Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews


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

Gut microbiota differences between healthy older adults and individuals T


with Parkinson’s disease: A systematic review
Nathan D. Nuzuma,*, Amy Loughmanb, Ewa A. Szymlek-Gaya, Ashlee Hendya, Wei-Peng Teoa,c,
Helen Macphersona
a
Deakin University, Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Geelong, Australia
b
Deakin University, Food & Mood Centre, IMPACT Strategic Research Centre, School of Medicine, Geelong, Australia
c
Physical Education and Sports Science Academic Group, National Institute of Education, Nanyang Technological University, Singapore

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

Keywords: The ‘Dual Hit’ hypothesis, stating that Parkinson’s disease (PD) begins via olfactory pathways and the gut, and
Short chain fatty acid the gastrointestinal symptoms PD individuals face, have largely driven the interest of the gut’s involvement in
Butyrate PD. Studies have since observed gut microbiota differences between PD groups and controls, with these al-
Microbiota-gut-brain axis terations potentially relating to PD pathophysiology. However, differences in the studies’ methodologies pre-
Alpha-diversity
cludes unanimity on the relationships of gut microbiota to PD.
Beta-diversity
Thirteen observational case-control studies investigating gut microbiota in PD and controls were reviewed to
Cognition
Motor-symptoms assess how microbiota abundance and diversity relates to PD. Nine studies showed butyrate producing gut
Non-motor symptoms microbiota had lower abundances in PD compared to controls. Three studies reported α-diversity was higher,
with one reporting it was lower, in PD compared to controls.
Given most studies show abundance, not diversity, differences of butyrate producing bacteria between
groups, we propose abundance differences are more associated with PD than microbiota diversity. As current
research is observational, investigating how specific bacteria and their metabolites may alter throughout PD
progression is warranted.

Information Box 1 Microbiome: The entire community of microorganisms, their ge-


The high rate of gastrointestinal issues in Parkinson’s disease (PD), netic material and their environment (Marchesi & Ravel, 2015).
and beforehand which may predict increased likelihood of PD devel- Dysbiosis: Changes to the typical bacterial composition of a com-
opment, has led to research into the gut microbiota of this population. munity either by introduction or elimination of specific bacterium
Technological advancements in next generation sequencing (NGS) in types, or abundance shifts of existing bacteria that could lead to ne-
the last decade have enabled this research (Sekirov et al., 2010). For an gative health outcomes (Holzapfel et al., 1998).
overview of NGS methods, including 16s rRNA sequencing and shotgun Alpha-diversity (α-diversity): The number (‘richness’) and distribu-
sequencing, see Hamady and Knight (2009). These techniques, in ad- tion (‘evenness’) of bacteria within a single population or community.
dition to culture dependent techniques, allow for the investigation of Beta-diversity (β-diversity): Similarity or dissimilarity between two
gut bacteria and their metabolites (E.g. short chain fatty acids). or more sets of microbial communities.
Short chain fatty acids (SCFAs), most commonly butyrate, acetate In this document, ‘gut microbiome’ or ‘gut microbiota’ refer to the
and propionate, have numerous purported effects in relation to the faecal microbiome, which is the most common method of sampling ‘the
microbiota-gut-brain axis. Namely, this includes the anti-inflammatory gut microbiome’ due to the difficulty in obtaining intestinal mucosal
effects within the gut and the role SCFAs have in maintaining intestinal samples.
wall health and function. For a review of SCFAs and their purported ;1;
effects, see Lopetuso et al. (2013), Mayer et al. (2015), Tan et al.
(2014b), and Westfall et al. (2017). 1. Introduction
Gut microbiota: The microorganisms of the gut (in the case of this
article, bacteria specifically). Parkinson’s disease (PD) is a movement disorder most commonly


Corresponding author at: School of Exercise and Nutrition Sciences, Deakin University, 221 Burwood Highway, Burwood, 3125, VIC, Australia.
E-mail address: n.nuzum@deakin.edu.au (N.D. Nuzum).

https://doi.org/10.1016/j.neubiorev.2020.02.003
Received 21 October 2019; Received in revised form 15 January 2020; Accepted 3 February 2020
Available online 04 February 2020
0149-7634/ © 2020 Elsevier Ltd. All rights reserved.
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

recognised through its cardinal motor impairments including muscle systematically review these studies to examine what is currently known
rigidity, postural instability and gait disturbances, bradykinesia and about gut microbiota differences between these groups and to infer
resting tremors (Jankovic, 2008). However, some of the most prevalent what role gut microbiota may play in PD onset or progression.
PD symptoms are non-motor, including cognitive deficits, from mild- This review aimed to synthesise the literature on the differences in
cognitive impairment to PD-related dementia, along with a range of gut microbiota profiles across PD and healthy control groups and to
gastrointestinal (GI) symptoms involving increased transit time, such as investigate the associations between gut microbiota and PD. The spe-
constipation (Fasano et al., 2015). Given the devastating nature of PD cific objectives of this systematic review were to 1) assess whether there
and its associated symptoms, understanding PD aetiology and pro- was an association between measures of gut microbiota diversity (in-
gression is crucial to developing preventative measures or therapeutic cluding both α- and β-diversity measures) and PD, 2) describe the
options. differences in specific bacterial taxa and their metabolites (E.g. SCFA)
Gastrointestinal symptoms are one of the most prevalent non-motor between PD and neurologically healthy controls and 3) assess whether
symptoms in individuals with PD, with up to 80 % of individuals af- the gut microbiota differences between groups related to PD, PD se-
fected (Martinez-Martin, 2011). These symptoms are common, and can verity, PD duration and/or to motor and non-motor symptoms of PD.
precede motor symptoms and clinical PD diagnosis. For example, con-
stipation may represent an increased risk for future diagnosis of PD 2. Methods
(Abbott et al., 2001). This indicates that the GI tract may be relevant to
the onset of PD. Additionally; impaired olfaction is an early non-motor This systematic review was completed per the PRISMA guidelines
symptom of PD (Haehner et al., 2009). One hypothesis to explain the (Moher et al., 2015) and was registered on PROSPERO (Registration
role of olfactory and GI tract pathways in PD aetiology includes the number: CRD42018095211, registration date 1/6/2018). The studies
‘Dual Hit’ hypothesis (Hawkes et al., 2009). Hawkes and colleagues included in the review were original, observational case-control studies
propose that an unknown neurotropic pathogen gains entry via the with human participants only. Accepted articles required a PD group to
nasal cavity and through swallowing of infected saliva/mucous leads to have an age- and sex-matched control group, or similar (E.g. a spousal
the pathogen passing through the mucosal and epithelial intestinal control group), and to have assessed the gut microbiota composition of
barrier to the underlying enteric nervous system, potentially resulting each group. No limitations were placed on the date of publication of the
in or involving a-synuclein misfolding. The misfolded α-synuclein included studies, but articles were restricted to those available in
subsequently propagates up the vagus nerve (Holmqvist et al., 2014; English and published in peer-reviewed journals. Conference abstracts,
Kim et al., 2019), and this retrograde path of ‘attack’ leads to neuronal book chapters and reviews were excluded, and reference lists of re-
cell death in the substantia nigra (among other brain regions) which is a levant reviews were manually checked for potential articles that fit the
pathological hallmark of PD. A comparison between the intestines of criteria.
early-stage PD individuals and healthy controls showed PD participants A systematic computerised search was performed from the 27/05/
had greater intestinal permeability, which significantly correlated to 18 until the 24/05/19 when the final search was conducted, through
both aggregation of misfolded α-synuclein at the intestines and greater the following databases: PubMed, Embase, Scopus, CINAHL (through
inflammation in the PD group, providing some support for the above EBSCOhost), Global Health (through EBSCOhost) and CENTRAL
hypothesis (Forsyth et al., 2011; Shannon et al., 2012). However, the (Cochrane Library). The full search strategy used for all databases is
specificity of aggregated α-synuclein at the intestine as a biomarker for available in Appendix A (see Supplementary material). Briefly, the
PD is questionable, as research shows aggregated α-synuclein in control overall search strategy included terms such as ‘gut bacteria’, ‘micro-
participants (Visanji et al., 2015). Additional research shows that α- biota’, ‘neurodegenerative diseases’, ‘PD’, as well as variations on these
synuclein aggregation, and oxidative stress, causes microglia over ac- terms (e.g. gut flora for gut bacteria). Mesh terms, or their equivalents,
tivation (Block and Hong, 2005), leading to neuroinflammation (Shults, were also used in the search strategy.
2006; Su et al., 2008), which could result in an increased risk for de- Title and abstract screening were completed in duplicate, with a
veloping PD, or could be the result of PD development (Hald and third researcher available in the event of discrepancies in accepted
Lotharius, 2005; Teismann and Schulz, 2004). In addition to the po- articles between researchers. Full text screening was completed in the
tential involvement of the GI tract in PD pathophysiology via the ‘Dual same manner. After screening was completed, a data extraction form
Hit’ hypothesis (Hawkes et al., 2009), systemic inflammation is also (an adapted version of the Cochrane data collection form for RCTs and
implicated in PD progression (Vivekanantham et al., 2015), and gives NRS) was utilised to extract all relevant information from the accepted
additional credence for investigating the GI tract. articles, by one researcher (NN). As part of the data extraction process,
Due to the potential effects of inflammation/neuroinflammation, a risk of bias assessment was conducted following the validated ‘Risk of
oxidative stress and increased intestinal permeability in the develop- Bias Assessment tool for Non-randomised Studies’ (RoBANS) protocol
ment of PD (Westfall et al., 2017), it is important to assess the gut (Kim et al., 2013).
microbiota and the metabolites they produce, as many are responsible
for maintaining intestinal wall integrity and are anti-inflammatory (Tan 2.1. Data analysis
et al., 2014b). Short chain fatty acids (SCFAs) are a prominent meta-
bolite produced through commensal gut microbiota that fulfil such Due to the high degree of heterogeneity between data across the
roles (Tan et al., 2014b). Examples of important SCFAs include pro- included studies, we were unable to synthesis the results quantitatively.
pionate, acetate and butyrate. While all SCFAs have anti-inflammatory Therefore, a qualitative, narrative synthesis of the results was con-
capabilities (Tan et al., 2014b), butyrate’s anti-inflammatory effects ducted.
seem to be the most potent. Butyrate also helps maintain intestinal wall
health through mucin production (Brown et al., 2011; Hatayama et al., 3. Results
2007), and is a major energy source for colonocytes (Canani et al.,
2016). Given the important physiological roles of SCFAs, dysbiosis of an This systematic review included 13 studies (See PRISMA Flow
individual’s gut microbiome, whereby many of the beneficial SCFA Diagram Fig. 1). Basic study characteristics are shown in Table 1. In
producing bacteria are potentially reduced, could result in an increased total, the 13 included studies provided data from 904 individuals with
risk for developing PD. PD and 683 control subjects. Of the 13 studies, two did not conduct next
As a result, multiple studies have now used culture-independent, generation sequencing (Hasegawa et al., 2015; Unger et al., 2016), but
next generation sequencing techniques to compare the gut microbiomes were included because the bacterial species they chose to investigate
of healthy matched controls to individuals with PD. There is a need to were reported in other studies, and because one compared SCFAs across

