You are on page 1of 10

Journal of Alzheimer’s Disease 63 (2018) 1337–1346 1337

DOI 10.3233/JAD-180176
IOS Press

Gut Microbiota is Altered in Patients


with Alzheimer’s Disease
Zhen-Qian Zhuanga , Lin-Lin Shena , Wei-Wei Lia , Xue Fub , Fan Zenga , Li Guic , Yang Lüb , Min Caid ,
Chi Zhua , Yin-Ling Tane , Peng Zhengf , Hui-Yun Lia , Jie Zhua , Hua-Dong Zhoua , Xian-Le Bua,∗
and Yan-Jiang Wanga,∗
a Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
b Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
c Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, China
d Department of Neurology, Chongqing General Hospital, Chongqing, China
e Department of Microbiology, Third Military Medical University, Chongqing, China
f Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China

Handling Associate Editor: Jin-Tai Yu

Accepted 26 March 2018

Abstract. Previous studies suggest that gut microbiota is associated with neuropsychiatric disorders, such as Parkinson’s
disease, amyotrophic lateral sclerosis, and depression. However, whether the composition and diversity of gut microbiota is
altered in patients with Alzheimer’s disease (AD) remains largely unknown. In the present study, we collected fecal samples
from 43 AD patients and 43 age- and gender-matched cognitively normal controls. 16S ribosomal RNA sequencing technique
was used to analyze the microbiota composition in feces. The composition of gut microbiota was different between the two
groups. Several bacteria taxa in AD patients were different from those in controls at taxonomic levels, such as Bacteroides,
Actinobacteria, Ruminococcus, Lachnospiraceae, and Selenomonadales. Our findings suggest that gut microbiota is altered
in AD patients and may be involved in the pathogenesis of AD.

Keywords: Alzheimer’s disease, amyloid-␤ peptide, gut microbiota, 16S ribosomal RNA sequencing

INTRODUCTION dementia [3]. Although tremendous efforts have been


made for the treatment of AD, no efficient disease-
Alzheimer’s disease (AD) is the most common modifying therapeutics are available, partially due to
form of dementia, causing a tremendous social and the limited understanding of the disease pathogene-
economic burden [1, 2]. Senile plaques are thought sis [4].
to be the pathological hallmark of AD, consisting of There is a large number of microbiota (most of
amyloid-␤ protein (A␤), which can cause secondary them are bacteria) in gut, which is ten times more
pathological changes such as hyperphosphoryla- than the number of cells in human body [5], and
tion of tau, neuroinflammation, oxidative stress, the total genome of gut microbiota are 150 times
and neurite degeneration, and eventually leading to more than genome of human body [6]. Amount-
ing evidence suggested that gut microbiota plays an
important role in the development of brain. And there
∗ Correspondence
is a bidirectional relationship between the brain, gut,
to: Yan-Jiang Wang and Dr. Xian-Le Bu,
Department of Neurology, Daping Hospital, Third Military Med-
and the microbiota within the gut, which is referred
ical University, Chongqing, China. E-mails: yanjiang wang@ as the microbiota-gut-brain axis [7]. The alterations
tmmu.edu.cn; buxianle@sina.cn. in the gut microbiota composition were proven linked

ISSN 1387-2877/18/$35.00 © 2018 – IOS Press and the authors. All rights reserved
1338 Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease

