Showing content from https://pmc.ncbi.nlm.nih.gov/articles/PMC7430034/ below:
The Microbiome as a Modifier of Neurodegenerative Disease Risk
. Author manuscript; available in PMC: 2021 Aug 12.
Abstract
The gut microbiome is increasingly implicated in modifying susceptibility to and progression of neurodegenerative diseases (NDs). In this review, we discuss roles for the microbiome in aging and in NDs. In particular, we summarize findings from human studies on microbiome alterations in Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis and Huntington’s disease. We assess animal studies of genetic and environmental models for NDs that investigate how manipulations of the microbiome causally impact the development of behavioral and neuropathological endophenotypes of disease. We additionally evaluate the likely immunological, neuronal and metabolic mechanisms for how the gut microbiota may modulate risk for NDs. Finally, we speculate on cross-cutting features for microbial influences across multiple NDs and consider the potential for microbiome-targeted interventions for NDs.
In Brief
In this Review, Fang and colleagues discuss emerging interest in the microbiome as a modulator of risk for neurodegenerative diseases. They summarize microbiome alterations reported in patients and animal models and discuss microbial influences on neuroimmunity, amyloid aggregation and neuronal activity in Parkinson’s disease, Alzheimer’s disease and amyotrophic lateral sclerosis.
Introduction
Neurodegenerative diseases (NDs) are characterized by progressive functional loss of neurons in the central nervous system (CNS) or peripheral nervous system (PNS), which leads to long-term motor and/or cognitive impairments. While genetic susceptibilities are major risk factors for developing NDs, environmental factors experienced throughout life also impact the onset, progression and severity of NDs. For example, Parkinson’s disease (PD) has been positively associated with pesticide exposure (Menegon et al., 1998) and gastrointestinal infection (Fasano et al., 2015), and Alzheimer’s disease (AD) has been increasingly associated with herpes simplex virus (HSV) (Itzhaki, 2017). Exactly how genetic and environmental risk factors are conveyed to together predispose to NDs remains poorly understood. However, emerging evidence suggests that genetically and environmentally-induced alterations in the gut microbiome may contribute in part to ND risk.
The gastrointestinal tract harbors trillions of microorganisms, collectively called the gut microbiota, that is shaped by both host genetics and environmental exposures (Lozupone et al., 2012), and that profoundly modulates the development and function of the CNS and PNS across model organisms (Fung et al., 2017). Relevant to aging-related disorders, like some NDs, the composition and activity of the gut microbiota changes over the lifespan, and manipulating the gut microbiota in animal models alters aging-associated immune function, metabolic activity and ultimately longevity (Kundu et al., 2017). Additional environmental risk factors for NDs, such as diet, chemical exposures and infection, also shape the gut microbiota in humans and animal models (Sherwin et al., 2018). Consistent with this, an increasing number of studies report changes in the microbiota in individuals with PD, AD, amyotrophic lateral sclerosis (ALS) and Huntington’s disease (HD) (Table 1), as well as in a subset of animal models for these disorders (Table 2). Across animal models for NDs, disruptions in the gut microbiome can positively or negatively impact the manifestation of neuropathological and behavioral endophenotypes of disease. While the precise mechanisms underlying these phenomena remain unclear, increasing evidence supports the notion of a “microbiota-gut-brain axis”, whereby intestinal microorganisms influence the central nervous system by modulation of neuroimmune function, sensory neuronal signaling and metabolic activity. Altogether, these findings raise timely interest in how the microbiome is shaped by risk factors for NDs, how functional activities of the microbiome impact the nervous system and how microbiome-gut-nervous system interactions may predispose to NDs.
Table 1.
Microbiome alterations in clinical cohorts for NDs
Parkinson’s disease: Microbiota alterations in humans Subject Microbiota Alterations Sample Other Variables Method Reference Human, PD Patients Increased: Bifidobacterium. Decreased: Prevotellaceae, Roseburia n = 64, feces Sex matched
Age matched
Diet: no difference
Medication: variable
16S rRNA gene sequencing (Aho et al., 2019) Human, L-Dopa Naïve PD Patients Family/Genus Increased: Verrumicrobiaceae (genus Akkermansia), unclassified Bacteria, Firmicutes
Family/Genus Decreased: Prevotellaceae (Prevotella), Erysipelotrichaceae (Eubacterium)
Species Increased: Akkermansia muciniphila, Alistipes shahii
Species Decreased: Prevotella copri, Eubacterium bioforme, Clostridium saccharolyticum n = 31, feces Males, age matched
Diet: no difference
Medication: no L-DOPA therapy
Shotgun metagenomic sequencing (Bedarf et al., 2017) Human, PD Patients Increased: Lactobacillus
Decreased: Clostridium coccoides, Bacteroides fragilis n = 52, feces Sex unmatched
Age matched
Diet: no difference
Medication: unknown
16S rRNA gene qPCR (Hasegawa et al., 2015) Human, PD Patients Increased Akkermansia n = 76, feces Sex unmatched
Age matched
Diet: unknown
Medication: variable
16S/18S rRNA gene sequencing (Heintz-Buschart et al., 2018) Human, PD Patients Family Increased: Bifidobacteriaceae, Lactobacillaceae, Tissierellaceae, Christensenellaceae, Verrucomicrobiaceae
Family Decreased: Lachnospiraceae, Pasteurellaceae
Genus Increased: Akkermansia, Christensenellaceae, Lactobacillus, Bifidobacterium
Genus Decreased: Blautia, Roseburia, Lachnospiraceae, Faecalibacterium n = 197, feces Sex adjusted