228
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

Fig. 1. PRISMA Flow Diagram of records identified through searching and the screening process (Moher et al., 2009).

a PD group and healthy control group (Unger et al., 2016). 3.2. Participant demographics and PD symptoms

Table 1 shows the included studies were geographically diverse and


3.1. Risk of Bias assessment all but one healthy control group included participants with a mean age
above 60 years (Keshavarzian et al., 2015), to match that of the PD
The most common source of bias across the included studies came population. Across studies, participants’ sex was similarly represented
from the ‘Confounding Variables’ domain with 10/13 studies scoring with most studies having a greater representation of men in PD groups.
‘High’ as they did not account for various potential confounding factors Five of the 13 studies showed the PD group had significantly more
(E.g. BMI, medications, diet etc. which are known to be related to gut severe or higher prevalence of GI symptoms compared to healthy
microbiota (David et al., 2014; Jackson et al., 2018). Additionally, the controls (all p < 0.05) (Hasegawa et al., 2015; Heintz‐Buschart et al.,
‘Selective Outcome reporting’ domain had 9/13 studies scoring ‘High’. 2018; Hill-Burns et al., 2017; Li et al., 2017; Qian et al., 2018), four
This was mostly due to studies either not conducting or reporting studies reported no difference (all p > 0.05) (Bedarf et al., 2017;
measures of α- and/or β-diversity measures (Heintz‐Buschart et al., Hopfner et al., 2017; Lin et al., 2018; Scheperjans et al., 2015) and one
2018), conducting only α- or only β-diversity measures (Bedarf et al., did not report on the differences between PD and control groups, de-
2017; Hill-Burns et al., 2017; Keshavarzian et al., 2015), or when they spite conducting the analysis (Barichella et al., 2019). The remaining
had conducted these measures they had only used one type of measure three studies did not measure GI symptoms in either group
for either α- or β-diversity (Hopfner et al., 2017; Li et al., 2017; Petrov (Keshavarzian et al., 2015; Petrov et al., 2017; Unger et al., 2016). Four
et al., 2017). The study by Barichella et al. (2019) scored uncertain for of the included studies measured PD severity and/or PD related
the ‘Selective Outcome reporting’ domain as it is not clear from their symptoms in controls, to rule out presence of PD and PD symptoms. Of
paper if they used appropriate corrections to adjust for multiple com- these four, one study conducted the assessment but did not report on
parisons when assessing specific bacterial abundance. For the ‘Blinding significance levels (Li et al., 2017), another did not compare groups but
of Outcome Assessments’ domain, the blinding of PD status for re- looked at the associations of the variable on microbial composition (Lin
searchers administering behavioural assessments would not be possible. et al., 2018), while two showed statistically significant differences be-
Only one study reported that the researcher was blinded when con- tween groups (Bedarf et al., 2017; Heintz‐Buschart et al., 2018). Higher
ducting the bacterial analysis (Hasegawa et al., 2015). The other studies PD severity was evident in the PD group (measured through the Hoen
have been rated as ‘Uncertain’ for this domain (See Supplementary and Yahr scale (H&Y) and the Unified Parkinson’s Disease Rating Scale-
Table 3 in Appendix B for detail). I and –III (UPDRS-I and III) (all p < 0.05)) (Bedarf et al., 2017;

229
Table 1
Group demographics from each study (mean ± SD; or median (IQR) indicated by a) including number of participants and sex (m = male, f = female), age (years), body mass index (BMI), gastrointestinal (GI) symptoms
and Parkinson’s disease (PD) severity according to various measures.
Study Location PD group demographics Control group demographics
N.D. Nuzum, et al.

N Age (years) BMI GI Symptoms PD severity N Age (years) BMI GI Symptoms PD severity

Hasegawa et al. Japan 52 (21 m, 68.9 ± 6.8 20.2 ± 2.8* Stool PD duration 36 (spousal 68.4 ± 9.7 22.6 ± 3.7 Stool NR
(2015) 31f) frequency/week (years) = 9.5 ± 5.4 controls:21 m, 15f) frequency/week
3.1 ± 1.2* H&Y stage = 2.7 ± 0.9 7.6 ± 4.6
UPDRS-I = 2.9 ± 2.3
UPDRS-II = 11.7 ± 6.8
UPDRS-III = 25.6 ± 11.8
UPDRS-IV = 3.4 ± 2.4
Keshavarzian et al. U.S.A 38 (24 m, 61.6 ± 9.4* 26.0 ± 3.2 Constipation PD duration (years) 34 (18 m, 16f) 45.1 ± 14.4 27.6 ± 6.0 NR NR
(2015) 14f) according to VAL 6.4 ± 4.7
2.8 ± 3.4 UPDRS score
29.7 ± 11.0(not stated which
section of UPDRS was conducted)

H&Y stage
2.1 ± 0.4
Scheperjans et al. Finland 72 (37 m, 65.3 ± 5.5 26.3 (23.8–29.3)a Wexner NR 72 (36 m, 36f 64.5 ± 6.9 26.2 (23.7–28.1)a Wexner NR
(2015) 35f 5 (3–9)a 2 (1–4)a
Unger et al. (2016) Germany 34 (24 m, 67.7 ± 8.9 NR Constipated Disease duration 82 months 34 age matched 64.6 ± 6.6 NR NR NR
10f) 7/34 (20.6 %) H&Y stage 2.5 (18 m,16f) & 10 (age matched
young control 33.3 ± 11.6
(5 m,5f) (younger group)
Bedarf et al. Germany 31 (all 64.8 ± 9.5 NR GSRS UPDRS III- 12.6 ± 6.9* 28 (all male) 65.6 ± 10.4 NR GSRS 2.2 ± 2.0 0±0
(2017) male) 3.4 ± 2.9

230
Hill-Burns et al. U.S.A 197 68.4 ± 9.2* 26.4 ± 5.3* Constipation in past PD duration (years) 130 (spousal 70.3 ± 8.6 28.3 ± 5.7 Constipation in NR
(2017) (132 m, 3-months 82/197 13.7 ± 6.5 controls :51 m, past 3-months 6/
65f)* (43.4 %)* 79f) 130 (4.7 %)
Hopfner et al. Germany 29 (23 m, 69.2 ± 6.5 NR Constipation UPDRS I score 29 (13 m, 16f) 69.4 ± 6.7 NR Constipation NR
(2017) 6f)* 4/29 12.6 ± 5.3 20/29 (68.9 %)
(13.8 %) UPDRS III score 20.9 ± 6.2
Li et al. (2017) China 24 (16 m, 73.75 ± 6.26 22.96 ± 3.64 Wexner UPDRS score 14 (6 m, 8f) 74.64 ± 5.57 24.00 ± 2.66 Wexner UPDRS score
8f) 17.58 ± 5.43* 33.11 ± 15.75(significance not 1.29 ± 2.59 2.36 ± 2.93
reported)
H&Y stage and
PD duration (years) 5 (2.25- PD duration NR
8: median and IQR)
H&Y stage 2.5 (2-3: median
and IQR)
Petrov et al. Russia 89 (sex 63(61.5;67.5) 26.72(25.4;28.84) NR NR 66 (sex NS) 67(65;69.75) 26.0 (23.82;28.66) NR NR
(2017) NS)
Heintz‐Buschart Germany 26 (19 m, 68.0 ± 9.7 28.5 ± 4.5* SCOPA-AUT MDS H&Y stage 38 (25 m, 13f) 68.4 ± 6.7 26.6 ± 4.1 SCOPA-AUT MDS H&Y score
et al. (2018) 7f) 31* 2.14 ± 0.75* 7 0 ± 0 MDS-
MDS-UPDRS I score UPDRS I score
11.7 ± 7.2* 5.5 ± 4.3
MDS-UPDRS III score MDS-UPDRS III
30 ± 14* score 1.5 ± 2.8
Lin et al. (2018) China 75 (49 m, 60.48 ± 10.72 24.27 ± 2.29# Wexner Disease duration ≥ 45 (spousal 63.2 ± 6.0 24.44 ± 2.36# Wexner Disease duration
26f) 8.81 ± 5.03 5 years = 30 controls :23 m, 2.04 ± 1.02 ≥ 5 years = 0
< 5 years = 44 22f) < 5 years = 0
UPDRS III score UPDRS III score
33.73 ± 18.08 0±0
Qian et al. (2018) China 45 (22 m, 68.1 ± 8 22.8 ± 2.3 Rome III 24/45 H&Y stage: 2.2 ± 0.7 45 (spousal 67.9 ± 8 23.4 ± 2.1 Rome III 3/45 (6.7 NR
23f) (53.3 %)* Age of onset (years): controls:23 m, 22f) %)
62.6 ± 8.1
(continued on next page)
Neuroscience and Biobehavioral Reviews 112 (2020) 227–241
Table 1 (continued)

Study Location PD group demographics Control group demographics

N Age (years) BMI GI Symptoms PD severity N Age (years) BMI GI Symptoms PD severity
N.D. Nuzum, et al.

Disease duration (years):


5.7 ± 4.1
UPDRS total scores:
38.4 ± 18.8
UPDRS Part I scores:
3.1 ± 1.9
UPDRS Part II scores:
10.0 ± 5.7
UPDRS Part III scores:
22.9 ± 12.3
UPDRS Part IV scores: 2.0
(2.0) a
Barichella et al. Italy 193 67.6 ± 9.7 25.3 ± 4.6 Rome III 99 (51.3 7.7 years disease duration 113 (47 m, 66f) 65.9 ± 9.9 25.2 ± 4.6 Rome III 21 (18.6 NR
(2019) (115 m, %) H&Y stage 2.0 %)
78f) UPDRS stage I = 2.0
UPDRS stage II = 9.4
UPDRS stage III = 17.2
UPDRS stage IV = 1.9

* Indicates significant difference between groups for respective baseline measure. # indicates significant effect of measure on microbial composition. N = Number of Participants that had stool samples assessed for gut
microbiota composition, NR = Not Reported; IQR = Inter quartile range; UPDRS = Unified Parkinson’s disease rating scale; H&Y = Hoen and Yahr scale; GSRS = Gastrointestinal symptoms rating scale;
MDS = Movement disorder society; SCOPA-AUT = assessment of autonomic dysfunction in Parkinson’s disease questions 1–7 (gastrointestinal part); VAL = Visual analogue scale; Wexner = Wexner constipation scoring
system; Constipated = defined as fewer than three bowel movements a week, or hard, dry and small bowel movements that are painful or difficult to pass. Note that Petrov et al. (2017) does not state in-text what the
numbers for Age and BMI represent (E.g. median or mean? Range or IQR?).