to a number of neuropsychiatric disorders including (ADL). The subjects who were abnormal in MMSE
depression, amyotrophic lateral sclerosis (ALS) and assessment were further administered with neuropsy-
Parkinson’s disease (PD) [7]. Specifically, gut micro- chological tests, including Clinical Dementia Rating
biota can induce depression-like behavior through (CDR), Fuld Object Memory Evaluation for detecting
altering host metabolism [8]. The proinflammatory extensive cognitive dysfunction mainly composed
gut microbiota dysbiosis in PD patients could trig- of memory, Rapid Verbal Retrieve for detecting
ger inflammation-induced misfolding of ␣-synuclein. the function of semantic memory, Wechsler Adult
AD shared several common properties with other Intelligence Scale (Digit Span and Block Design sub-
neurodegenerative disorders: 1) accumulation of mis- tests) for evaluating immediate memory and function
folded proteins (A␤ and hyperphosphorylated tau); 2) of graphical recognition, Pfeiffer Outpatient Dis-
evidence for a prion-like spread of pathology with ability Questionnaire for assessing ability of social
misfolded proteins; and 3) neuroinflammation [9]. activities, Hamilton Depression Rating Scale for
However, whether gut microbiota participated the measuring emotional status, and Hachinski Ischemic
pathogenesis of AD remains largely unknown. In the Score (HIS) for evaluating significant vascular dis-
present study, we collected the feces from AD patients eases. The diagnosis of AD was made according
and cognitively normal controls and found that the gut to the criteria of the National Institute of Neuro-
microbiota composition in AD patients was different logical and Communicative Diseases and Stroke/AD
from that in normal controls. and Related Disorders Association. These procedures
were administered by trained interviewers who were
MATERIALS AND METHODS experienced neurologists. In addition, twelve people
were administered with A␤ positron emission tomog-
Subjects raphy (PET) inspection with Pittsburg compound B
to detect and quantify A␤ deposition in the brain.
Forty-three AD patients and 43 age- and gender-
matched controls with normal cognition were Fecal sample collection and DNA isolation
recruited from Daping Hospital and Southwest Hos-
pital of Third Military Medical University, First Fecal samples were collected from the recruited
Affiliated Hospital of Chongqing Medical University, AD patients and matched controls in the morning and
and Chongqing People’s Hospital. The subjects were stored at –80◦ C prior to analyses. DNA was extracted
excluded if they had 1) a family history of demen- using the standard Power Soil Kit according to the
tia; 2) any kind of other neurodegenerative disease product instructions [8]. The final DNA concentra-
(e.g., PD, ALS); 3) severe cardiac, pulmonary, hep- tion and purification were determined by NanoDrop
atic, renal diseases, or any kinds of tumor; 4) enduring 2000 UV-vis spectrophotometer (Thermo Scientific,
mental illness (e.g., schizophrenia); 5) a history of Wilmington, USA), and DNA quality was checked by
taking antibiotics within six months, 6) intestinal dis- 1% agarose gel electrophoresis. DNA samples were
eases (e.g., irritable bowel syndrome). This study was stored at –20◦ C until subsequent analyses.
approved by the Institutional Review Board of Dap-
ing Hospital. 16S rRNA gene sequencing and analysis

Clinical assessment and diagnosis of AD The V3-V4 region of the bacteria’s 16S ribosomal
RNA (rRNA) gene was amplified by thermocy-
The clinical assessment and diagnosis of AD cler PCR system (GeneAmp 9700, ABI, USA) with
dementia were performed following a previously barcode-indexed primers (338F and 806R) accord-
used protocol [10, 11]. In brief, the demographic data ing to previous protocols [14]. The PCR reactions
and medical history (such as hypertension, coronary were conducted using the following program: 3 min
heart disease, diabetes mellitus and gastrointestinal of denaturation at 95◦ C, 27 cycles of 30 s for denatu-
diseases) were collected. Cognitive and functional ration at 95◦ C, 30 s for annealing at 55◦ C, and 45 s for
status was assessed based on a neuropsychological elongation at 72◦ C, and a final extension at 72◦ C for
battery as described previously [12]. The cognitive 10 min. PCR reactions were performed in triplicate
and functional status was assessed using the Chinese 20 ␮L mixture containing 4 ␮L of 5 × FastPfu Buffer,
version of the Mini-Mental State Examination 2 ␮L of 2.5 mM dNTPs, 0.8 ␮L of each primer
(MMSE) and instrumental Activities of Daily Living (5 ␮M), 0.4 ␮L of FastPfu Polymerase and 10 ng
Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease 1339

Table 1
Characteristics of study population
Characteristics AD patients (n = 43) Normal controls (n = 43) p
Age, y (SD) 70.12 (8.78) 69.72 (9.24) 0.839
Female, n (%) 20 (46.5) 20 (46.5) 1
Education, y (SD) 9.23 (2.15) 9.81 (2.52) 0.253
Hypertension, n (%) 15 (34.9) 13 (30.2) 0.645
Diabetes mellitus, n (%) 7 (16.3) 5 (11.6) 0.534
Hypercholesterolemia, n (%) 11 (25.5) 10 (23.3) 0.802
Coronary heart disease, n (%) 8 (18.6) 6 (14) 0.559
MMSE scores, mean (SD) 14.70 (4.94) 27.78 (2.54) <0.001
CDR score, mean (SD) 1.67 (0.84) 0.00 (0.00) <0.001
ADL score, mean (SD) 48.63 (18.03) 21.18 (3.17) <0.001
APOE ␧4 carriers, n (%) 17 (39.5) 10 (23.3) 0.009
AD, Alzheimer’s disease; BMI, Body mass index; MMSE, Mini-Mental State Examination; CDR, Clinical
Dementia Rating; ADL, Activities of Daily Living; All data, except for gender, Hypertension, Diabetes melli-
tus, Hypercholesterolemia, Coronary heart disease and APOE genotype, are expressed as means (SD). pvalue,
two-dependent t test or Chi-squared test were as appropriate.