Age adjusted
Diet adjusted
Medication adjusted
16S rRNA gene sequencing (Hill-Burns et al., 2017) Human, PD Patients Increased: Lactobacillaceae, Barnesiellaceae, Enterococcaceae n = 29, feces Sex unmatched
Age matched
Diet: no difference
Medication: unknown
16S rRNA gene sequencing (Hopfner et al., 2017) Human, PD Patients Mucosal Genus Increased: Faecalibacterium, Ralstonia
Fecal Genus Increased: Blautia, Coprococcus, Roseburia. n = 38, sigmoid mucosal biopsies, feces Sex matched
Age: PD patients are older than HC
Diet: no difference
Medication: variable
16S rRNA gene sequencing (Keshavarzian et al., 2015) Human, PD Patients Increased: Escherichia-Shigella, Streptococcus, Proteus, Enterococcus
Decreased: Blautia, Faecalibacterium, Ruminococcus n = 24, feces Sex matched
Age matched
Diet: unknown
Medication: unknown
16S rRNA gene sequencing (Li et al., 2017) Human, PD Patients Increased: Ruminococcaceae, Verrucomicrobiaceae, Porphyromondaceae, Hydrogenoanaerobacterium, Lachnospiraceae
Decreased: Bacteroides, Prevotellaceae n = 10, feces Sex unmatched
Age matched
Diet: unknown
Medication: unknown
16S rRNA gene sequencing (Li et al., 2019) Human, PD Patients Increased: Eubacteriaceae, Bifidobacteriaceae, Aerococcaceae, Desulfovibrionaceae
Decreased: Streptococcaceae, Methylobacteriaceae, Comamonadaceae, Halmonadaceae, Brucellaceae, Xanthomonadaceae, Lachnospiraceae, Actinomycetaceae, Aphingomonadaceae, Pasturellaceae, Micrococcaceae, Intrasporangiacea, Methanobacteriacea, Idomarinaceae, Brevibacteriaceae, Gemellaceae n = 75, feces Sex matched
Age matched
Diet: no influence on results
Medication: variable
16S rRNA gene sequencing (Lin et al., 2018) Human, PD Patients Genus Increased: Christensenella, Catabacter, Lactobacillus, Oscillospira, Bifidobacterium
Genus Decreased: Dorea, Bacteroides, Prevotella, Faecalibacterium
Species Increased: Christensenella minuta, Catabacter hongkongensis, Lactobacillus mucosae, Ruminococcus bromii Palpillibacter cinnamivorans
Species Decreased: Bacteroides massiliensis, Stoquefichus massiliensis, Bacteroides coprocola, Blautia glucerasea, Dorea longicatena, Bacteroides dorei, Bacteroides plebeus, Prevotella copri, Coprococcus eutactus, Ruminococcus callidus n = 89, feces Sex unknown
Age matched
Diet unknown
Medication unknown
16S rRNA gene sequencing (Petrov et al., 2017) Human, PD Patients Increased: Clostridium IV, Aquabacterium, Holdemania, Sphingomonas, Clostridium XVII, Butyricicoccus, Anaetroncus n = 45, feces Sex matched
Age matched
Diet matched
Medication: antiparkinsonia n medications
16S rRNA gene sequencing (Qian et al., 2018) Human, PD Patients Family Increased: Enterobacteriaceae
Phylum Decreased: Bacteroidetes
Family Decreased: Prevotellaceae n = 43, feces Sex unmatched
Age matched
Diet: no difference
Medication: no dopaminergic drugs
16s RT-qPCR (Unger et al., 2016) Human, PD Patients Increased: Akkermansia, Bifidobacterium (trending),
Decreased: Prevotella (trending) n = 9, feces Sex unknown
Age: 35-95
Diet: unknown
Medication: unknown
16S rRNA gene sequencing (Vidal-Martinez et al., 2020) Human, PD Patients with normal cognition (NC) or mild cognitive impairment (MCI) PD-NC vs. PD-MCI and HC:
Increased: Blautia, Ruminococcus
PD-MCI vs PD-NC and HC:
Increased: Alistipes, Barnesiella, Butyricmonas, Odoribacter, Anaerotruncus n = 13 (MCI),
n = 14 (NC), feces
Sex adjusted
Age adjusted
Diet: no restriction
Medication: unknown
16s rRNA gene sequencing (Ren et al., 2020) Alzheimer’s disease: Microbiota alterations in humans Subject Microbiota Alterations Sample Other Variables Method Reference Human, AD Patients Phylum level: mild decrease in Bacteriodetes decrease in Verrucomicrobia
Class level: increase in Actinobacteria and Bacilli decrease in Negativicutes and Bacteroidia
Order level: increase in Lactobacillales decrease in Bacteroidales and Selenomonadales Family level: increase in Ruminococcaceae, Enterococcaceae, and Lactobacillaceae decrease in Lanchnospiracea, Bacteroidaceae and Veillonellaceae n=43, feces Sex matched
Age matched
Diet: unknown
Medication: unknown
16S rRNA gene sequencing (Zhuang et al., 2018) Human, AD Patients Phylum level: increase in Bacteriodetes decrease in Firmicutes and Actinobacteria
Family level: increase in Gemellaceae, Bacteroidaceae and Rikenellaceae decrease in Bifidobacteriaceae, Ruminococcaceae, Turicibacteraceae, Peptostreptococcaceae, Clostridiaceae, and Mogibacteriaceae
Genus level: increase in Bacteroides, Alistipes, Bilophila, Blautia, Phascolarctobacterium and Gemella
decrease in SMB53, Dialister, Clostridium, Turicibacter, cc115, Bifidobacterium and Adlercreutzia n=25, feces Sex matched
Age matched
Diet: no difference
Medication: variable
16S rRNA gene sequencing (Vogt et al., 2017) Amyotrophic lateral sclerosis: Microbiome alterations in humans Subject Microbiome Alterations Sample Other Variables Method Reference Human, ALS Patients Increased Methanobrevibacter
Decreased Faecalibacterium and Bacteroides n=8, feces Sex matched
Age matched
Diet: no difference
Medication: unknown
16S rRNA gene sequencing (Zhai et al., 2019) Human, ALS Patients No significant alterations n=25-32, feces Sex matched
Age matched
Diet unknown
Medication unknown
16S rRNA gene sequencing (Brenner et al., 2018) Human, ALS Patients Increased Dorea
Decreased Oscillibacter, Anaerostipes, Lachnospiraceae n=5-6, feces Sex unknown
Age unknown
Diet unknown
Medication unknown
16S rRNA gene sequencing (Fang et al., 2016) Human, ALS Patients Marginally increased Anaerostipes hadrus and Bacteroidales bacterium
Marginally decreased Bifidobacterium pseudocatenulatum, Clostridium leptum and Escherichia coli n=29-37, feces Sex mixed
Age matched
Diet unknown
Medication: no constipation medication
Shotgun metagenomic sequencing (Blacher et al., 2019) Table 2.
