pylori-specific antigens differentiate re- blind, placebo-controlled efficacy study, Helicobacter 10 (4) (2005) 276–287.
lapsing remitting from secondary progressive multiple sclerosis, Sci. Rep. 7 (1) [39] R.J. Dobbs, A. Charlett, S.M. Dobbs, C. Weller, A.I. MA, O. Iguodala, C. Smee,
(2017) 7929. J.M. Plant, A.J. Lawson, D. Taylor, I. Bjarnason, Leukocyte-subset counts in idio-
[14] E. Gavalas, J. Kountouras, M. Boziki, C. Zavos, S.A. Polyzos, E. Vlachaki, pathic parkinsonism provide clues to a pathogenic pathway involving small in-
I. Venizelos, D. Tsiptsios, G. Deretzi, Relationship between Helicobacter pylori in- testinal bacterial overgrowth. A surveillance study, Gut Pathog. 4 (1) (2012) 12.
fection and multiple sclerosis, Ann. Gastroenterol. 28 (3) (2015) 353–356. [40] C. Weller, A. Charlett, N.L. Oxlade, S.M. Dobbs, R.J. Dobbs, D.W. Peterson,
[15] F. Tsolaki, J. Kountouras, F. Topouzis, M. Tsolaki, Helicobacter pylori infection, I.T. Bjarnason, Role of chronic infection and inflammation in the gastrointestinal
dementia and primary open-angle glaucoma: are they connected? BMC Ophthalmol. tract in the etiology and pathogenesis of idiopathic parkinsonism. Part 3: predicted
15 (2015) 24. probability and gradients of severity of idiopathic parkinsonism based on H. Pylori
[16] F. Franceschi, A. Gasbarrini, S.A. Polyzos, J. Kountouras, Extragastric diseases and antibody profile, Helicobacter 10 (4) (2005) 288–297.
Helicobacter pylori, Helicobacter 20 (Suppl. 1) (2015) 40–46. [41] R.J. Dobbs, S.M. Dobbs, C. Weller, I.T. Bjarnason, N.L. Oxlade, A. Charlett, M.A. Al-
[17] R.R. Strang, The association of gastro-duodenal ulceration and Parkinson’s disease, Janabi, R.W. Kerwin, R.F. Mahler, A.B. Price, Role of chronic infection and in-
Med. J. Aust. 1 (23) (1965) 842–843. flammation in the gastrointestinal tract in the etiology and pathogenesis of idio-
[18] E. Altschuler, Gastric Helicobacter pylori infection as a cause of idiopathic pathic parkinsonism. Part 1: eradication of Helicobacter in the cachexia of idio-
Parkinson disease and non-arteric anterior optic ischemic neuropathy, Med. pathic parkinsonism, Helicobacter 10 (4) (2005) 267–275.
Hypotheses 47 (5) (1996) 413–414. [42] A. Fasano, F. Bove, M. Gabrielli, E. Ragazzoni, S. Fortuna, A. Tortora, M.A. Zocco,
[19] A. Charlett, R.J. Dobbs, S.M. Dobbs, C. Weller, P. Brady, D.W. Peterson, S. Marconi, A. Gasbarrini, A.R. Bentivoglio, Liquid melevodopa versus standard
Parkinsonism: siblings share Helicobacter pylori seropositivity and facets of syn- levodopa in patients with Parkinson disease and small intestinal bacterial over-
drome, Acta Neurol. Scand. 99 (1) (1999) 26–35. growth, Clin. Neuropharmacol. 37 (4) (2014) 91–95.
[20] M. Pierantozzi, A. Pietroiusti, L. Brusa, S. Galati, A. Stefani, G. Lunardi, E. Fedele, [43] C. Blaecher, A. Smet, B. Flahou, F. Pasmans, R. Ducatelle, D. Taylor, C. Weller,
G. Sancesario, G. Bernardi, A. Bergamaschi, A. Magrini, P. Stanzione, A. Galante, I. Bjarnason, A. Charlett, A.J. Lawson, R.J. Dobbs, S.M. Dobbs, F. Haesebrouck,
Helicobacter pylori eradication and l-dopa absorption in patients with PD and Significantly higher frequency of Helicobacter suis in patients with idiopathic
motor fluctuations, Neurology 66 (12) (2006) 1824–1829. parkinsonism than in control patients, Aliment. Pharmacol. Ther. 38 (11-12) (2013)
[21] W.Y. Lee, W.T. Yoon, H.Y. Shin, S.H. Jeon, P.L. Rhee, Helicobacter pylori infection 1347–1353.
and motor fluctuations in patients with Parkinson’s disease, Mov. Disord. 23 (12) [44] X.L. Bu, X. Wang, Y. Xiang, L.L. Shen, Q.H. Wang, Y.H. Liu, S.S. Jiao, Y.R. Wang,
(2008) 1696–1700. H.Y. Cao, X. Yi, C.H. Liu, B. Deng, X.Q. Yao, Z.Q. Xu, H.D. Zhou, Y.J. Wang, The
[22] W.Y. Nafisah, N. Mohamed Ibrahim, H. Najman Achok, R. Hamizah, S. Azmin, association between infectious burden and Parkinson’s disease: a case-control study,