of template DNA. The resulted PCR products were (Beta diversity) were generated based on weighted
extracted from a 2% agarose gel and further purified and non-weighted algorithms and generated prin-
using the AxyPrep DNA Gel Extraction Kit (Axygen cipal coordinate analysis (PCoA) in R software
Biosciences, Union City, CA, USA) and quantified (Version 2.15.3). PLS-DA was performed using
using QuantiFluor™-ST (Promega, USA) according SIMCA-P software to cluster the sample plots
to the manufacturer’s protocol. Purified amplicons across groups. To perform the UniFrac analysis,
were pooled in equimolar and paired-end sequenced representative sequences for each OTU were aligned
(2 × 300) on an Illumina MiSeq platform (Illumina, using PyNAST and a phylogenetic tree from this
San Diego, USA) according to the standard protocols alignment was constructed with Fast Tree. Random
by Majorbio Bio-Pharm Technology Co. Ltd. (Shang- Forest algorithm was carried out to identify the key
hai, China). The raw reads were deposited into the discriminatory OTUs which assigns an importance
NCBI Sequence Read Archive (SRA) database. score to each OTU by estimating the increase in
error caused by removing that OTU from the set
Sequence analysis of predictors. Linear discriminant analysis with
effect size (LEfSe) and phylogenetic investigation
The final 86 samples from AD patients and healthy of communities by reconstruction of unobserved
group were pooled into four libraries according to 16S states (PICRUSt) were performed online (http://
rRNA sequencing data. The 3273141 qualified reads huttenhower.sph.harvard.edu/galaxy/), based on
from 6546282 raw reads were filtered for downstream the taxonomic files obtained from the QIIME
analysis. After filtering, an average of 38060 reads analysis.
per sample was obtained (minimum 30460; maxi-
mum 44928). Then all remaining reads were assigned
to operational taxonomic units (OTUs) with a 97%
threshold of distance-based similarity and classified RESULTS
according to the RDP reference database. Summaries
of the taxonomic distributions of OTUs were con- Subject characteristics
structed with these taxonomies and were used to
calculate the relative abundances of microbiota at The characteristics of AD patients and age and
different taxonomic levels. gender-matched cognitively normal controls are
shown in Table 1. AD patients had higher frequency
Statistical analysis of APOE ␧4 carriers, lower MMSE score, and higher
CDR and ADL score. No significant difference was
Alpha diversity, including the Shannon Index, found in education and comorbidities of hyperten-
observed species, Simpson and Chao1 indices, sion, diabetes mellitus, hypercholesterolemia, and
were investigated in QIIME, and the significance coronary heart disease between AD patients and nor-
was determined using a t test. Distance matrices mal controls.
1340 Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease

Fig. 1. Beta diversity measures between AD and control group. a–d: PLS-DA analysis at the family (a), genus (b), species (c) and OTU (d)
level.

Bacterial community structure in AD and control dominant bacteria (Fig. 3a). A mild decrease was
group observed in the abundance of Bacteroidetes among
AD patients (p = 0.039), whereas Actinobacteria
To examine the bacterial community structure in were slightly more abundant. While Verrucomicrobia
the stool samples, the dataset were analyzed by Par- decreased in AD patients, yet it is not statistically sig-
tial Least Squares Discriminant Analysis (PLSDA). nificant (p = 0.15). Firmicutes were almost the same
Fecal microbial communities separated between AD between the two groups. At the class level, Clostridia,
and control group on family, genus, species and OTU Gammaproteobacteria, Bacteroidia, Bacilli, Neg-
level, suggesting a unique gut microbiota in AD ativicutes and Actinobacteria were the dominant
(Fig. 1). bacteria (Fig. 3b). We found that the relative abun-
The subgroup analysis including AD patient who dance of Actinobacteria and Bacilli increased, while
underwent PET and cognitively normal controls, also that of Negativicutes and Bacteroidia decreased
showed an obvious unique gut microbiota in AD with significantly in the AD group compared with the
PLSDA. In this subgroup analysis, twelve age- and control group. At the order level, Clostridiales,
gender-matched cognitively normal subjects were Enterobacteriales, Bacteroidales, Lactobacillales,
selected as controls. These results were essentially and Selenomonadales were the dominant bacteria
the same as that from the whole subjects (Fig. 2). (Fig. 3c). The relative abundance of Lactobacillales
increased, while Bacteroidales and Selenomonadales
Composition of gut microbiota in AD and control decreased in the AD group compared with the control
groups group.
At the family level, Lachnospiraceae, Ruminococ-
The overall microbial compositions of AD and caceae, Enterobacteriaceae, Bacteroidaceae, Veil-
control groups were examined at different taxonomic lonellaceae, Erysipelotrichaceae, and Enterococ-
levels. At the phylum level, Firmicutes, Proteobac- caceae were the dominant bacteria (Fig. 3d). In AD
teria, Bacteroidetes and Actinobacteria were the patients, the relative abundance of Ruminococcaceae,
Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease 1341