Microbiome alterations in animal models for NDs
Parkinson’s disease: Microbiota alterations in animal models Subject Microbiota Alterations Sample Method Reference Mice, Thy1-A30P:SNCA Decreased: Verrumicrobiae n=26, feces 16S rRNA gene sequencing (Gorecki et al., 2019) Mice; Rotenone No observed differences with FDR-P. With less stringent analysis,
Decreased: Actinobacteria
n = 14, feces 16S rRNA gene sequencing (Dodiya et al., 2020) Rats, Rotenone (RT-qPCR) Increased: Bifidobacterium (Sequencing, SI) Phylum Increased: Actinobacteria, Proteobacteria
(Sequencing, SI) Phylum Decreased: Bacteroidetes, Cyanobacteria, Firmicutes
(Sequencing, colon) Phylum Decreased: Cyanobacteria, Firmicutes
(Sequencing, SI/colon) Family Increased: Bifidobacteriaceae, Alcaligenaceae, Clostridiaceae
(Sequencing, SI/colon) Family Decreased: S24-7, Prevotellaceae, Paraprevotellaceae, Lachnospiraceae
(Sequencing, SI/Colon) Genus Increased: Bifidobacterium, Lactobacillus, Turicibacter, Sutterella
(Sequencing, SI/Colon) Genus Decreaased: Prevotella, S24-7, Oscillospira n=12, distal small intestine and colon contents qPCR,
shotgun
metagenomic
sequencing (Johnson et al., 2018) Mice, MPTP Phylum Decreased: Proteobacteria (2 days and 3 weeks following treatment)
Order Increased: Erysipelotrichales (3 weeks following treatment)
Order Decreased: Clostridiales (3 weeks following treatment)
Family Increased: Prevotellaceae (2 days following treatment), Erysipelotrichaceae (3 weeks following treatment)
Family Decreased: Lachnospiraceae (2 days and 3 weeks following treatment) n = 10, feces 16S rRNA gene sequencing (Lai et al., 2018) Mice, MPTP Mucosa Phylum Increased: Bacteroidetes, Firmicutes
Mucosa Phylum Decreased: Actinobacteria
Lumenal Content Phylum Increased: Proteobacteria
Lumenal Content Phylum Decreased: Actinobacteria
Mucosa and Lumenal Content Family Increased: Rikenellaceae, S24-7, Clostridiales (unclassified), Ruminococcaceae
Mucosa and Lumenal Content Family Decreased: Bifidobacterium
Mucosa Genus Increased: Allobaculum n = 9-10, cecum mucosal-associated and lumenal contents 16S rRNA gene sequencing (Perez-Pardo et al., 2018) Mice, MPTP Phylum Increased: Proteobacteria
Phylum Decreased: Firmicutes
Order Increased: Turicibacterales, Enterobacteriales
Order Decreased: Clostridiales n = 7, feces 16S rRNA gene sequencing (Sun et al., 2018) Mice, Rotenone, within-comparison over 4 weeks Phylum Increased: Firmicutes (3w, 4w of treatment)
Phylum Decreased: Bacteroidetes(3w, 4w of treatment)
Genus Increased: Lactobacillus (3 and 4w of treatment)
Genus Decreased: Clostridium (1-4w of treatment), Sutterella (1-4w of treatment), Lactococcus(1-3w of treatment), Desulfovibrio (2-4w of treatment), Aldercreutzia (2w of treatment), Paraprevotella (1 and 3w of treatment) n = 5-8, feces 16S rRNA gene sequencing (Yang et al., 2017) Alzheimer’s disease: Microbiota alterations in animal models Subject Microbiota Alterations Sample Method Reference Mice, Tau P301L Phylum level: increase in Bacteroidetes decrease in Firmicutes, Actinobacteria and Tenericutes
Genus level: increase in
Ruminococcaceae NK4A214_group, Bacteroides, Anaeroplasma,
norank_f_Clostridiales_vadinBB60_group, norank_f_Bacteroidales_24_7_group, Phascolarctobacterium, Parabacteroides, and unclassified_o_Bacteroidales, Anaerovorax, Peptococcus, Caproiciproducens, Ruminoclostridium, and Oscillibacter decrease in Lactobacillus, Enterorhabdus, Staphylococcus, Bifidobacterium, Gemella, Roseburia, norank_f_Mycoplasmataceae, Lachnoclostridium, Klebsiella, and Streptococcus n=32, feces 16S rRNA gene sequencing (Sun, B.L. et al., 2019) Mice, APP/PS1 Phylum level: increase in Proteobacteria and Verrucomicrobia
Genus level: decrease in Ruminococcus and Butricicocus n=24, feces 16S rRNA gene sequencing (Zhang, L. et al., 2017) Mice, 5xFAD Phylum level: increase in Firmicutes decrease in Bacteriodetes n=18, feces 16S rRNA gene qPCR (Brandsche id et al., 2017) Amyotrophic lateral sclerosis: Microbiome alterations in animal models Subject Microbiome Alterations Sample Method Reference Mice, SOD1G93A Decreased Butyrivibrio Fibrisolvens n=3, feces 16S rRNA gene sequencing (Zhang, Y.G. et al., 2017) Mice, SOD1G93A Decreased Akkermansia muciniphila, Parabaceroides distasonis, Rikenellaceae, Prevotella, Lactobacillus murinus, Alistipes unclassified, Eggertella unclassified,
Increased Sutterella, Allobaculum, Desulfovibrionaceae, Coprococcus, Oscillospira, Bifidobacterium, Helicobacter hepaticus, Lactobacillus johnsonii and Lactobacillus reuteri n=6, feces 16S rRNA gene sequencing, Shotgun metagenomic sequencing (Blacher et al., 2019) Huntington’s disease: Microbiota alterations in animal models Subject Microbiota Alterations Sample Method Reference Mice, R6/1 Sex difference in the bacterial signature of R6/1 mice.