231
Neuroscience and Biobehavioral Reviews 112 (2020) 227–241
N.D. Nuzum, et al.

Table 2
Studies that conducted α- and/or β-diversity measures across cases (Parkinson’s disease) versus controls, and the types of methods for each. Where studies completed the measure, results are included.
Study α-diversity β-diversity

Shannons Simpsons Observed species Chao1 Phylogenetic Other Unweighted-UniFrac Weighted UniFrac Other
/OTUs diversity Whole tree

Keshavarzian et al. At phylum level At phylum level p = 0.01# NM NM NM Margalef at phylum level: Not measured
(2015) p = .01# (note; authors conducted the p = 0.01# and at genus: level
Gini-Simpsons index) p = 0.03#
Scheperjans et al. p > 0.05 Inverse Simpson p > 0.05 NM p > 0.05 NM ACE p > 0.05 p < 0.02 p < 0.001 NM
(2015)
Bedarf et al. (2017) p = 0.55 p = 0.55 p = 0.87 P = 0.97 NM NM Not measured
Hill-Burns et al. (2017) Not Measured P < 0.01 P < 0.01 Canberra distance
P < 0.01
2

232
Hopfner et al. (2017) p > 0.05 p > 0.05 NM p > 0.05 NM Faith’s phylogenetic distance R = 0.0241, p = 0.0491 NM NM
p > 0.05
Li et al. (2017) p = 0.248 NM NM p = 0.140 NM NM HC and PD separated NM NM
according to PC1 and PC2
(8.03 % and 6.32 %)
Petrov et al. (2017) NM NM NM p = 0.001* NM NM NM R = 0.214 NM
p = 0.0001
Heintz‐Buschart et al. Not Measured
(2018)
Lin et al. (2018) p = 0.74 NM p = 0.233 p = 0.022 p = 0.162 NM R2 = 0.02136 p = 0.001 NM NM
Qian et al. (2018) p = 0.005# p = 0.010# p = 0.009# p = 0.017# p = 0.010# NM R = 0.027 R = 0.008 NM
p = 0.038 p = 0.181
Barichella et al. (2019) NM NM NM p = 0.012 # NM NR P = 0.002 NM
Not stated for which measure
though
Total Number 6 4 3 6 2 2 7 5 1
Neuroscience and Biobehavioral Reviews 112 (2020) 227–241
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

Heintz‐Buschart et al., 2018). The other eight studies did not assess PD Megasphaera (Lin et al., 2018) and Butyricicoccus (Qian et al., 2018)
symptoms or severity in controls. (purported butyrate producers, although Megasphaera eldesnii is speci-
fically a butyrate producer with only the genera reported here). Two
3.3. Gut microbiota measures articles did not report any abundance differences at genus or species
levels (Hopfner et al., 2017; Scheperjans et al., 2015). Note that it is
3.3.1. Measures of gut microbiota diversity unclear if Barichella et al. (2019) or Hopfner et al. (2017) performed
A range of α- and β-diversity measures were utilised to assess gut adequate FDR corrections on their comparison of bacterial abundance
microbiota diversity within and between groups (E.g. Shannon Index, between groups to adjust for multiple comparisons. Additionally,
Chao1 Index, and weighted and unweighted UniFrac distances. For a Scheperjans et al. (2015) reported 6 different bacterial family groups to
full list of these measures, see Table 2). Two studies used real time- be differently abundant however only one (Prevotellaceae) had a Q-
quantitative polymerase chain reaction instead of next-generation se- value lower than 0.05, while the bacteria reported at the species level
quencing (e.g. 16 s rRNA sequencing) techniques, a selective sequen- for Bedarf et al. (2017) were not significant after they applied the FDR
cing method that is not amenable to measuring overall microbial di- correction (See Supplementary Table 2). Therefore, these results should
versity (Hasegawa et al., 2015; Unger et al., 2016). be considered with some caution. Various lactate producing Lactoba-
Nine studies measured α-diversity (See Table 2), with four studies cillus bacteria were more abundant in PD groups (Hasegawa et al.,
showing a significant difference between groups. Three of the four 2015; Hill-Burns et al., 2017; Petrov et al., 2017). However, studies
studies showed the PD group to have higher α-diversity (Barichella showing this either did not exclude participants taking probiotics or did
et al., 2019; Keshavarzian et al., 2015; Qian et al., 2018), with one not adequately collect information on frequency of consumption, do-
study showing the healthy control group had higher α-diversity (Petrov sage/number of probiotics taken, time between probiotic ingestion and
et al., 2017). All eight studies that measured β-diversity (Using faecal sample collection, and the type of probiotic taken.
Weighted and/or Unweighted UniFrac measures or Canberra distance), While Hasegawa et al. (2015) and Unger et al. (2016) did not
indicated that the community of gut microbiota in the PD and control conduct NGS, they still revealed some significant differences in bac-
groups were different (See Table 2). Note that Qian et al. (2018) found terial abundance. Hasegawa et al. (2015) showed counts of Lactobacillus
significant differences in Unweighted UniFrac (p = 0.038), but not for were significantly more abundant in the PD group (p = 0.003), whilst
the Weighted UniFrac distance (p = 0.181), indicating the presence/ counts of Clostridium coccoides and Clostridium leptum (i.e. Clostridial
absence of different bacteria but not necessarily in different abundances cluster XIVa and IV, with both known to contain butyrate producers
in the patient and control groups. However, four studies did show (Collins et al., 1994; Liu et al., 2008)), and Bacteroides fragilis were all
significance at weighted UniFrac measures, indicating differential significantly less abundant in the PD group (p < 0.009) (Hasegawa
abundance of bacteria that were present. et al., 2015). Unger et al. (2016) found that the bacterial family En-
terobacteriaceae, and the genus Bifidobacterium were significantly more
3.3.2. Abundance of SCFA producing bacteria and SCFA concentrations abundant in PD compared to controls, while bacteria from the phylum
All but two studies used NGS, revealing many bacterial abundance Bacteroidetes and from the families of Lactobacillaceae and En-
differences between controls and the PD group reported across many terococcaceae, were significantly less abundant in the PD group. Ad-
levels of classification. As such, the most important results have been ditionally Unger et al. (2016) showed Faecalibacterium prausnitzii, a
highlighted here (See Fig. 2A and B, and Table 2 in Appendix B of the known butyrate producer (Duncan et al., 2002), was significantly less
Supplementary material, for data summary). Bacterial abundance data abundant in the PD group compared to controls.
from the studies presented below is relative abundance (bacterial taxa
expressed as a percentage of the total bacterial taxa found) in all but 3.3.3. SCFA concentrations
one study (Hasegawa et al., 2015), where absolute abundance was Only one study investigated SCFA concentrations from faecal sam-
presented (bacterial taxa presented as counts of the gut bacteria from ples of individuals with PD compared to healthy age-matched controls
sequencing). (Unger et al., 2016). Researchers used gas chromatography to assess
Across the 11 studies using NGS, eight showed five different puta- SCFA concentrations and found that absolute concentrations (mmol/g)
tive butyrate producing species and genera of bacteria that had lower of the three most common SCFAs, acetate, propionate and butyrate,
abundance in the PD group compared to controls. These included the were significantly lower in the PD group (all p < 0.01). The relative
Faecalibacterium genera and/or species Faecalibacterium prausnitzii (Hill- concentration (% of all SCFAs assessed) of butyrate was also sig-
Burns et al., 2017; Li et al., 2017; Lin et al., 2018; Petrov et al., 2017; nificantly lower in the PD group (p < 0.05) (Unger et al., 2016).
Unger et al., 2016), Eubacterium biforme (Bedarf et al., 2017), Copro-
coccus eutactus (Petrov et al., 2017), Roseburia faecis (Bedarf et al., 3.4. Gut microbiota differences in relation to PD progression and symptoms
2017; Heintz‐Buschart et al., 2018; Hill-Burns et al., 2017;
Keshavarzian et al., 2015; Lin et al., 2018), and the Blautia genera Seven studies investigated the relationship between bacterial dif-
(Bedarf et al., 2017; Hill-Burns et al., 2017; Keshavarzian et al., 2015; Li ferences of control and PD groups and PD, PD severity and/or PD re-
et al., 2017; Lin et al., 2018). Other bacteria found to be lower in the PD lated symptoms. Summaries of their findings are reported in the below
group compared to controls that potentially produce the SCFA propio- paragraphs (statistical values are stated where available) (Barichella
nate, included Prevotella copri (Bedarf et al., 2017; Petrov et al., 2017), et al., 2019; Heintz‐Buschart et al., 2018; Li et al., 2017; Lin et al., 2018;
Veillonella parvula (Lin et al., 2018), Roseburia faecis and Blautia (both Qian et al., 2018; Scheperjans et al., 2015; Unger et al., 2016). An
Roseburia faecis and Blautia reportedly produce both butyrate and pro- overview of the PD symptoms of primary interest to this review, and
pionate). Bacteria which have the potential to produce the SCFA symptoms most commonly assessed and reported by authors of the
acetate, that were found to be less abundant in PD groups included included studies, can be seen in the description of Table 1. Additional
Ruminococcus torques (Bedarf et al., 2017), and Veillonella parvula symptoms not mentioned in the table (and measures assessing the
(produces both acetate and propionate) (See Supplementary Table 2 for symptoms) included cognition (Montreal Cognitive Assessment and
full details). In contrast, across the included studies, eight showed that Mini-Mental State Examination) and depression (Hamilton Depression
four different putative SCFA producing bacteria (any of butyrate, and Montgomery-Asberg depression scales).
acetate or propionate) were higher in the PD group compared to con-
trols. These included Akkermansia muciniphila (Bedarf et al., 2017; Lin 3.4.1. PD severity and disease duration
et al., 2018), Bifidobacterium adolescentis (propionate and acetate, and To investigate how gut microbiota abundance related to PD se-
acetate and lactate producers respectively) (Hill-Burns et al., 2017), verity, Li et al. (2017) separated their PD cohort into mild (1–2.5) and

233
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

Fig. 2. A. Individual bacteria shown to be significantly higher in the PD group compared to controls across the taxonomic levels of Phylum, Class, Order, Family, Genus
and Species. Number in brackets following bacteria denotes the number of studies showing significance for that bacteria, at the indicated classification level. Archaea
are denoted with *. Tree generated using PhyloT (database version 2019.1) based on the NCBI taxonomic database and visualised in iTOL (Letunic and Bork, 2016).
B. Individual bacteria shown to be significantly lower in the PD group compared to controls across the taxonomic levels of Phylum, Class, Order, Family, Genus and
Species. Number in brackets following bacteria denotes the number of studies showing significance for that bacteria, at the indicated classification level. Archaea are
denoted with * and Eukaryotes are denoted with #. Tree generated using PhyloT and iTOL based on NCBI taxonomic database. Note that the bacterial family
Coprobacillaceae is presented under its reclassification, Erysipelotrichaceae, and that Clostridium coccoides is presented as its reclassified name Blautia coccoides.

severe (3–5) PD severity based on their Hoen & Yahr scores (rated on a While only two studies have analysed PD severity and gut micro-
1–5 scale). Comparing these two groups to the control group revealed biota, four studies have assessed the relationship between PD duration
that the relative abundance of Faecalibacterium was inversely associated and gut microbiota. Analysis of β-diversity revealed that the distances
with PD severity (5.14 % for controls, 3.17 % in mild PD and 1.22 % for between the microbial communities of the PD and control groups in-
severe PD; significant difference between controls and severe PD, creased with increasing disease duration (Barichella et al., 2019).
p = 0.048). Additionally, the relative abundance of Megasphaera was Analysis of bacterial abundance data and disease duration revealed
positively associated with increasing PD severity (0.04 % in controls, significant trends for decreasing Lachnospiraceae, and increasing Ak-
2.52 % in mild PD and 5.50 % in severe PD group; significant difference kermansia and Lactobacillus with increasing disease duration (all
between controls and severe PD group, p = 0.003). Qian et al. (2018), p < 0.05) (Barichella et al., 2019). While Qian et al. (2018) showed
conducted spearman correlations (with GLM adjustments) and found no that Eschericia/Shigella genera were significantly (p = 0.028) negatively
correlations between microbiota and Hoen & Yahr staging. correlated to disease duration, Li et al. (2017), in contrast, found