Fig. 2. Beta diversity measures between PET-AD and control group. a–d: PLS-DA analysis at the family (a), genus (b), species (c) and OTU
(d) level.

Fig. 3. Composition of the gut microbiota in AD and control group. Relative abundance of phylum-level (a), class-level (b), order-level
(c) and family-level (d) gut microbial taxa.
1342 Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease

Fig. 4. Differences of bacterial taxa between AD and control group. a) Taxonomic represents statistically differences between AD and
control group. Differences are represented by the color of the most abundant class. The diameter of each circle’s diameter is proportional to
the taxon’s abundance. b) Histogram of the LDA scores for different abundant genera. Positive LDA scores indicate the enrichment of taxa
in AD group (red) relative to control group (green), and negative LDA scores indicate the depletion of taxa in AD group relative to control
group. Box shows statistically significant different bacteria. On the phylum level (c), on the class level (d), on the genus level-selected major
taxa (e), on the species-selected major taxa (f). Each box represents the 25th to 75th percentiles. (Pairwise comparisons using Wilcoxon rank
sum test; ∗ p < 0.05; ∗∗ p < 0.01; ∗∗∗ p < 0.001).
Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease 1343

Enterococcaceae, and Lactobacillaceae increased, Ruminococcaceae, Selenomonadales, and Lachno-


while that of Lanchnospiraceae, Bacteroidaceae, clostridium, may contribute to the differences.
and Veillonellaceae decreased significantly com- The balance of bacteria in the gut plays an
pared with the control group. important role in human health. Increasing evidence
The microbial composition for PET subgroup was showed that gut microbes can influence brain function
shown in Supplementary Figure 1 in the supplemental and behavior via the microbiota-gut-brain axis [16].
material. They were essentially the same with that of Using germ-free animals, antibiotics or probiotics
the overall subjects. intervention and fecal microbiota transplantation sug-
gested a role for the gut microbiota in the regulation
Crucial bacteria associated with AD of mood, anxiety, pain and cognition [17]. The gut
microbiota may communicate with brain through
A representative cladogram of fecal microbial neural, endocrine and immune pathways [17, 18].
structure indicated a significant gut microbial imbal- A number of neuropsychiatric disorders including
ance in AD. We used LEfSe to identify the taxa depression, ALS, and PD have been proved to be
that were significantly different between groups. In related to gut microbiota [7]. Moreover, gut micro-
our study, taxa with an average relative abundance biota was reported to modulate brain inflammatory
over 0.0001 were selected for LEfSe. This threshold pathways via the inflammasome signaling [19]. Some
ensured that meaningful taxa were retained and the probiotics in gut could modulate the expression of
rarest taxa were removed. The characteristics of these specific genes in the brain [20]. Gut microbiota could
comparisons are presented in Fig. 4. The LEfSe for also influence neural development [16]. Moreover,
PET subgroup were shown in Supplementary Fig- recent study indicated that microbial products can be
ure 2 in the supplemental material. translocated into the brain and modulate brain gene
We further analyzed the variant taxa that occupy expression [21].
high abundance at different taxonomic levels. At the Emerging evidence suggested a link between gut
phylum level, we found differences between groups microbiota, cognition, and AD. Previous animal stud-
in Bacteroidetes and Actinobacteria. The subspecies ies show that Bifidobacteria has a positive impact on
of Bacteroidetes in other taxonomic levels were also cognition of an anxious mouse strain after daily feed
different. Although Firmicutes were almost the same for 11 weeks [22], and the composition and diversity
between the two groups, there still was a difference of gut microbiota were changed in AD transgenic
in Firmicutes/Bacteroidetes ratio, which was reported mice compared with wild-type mice [23]. A recent
increased in subjects with obesity [15]. study showed that gut microbiota was altered in AD
At the class level, the results were almost the same patients [24]. The present study further provided the
as phylum level, but we found a significant decrease human evidence that the gut microbiota composition
of Negativicutes in AD group in comparison to nor- was altered in AD patients, especially in PiB-PET
mal controls. positive AD patients. It is worthy of noting that the
At the family level, while Lachnospiraceae detailed changes of the gut microbiota of AD patients
was decreased in AD group, there was an in this study is different from ours. This discrep-
increase in Ruminococcaceae. Lachnospiraceae, and ancy might be due to the sample size, population,
Ruminococcaceae are two high abundance taxa lifestyle, dietary habits, RNA sequencing area, and
in Clostridiales, and are two important digestive comorbidity burden [25], which is associated with gut
bacteria in human body. Additionally, the change microbiota, between the two studies. However, the
of Lachnoclostridium and Subdoligranulum, which underlying mechanisms remained poorly understood.
belonged to their subspecies respectively at the genus The gut microbiota emerged as an important reg-
level, showed the similar results. ulator of host immune function and metabolism.
Changes in the composition of the gut microbiota
could increase intestinal permeability and induce
DISCUSSION inflammation that has been proved to increase the
risk of AD [17]. In this study, we found a lower
In the present study, the gut microbiota composi- Bacteroides abundance in AD patients, which is
tion and diversity were found altered in AD patients consistent with a previous study showing that Bac-
compared with cognitively normal controls. Several teroides fragilis was lower in patients with cognitive
bacterial taxa, such as Actinobacteria, Bacteroidales, impairment and brain amyloidosis [26]. However, the
1344 Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease

Bacteroides fragilis was not detected in this study, erate amyloid peptides [41, 42]. As to our finding
partially due to its low abundance which it is esti- that peripheral A␤ can enter brain and cause AD-
mated as only 0.5% of the Bacteroides species in the type pathologies [43], gut microbiota might directly
gut and make it technically difficult to be detected contribute to brain A␤ deposition with bacterial
[27]. Bacteroides fragilis can be classified into two amyloids.
strains: non-toxigenic Bacteroides fragilis (NTBF) Food style and comorbidities such as hypertension,
and enterotoxigenic Bacteroides fragilis (ETBF). hyperlipidemia, and diabetes mellitus may influence
ETBF, a kind of anti-inflammatory bacteria, has the the composition of gut microbiota. In the present
ability to strengthen intestinal barrier and reverse study, all AD patients and control subjects were
gut leakiness [28]. On the other hand, Ruminococ- recruited from same area and shared similar food
cus species (increased in AD patients in the present habit. In addition, there were no difference on the inci-
study), a Gram-positive bacteria, need fermentable dence of hypertension, hyperlipidemia, and diabetes
carbohydrates for growth, and can degrades mucus mellitus between AD patients and controls. Thus, the
by expressing intramolecular trans-sialidases [29]. It alteration of gut microbiota in AD patients may not be
is worthy of noting that bacterial components were resulted from the differences in food style and comor-
found in the brain of AD patients [30, 31]. It is bidities. A major limitation of the present study is that
intriguing to speculate that gut leakiness due to gut our study is a cross-sectional study. We cannot draw
microbiota alteration may facilitate the entrance of conclusion about the causal relationship between the
gut bacteria and their components into the body. alteration of gut microbiota and AD. Longitudinal
It has been proved that some metabolites of studies are needed to better clarify the impact of gut
gut microbiota can induce neuroinflammation [32]. microbiota on AD in the future.
Recent study showed that changing the gut microbial In summary, our study revealed the alternation of
composition and diversity of APP/PS1 mice by long- gut microbiota composition in AD patients. Modu-
term broad-spectrum antibiotics could decrease brain lation of gut microbiota through personalized diet or
A␤ burden via regulating host innate immunity [33, beneficial microbiota intervention may be a poten-
34]. We previously found that AD is associated with tial strategy for the prevention treatment of AD. Our
infectious burden [12]. Infection with pathogenic study also suggests that AD might not be a disease of
microbes like Helicobacter pylori may exacerbate brain itself, and brain health is closely associated with
brain A␤ production and tau phosphorylation and our whole body. We need to understand the disease
increase the risk of AD [35, 36]. pathogenesis and develop therapies systemically for
Gut microbiota play important roles in the synthe- AD and other neurodegenerative diseases [44, 45].
sis of brain neurotransmitters such as ␥-aminobutyric
acid (GABA), serotonin, and brain-derived neu-
ACKNOWLEDGMENTS
rotrophic factor which are all related with AD [37].
Some gut microbes can also produce metabolites such
This study was supported by National Natural
as neurotransmitters, neuropeptides, endocrine hor-
Science Foundation of China (NSFC) Grant No.
mones, and immunomodulators [38]. Ruminococcus
91749206, 81701043 and 81625007.
in gut, which was increased in AD patients of the
Authors’ disclosures available online (https://
present study, was suggested to predict lower brain
www.j-alz.com/manuscript-disclosures/18-0176r1).
N-acetylaspartate, a biochemical indicator of neu-
ronal health and is decreased in neuron metabolic
disturbance [39]. The increase of Ruminococcus SUPPLEMENTARY MATERIAL
species in AD patients provided a proof of interac-
tion between gut microbiota and brain metabolites. The supplementary material is available in the
Metabolic diseases including obesity, insulin resis- electronic version of this article: http://dx.doi.
tance, hypertension, and hyperlipidemia are related org/10.3233/JAD-180176.
to AD development [40]. Meanwhile, accumulating
evidence indicated a link between gut microbiota
REFERENCES
and metabolic disease [38]. So the alternations of
gut microbiota might contribute to AD pathogene-
[1] Masters CL, Bateman R, Blennow K, Rowe CC, Sperling
sis through metabolic pathways. Additionally, a large RA, Cummings JL (2015) Alzheimer’s disease. Nat Rev Dis
amount of bacteria in human intestine could gen- Primers 1, 15056.
Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease 1345