Male: an increase in Bacteroidales, Lactobacillales and a decrease in Clostridiales
Female: an increase in Coriobacteriales, Erysipelotrichales, Bacteroidales, Burkholderiale and a decrease in Clostridiales n=7-8, feces 16S rRNA gene sequencing (Kong et al., 2018) The Gut Microbiome on Aging and Longevity
Preceding interest in the microbiome and aging-related NDs were several pioneering studies on the gut microbiome and healthy aging. Aging is commonly accompanied by chronic inflammation, increased intestinal permeability, impaired digestion and disrupted nutrient absorption, each of which exhibits bidirectional interactions with the gut microbiome (An et al., 2018). Consistent with these interactions, a preponderance of sequencing studies reports shifts in the composition of the gut microbiota from adulthood to old age (Langille et al., 2014; O’Toole and Jeffery, 2015). However, there is little consensus on the particular aging-associated taxa found across these studies. For example, increases in Bacteroidetes were correlated with aging in some studies (Claesson et al., 2011; Odamaki et al., 2016), whereas decreases in Bacteroidaceae, Lachnospiraceae and Ruminococcaceae, and increases in Akkermansia, Bifidobacterium and Christensenellaceae were reported in another (Biagi et al., 2016). Aside from differences in study design and technical approaches, several confounding factors are likely to contribute to the lack of study consensus. In a study of hospitalized elderly and healthy controls, for example, medication-intake correlated negatively with Massilia and Lachnospiraceae and positively with Bradyrhizobium, Coprobacter, Helicobacter and Prevotella (Ticinesi et al., 2017). In a sixty-year longitudinal study, married individuals exhibited greater microbial diversity and richness compared to individuals living alone (Dill-McFarland et al., 2019), which may be related to reported influences of social interaction on the gut microbiota.
Given the technical challenges of conducting mechanistic aging-related research in humans, animal models have been particularly useful for demonstrating proof-of-principle for microbial effects on aging. Manipulation of the gut microbiota can modify lifespan across rodents, fish, flies and worms (Gordon et al., 1966; Lee and Hase, 2014; Obata et al., 2018; Smith et al., 2017; Thevaranjan et al., 2017). In Drosophila melanogaster, age-related alterations in the gut microbiota preceded and predicted age-associated intestinal barrier dysfunction and subsequent decline (Clark et al., 2015). Specific depletion of Acetobacter from the gut in response to early life oxidant exposure extended longevity (Obata et al., 2018), whereas overgrowth of Lactobacillus plantarum shortened lifespan via lactic acid-mediated overproduction of reactive oxygen species (Iatsenko et al., 2018). In C. elegans, gut colonization of Bacillus subtilis extended lifespan via biofilm formation and the production of nitric oxide and quorum-sensing pentapeptide (Donato et al., 2017). In the African turquoise killifish, transplanting microbiota from young donors into middle-aged recipients extended lifespan, which correlated with engraftment of Exiguobacterium, Planococcus, Propionigenium and Psychrobacter (Smith et al., 2017). Consistent with reported age-associated reductions in Akkermansia muciniphila in mice and humans (Fransen et al., 2017; Langille et al., 2014; Sovran et al., 2019), supplementation of A. muciniphila to the LmnaG609G/G609G mouse model of progeria significantly extended lifespan (Barcena et al., 2019). While little is known regarding the exact molecular and cellular mechanisms underlying these distinct phenomena, several studies suggest that microbial modulation of immune homeostasis may be involved. Germ-free mice colonized with microbiota from old, but not young, conventional mice displayed elevated inflammatory cytokines in the serum and higher intestinal permeability, suggesting that aging-associated changes in the gut microbiota enhance systemic inflammation (Thevaranjan et al., 2017). Consistent with this, impaired proliferation of B cells in the Peyer’s patches of aged mice was corrected by fecal microbiota transplantation from young mice (Stebegg et al., 2019), indicating protective effects of the microbiota from young mice against aging-associated immune dysregulation. These foundational studies on roles for the microbiome during normal aging highlight temporal changes in the microbiome that impact healthspan and lifespan, likely through immune and stress responses. Given that aging is a major risk factor for many NDs, these studies further pave the way for active interest in microbial influences on aging-related neurodegenerative disorders, such as PD and AD.