R. Remli, S.A. Shah, High prevalence of Helicobacter pylori infection in Malaysian Parkinsonism Relat. Disord. 21 (8) (2015) 877–881.
Parkinson's disease patients, J. Park. Restless Legs Syndr. (2013) 63. [45] A. Charlett, R.J. Dobbs, S.M. Dobbs, C. Weller, M.A. Ibrahim, T. Dew, R. Sherwood,
[23] H.H. Nielsen, J. Qiu, S. Friis, L. Wermuth, B. Ritz, Treatment for Helicobacter pylori N.L. Oxlade, J.M. Plant, J. Bowthorpe, A.J. Lawson, A. Curry, D.W. Peterson,
infection and risk of Parkinson’s disease in Denmark, Eur. J. Neurol. 19 (6) (2012) I.T. Bjarnason, Blood profile holds clues to role of infection in a premonitory state
864–869. for idiopathic parkinsonism and of gastrointestinal infection in established disease,
[24] H. Hashim, S. Azmin, H. Razlan, N.W. Yahya, H.J. Tan, M.R. Manaf, N.M. Ibrahim, Gut Pathog. 1 (1) (2009) 20.
Eradication of Helicobacter pylori infection improves levodopa action, clinical [46] A. Fasano, F. Bove, M. Gabrielli, M. Petracca, M.A. Zocco, E. Ragazzoni, F. Barbaro,
symptoms and quality of life in patients with Parkinson’s disease, PLoS One 9 (11) C. Piano, S. Fortuna, A. Tortora, R. Di Giacopo, M. Campanale, G. Gigante,
(2014) e112330. E.C. Lauritano, P. Navarra, S. Marconi, A. Gasbarrini, A.R. Bentivoglio, The role of
[25] A. Hamlet, A.C. Thoreson, O. Nilsson, A.M. Svennerholm, L. Olbe, Duodenal small intestinal bacterial overgrowth in Parkinson’s disease, Mov. Disord. 28 (9)
Helicobacter pylori infection differs in cagA genotype between asymptomatic sub- (2013) 1241–1249.
jects and patients with duodenal ulcers, Gastroenterology 116 (2) (1999) 259–268. [47] R.J. Dobbs, A. Charlett, S.M. Dobbs, C. Weller, D.W. Peterson, Parkinsonism: dif-
[26] M. Pierantozzi, A. Pietroiusti, G. Sancesario, G. Lunardi, E. Fedele, P. Giacomini, ferential age-trend in Helicobacter pylori antibody, Aliment. Pharmacol. Ther. 14
S. Frasca, A. Galante, M.G. Marciani, P. Stanzione, Reduced L-dopa absorption and (9) (2000) 1199–1205.
increased clinical fluctuations in Helicobacter pylori-infected Parkinson’s disease [48] A.H. Tan, S. Mahadeva, C. Marras, A.M. Thalha, C.K. Kiew, C.M. Yeat, S.W. Ng,
patients, Neurol. Sci. 22 (1) (2001) 89–91. S.P. Ang, S.K. Chow, M.F. Loke, J.S. Vadivelu, N. Ibrahim, H.S. Yong, C.T. Tan,
[27] E. Narozanska, M. Bialecka, U. Adamiak-Giera, B. Gawronska-Szklarz, W. Soltan, S.H. Fox, A.E. Lang, S.Y. Lim, Helicobacter pylori infection is associated with worse
M. Schinwelski, P. Robowski, M.H. Madalinski, J. Slawek, Pharmacokinetics of le- severity of Parkinson’s disease, Parkinsonism Relat. Disord. 21 (3) (2015) 221–225.
vodopa in patients with Parkinson disease and motor fluctuations depending on the [49] H. Liu, W. Su, S. Li, W. Du, X. Ma, Y. Jin, K. Li, H. Chen, Eradication of Helicobacter
presence of Helicobacter pylori infection, Clin. Neuropharmacol. 37 (4) (2014) pylori infection might improve clinical status of patients with Parkinson’s disease,
96–99. especially on bradykinesia, Clin. Neurol. Neurosurg. 160 (2017) 101–104.
[28] K.R. Mridula, R. Borgohain, V. Chandrasekhar Reddy, V. Bandaru, T. Suryaprabha, [50] M. Pierantozzi, A. Pietroiusti, A. Galante, G. Sancesario, G. Lunardi, E. Fedele,
Association of Helicobacter pylori with Parkinson’s disease, J. Clin. Neurol. 13 (2) P. Giacomini, P. Stanzione, Helicobacter pylori-induced reduction of acute levo-
(2017) 181–186. dopa absorption in Parkinson’s disease patients, Ann. Neurol. 50 (5) (2001)
[29] X. Shen, H. Yang, Y. Wu, D. Zhang, H. Jiang, Association of Helicobacter pylori 686–687.
infection with Parkinson’s diseases: a meta-analysis, Helicobacter (2017). [51] D.J. McGee, X.H. Lu, E.A. Disbrow, Stomaching the possibility of a pathogenic role
[30] S.M. Dobbs, R.J. Dobbs, C. Weller, A. Charlett, I.T. Bjarnason, A.J. Lawson, for Helicobacter pylori in Parkinson’s disease, J. Parkinsons Dis. 8 (3) (2018)