[2] Lim SL, Rodriguez-Ortiz CJ, Kitazawa M (2015) Infection, SL, Rogers GB, Licinio J (2016) Inflammasome signal-
systemic inflammation, and Alzheimer’s disease. Microbes ing affects anxiety- and depressive-like behavior and gut
Infect 17, 549-556. microbiome composition. Mol Psychiatry 21, 797-805.
[3] Hardy J, Selkoe DJ (2002) The amyloid hypothesis of [20] Distrutti E, O’Reilly JA, McDonald C, Cipriani S, Renga
Alzheimer’s disease: Progress and problems on the road to B, Lynch MA, Fiorucci S (2014) Modulation of intestinal
therapeutics. Science 297, 353-356. microbiota by the probiotic VSL#3 resets brain gene expres-
[4] Klein HU, Bennett DA, De Jager PL (2016) The epigenome sion and ameliorates the age-related deficit in LTP. PLoS
in Alzheimer’s disease: Current state and approaches for One 9, e106503.
a new path to gene discovery and understanding disease [21] Arentsen T, Qian Y, Gkotzis S, Femenia T, Wang T,
mechanism. Acta Neuropathol 132, 503-514. Udekwu K, Forssberg H, Diaz Heijtz R (2017) The bacterial
[5] Parashar A, Udayabanu M (2017) Gut microbiota: Implica- peptidoglycan-sensing molecule Pglyrp2 modulates brain
tions in Parkinson’s disease. Parkinsonism Relat Disord 38, development and behavior. Mol Psychiatry 22, 257-266.
1-7. [22] Savignac HM, Tramullas M, Kiely B, Dinan TG, Cryan JF
[6] Jin Y, Wu S, Zeng Z, Fu Z (2017) Effects of environmental (2015) Bifidobacteria modulate cognitive processes in an
pollutants on gut microbiota. Environ Pollut 222, 1-9. anxious mouse strain. Behav Brain Res 287, 59-72.
[7] Rieder R, Wisniewski PJ, Alderman BL, Campbell SC [23] Zhang L, Wang Y, Xiayu X, Shi C, Chen W, Song N, Fu
(2017) Microbes and mental health: A review. Brain Behav X, Zhou R, Xu YF, Huang L, Zhu H, Han Y, Qin C (2017)
Immun 66, 9-17. Altered gut microbiota in a mouse model of Alzheimer’s
[8] Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L, disease. J Alzheimers Dis 60, 1241-1257.
Chen J, Fan S, Du X, Zhang X, Yang D, Yang Y, Meng [24] Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Mer-
H, Li W, Melgiri ND, Licinio J, Wei H, Xie P (2016) Gut luzzi AP, Johnson SC, Carlsson CM, Asthana S, Zetterberg
microbiome remodeling induces depressive-like behaviors H, Blennow K, Bendlin BB, Rey FE (2017) Gut microbiome
through a pathway mediated by the host’s metabolism. Mol alterations in Alzheimer’s disease. Sci Rep 7, 13537.
Psychiatry 21, 786-796. [25] Wang QH, Wang X, Bu XL, Lian Y, Xiang Y, Luo HB,
[9] Ahmad K (2013) Identification of common target proteins Zou HQ, Pu J, Zhou ZH, Cui XP, Wang QS, Shi XQ, Han
for multiple neurodegenerative disorders and reconstruction W, Wu Q, Chen HS, Lin H, Gao CY, Zhang LL, Xu ZQ,
of disease pathway. Physical Review B 67, 1393-1406. Zhang M, Zhou HD, Wang YJ (2017) Comorbidity bur-
[10] Wang JY, Zhou DH, Li J, Zhang M, Deng J, Tang M, Gao C, den of dementia: A hospital-based retrospective study from
Li J, Lian Y, Chen M (2006) Leisure activity and risk of cog- 2003 to 2012 in seven cities in China. Neurosci Bull 33,
nitive impairment: The Chongqing aging study. Neurology 703-710.
66, 911-913. [26] Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N,
[11] Li J, Wang YJ, Zhang M, Xu ZQ, Gao CY, Fang CQ, Yan JC, Festari C, Ferrari C, Guerra UP, Paghera B, Muscio C,
Zhou HD, Chongqing Ageing Study Group (2011) Vascular Bianchetti A, Volta GD, Turla M, Cotelli MS, Gennuso