The Gut Microbiome and Parkinson’s Disease
Of all NDs, PD has the richest history of interest in peripheral contributions to central neural dysfunction. PD is a multifactorial neurodegenerative disease, believed to be caused by both environmental and genetic risk factors. It is characterized by the presence of neurotoxic alpha-synuclein inclusions that result in striatal dopaminergic cell death and motor deficits. A role for the GI tract in the pathogenesis of PD has been hypothesized since the 1980’s when alpha-synuclein inclusions were first observed in enteric tissues from PD patients (Wakabayashi et al., 1988). In the years to follow, Braak and colleagues went on to characterize the topographical spread of alpha-synuclein inclusions, noting that regions receiving input from peripheral nerve fibers such as the dorsal vagal brainstem motor nuclei and olfactory bulb are affected early on (Braak and Braak, 2000; Braak et al., 2002; Braak et al., 2000). These observations lead to the development of the “dual-hit” staging hypothesis, suggesting that PD pathology originates from insults in peripheral organs where alpha-synuclein is seeded, such as the gastrointestinal tract and nasal cavity, before translocating to the brain (Hawkes et al., 2007). In line with this hypothesis, many PD patients experience hyposmia and gastrointestinal issues prior to the onset of motor symptoms, and patients with inflammatory bowel disease (IBD) are at a greater risk of developing PD.
In the past decade, this interest in gastrointestinal associations with PD has extended to include alterations in the gut microbiota as a potential disease biomarker and/or disease modifier. Clinical studies profiling the microbiota of PD patients reported many alterations relative to healthy controls, including increases in the relative abundance of Bifidobacterium (Hasegawa et al., 2015; Peng et al., 2018), Lactobacillus (Hasegawa et al., 2015; Petrov et al., 2017), and Verrucomicrobiaceae (Hasegawa et al., 2015; Petrov et al., 2017; Unger et al., 2016). Consistent with the latter, increases in the abundance of Akkermansia have been widely reported in individuals with PD (Hill-Burns et al., 2017; Keshavarzian et al., 2015; Scheperjans et al., 2015; Unger et al., 2016). Decreases in Blautia and Coprococcus (Bedarf et al., 2017; Hasegawa et al., 2015; Unger et al., 2016), as well as Prevotellaceae (Hasegawa et al., 2015; Heintz-Buschart et al., 2018; Hill-Burns et al., 2017) have also appeared across studies. One metagenomic study reported that increased abundance of A. municiphila, Prevotella copri, and Eubacterium in L-DOPA-naïve patients correlated with alterations in microbial metabolism of tryptophan and beta-glucuronide (Bedarf et al., 2017). Interestingly, longitudinal studies assessing the temporal dynamics of microbiota composition in PD patients indicate that microbial changes remain relatively stable over years following the onset of PD symptoms (Aho et al., 2019; Minato et al., 2017). However, as the samples were collected 5-9 years after diagnosis, it remains unclear whether there are PD-associated changes in the microbiota on a shorter temporal scale. Despite these advances, there remain many contradictory findings likely due to inter-study variability in patient cohort design, sample preparation and sequencing methodology, and data analysis relative to confounding variables. Rigorous, standardized methodology and study design are thus required in order to draw stronger conclusions regarding whether there exist common microbiome signatures for PD.
Changes in the gut microbiota are also seen in some genetic and pharmacological animal models with face and construct validity for PD. In one study, mice overexpressing human alpha-synuclein (ASO) exhibited reduced Verrucomicrobiaceae, which contrasts the increased levels of this taxon reported in some aforementioned human studies (Gorecki et al., 2019). When raised as germ-free or treated with antibiotics in order to deplete the microbiota during adulthood, ASO mice displayed improved motor function compared to transgenic littermates harboring a complex microbiota (Sampson et al., 2016), which suggested that ASO-associated alterations in the microbiota contribute to motor symptoms of PD. Furthermore, transplantation of ASO mice with human microbiota from PD patients induced more severe motor dysfunction and reactive microglia relative to mice transplanted with human microbiota from healthy controls, suggesting that select human-derived microbes from PD patients can exacerbate motor deficits and neuroinflammation in mice (Sampson et al., 2016). In a rat model for PD, chronic treatment with the pesticide, rotenone, altered the small intestinal and colonic microbiota, with significant increases in Actinobacteria and Proteobacteria, but decreases in Bacteroidetes and Cyanobacteria. Alongside these observed shifts, their model was able to reproduce symptoms of gastroparesis before the development of nigrostriatal pathology (Johnson et al., 2018). Mice receiving similar exposures to rotenone show decreases in the relative abundance of Bifidobacterium and alterations in associated metabolic pathways such as glycosaminoglycan degradation in the cecum (Perez-Pardo et al., 2018). Another neurotoxin model for PD involving low-dose, chronic administration of methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) also alters the microbiota, increasing Prevotellaceae and Erysipelotrichaceae while decreasing Lachnospiraceae (Lai et al., 2018). Another group reported similar changes, where MPTP-treated mice exhibited decreased Firmicutes and Clostridiales and increased Proteobacteria, Turicibacterales, and Enterobacteriales (Sun et al., 2018). Notably, transplantation of the MPTP-associated microbiota into conventional mice impaired motor function and reduced striatal dopamine levels relative to controls. Additionally, administration of a single bacterium, Proteus mirabilis, to mice was able to exacerbate MPTP-induced motor deficits, dopaminergic neuronal damage, and inflammation in brain structures relevant to PD, such as the substantia nigra (Choi et al., 2018). Altogether, these studies indicate that alterations in the gut microbiome can contribute to the presentation of PD-associated endophenotypes in animals.