234
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

Fig. 2. (continued)

significantly positive correlations between PD duration and Escherichia/ diversity did not differ between groups. Lin et al. (2018) subsequently
Shigella genera (r = 0.52, p = 0.001), as well as with Proteus (r = 0.46, compared specific bacterial taxa and found that Rikenellaceae (adjusted
p = 0.003), Enterococcus (r = 0.56, p < 0.001), and Megasphaera p value = 0.0027) and Deferribacteraceae (adjusted p value = 0.016)
(r = 0.41, p = 0.012). Li et al. (2017) also found negative correlations were more abundant in the > 5 year PD group.
with PD duration and the bacteria Blautia (r = −0.40, p = 0.013),
Ruminococcus (r = −0.58, p < 0.001), Sporobacter (r = −0.40,
3.4.2. PD motor symptoms
p = 0.013) and Haemophilus (r = −0.47, p = 0.003). In both faecal and
The relationship between PD related motor symptoms and gut mi-
GI mucosal microbiota samples (Keshavarzian et al., 2015), Firmicutes
crobiota is not clear from the reviewed studies. One study revealed that
were significantly negatively correlated with PD duration (r = −0.38,
the abundance of Anaerotruncus spp., Clostridium XIVa, and
p = 0.04 in faecal samples and r = −0.58, p < 0.01 in mucosal
Lachnospiraceae were significantly positively associated to motor
samples). Lin et al. (2018) assessed PD duration by dividing the ana-
symptoms (higher UPDRS-III scores) (Heintz‐Buschart et al., 2018).
lyses of their PD cohort into those with a PD duration of less than or
Additionally, significant positive associations between worse motor
greater than 5 years. Between these groups α-diversity was significantly
symptoms and the Lactobacillaceae family and Lactobacillus genus of
different across three of four measures (Chao1, p = 0.045; observed
bacteria was demonstrated (Barichella et al., 2019). Scheperjans et al.
Operational Taxonomic Units (OTUs), p = 0.023; PD-whole-tree,
(2015) indicated that Prevotellaceae abundance was significantly as-
p = 0.018, Shannon, p = 0.194), however, authors did not report the
sociated with UPDRS-III scores, but only when patients with motor
direction of these significant differences, whilst also stating that β-
fluctuations who were on medication, or using deep brain stimulation,

235
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

during motor-symptom examination were included. An analysis ex- that were associated with PD patients after adjusting for confounding
cluding these individuals, revealed no association between Pre- factors. Li et al. (2017) only presented a visual representation of
votellaceae abundance and UPDRS-III scores. Qian et al. (2018) also associations (Wexner scores) between constipation and the top 65
found no correlations between motor symptoms and the gut microbiota abundant genera finding negative correlations to bacteria such as
(adjusted for confounders). However, Li et al. (2017) who assessed Blautia and Ruminococcus and positive correlations to Megasphaera
correlations between PD symptoms (whether this was for specific motor and Escherichia/Shigella (no r or p values were reported).
or non-motor symptoms is not specified) and bacterial taxa, found
worse symptoms positively correlated with Enterococcus (r = 0.336, 4. Discussion
p = 0.039), Proteus (r = 0.45, p = 0.005) and Escherichia/Shigella
(r = 0.383, p = 0.018). Li et al. (2017), also observed that worse motor This review aimed to synthesise the available evidence regarding
symptoms negatively correlated with Blautia (r = −0.50, p = 0.002), gut microbiota composition in PD, and its relationship to PD diagnosis,
Faecalibacterium (r = −0.357, p = 0.028), Ruminococcus (r = −0.51, disease severity and motor and non-motor symptoms. We found nine
p = 0.001), Haemophilus (r = −0.46, p = 0.004) and Odoribacter studies that directly reported butyrate producing bacteria, at either the
abundance (r = −0.41, p = 0.011), indicating these bacteria are as- genus or species level, were in lower abundances in PD compared to
sociated with less severe PD symptoms. control groups. Additionally, α-diversity was not reliably shown to
Three studies examined PD phenotypes (Lin et al., 2018; differ between individuals with PD and healthy controls, across studies,
Scheperjans et al., 2015; Unger et al., 2016). Lin et al. (2018) divided indicating that α-diversity may be less associated with PD compared to
participants into tremor dominant (n = 18) and non-tremor dominant the abundance of SCFA, specifically butyrate, producing bacteria.
(n = 56) groups. No significant difference in α- or β-diversity was seen
between groups. However, Leptotrichia abundance was significantly 4.1. SCFA, particularly butyrate, producing bacteria are likely important
higher in patients with the tremor phenotype (adjusted p factors regarding PD onset/progression
value = 0.022). Roseburia was significantly more abundant in the pa-
tients with the non-tremor phenotype (adjusted p value = 0.028). The One theory by which gut microbiota may influence PD onset or
abundance of Enterobacteriaceae did not differ between the pheno- progression relates to lower abundances of beneficial SCFAs (Hill-Burns
types. Similarly, Unger et al. (2016) found Enterobacteriaceae abun- et al., 2017; Li et al., 2017), particularly butyrate, and the bacteria that
dance was not significantly different across phenotypes although produce them. Nine studies found five different butyrate-producing
sample sizes between groups were small: tremor dominant (n = 6), bacteria at the genus and species level that had lower abundances in PD
hypokinetic-rigid (n = 15) or mixed (n = 13). Scheperjans et al. (2015) groups compared to controls, and assessment of SCFAs showed lower
assessed slightly different phenotypes; postural instability and gait concentrations of butyrate in PD individuals (Unger et al., 2016). These
difficulty phenotype (n = 40), tremor dominant phenotype (n = 23) bacteria include Faecalibacterium prausnitzii, Eubacterium biforme, Co-
and mixed phenotypes (n = 9), finding that Enterobacteriaceae were prococcus eutactus, Roseburia faecis and the Blautia genera. Studies
significantly more abundant in patients with the postural instability and showing butyrate producing bacteria to be lower are strengthened by
gait difficulty phenotype (assessed through a GLM, p < 0.001). their large sample sizes (Barichella et al., 2019; Hill-Burns et al., 2017),
and by the range of confounding factors they accounted for including
3.4.2.1. PD non-motor symptoms: psychological. Heintz‐Buschart et al. sex, BMI, medication, age and constipation (Barichella et al., 2019; Hill-
(2018) showed Anaerotruncus and Akkermansia were significantly Burns et al., 2017; Lin et al., 2018). Findings from studies with smaller
related to greater presence of non-motor symptoms (UPDRS-I). sample sizes, and/or that did not consider relevant confounding factors,
Specific bacteria that were related to depression in the PD cohort should be interpreted with greater caution. Additionally, given the
included Anaerotruncus spp., however, authors reported no association variation in methodologies used between studies in the analysis of the
between cognition (Montreal Cognitive Assessment and Mini Mental gut microbiota, the interpretations drawn from them need to be con-
State Examination scores) and the microbiota. Qian et al. (2018) sidered with some caution as these variations may bias results (Hamady
showed that the genera Butyricicoccus (p = 0.013) and Clostridium and Knight, 2009; Thomas et al., 2015). Although methodological dif-
XIVb (p = 0.0003) were significantly positively associated with ferences may not bias microbiota results as much as previously thought
cognitive scores on the Mini Mental State Examination, but no (Lauber et al., 2010; Vogtmann et al., 2017).
significant correlations existed between gut microbiota and Two pathophysiological factors we speculate may be involved in PD
depression scores or cognition assessed through the Montreal onset/progression are gut permeability and gut inflammation leading to
Cognitive Assessment (confounding factors adjusted for). Barichella neuroinflammation. This is due to butyrate being involved in pre-
et al. (2019) found no significant differences between controls and PD venting increased gut inflammation and GI permeability (Canani et al.,
individuals in cognition (measured via Mini Mental State Examination). 2016; Tan et al., 2014b), and the current reviews finding that butyrate
They however reported greater Lactobacillus genera to be associated producing bacteria may be lower in PD. With increased GI permeability,
with intellectual impairment (OR: 1.63, 95 % CI: 1.04–2.56). which PD individuals have been shown to have (Forsyth et al., 2011), it
could be easier for the neurotropic pathogen to pass through the GI
3.4.2.2. PD non-motor symptoms: GI. Hasegawa et al. (2015) showed wall, as proposed in the Dual Hit hypothesis (Hawkes et al., 2009),
negative correlations between transit time (i.e. time taken for food to where α-synuclein misfolds and aggregates in the enteric nervous
move from ingestion through to the end of the digestive tract) and PD system. Regarding inflammation, neuroinflammation is typical of PD
duration (r = −0.34) and UPDRS-II (r = −0.40), which indicates a progression (Hald and Lotharius, 2005; Teismann and Schulz, 2004),
worsening of constipation with PD progression. However, the authors and GI inflammation is involved in this process (Devos et al., 2013).
did not assess the relationship between transit time and bacterial Therefore, GI inflammation and increased gut permeability, may be two
diversity in their study. While data are not shown, Heintz‐Buschart general pathways where gut microbiota may be involved in PD pa-
et al. (2018) state that few taxa with different abundances were found thophysiology and as such are discussed in detail below.
between constipated and non-constipated individuals. Hill-Burns et al. The role of GI inflammation in PD involves the immune cells of the
(2017) used 13 covariates, including GI-discomfort and constipation, to brain, known as microglia. When microglia are activated they con-
adjust for analyses of differentially abundant taxa between controls and tribute to neuroinflammation (Forsyth et al., 2011; Hald and Lotharius,
PD and found no change relative to the unadjusted analyses. Qian et al. 2005), and GI inflammation has been implicated in this process (Devos
(2018) used the Rome-III criteria to assess constipation, using this as a et al., 2013). Additionally, both the presence of α-synuclein (Su et al.,
confounding factor in their general linear models to determine bacteria 2008), and peripheral or systemic pro-inflammatory factors that enter