risk factors promote conversion from mild cognitive impair- M, Prelle A, Zanetti O, Lussignoli G, Mirabile D, Bellandi
ment to Alzheimer disease. Neurology 76, 1485-1491. D, Gentile S, Belotti G, Villani D, Harach T, Bolmont T,
[12] Bu XL, Yao XQ, Jiao SS, Zeng F, Liu YH, Xiang Y, Liang Padovani A, Boccardi M, Frisoni GB, Group I-F (2017)
CR, Wang QH, Wang X, Cao HY, Yi X, Deng B, Liu CH, Xu Association of brain amyloidosis with pro-inflammatory
J, Zhang LL, Gao CY, Xu ZQ, Zhang M, Wang L, Tan XL, gut bacterial taxa and peripheral inflammation markers in
Xu X, Zhou HD, Wang YJ (2015) A study on the association cognitively impaired elderly. Neurobiol Aging 49, 60-68.
between infectious burden and Alzheimer’s disease. Eur J [27] Polk BF, Kasper DL (1977) Bacteroides fragilis subspecies
Neurol 22, 1519-1525. in clinical isolates. Ann Intern Med 86, 569-571.
[13] Adetutu EM, Thorpe K, Bourne S, Cao X, Shahsavari E, [28] Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER,
Kirby G, Ball AS (2011) Phylogenetic diversity of fun- McCue T, Codelli JA, Chow J, Reisman SE, Petrosino JF,
gal communities in areas accessible and not accessible to Patterson PH, Mazmanian SK (2013) Microbiota modulate
tourists in Naracoorte Caves. Mycologia 103, 959-968. behavioral and physiological abnormalities associated with
[14] Andersson AF, Lindberg M, Jakobsson H, Backhed F, Nyren neurodevelopmental disorders. Cell 155, 1451-1463.
P, Engstrand L (2008) Comparative analysis of human [29] Hynonen U, Rasinkangas P, Satokari R, Paulin L, de Vos
gut microbiota by barcoded pyrosequencing. PLoS One 3, WM, Pietila TE, Kant R, Palva A (2016) Isolation and
e2836. whole genome sequencing of a Ruminococcus-like bac-
[15] Hou YP, He QQ, Ouyang HM, Peng HS, Wang Q, Li J, Lv terium, associated with irritable bowel syndrome. Anaerobe
XF, Zheng YN, Li SC, Liu HL, Yin AH (2017) Human gut 39, 60-67.
microbiota associated with obesity in Chinese children and [30] Zhan X, Stamova B, Jin LW, DeCarli C, Phinney B, Sharp
adolescents. Biomed Res Int 2017, 7585989. FR (2016) Gram-negative bacterial molecules associate
[16] Rogers GB, Keating DJ, Young RL, Wong ML, Licinio J, with Alzheimer disease pathology. Neurology 87, 2324-
Wesselingh S (2016) From gut dysbiosis to altered brain 2332.
function and mental illness: Mechanisms and pathways. Mol [31] Emery DC, Shoemark DK, Batstone TE, Waterfall CM,
Psychiatry 21, 738-748. Coghill JA, Cerajewska TL, Davies M, West NX, Allen
[17] Cryan JF, Dinan TG (2012) Mind-altering microorganisms: SJ (2017) 16S rRNA next generation sequencing analy-
The impact of the gut microbiota on brain and behaviour. sis shows bacteria in Alzheimer’s post-mortem brain. Front
Nat Rev Neurosci 13, 701-712. Aging Neurosci 9, 195.
[18] Fung TC, Olson CA, Hsiao EY (2017) Interactions between [32] Lukiw WJ (2016) Bacteroides fragilis lipopolysaccharide
the microbiota, immune and nervous systems in health and and inflammatory signaling in Alzheimer’s disease. Front
disease. Nat Neurosci 20, 145-155. Microbiol 7, 1544.
[19] Wong ML, Inserra A, Lewis MD, Mastronardi CA, Leong [33] Minter MR, Zhang C, Leone V, Ringus DL, Zhang X,
L, Choo J, Kentish S, Xie P, Morrison M, Wesselingh Oyler-Castrillo P, Musch MW, Liao F, Ward JF, Holtzman
1346 Z.-Q. Zhuang et al. / Gut Microbiota and Alzheimer’s Disease