The Microbiome and Intestinal Inflammation in PD
Exactly how the microbiome may impact PD-related symptoms remains unclear. However, several lines of inquiry are now converging on the notion that microbial regulation of intestinal inflammation may be involved. Some PD patients experience gastrointestinal disturbances, such as chronic gastritis and constipation, several years prior to the onset of motor symptoms, leading scientists to consider gastrointestinal issues as early prodromal features of PD in subsets of patients (Cersosimo and Benarroch, 2012; Pfeiffer, 2011). In a large Danish cohort where individuals with inflammatory bowel disease (IBD) were tracked over 30 years, IBD patients exhibited a 22% increase in PD incidence compared to the unaffected controls (Villumsen et al., 2019). In this clinical investigation, patients with ulcerative colitis exhibited a significantly higher risk of PD compared to patients with Crohn’s disease (Villumsen et al., 2019). In a nationwide Swedish cohort, the incidence of PD was 30% higher in the individuals with IBD compared to those without IBD (Weimers et al., 2019). Another study of an American cohort also observed elevated risk of PD in patients with IBD compared to non-IBD individuals (Peter et al., 2018). Accordingly, alpha-synuclein accumulation was observed in the submucosal layer of the intestine in PD patients with colitis (Grathwohl et al., 2019), providing a potential link between IBD and PD pathology. Notably, the IBD patients who received anti-TNF therapy had a 78% reduction in the incidence of PD, suggesting that suppressing peripheral inflammation may protect against PD (Peter et al., 2018). While the biological links between intestinal autoimmune disease and PD remain elusive, genetic studies of IBD and PD have converged on some shared genetic risk factors. For example, mutations in the kinase domain of leucine-rich repeat kinase 2 (LRRK2) has been identified in both the IBD patients and PD patients, with the N2081D mutation found in IBD and the G2019S mutation representing the most common monogenic cause of PD (Hui et al., 2018). Notably, both N2081D and G2019S mutations result in increased kinase activity of LRRK2, which is associated with impaired function of autophagy and lysosomal formation (Hui et al., 2018). Previous studies had found decreased Firmicutes and increased Enterobacteriaceae in the microbiome of IBD patients (Frank et al., 2007; Schirmer et al., 2019). These microbial features in IBD patients align with some microbial alterations reported across several studies of PD patients (Unger et al., 2016; Vogt et al., 2017). Other specific bacteria implicated in immune tolerance and IBD such as Bacteroides fragilis and B. vulgatus have not been specifically investigated in PD. Overall, these studies reveal associations between PD and IBD, a condition for which the gut microbiome is strongly implicated (Johnson et al., 2019).
An increasing number of animal studies provide causal evidence that intestinal disruptions can trigger PD-related pathology. For example, intestinal injury by dextran sodium sulfate (DSS) triggered the aggregation of alpha-synuclein in enteric neurons and dopaminergic neurons of the substantia nigra, ultimately leading to neuronal degeneration in aged alpha-synuclein transgenic mice (Grathwohl et al., 2019). In ASO mice, lipopolysaccharide injection reduced intestinal barrier integrity and expedited the onset of motor symptoms (Gorecki et al., 2019). Intestinal infection of Pink1−/− mice with Citrobacter rodentium induced severe motor deficits and loss of striatal dopaminergic axons as compared to uninfected Pink1−/− mice and infected wildtype littermates (Matheoud et al., 2019), suggesting interactions between intestinal inflammation and genetic risk for PD. Moreover, in rotenone-treated mice, depletion of Toll-like receptor (TLR) 4 ameliorates rotenone-induced intestinal infiltration of CD3+ T cells and intestinal dysfunction, as well as rotenone-induced motor deficits and loss of dopaminergic neurons (Perez-Pardo et al., 2019). These studies provide proof-of-principle that intestinal inflammation can induce or exacerbate behavioral and pathological symptoms in mouse models of PD. Additional research is needed to examine potential relationships between the microbiome, intestinal disease and PD, and to further determine whether microbial regulation of peripheral immunity may underlie any of the existing links between the microbiome and PD in humans and animal models.
The Microbiome and Amyloidogenesis in PD
Beyond potential immune-mediated pathways linking the microbiome to PD-related neurophysiological and behavioral symptoms, several studies suggest that select microbes and microbial products could stimulate alpha-synuclein aggregation, leading to the presence of neuronal inclusions comprised of misfolded alpha-synuclein protein. Early clinical characterization of amyloid pathology in postmortem brains of sporadic PD patients by Braak and colleagues uncovered particular peripheral organs susceptible to aggregates at different stages in the development of PD (Braak and Braak, 2000; Braak et al., 2002). Histological examination of post-mortem intestinal tissue from non-symptomatic elderly patients uncovered the early presence of alpha-synuclein inclusions in the intestine, in the absence of Parkinsonian motor symptoms (Bloch et al., 2006; Braak et al., 2006; Shannon et al., 2012). Subsequent studies in animal models of PD have demonstrated the presence of intestinal alpha-synuclein during disease (Drolet et al., 2009; Kelly et al., 2014; Wang et al., 2012) that coincides with alterations in gastric motility and permeability (Kelly et al., 2014). Further, intestinal injection of alpha-synuclein pre-formed fibrils (PFFs) (Pan-Montojo et al., 2010) or alpha-synuclein from human brain isolates (Holmqvist et al., 2014) results in the appearance of alpha-synuclein inclusions in the CNS. Duodenal inoculation of alpha-synuclein PFFs to aged ASO mice leads to loss of striatal dopamine in the mid brain (Challis et al., 2020). Together, these data implicate the ability for peripheral alpha-synuclein aggregation to propagate to the CNS and induce PD-related pathology. Alpha-synuclein overexpression is associated with alterations in vagal neuronal gene expression (Noorian et al., 2012), the dorsal motor vagal nucleus is one of the first brain regions in which alpha-synuclein inclusions appear, and there is a decreased risk of developing PD in vagotomized patients (Svensson et al., 2015). The vagus nerve possesses dense varicose projections to the intestinal villi of the small intestine, situating its terminals immediately adjacent to the intestinal lumen whereby the members of the host microbiota reside. One proposed mode of transport is via trans-neuronal propagation of alpha-synuclein along the vagus nerve. Indeed, alpha-synuclein released from enteric neurons or alpha-synuclein PFFs injected into the gastrointestinal tract are unable to reach the brain following vagotomy (Kim et al., 2019; Pan-Montojo et al., 2012; Uemura et al., 2018), suggesting a role for the vagus nerve in propagating alpha-synuclein spread from the periphery to the brain in a prion-like manner.