236
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

the brain (Hald and Lotharius, 2005; Qin et al., 2007), are thought to presence of pro-inflammatory bacteria and lipopolysaccharides), small
activate microglia. The role of these peripheral pro-inflammatory fac- intestinal bowel overgrowth (SIBO) may contribute to a greater amount
tors have been demonstrated in a mouse model when peripheral ad- of GI inflammation, intestinal epithelial permeability and potentially to
ministration of tumor necrosis factor-α increased the expression of pro- neuroinflammation (Devos et al., 2013; Quigley and Quera, 2006). The
inflammatory factors in the brain (Qin et al., 2007). In conjunction with prevalence of SIBO is significantly higher in PD individuals compared to
lower abundance of SCFAs, which help create a pro-inflammatory en- controls (Gabrielli et al., 2011), most likely due to the greater occur-
vironment, the presence of pro-inflammatory pathobionts such as Es- rence of GI symptoms in PD, like constipation, which creates favourable
cherichia/Shigella, Ralstonia and Enterococcus genera (among others) conditions for SIBO to occur (Gasbarrini et al., 2007). As GI issues likely
(Barichella et al., 2019; Heintz‐Buschart et al., 2018; Li et al., 2017; Lin contribute to SIBO occurring it is unlikely this condition relates to PD
et al., 2018; Qian et al., 2018), may contribute to an increased in- onset, but perhaps SIBO relates to PD progression, as research shows it
flammatory state, with these pathobionts only found to have a greater potentially relates to worse motor functioning (Tan et al., 2014a).
abundance in PD groups of the included studies. It is noted that other Considering that butyrate is anti-inflammatory and helps maintain
Gram-negative bacteria were found to have greater abundance in con- intestinal wall integrity in ways other SCFAs do not, the presence and
trols across some studies (Heintz‐Buschart et al., 2018; Lin et al., 2018); abundance of bacteria that produce butyrate in particular could be of
therefore, the role of these pro-inflammatory bacteria need to be in- importance to PD onset and progression. The lower abundances of
terpreted with caution. These Gram-negative bacteria contain lipopo- butyrate producing bacteria that were evident across PD groups of
lysaccharide in their outer membranes, an endotoxin implicated as an multiple included studies provides some support for this claim.
environmental trigger for PD and used via systemic injections to create Additionally, although other studies have observed lower SCFA con-
animal models of PD (Qin et al., 2007). Lipopolysaccharide is also in- centrations between controls and other disease states (Pozuelo et al.,
volved in microglia activation as demonstrated through systemic in- 2015; Qin et al., 2012, 2014), this does not exclude the importance of
jections (Qin et al., 2007). Additionally, animal models indicate the SCFAs to PD onset. It instead indicates that additional mechanisms, like
substantia nigra has approximately 4.5 times more microglia than other greater abundance of mucin degrading bacteria, pro-inflammatory
brain regions (Kim et al., 2000), which may help explain the specific bacteria, presence of SIBO, and the presence of a neurotropic pathogen,
disease progression of PD in humans, as this brain region would be are relevant to PD pathophysiology as well.
more susceptible to microglia activation, neuroinflammation and sub-
sequent dopaminergic cell death. Overall, the greater presence of per- 4.2. α- and β-diversity measures are not reliable markers of PD or its
ipheral pro-inflammatory factors, lipopolysaccharide and α-synuclein progression
may all be contributing to neuroinflammation and by extension PD
onset or progression. Greater bacterial diversity has been thought to be indicative of good
In isolation, the presence of a local inflammatory environment and/ health, while low bacterial diversity was thought to relate to disease
or pathogenic bacteria containing lipopolysaccharide in the GI tract are states and poor health, with recent research still claiming this is the
not likely to be sufficient to cause neuroinflammation or α-synuclein case (McBurney et al., 2019). This line of thinking has led to hypotheses
misfolding and aggregation. Multiple bacterial endotoxins are found that gut microbiota diversity would differ between control and PD
within the luminal environment of healthy individuals, whom do not groups (Scheperjans et al., 2015), and that α-diversity would be lower
have neuroinflammation or α-synuclein misfolding and aggregation in PD. However, the reviews findings show α-diversity does not reliably
(Forsyth et al., 2011). Therefore, increased intestinal permeability and differ between groups. Therefore, these results do not support the no-
degradation of the mucosal barrier are likely additional contributors, as tion that greater diversity is better for health in all circumstances, and
inflammatory factors and lipopolysaccharide need to be translocated instead the relationships between gut microbiota and the progression of
across this barrier before contacting with epithelial cells and promoting various diseases are likely more complex than just altered bacterial
α-synuclein misfolding (Forsyth et al., 2011), or leading to neuroin- diversity, considering different diseases have different pathological
flammation. Butyrate is important to this process, as butyrate helps to processes and likely involve various microbiota-host interactions
prevent intestinal permeability through provision of energy to colono- (Gerritsen et al., 2011). Additionally, we mention bacterial diversity
cytes (Canani et al., 2011; Donohoe et al., 2011), and because butyrate secondarily to the abundance of specific butyrate producing bacteria in
is key to mucin production (Brown et al., 2011; Hatayama et al., 2007). this review as we find the latter to have stronger evidence regarding its
With butyrate absent, bacterial translocation across the epithelium may role in PD pathophysiology, with the evidence for the potential roles of
be more likely. Tight junction proteins like occludin are also important bacterial diversity (specifically α-diversity) in PD pathophysiology
for maintaining the GI epithelium and preventing GI permeability being weaker.
(Rocha et al., 2014). The butyrate producing Faecalibacterium prausnitzii It is typical for a range of α-diversity indices to be measured as each
species has been shown to be protective against models of intestinal index measures a different aspect of α-diversity and there is no clear
permeability (Laval et al., 2015), through increased expression of oc- consensus as to the most informative index to use (Beck and
cludin. The Faecalibacterium prausnitzii species was lower in PD groups Schwanghart, 2010; Morris et al., 2014). While a range of measures
across six studies in this review (Barichella et al., 2019; Hill-Burns et al., were used across most studies Petrov et al. (2017), who showed α-di-
2017; Li et al., 2017; Lin et al., 2018; Petrov et al., 2017; Unger et al., versity was lower in PD groups, only used a single measure of α-di-
2016). Additionally, degradation of mucin is considered pathogenic versity (Chao1 Index). The other studies that found PD individuals had
(Cohen and Laux, 1995; Smith et al., 1995), with one such mucin de- higher α-diversity (Barichella et al., 2019; Keshavarzian et al., 2015;
grading bacteria being Akkermansia muciniphila (Derrien et al., 2004). Qian et al., 2018), or that found no between-group differences (Bedarf
Six of the included studies showed the abundance of Akkermansia et al., 2017; Hopfner et al., 2017; Li et al., 2017; Lin et al., 2018;
bacteria were greater in PD groups at either the genus level (Barichella Scheperjans et al., 2015), utilised multiple measures (See Table 2 for a
et al., 2019; Heintz‐Buschart et al., 2018; Hill-Burns et al., 2017; full list of α-diversity measures used across studies). The fact that
Keshavarzian et al., 2015), or species level (Bedarf et al., 2017; Lin Petrov et al. (2017) reported only a single measure diminishes the re-
et al., 2018). Overall, a combination of lowered levels of butyrate liability of their finding, and the multiple measures used across the
producing bacteria and increased pathogenic mucin degrading bacteria, other studies suggests that there may in fact be no reliable α-diversity
could be leading to increased GI permeability and contributing to PD differences between groups, or that PD groups have greater α-diversity.
onset as a result. Additionally, some studies who reported differences in bacterial di-
In addition to the above-mentioned mechanisms (lower SCFA pro- versity had larger sample sizes (n = 34–193) (Barichella et al., 2019;
ducing bacteria, greater mucin degrading bacteria, and greater Keshavarzian et al., 2015; Petrov et al., 2017; Qian et al., 2018), than

237
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

those reporting no differences, suggesting a possible lack of power in utilised, which only included the Montreal Cognitive Assessment and
latter studies (n = 14–74) (Bedarf et al., 2017; Hopfner et al., 2017; Li the Mini-Mental State Examination, with these tests used as screening
et al., 2017; Lin et al., 2018; Scheperjans et al., 2015). Finally, while tools to rule out cognitive impairment rather than being sensitive
many factors are known to influence gut microbiota diversity, only one measures of variations in cognitive function (Hoops et al., 2009). Future
study assessed any potential confounding factors in relation to α-di- research should use measures targeting various cognitive domains that
versity, in this case age and sex (Hopfner et al., 2017). are sensitive to cognitive performance changes. Doing so would help
While the current literature does not present strong evidence to elucidate whether relationships between gut microbiota and cognition
support a-diversity differences in PD pathophysiology, it remains pos- are apparent, and whether these relationships differ from normal
sible that the lack of consensus is derived from a combination of factors ageing to PD. As the MGB-axis field is newly emerging, it is not sur-
such as lack of power, differences in α-diversity measures used, and prising that few of the included studies have used cognitive testing in
measurement error due to confounding. This review supports the idea their protocols. This is generally the case across other neurodegenera-
that mechanisms other than lower microbiota diversity (described tive conditions, and even in healthy controls research is limited. For
through α-diversity indices), such as decreased abundance of SCFA- example, in healthy older adults, gut microbiota composition was as-
producing bacteria and increased GI permeability, are associated with sociated with cognitive test performance across a brief cognitive test
PD. battery (Manderino et al., 2017), however associations were only at the
phylum level and without identifying specific genera or species of
4.3. The optimal range hypothesis: too little or too many, the optimal bacteria, relating cognition to bacteria and their functions cannot be
number of gut microbiota determined. Additionally, Mini Mental State Examination scores in in-
dividuals with Alzheimer’s disease improved after probiotic ingestion
Our bodily functions exist across a normal homeostatic range, with which contained various Lactobacillus species as well as Bifidobacterium
large fluctuations outside of this leading to health consequences. Given bifidum (Akbari et al., 2016). However, authors did not measure gut
gut microbiota are responsible for a plethora of key functions that microbiota differences pre- or post-intervention, meaning associations
specifically relate to our overall homeostasis (E.g. maintenance of in- between abundances of specific gut microbiota, or gut microbiota di-
testinal wall integrity), we should similarly consider individual bacteria versity, and cognition cannot be made (Akbari et al., 2016). These
abundance to have negative health outcomes when they decrease or findings suggest that while a link between cognition and gut microbiota
increase outside of a certain range, whereby dysfunction of key bac- may be apparent, the specific links between various cognitive domains
terial functions may occur. I.e. there may be an optimal range that key and presence/absence and abundance of specific bacteria are still un-
bacteria or bacterial groups should ideally remain within, to ensure known and require further investigation. Without understanding how
optimal host health. Butyrate producing bacteria were generally shown cognition relates to gut microbiota in healthy individuals, it is difficult
to be lower in PD, with lower levels of butyrate reported as well (Unger to describe how this would then relate to individuals with neurode-
et al., 2016). In contrast, Akkermansia bacteria were demonstrated to be generative conditions and in PD.
in greater abundance in PD groups (Barichella et al., 2019; Bedarf et al.,
2017; Heintz‐Buschart et al., 2018; Hill-Burns et al., 2017; 4.5. Measuring diet and additional lifestyle factors
Keshavarzian et al., 2015; Lin et al., 2018), with these bacteria known
pathogenic mucin degraders. If the observed abundance levels of these Given the variation in individuals’ gut microbiota, and the factors
bacteria have decreased, or increased outside of a ‘normal’ range it is that contribute to this variability (Gerritsen et al., 2011), controlling for
possible that dysfunctional physiological processes would result, po- these confounding factors is essential to adequately determine and
tentially leading to PD onset. We propose that the presence/absence understand the role of gut microbiota in PD. While numerous con-
and abundance of specific bacteria, or groups of specific bacteria, who founding factors were assessed across some studies, lifestyle factors like
perform critical functions, are key to our overall health and should be diet, physical activity and sleep were not. Some studies accounted for
considered more important than bacterial diversity (α-diversity). Ad- diet by using spousal controls (Hasegawa et al., 2015; Lin et al., 2018;
ditionally, depending on the bacteria and their purported functions, Qian et al., 2018), as spouses would likely share similar diets to PD
either increased or decreased abundance of those bacteria may be individuals, although this is unlikely to be as effective as directly
detrimental. measuring diet. Lin et al. (2018) collected dietary data via a ques-
tionnaire, although no details regarding the questionnaire or its validity
4.4. Parkinson’s disease symptoms and severity are available. Additionally, authors only categorised participants as
‘omnivorous’, ‘meat (-eating)’, or ‘vegetable (-eating)’, providing lim-
In establishing the relationships gut microbiota may have to PD, it is ited characterisation of the actual nutritional intakes of participants.
important to assess how gut microbiota differences may relate to PD Keshavarzian et al. (2015) collected dietary data with a modified
symptoms, severity and duration. Duration of PD and worse PD motor Harvard food frequency questionnaire, and for total intakes of energy,
symptoms were positively correlated to Escherichia/Shigella, with ne- macronutrients and dietary fibre no significant differences were seen
gative correlations found between putative butyrate producing bac- between groups. However, this was only investigated in a subsample of
teria, Blautia, and PD duration as well as motor-symptoms (Li et al., participants (24/38 PD and 15/34 controls). Barichella et al. (2019)
2017). These correlations do align with the above stated hypothesis that also assessed dietary intake, using a 10-day food diary and 24-h food
these bacteria may be involved in PD pathophysiology due to them recall, assessing intake of calories, protein (vegetable or animal), fibre,
being pro-inflammatory, and it may be that Escherichia/Shigella cause water and alcohol. While fibre ingestion related to increased Lactoba-
worse motor-symptoms and/or potentially increase with increasing PD cillaceae abundance across both groups, authors mostly used nutri-
duration. However, a lack of replication across studies means con- tional information as a confounding factor in their analysis of bacterial
sistently identifying gut microbiota that relate to PD duration and abundance and diversity, and baseline measures of fibre between con-
motor-symptoms is not possible at this time. trols and PD individuals were not significantly different.
As for non-motor symptoms, relationships between gut microbiota No other included studies assessed nutritional intake of participants,
and PD non-motor symptoms, namely cognition, are not clear. Across and across all studies resistant starch intake (present in e.g. legumes
three included studies, two found no associations between cognitive and whole grains), which can help commensal bacteria proliferate, and
measures and gut microbiota (Barichella et al., 2019; Heintz‐Buschart in turn produce SCFAs (Bourassa et al., 2016; Topping and Clifton,
et al., 2018), with only one study showing an association (Qian et al., 2001), was not assessed. Additionally, the diversity of plants consumed
2018). The lack of association could in part be due to the cognitive tests was not assessed despite research indicating a larger variety of plants