DM, Chang EB, Tanzi RE, Sisodia SS (2016) Antibiotic- [40] Cai H, Cong WN, Ji S, Rothman S, Maudsley S, Martin B
induced perturbations in gut microbial diversity influences (2012) Metabolic dysfunction in Alzheimer’s disease and
neuro-inflammation and amyloidosis in a murine model of related neurodegenerative disorders. Curr Alzheimer Res 9,
Alzheimer’s disease. Sci Rep 6, 30028. 5-17.
[34] Minter MR, Hinterleitner R, Meisel M, Zhang C, Leone [41] Chapman MR, Robinson LS, Pinkner JS, Roth R, Heuser
V, Zhang X, Oyler-Castrillo P, Zhang X, Musch MW, J, Hammar M, Normark S, Hultgren SJ (2002) Role of
Shen X, Jabri B, Chang EB, Tanzi RE, Sisodia SS (2017) Escherichia coli curli operons in directing amyloid fiber
Antibiotic-induced perturbations in microbial diversity dur- formation. Science 295, 851-855.
ing post-natal development alters amyloid pathology in an [42] Zhao Y, Dua P, Lukiw WJ (2015) Microbial sources of
aged APPSWE/PS1DeltaE9 murine model of Alzheimer’s amyloid and relevance to amyloidogenesis and Alzheimer’s
disease. Sci Rep 7, 10411. Didease (AD). J Alzheimers Dis Parkinsonism 5, 177.
[35] Wang XL, Zeng J, Feng J, Tian YT, Liu YJ, Qiu M, Yan [43] Bu XL, Xiang Y, Jin WS, Wang J, Shen LL, Huang ZL,
X, Yang Y, Xiong Y, Zhang ZH, Wang Q, Wang JZ, Liu Zhang K, Liu YH, Zeng F, Liu JH, Sun HL, Zhuang ZQ,
R (2014) Helicobacter pylori filtrate impairs spatial learn- Chen SH, Yao XQ, Giunta B, Shan YC, Tan J, Chen XW,
ing and memory in rats and increases beta-amyloid by Dong ZF, Zhou HD, Zhou XF, Song W, Wang YJ (2017)
enhancing expression of presenilin-2. Front Aging Neurosci Blood-derived amyloid-beta protein induces Alzheimer’s
6, 66. disease pathologies. Mol Psychiatry, doi: 10.1038/mp.
[36] Wang XL, Zeng J, Yang Y, Xiong Y, Zhang ZH, Qiu M, Yan 2017.204
X, Sun XY, Tuo QZ, Liu R, Wang JZ (2015) Helicobacter [44] Wang J, Gu BJ, Masters CL, Wang YJ (2017) A systemic
pylori filtrate induces Alzheimer-like tau hyperphospho- view of Alzheimer disease - insights from amyloid-beta
rylation by activating glycogen synthase kinase-3beta. metabolism beyond the brain. Nat Rev Neurol 13, 612-623.
J Alzheimers Dis 43, 153-165. [45] Lionnet A, Leclair-Visonneau L, Neunlist M, Murayama S,
[37] Hu X, Wang T, Jin F (2016) Alzheimer’s disease and gut Takao M, Adler CH, Derkinderen P, Beach TG (2018) Does
microbiota. Sci China Life Sci 59, 1006-1023. Parkinson’s disease start in the gut? Acta Neuropathol 135,
[38] Alkasir R, Li J, Li X, Jin M, Zhu B (2017) Human gut 1-12.
microbiota: The links with dementia development. Protein
Cell 8, 90-102.
[39] Mudd AT, Berding K, Wang M, Donovan SM, Dilger RN
(2017) Serum cortisol mediates the relationship between
fecal Ruminococcus and brain N-acetylaspartate in the
young pig. Gut Microbes 8, 589-600.

You might also like