CNS alpha-synuclein burden is decreased in germ-free animals lacking a microbiome compared to specific-pathogen free mice harboring a complex microbiome (Sampson et al., 2016). One possible mechanism by which the microbiota may contribute to the development of PD alpha-synuclein pathology is through pathogenic cross-seeding, whereby one amyloidogenic protein causes another to adopt a beta-sheet structure, between alpha-synuclein and bacterial amyloids, such as curli in Escherichia coli in the gut. Curli are highly-conserved surface organelles that mediate binding of bacteria to soluble matrix proteins (Olsen et al., 1989). Additionally, the key element, CsgA, contains amyloidogenic peptide repeat motifs shared by human prions (Miraglia et al., 2018). Two genes in the curli operon, CsgE and CsgC, have been shown to modulate alpha-synuclein aggregation in vitro (Chorell et al., 2015) and oral administration of curli-producing bacteria, but not curli-deficient mutant strains, to wild-type rats results in intestinal accumulation of alpha-synuclein (Chen et al., 2016). A recent study reproduced these findings in mice overexpressing alpha-synuclein, and additionally demonstrated that introduction of curli-producing bacteria to a healthy, complex human microbiota exacerbated alpha-synuclein pathology and behavioral abnormalities in a CsgA-dependent manner (Sampson et al., 2020). These findings correlated with morphological alterations in midbrain microglia, suggesting an inflammatory state. However, curli are also considered microbial-associated molecular patterns and are recognized by TLRs to induce activation of the innate immune system (Rapsinski et al., 2015). Due to the role of inflammation in facilitating alpha-synuclein aggregation, more research is needed to determine whether bacterial amyloids from the gut microbiome may act to either directly or indirectly to increase peripheral alpha-synuclein aggregation. This amyloid hypothesis has also recently been extended to non-proteinaceous metabolites that are able to form fibrillar amyloid-like assemblies that seed alpha-synuclein aggregation in vitro (Tavassoly et al., 2018). Due to the vast array of metabolic biotransformations carried out by the microbiota, additional efforts are warranted to evaluate effects of microbially-modulated metabolites on amyloidogenesis.
Most recently, alpha-synuclein was found to be cleaved and enriched particularly in the appendices of healthy individuals (Gray et al., 2014; Killinger et al., 2018), suggesting potential biological relevance to PD pathology. This may be relevant as the appendix has been proposed to serve as a reservoir for the microbiota (Lautenschlager et al., 2017). However, whether appendectomy associates with decreased risk of PD remains controversial. Several epidemiological studies show no or limited correlation between appendectomy and the incidence of PD (Palacios et al., 2018; Svensson et al., 2016; Yilmaz et al., 2017), whereas one very large study indicates that removal of the appendix is associated with lower risk of PD, particularly for people living in rural areas (Killinger et al., 2018).
Notably, subsets of PD patients do not exhibit PNS-CNS spread of alpha synuclein, with some completely lacking alpha-synuclein pathology in the dorsal motor nucleus of the vagus. This has led to the development of a hypothesis for a subtype of PD that is “CNS-first” (Borghammer and Van Den Berge, 2019). In these cases, it is possible that efferent signaling from the CNS during pathology may drive alterations in the microbiota, as both stress and immune signaling have been demonstrated to alter its composition (Bonaz et al., 2018). However, more evidence is needed to clearly determine the effects of efferent-induced alterations in the microbiota on PD outcomes. Similarly, additional understanding is needed for how comorbidities during the prodrome may alter the microbiota to impact the development of PD. Rapid eye movement (REM) sleep behavioral disorder, anxiety and depression are of particular interest given their prevalence during the PD prodrome (Borghammer and Van Den Berge, 2019) and strong evidence linking both circadian issues and stress to alterations in the microbiome (Gubert et al., 2020; Heintz-Buschart et al., 2018; Voigt et al., 2014). Additionally, a recent study demonstrated alterations in the microbiota compositions of Multiple System Atrophy (MSA) patients (Engen et al., 2017), highlighting the importance of considering the role of the microbiome in additional synuclein-based neurodegenerative diseases such as MSA and Lewy Body Dementia (LBD).