238
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

consumed is associated with gut microbial diversity (McDonald et al., section of the qualitative analysis to do with measures of diversity,
2018), with this factor potentially explaining differences in bacteria presence/absence and abundance as well as sections in the discussion
diversity measures across included studies. Other lifestyle factors like relating to gut microbiota specifically. HM provided multiple revision
physical activity and sleep were not assessed, despite links to the gut on the overall discussion section, and helped generate the overall dis-
microbiota indicating these factors can alter bacterial diversity and cussion section, as well as revising the overall document. All authors
bacterial abundance as well as bacterial metabolites (Allen et al., 2018; read and accepted the final version of the manuscript for submission.
Benedict et al., 2016). Given diet, physical activity and sleep may all
alter bacterial diversity and/or bacterial abundance they need to be Financial disclosures
accounted for in studies to properly determine the roles gut bacteria
have in PD onset or progression, and to identify relationships between An Australian Government Research Training Program Scholarship
gut microbiota and PD severity and symptoms. supports NN.

4.6. Future directions Declaration of Competing Interest

Given the proposed relationships between lifestyle factors, like diet, The authors report no conflicts of interest.
physical activity and sleep, and the gut microbiota (Allen et al., 2018;
Benedict et al., 2016; David et al., 2014), it is important future studies Appendix A. Supplementary data
include these as part of their analyses. Probiotic use should also be
considered. While outside the scope of this review, additional factors Supplementary material related to this article can be found, in the
including environmental toxins (Ball et al., 2019; Betarbet et al., 2000; online version, at doi:https://doi.org/10.1016/j.neubiorev.2020.02.
Di Monte, 2001; Gorell et al., 1999) and genetics (Ball et al., 2019; 003.
Breydo et al., 2012; Cookson, 2005), and their potential effects on gut
microbiota composition, still need to be considered in PD pathogenesis. References
Future studies would also benefit from discussing the functional re-
lationships of bacteria together, due to the influence of certain bacteria Abbott, R.D., Petrovitch, H., White, L., Masaki, K., Tanner, C., Curb, J., Grandinetti, A.,
on other bacteria. For example in ‘cross-feeding’ whereby the metabo- Blanchette, P., Popper, J., Ross, G., 2001. Frequency of bowel movements and the
future risk of Parkinson’s disease. Neurology 57 (3), 456–462.
lites of one bacteria are used by other bacteria to produce another, Akbari, E., Asemi, Z., Daneshvar Kakhaki, R., Bahmani, F., Kouchaki, E., Tamtaji, O.R.,
different, metabolite (For more on ‘cross-feeding’ see Duncan et al., Hamidi, G.A., Salami, M., 2016. Effect of probiotic supplementation on cognitive
2004; Flint et al., 2012). Additionally, to increase the reliability of the function and metabolic status in Alzheimer’s disease: A randomized, double-blind
and controlled trial. Front. Aging Neurosci. 8 (256).
reported results, gut microbiota analysis should not be limited to the Allen, J.M., Mailing, L.J., Niemiro, G.M., Moore, R., Cook, M.D., White, B.A., Holscher,
surrogate measure of using faecal samples, but additional use of GI H.D., Woods, J.A., 2018. Exercise alters gut microbiota composition and function in
mucosal microbiota samples, as was conducted by Keshavarzian et al. lean and obese humans. Med. Sci. Sports Exerc. 50 (4), 747–757.
Ball, N., Teo, W.P., Chandra, S., Chapman, J., 2019. Parkinson’s disease and the en-
(2015), would bolster current results. The observational nature of the vironment. Front. Neurol. 10, 218.
included studies also makes it impossible to state whether gut micro- Barichella, M., Severgnini, M., Cilia, R., Cassani, E., Bolliri, C., Caronni, S., Ferri, V.,
biota differences, particularly lowered abundance of butyrate produ- Cancello, R., Ceccarani, C., Faierman, S., 2019. Unraveling gut microbiota in
Parkinson’s disease and atypical parkinsonism. Mov. Disord. 34 (3), 396–405.
cing bacteria, contribute to PD, or if gut microbiota differences result
Beck, J., Schwanghart, W., 2010. Comparing measures of species diversity from in-
from PD. Longitudinal studies assessing gut microbiota changes over complete inventories: an update. Methods Ecol. Evol. 1 (1), 38–44.
time in relation to disease onset, pathophysiology and symptom se- Bedarf, J.R., Hildebrand, F., Coelho, L.P., Sunagawa, S., Bahram, M., Goeser, F., Bork, P.,
verity of PD, that control for the appropriate confounding factors, are Wüllner, U., 2017. Functional implications of microbial and viral gut metagenome
changes in early stage L-DOPA-naïve Parkinson’s disease patients. Genome Med. 9
therefore warranted. (1), 39-.
Benedict, C., Vogel, H., Jonas, W., Woting, A., Blaut, M., Schürmann, A., Cedernaes, J.,
5. Conclusion 2016. Gut microbiota and glucometabolic alterations in response to recurrent partial
sleep deprivation in normal-weight young individuals. Mol. Metab. 5 (12),
1175–1186.
The current review indicates that gut microbiota’s role in the mi- Betarbet, R., Sherer, T.B., MacKenzie, G., Garcia-Osuna, M., Panov, A.V., Greenamyre,
crobiota-gut-brain axis and association to PD may involve lowered J.T., 2000. Chronic systemic pesticide exposure reproduces features of Parkinson’s
disease. Nat. Neurosci. 3 (12), 1301.
abundance of a range of specific SCFA, particularly butyrate, producing Block, M.L., Hong, J.-S., 2005. Microglia and inflammation-mediated neurodegeneration:
bacteria (whereby these bacteria may be reduced outside of an ‘optimal multiple triggers with a common mechanism. Prog. Neurobiol. 76 (2), 77–98.
range’). However, the mechanisms of PD pathophysiology are unlikely Bourassa, M.W., Alim, I., Bultman, S.J., Ratan, R.R., 2016. Butyrate, neuroepigenetics and
the gut microbiome: can a high fiber diet improve brain health? Neurosci. Lett. 625,
to be solely a lack of SCFAs. Rather, a combination of factors including
56–63.
increased pro-inflammatory bacteria, greater intestinal permeability, Breydo, L., Wu, J.W., Uversky, V.N., 2012. α-Synuclein misfolding and Parkinson’s dis-
introduction of a neurotropic pathogen and small intestinal bowel ease. Biochimica et Biophysica Acta (BBA)-Mol. Basis Dis. 1822 (2), 261–285.
Brown, C.T., Davis-Richardson, A.G., Giongo, A., Gano, K.A., Crabb, D.B., Mukherjee, N.,
overgrowth, are likely additionally responsible (Canani et al., 2011;
Casella, G., Drew, J.C., Ilonen, J., Knip, M., Hyöty, H., Veijola, R., Simell, T., Simell,
Tan et al., 2014b). Despite this, additional research is needed to address O., Neu, J., Wasserfall, C.H., Schatz, D., Atkinson, M.A., Triplett, E.W., 2011. Gut
how these gut microbiota differences relate to PD duration and symp- microbiome metagenomics analysis suggests a functional model for the development
toms, and if these differences are still present when accounting for of autoimmunity for type 1 diabetes. PLoS One 6 (10), e25792.
Canani, R.B., Costanzo, M.D., Leone, L., Pedata, M., Meli, R., Calignano, A., 2011.
confounding lifestyle and health factors. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases.
World J. Gastroenterol. 17 (12), 1519–1528.
Author contributions Canani, R.B., Sangwan, N., Stefka, A.T., Nocerino, R., Paparo, L., Aitoro, R., Calignano, A.,
Khan, A.A., Gilbert, J.A., Nagler, C.R., 2016. Lactobacillus rhamnosus GG-supple-
mented formula expands butyrate-producing bacterial strains in food allergic infants.
NN completed the search of articles, the screening at each stage as ISME J. 10 (3), 742.
well as data extraction and risk of bias assessment. Authors WPT, AH, Cohen, P.S., Laux, D.C., 1995. Bacterial adhesion to and penetration of intestinal mucus in
vitro. Methods in Enzymology, vol. 253. Academic Press, pp. 309–314.
AL, HM, and ESG participated in the article screening process at the title Collins, M., Lawson, P., Willems, A., Cordoba, J., Fernandez-Garayzabal, J., Garcia, P.,
and abstract as well as full-text screening stages. Manuscript writing Cai, J., Hippe, H., Farrow, J., 1994. The phylogeny of the genus Clostridium: proposal
was completed by NN, with WPT, ESG, and AH revising the overall of five new genera and eleven new species combinations. Int. J. Syst. Evol. Microbiol.
44 (4), 812–826.
manuscript multiple times throughout the writing process. AL provided Cookson, M.R., 2005. The biochemistry of Parkinson’s disease. Annu. Rev. Biochem. 74,
comments and revisions on the cladogram figures, methods and results