23
E. Dardiotis et al. Clinical Neurology and Neurosurgery 175 (2018) 16–24

367–374. P. Katsinelos, E. Rapti, D. Chatzopoulos, D. Tzilves, C. Stergiopoulos,


[52] K. Kaur, J.S. Gill, P.K. Bansal, R. Deshmukh, Neuroinflammation - a major cause for K. Christodoulou, Helicobacter pylori infection and Parkinson’s disease: apoptosis
striatal dopaminergic degeneration in Parkinson’s disease, J. Neurol. Sci. 381 as an underlying common contributor, Eur. J. Neurol. 19 (6) (2012) e56.
(2017) 308–314. [58] A. Dahlstrom, A. Wigander, K. Lundmark, C.G. Gottfries, P.M. Carvey, A. McRae,
[53] G. Deretzi, J. Kountouras, S.A. Polyzos, C. Zavos, E. Giartza-Taxidou, E. Gavalas, Investigations on auto-antibodies in Alzheimer’s and Parkinson’s diseases, using
I. Tsiptsios, Gastrointestinal immune system and brain dialogue implicated in defined neuronal cultures, J. Neural Transm. Suppl. 29 (1990) 195–206.
neuroinflammatory and neurodegenerative diseases, Curr. Mol. Med. 11 (8) (2011) [59] B.J. Appelmelk, R. Negrini, A.P. Moran, E.J. Kuipers, Molecular mimicry between
696–707. Helicobacter pylori and the host, Trends Microbiol. 5 (2) (1997) 70–73.
[54] V.H. Perry, J.A. Nicoll, C. Holmes, Microglia in neurodegenerative disease, Nat. [60] A.A. Mentis, E. Dardiotis, N. Grigoriadis, E. Petinaki, G.M. Hadjigeorgiou, Viruses
Rev. Neurol. 6 (4) (2010) 193–201. and multiple sclerosis: from mechanisms and pathways to translational research
[55] J. Kountouras, C. Zavos, S.A. Polyzos, G. Deretzi, Potential impact of Helicobacter opportunities, Mol. Neurobiol. 54 (5) (2017) 3911–3923.
pylori-related human beta-defensin-1 on hepatic encephalopathy and neurodegen- [61] A.A. Mentis, E. Dardiotis, N. Grigoriadis, E. Petinaki, G.M. Hadjigeorgiou, Viruses
eration, Ann. Gastroenterol. 29 (1) (2016) 99. and endogenous retroviruses in multiple sclerosis: from correlation to causation,
[56] A. Ulusoy, R.J. Phillips, M. Helwig, M. Klinkenberg, T.L. Powley, D.A. Di Monte, Acta Neurol. Scand. 136 (6) (2017) 606–616.
Brain-to-stomach transfer of alpha-synuclein via vagal preganglionic projections, [62] K.S. Button, J.P. Ioannidis, C. Mokrysz, B.A. Nosek, J. Flint, E.S. Robinson,
Acta Neuropathol. 133 (3) (2017) 381–393. M.R. Munafo, Power failure: why small sample size undermines the reliability of
[57] J. Kountouras, C. Zavos, S.A. Polyzos, G. Deretzi, E. Vardaka, E. Giartza-Taxidou, neuroscience, Nat. Rev. Neurosci. 14 (5) (2013) 365–376.

24

You might also like