239
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

29–52. difference in susceptibility to lipopolysaccharide-induced neurotoxicity in the rat


David, L.A., Maurice, C.F., Carmody, R.N., Gootenberg, D.B., Button, J.E., Wolfe, B.E., brain: role of microglia. J. Neurosci. 20 (16), 6309–6316.
Ling, A.V., Devlin, A.S., Varma, Y., Fischbach, M.A., 2014. Diet rapidly and re- Kim, S.Y., Park, J.E., Lee, Y.J., Seo, H.-J., Sheen, S.-S., Hahn, S., Jang, B.-H., Son, H.-J.,
producibly alters the human gut microbiome. Nature 505 (7484), 559. 2013. Testing a tool for assessing the risk of bias for nonrandomized studies showed
Derrien, M., Vaughan, E.E., Plugge, C.M., de Vos, W.M., 2004. Akkermansia muciniphila moderate reliability and promising validity. J. Clin. Epidemiol. 66 (4), 408–414.
gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Kim, S., Kwon, S.H., Kam, T.I., Panicker, N., Karuppagounder, S.S., Lee, S., Lee, J.H., Kim,
Microbiol. 54 (5), 1469–1476. W.R., Kook, M., Foss, C.A., Shen, C., 2019. Transneuronal propagation of pathologic
Devos, D., Lebouvier, T., Lardeux, B., Biraud, M., Rouaud, T., Pouclet, H., Coron, E., α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 103 (4),
Bruley des Varannes, S., Naveilhan, P., Nguyen, J.-M., Neunlist, M., Derkinderen, P., 627–641.
2013. Colonic inflammation in Parkinson’s disease. Neurobiol. Dis. 50, 42–48. Lauber, C.L., Zhou, N., Gordon, J.I., Knight, R., Fierer, N., 2010. Effect of storage con-
Di Monte, D.A., 2001. The role of environmental agents in Parkinson’s disease. Clin. ditions on the assessment of bacterial community structure in soil and human-asso-
Neurosci. Res. 1 (6), 419–426. ciated samples. FEMS Microbiol. Lett. 307 (1), 80–86.
Donohoe, Dallas R., Garge, N., Zhang, X., Sun, W., O’Connell, Thomas M., Bunger, Laval, L., Martin, R., Natividad, J.N., Chain, F., Miquel, S., de Maredsous, C.D.,
Maureen K., Bultman, Scott J., 2011. The microbiome and butyrate regulate energy Capronnier, S., Sokol, H., Verdu, E.F., van Hylckama Vlieg, J.E.T., Bermúdez-
metabolism and autophagy in the mammalian colon. Cell Metab. 13 (5), 517–526. Humarán, L.G., Smokvina, T., Langella, P., 2015. Lactobacillus rhamnosus CNCM I-
Duncan, S.H., Hold, G.L., Harmsen, H.J.M., Stewart, C.S., Flint, H.J., 2002. Growth re- 3690 and the commensal bacterium Faecalibacterium prausnitzii A2-165 exhibit si-
quirements and fermentation products of Fusobacterium prausnitzii, and a proposal milar protective effects to induced barrier hyper-permeability in mice. Gut Microbes
to reclassify it as Faecalibacterium prausnitzii gen. nov., comb. Nov. Int. J. Syst. Evol. 6 (1), 1–9.
Microbiol. 52 (6), 2141–2146. Letunic, I., Bork, P., 2016. Interactive tree of life (iTOL) v3: an online tool for the display
Duncan, S.H., Louis, P., Flint, H.J., 2004. Lactate-utilizing bacteria, isolated from human and annotation of phylogenetic and other trees. Nucleic Acids Res. 44 (W1)
feces, that produce butyrate as a major fermentation product. Appl. Environ. W242-W5.
Microbiol. 70 (10), 5810–5817. Li, W., Wu, X., Hu, X., Wang, T., Liang, S., Duan, Y., Jin, F., Qin, B., 2017. Structural
Fasano, A., Visanji, N.P., Liu, L.W., Lang, A.E., Pfeiffer, R.F., 2015. Gastrointestinal changes of gut microbiota in Parkinson’s disease and its correlation with clinical
dysfunction in Parkinson’s disease. Lancet Neurol. 14 (6), 625–639. features. Sci. China Life Sci. 60 (11), 1223–1233.
Flint, H.J., Scott, K.P., Duncan, S.H., Louis, P., Forano, E., 2012. Microbial degradation of Lin, A., Zheng, W., He, Y., Tang, W., Wei, X., He, R., Huang, W., Su, Y., Huang, Y., Zhou,
complex carbohydrates in the gut. Gut Microbes 3 (4), 289–306. H., 2018. Gut microbiota in patients with Parkinson’s disease in southern China.
Forsyth, C.B., Shannon, K.M., Kordower, J.H., Voigt, R.M., Shaikh, M., Jaglin, J.A., Estes, Parkinsonism Relat. Disord. 53, 82–88.
J.D., Dodiya, H.B., Keshavarzian, A., 2011. Increased intestinal permeability corre- Liu, C., Finegold, S.M., Song, Y., Lawson, P.A., 2008. Reclassification of Clostridium
lates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers coccoides, Ruminococcus hansenii, Ruminococcus hydrogenotrophicus,
in early Parkinson’s disease. PLoS One 6 (12), 1–10. Ruminococcus luti, Ruminococcus productus and Ruminococcus schinkii as Blautia
Gabrielli, M., Bonazzi, P., Scarpellini, E., Bendia, E., Lauritano, E.C., Fasano, A., Ceravolo, coccoides gen. nov., comb. nov., Blautia hansenii comb. nov., Blautia hydro-
M.G., Capecci, M., Rita Bentivoglio, A., Provinciali, L., Tonali, P.A., Gasbarrini, A., genotrophica comb. nov., Blautia luti comb. nov., Blautia producta comb. nov.,
2011. Prevalence of small intestinal bacterial overgrowth in Parkinson’s disease. Blautia schinkii comb. nov. and description of Blautia wexlerae sp. nov., isolated
Mov. Disord. 26 (5), 889–892. from human faeces. Int. J. Syst. Evol. Microbiol. 58 (8), 1896–1902.
Gasbarrini, A., Lauritano, E.C., Gabrielli, M., Scarpellini, E., Lupascu, A., Ojetti, V., Lopetuso, L.R., Scaldaferri, F., Petito, V., Gasbarrini, A., 2013. Commensal Clostridia:
Gasbarrini, G., 2007. Small intestinal bacterial overgrowth: diagnosis and treatment. leading players in the maintenance of gut homeostasis. Gut Pathog. 5 (1), 23.
Dig. Dis. 25 (3), 237–240. Manderino, L., Carroll, I., Azcarate-Peril, M.A., Rochette, A., Heinberg, L., Peat, C.,
Gerritsen, J., Smidt, H., Rijkers, G.T., de Vos, W.M., 2011. Intestinal microbiota in human Steffen, K., Mitchell, J., Gunstad, J., 2017. Preliminary evidence for an association
health and disease: the impact of probiotics. Genes Nutr. 6 (3), 209–240. between the composition of the gut microbiome and cognitive function in neurolo-
Gorell, J., Johnson, C., Rybicki, B., Peterson, E., Kortsha, G., Brown, G., Richardson, R., gically healthy older adults. J. Int. Neuropsychol. Soc.: JINS 23 (8), 700–705.
1999. Occupational exposure to manganese, copper, lead, iron, mercury and zinc and Marchesi, J.R., Ravel, J., 2015. The Vocabulary of Microbiome Research: a Proposal.
the risk of Parkinson’s disease. Neurotoxicology 20 (2–3), 239–247. BioMed Central, pp. 2049–2618.
Haehner, A., Hummel, T., Reichmann, H., 2009. Olfactory dysfunction as a diagnostic Martinez-Martin, P., 2011. The importance of non-motor disturbances to quality of life in
marker for Parkinson’s disease. Expert Rev. Neurother. 9 (12), 1773–1779. Parkinson’s disease. J. Neurol. Sci. 310 (1), 12–16.
Hald, A., Lotharius, J., 2005. Oxidative stress and inflammation in Parkinson’s disease: is Mayer, E.A., Tillisch, K., Gupta, A., 2015. Gut/brain axis and the microbiota. J. Clin.
there a causal link? Exp. Neurol. 193 (2), 279–290. Invest. 125 (3), 926.
Hamady, M., Knight, R., 2009. Microbial community profiling for human microbiome McBurney, M.I., Davis, C., Fraser, C.M., Schneeman, B.O., Huttenhower, C., Verbeke, K.,
projects: tools, techniques, and challenges. Genome Res. 19 (7), 1141–1152. Walter, J., Latulippe, M.E., 2019. Establishing what constitutes a healthy human gut
Hasegawa, S., Goto, S., Tsuji, H., Okuno, T., Asahara, T., Nomoto, K., Shibata, A., microbiome: state of the science, regulatory considerations, and future directions. J.
Fujisawa, Y., Minato, T., Okamoto, A., Ohno, K., Hirayama, M., 2015. Intestinal Nutr. 149 (11), 1882–1895.
dysbiosis and lowered serum lipopolysaccharide-binding protein in Parkinson’s dis- McDonald, D., Hyde, E., Debelius, J.W., Morton, J.T., Gonzalez, A., Ackermann, G.,
ease. PLoS One 10 (11), e0142164. Aksenov, A.A., Behsaz, B., Brennan, C., Chen, Y., DeRight Goldasich, L., Dorrestein,
Hatayama, H., Iwashita, J., Kuwajima, A., Abe, T., 2007. The short chain fatty acid, P.C., Dunn, R.R., Fahimipour, A.K., Gaffney, J., Gilbert, J.A., Gogul, G., Green, J.L.,
butyrate, stimulates MUC2 mucin production in the human colon cancer cell line, Hugenholtz, P., Humphrey, G., Huttenhower, C., Jackson, M.A., Janssen, S., Jeste,
LS174T. Biochem. Biophys. Res. Commun. 356 (3), 599–603. D.V., Jiang, L., Kelley, S.T., Knights, D., Kosciolek, T., Ladau, J., Leach, J., Marotz, C.,
Hawkes, C.H., Del Tredici, K., Braak, H., 2009. Parkinson’s disease: the dual hit theory Meleshko, D., Melnik, A.V., Metcalf, J.L., Mohimani, H., Montassier, E., Navas-
revisited. Ann. N. Y. Acad. Sci. 1170 (1), 615–622. Molina, J., Nguyen, T.T., Peddada, S., Pevzner, P., Pollard, K.S., Rahnavard, G.,
Heintz‐Buschart, A., Pandey, U., Wicke, T., Sixel‐Döring, F., Janzen, A., Sittig‐Wiegand, Robbins-Pianka, A., Sangwan, N., Shorenstein, J., Smarr, L., Song, S.J., Spector, T.,
E., Trenkwalder, C., Oertel, W.H., Mollenhauer, B., Wilmes, P., 2018. The nasal and Swafford, A.D., Thackray, V.G., Thompson, L.R., Tripathi, A., Vázquez-Baeza, Y.,
gut microbiome in Parkinson’s disease and idiopathic rapid eye movement sleep Vrbanac, A., Wischmeyer, P., Wolfe, E., Zhu, Q., Knight, R., 2018. American gut: an
behavior disorder. Mov. Disord. 33 (1), 88–98. open platform for citizen science microbiome research. mSystems 3 (3), e00031–18.
Hill-Burns, E.M., Debelius, J.W., Morton, J.T., Wissemann, W.T., Lewis, M.R., Wallen, Moher, D., Liberati, A., Tetzlaff, J., Altman, D.G., The, P.G., 2009. Preferred reporting
Z.D., Peddada, S.D., Factor, S.A., Molho, E., Zabetian, C.P., Knight, R., Payami, H., items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 6
2017. Parkinson’s disease and Parkinson’s disease medications have distinct sig- (7), e1000097.
natures of the gut microbiome. Mov. Disord. 32 (5), 739–749. Moher, D., Shamseer, L., Clarke, M., Ghersi, D., Liberati, A., Petticrew, M., Shekelle, P.,
Holmqvist, S., Chutna, O., Bousset, L., Aldrin-Kirk, P., Li, W., Björklund, T., Wang, Z.-Y., Stewart, L.A., 2015. Preferred reporting items for systematic review and meta-ana-
Roybon, L., Melki, R., Li, J.-Y., 2014. Direct evidence of Parkinson pathology spread lysis protocols (PRISMA-P) 2015 statement. Syst. Rev. 4 (1), 1.
from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 128 (6), Morris, E.K., Caruso, T., Buscot, F., Fischer, M., Hancock, C., Maier, T.S., Meiners, T.,
805–820. Müller, C., Obermaier, E., Prati, D., Socher, S.A., Sonnemann, I., Wäschke, N., Wubet,
Holzapfel, W.H., Haberer, P., Snel, J., Schillinger, U., in’t Veld, J.H.H., 1998. Overview of T., Wurst, S., Rillig, M.C., 2014. Choosing and using diversity indices: insights for
gut flora and probiotics. Int. J. Food Microbiol. 41 (2), 85–101. ecological applications from the German Biodiversity Exploratories. Ecol. Evol. 4
Hoops, S., Nazem, S., Siderowf, A.D., Duda, J.E., Xie, S.X., Stern, M.B., Weintraub, D., (18), 3514–3524.
2009. Validity of the MoCA and MMSE in the detection of MCI and dementia in Petrov, V., Saltykova, I., Zhukova, I., Alifirova, V., Zhukova, N., Dorofeeva, Y., Tyakht, A.,
Parkinson disease. Neurology 73 (21), 1738. Kovarsky, B., Alekseev, D., Kostryukova, E., Mironova, Y., Izhboldina, O., Nikitina,
Hopfner, F., Künstner, A., Müller, S.H., Künzel, S., Zeuner, K.E., Margraf, N.G., Deuschl, M., Perevozchikova, T., Fait, E., Babenko, V., Vakhitova, M., Govorun, V., Sazonov,
G., Baines, J.F., Kuhlenbäumer, G., 2017. Gut microbiota in Parkinson disease in a A., 2017. Analysis of gut microbiota in patients with Parkinson’s disease. Bull. Exp.
northern German cohort. Brain Res. 1667, 41–45. Biol. Med. 162 (6), 734–737.
Jackson, M.A., Verdi, S., Maxan, M.-E., Shin, C.M., Zierer, J., Bowyer, R.C., Martin, T., Pozuelo, M., Panda, S., Santiago, A., Mendez, S., Accarino, A., Santos, J., Guarner, F.,
Williams, F.M., Menni, C., Bell, J.T., 2018. Gut microbiota associations with common Azpiroz, F., Manichanh, C., 2015. Reduction of butyrate- and methane-producing
diseases and prescription medications in a population-based cohort. Nat. Commun. 9. microorganisms in patients with Irritable Bowel Syndrome. Sci. Rep. 5, 12693.
Jankovic, J., 2008. Parkinson’s disease: clinical features and diagnosis. J. Neurol. Qian, Y., Yang, X., Xu, S., Wu, C., Song, Y., Qin, N., Chen, S.-D., Xiao, Q., 2018. Alteration
Neurosurg. Psychiatr. 79 (4), 368–376. of the fecal microbiota in Chinese patients with Parkinson’s disease. Brain Behav.
Keshavarzian, A., Green, S.J., Engen, P.A., Voigt, R.M., Naqib, A., Forsyth, C.B., Mutlu, E., Immun. 70, 194–202.
Shannon, K.M., 2015. Colonic bacterial composition in Parkinson’s disease. Mov. Qin, L., Wu, X., Block, M.L., Liu, Y., Breese, G.R., Hong, J.S., Knapp, D.J., Crews, F.T.,
Disord. 30 (10), 1351–1360. 2007. Systemic LPS causes chronic neuroinflammation and progressive neurode-
Kim, W.-G., Mohney, R.P., Wilson, B., Jeohn, G.-H., Liu, B., Hong, J.-S., 2000. Regional generation. Glia 55 (5), 453–462.

240
N.D. Nuzum, et al. Neuroscience and Biobehavioral Reviews 112 (2020) 227–241

Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., Zhang, F., Liang, S., Zhang, W., Guan, Y., Shen, D., Synuclein activates microglia in a model of Parkinson’s disease. Neurobiol. Aging 29
Peng, Y., Zhang, D., Jie, Z., Wu, W., Qin, Y., Xue, W., Li, J., Han, L., Lu, D., Wu, P., (11), 1690–1701.
Dai, Y., Sun, X., Li, Z., Tang, A., Zhong, S., Li, X., Chen, W., Xu, R., Wang, M., Feng, Tan, A.H., Mahadeva, S., Thalha, A.M., Gibson, P.R., Kiew, C.K., Yeat, C.M., Ng, S.W.,
Q., Gong, M., Yu, J., Zhang, Y., Zhang, M., Hansen, T., Sanchez, G., Raes, J., Falony, Ang, S.P., Chow, S.K., Tan, C.T., 2014a. Small intestinal bacterial overgrowth in
G., Okuda, S., Almeida, M., LeChatelier, E., Renault, P., Pons, N., Batto, J.-M., Zhang, Parkinson’s disease. Parkinsonism Relat. Disord. 20 (5), 535–540.
Z., Chen, H., Yang, R., Zheng, W., Li, S., Yang, H., Wang, J., Ehrlich, S.D., Nielsen, R., Tan, J., McKenzie, C., Potamitis, M., Thorburn, A.N., Mackay, C.R., Macia, L., 2014b. The
Pedersen, O., Kristiansen, K., Wang, J., 2012. A metagenome-wide association study role of short-chain fatty acids in health and disease. In: In: Alt, F.W. (Ed.), Advances
of gut microbiota in type 2 diabetes. Nature 490, 55. in Immunology, vol. 121. Academic Press, pp. 91–119.
Qin, N., Yang, F., Li, A., Prifti, E., Chen, Y., Shao, L., Guo, J., Le Chatelier, E., Yao, J., Wu, Teismann, P., Schulz, J.B., 2004. Cellular pathology of Parkinson’s disease: astrocytes,
L., Zhou, J., Ni, S., Liu, L., Pons, N., Batto, J.M., Kennedy, S.P., Leonard, P., Yuan, C., microglia and inflammation. Cell Tissue Res. 318 (1), 149–161.
Ding, W., Chen, Y., Hu, X., Zheng, B., Qian, G., Xu, W., Ehrlich, S.D., Zheng, S., Li, L., Thomas, V., Clark, J., Doré, J., 2015. Fecal microbiota analysis: an overview of sample
2014. Alterations of the human gut microbiome in liver cirrhosis. Nature 513, 59. collection methods and sequencing strategies. Future Microbiol. 10 (9), 1485–1504.
Quigley, E.M., Quera, R., 2006. Small intestinal bacterial overgrowth: roles of antibiotics, Topping, D.L., Clifton, P.M., 2001. Short-chain fatty acids and human colonic function:
prebiotics, and probiotics. Gastroenterology 130 (2), S78–S90. roles of resistant starch and nonstarch polysaccharides. Physiol. Rev. 81 (3),
Rocha, B., Correia, M., Barbosa, R., Laranjinha, J., 2014. A dietary-driven redox mod- 1031–1064.
ulation of gut microbiome-host interactions: the rescue of epithelial barrier and Unger, M.M., Spiegel, J., Dillmann, K.-U., Grundmann, D., Philippeit, H., Bürmann, J.,
mucus production during dysbiosis by dietary nitrate. Paper Presented to Free Faßbender, K., Schwiertz, A., Schäfer, K.-H., 2016. Short chain fatty acids and gut
Radical Biology and Medicine 2014/10/01/. http://www.sciencedirect.com/ microbiota differ between patients with Parkinson’s disease and age-matched con-
science/article/pii/S0891584914012805. trols. Parkinsonism Relat. Disord. 32, 66–72.
Scheperjans, F., Aho, V., Pereira, P.A., Koskinen, K., Paulin, L., Pekkonen, E., Haapaniemi, Visanji, N.P., Marras, C., Kern, D.S., Al Dakheel, A., Gao, A., Liu, L.W., Lang, A.E., Hazrati,
E., Kaakkola, S., Eerola‐Rautio, J., Pohja, M., 2015. Gut microbiota are related to L.-N., 2015. Colonic mucosal α-synuclein lacks specificity as a biomarker for
Parkinson’s disease and clinical phenotype. Mov. Disord. 30 (3), 350–358. Parkinson disease. Neurology 84 (6), 609–616.
Sekirov, I., Russell, S.L., Antunes, L.C.M., Finlay, B.B., 2010. Gut microbiota in health and Vivekanantham, S., Shah, S., Dewji, R., Dewji, A., Khatri, C., Ologunde, R., 2015.
disease. Physiol. Rev. 90 (3), 859–904. Neuroinflammation in Parkinson’s disease: role in neurodegeneration and tissue re-
Shannon, K.M., Keshavarzian, A., Mutlu, E., Dodiya, H.B., Daian, D., Jaglin, J.A., pair. Int. J. Neurosci. 125 (10), 717–725.
Kordower, J.H., 2012. Alpha‐synuclein in colonic submucosa in early untreated Vogtmann, E., Chen, J., Amir, A., Shi, J., Abnet, C.C., Nelson, H., Knight, R., Chia, N.,
Parkinson’s disease. Mov. Disord. 27 (6), 709–715. Sinha, R., 2017. Comparison of collection methods for fecal samples in microbiome
Shults, C.W., 2006. Lewy bodies. Proc. Natl. Acad. Sci. U. S. A. 103 (6), 1661. studies. Am. J. Epidemiol. 185 (2), 115–123.
Smith, C.J., Kaper, J.B., Mack, D.R., 1995. Intestinal mucin inhibits adhesion of human Westfall, S., Lomis, N., Kahouli, I., Dia, S.Y., Singh, S.P., Prakash, S., 2017. Microbiome,
enteropathogenic Escherichia coli to HEp-2 cells. J. Pediatr. Gastroenterol. Nutr. 21 probiotics and neurodegenerative diseases: deciphering the gut brain axis. Cell. Mol.
(3), 269–276. Life Sci.: CMLS 74 (20), 3769–3787.
Su, X., Maguire-Zeiss, K.A., Giuliano, R., Prifti, L., Venkatesh, K., Federoff, H.J., 2008.

241

You might also like