The Gut Microbiome and Alzheimer’s Disease
In addition to PD, the gut microbiome is increasingly implicated in the manifestation of AD-related symptoms. AD is a complex neurodegenerative disorder that is characterized by the progressive loss of cognition, memory, and motor ability. A hallmark of AD is the accumulation of extracellular Aβ plaques and intracellular hyperphosphorylated tau tangles in the brain while other neuropathological signs include neuronal loss and brain atrophy. The lack of effective single-target therapeutic treatments and failures of clinical trials, particularly those targeting the amyloid pathways, are evidence that the current understanding of the pathogenesis of AD must be extended (Bullain and Doody, 2020). A few lines of evidence motivate interest in the microbiome and AD. From fundamental behavioral observations in laboratory animals, several studies reveal that manipulations of the microbiome, like germ-free rearing, antibiotic-treatment and severe dietary alterations, alter learning and memory-related behavior (Abbott et al., 2019; Frohlich et al., 2016; Luczynski et al., 2016). In addition, the microbiome is emerging as an important regulator of the development and function of the neuroimmune system (Fung et al., 2017), which is implicated in AD among several other neurological disorders (Giau et al., 2018; Kowalski and Mulak, 2019). As yet, however, there is only some evidence of microbiome alterations in human AD. In one study, decreases in the Negativicutes and Lachnospirceae and an increase in Ruminococcaceae were seen in AD participants compared to healthy controls (Zhuang et al., 2018). In another study, analysis of fecal samples obtained from AD participants reported alterations in the gut microbiota with decreased relative abundance of Ruminococcaceae, Turicibacteraceae, Peptostreptococcaceae, Clostridiaceae, and Mogibacteriaceae and increased relative abundance of Bacteroidaceae and Rikenellaceae compared to the healthy control group (Vogt et al., 2017). Notably, increases in bacterial abundance were correlated with alterations in cerebrospinal fluid (CSF) biomarkers suggestive of increased amyloid burden in the brain. In particular, elevated abundance of Bacteroides and Blautia corresponded to a decrease in CSF Aβ42/Aβ40 and increase in CSF p-tau and p-tau/Aβ42 (Vogt et al., 2017). Microbiome profiling of mouse models for AD has also suggested alterations in the gut microbiota (Brandscheid et al., 2017; Zhang, L. et al., 2017a). For the 5xFAD mouse model, which expresses human transgenes for amyloid precursor protein (APP) and presenilin-1 (PS1) containing three familiar AD mutations in APP and two mutation in PS1, Firmicutes increased from 6 weeks of age to 18 weeks while Bacteroidetes decreased (Brandscheid et al., 2017). In the APP/PS1 mouse model, which expresses the human transgenes containing the Swedish mutation in APP and lacking exon 9 in PS1, Proteobacteria doubled in relative abundance by 6 months of age, and Verrucomicrobia increased by 6-fold by 12 months of age (Zhang et al., 2017a). Increases in Firmicutes and decreases in Bacteroidetes were also reported in the APP/PS1 line, consistent with findings from 5xFAD mice (Zhang et al., 2017a). However, a longitudinal study of the microbiota in the P301L tau mouse model of AD reported the opposite relationship, wherein the abundance in Bacteriodetes increased starting at 3 months of age while Firmicutes decreased (Sun, B.L. et al., 2019). By 10 months of age, Candidatus_saccharimonas, Alistipes, Rikenella, Odoribacter, Blautia, Ruminococcaceae, Eubacterium_xylanophilum, Paraprevotella, Butyricicoccus, and Parvibacter were among the bacteria that were negatively associated with tau pathology in the brain. In contrast, Bacteroides, Parabacteroides, Escherichia-Shigella, and Clostridium_innocuum were positively correlated with tau pathology. Across these studies examining the microbiota in human AD and AD mouse models, there is as yet no clear microbial signature for AD.
The Microbiome and Neuroinflammation in AD
Despite limited evidence for a defined shift in the composition of the microbiota in human AD and AD mouse models, a growing number of studies report that manipulating the gut microbiota impacts the severity of Aβ pathology and cognitive impairment in genetic models for AD. Several of these studies suggest that microbial regulation of neuroimmunity may contribute to AD symptoms. For example, rearing APP/PS1 mice as germ-free resulted not only in reduction of Aβ aggregates in the brain, but also a 40% decrease in Iba-1 positive microglia leading to an overall decrease in neuroinflammation compared to conventionalized mice (Harach et al., 2017). The pro-inflammatory cytokine IL-1β was also significantly decreased by 36% in germ-free APP/PS1 mice compared to conventionally colonized APP/PS1 controls. Another study using antibiotics to acutely deplete the microbiota of APP/PS1 mice saw similar results. One week of antibiotic treatment led to a decrease in Aβ load and down regulation of pro-inflammatory cytokine IL-6 compared to the vehicle-treated group (Minter et al., 2017). These alterations corresponded with reductions in numbers of plaque-associated microglia and astrocytes, suggesting that depletion of the microbiome reduced neuroinflammation and gliosis in the brain. In a separate study, treatment of APP/PS1 mice with an antibiotic cocktail led to a reduction in Aβ plaques and astrogliosis in male mice only (Dodiya et al., 2019). Tg2576 mice, which express a variant of human APP, exhibited an aging-related increase in intestinal Bacteroides fragilis relative to wildtype controls. Notably, administering B. fragilis to APP/PS1 female mice promoted Aβ plaque deposition in the cortex, which was rescued by calorie restriction (Cox et al., 2019). While exact mechanisms are unknown, the findings appear to counter existing literature reporting that B. fragilis elicits immunosuppressive effects that may ameliorate symptoms of other neurological diseases. Together these two studies suggest that microbiome alterations may modulate AD pathology in a sex-dependent manner. Overall, these studies provide some evidence that large alterations in the gut microbiota can influence the progression and manifestation of AD-related features in genetically susceptible animals.
Infection and AD
While many studies on the microbiota and AD focus on symbiotic microorganisms indigenous to the gut, a growing body of literature links AD to bacterial and viral pathogens. RNA sequencing of brain tissue from deceased patients with a variety of neurodegenerative diseases revealed the presence of infectious agents in the CNS (Bennett et al., 2019). For AD in particular, reads aligning to Toxoplasmosis gondii, Trichinella sp. T6, Babesia microti, Borrella burgdorferi, Porphyromonas gingivalis, and Treponema denticola were observed in samples of the frontal cortex from post-mortem AD brains. Similarly, another transcriptomics study mapped HSV6 and 7 to multiple brain regions in AD patients (Readhead et al., 2018), which aligns with multiple case and epidemiological studies on HSV and AD. This could be due to direct interactions between neurotropic viruses and amyloids, as HSV1 was capable of catalyzing Aβ plaque formation by binding Aβ-peptides using the protein corona (Ezzat et al., 2019). In addition, data from the National Health and Nutrition Examination Surveys database revealed a positive association between AD mortality and H. pylori (Beydoun et al., 2018). One study showed that H. pylori is able to induce tau hyperphosphorylation by activating the tau kinase glycogen synthase kinase-3β (Wang et al., 2015). Additional studies reported that Aβ precursor was upregulated in tissues from AD patients with periodontitis relative to unaffected AD controls (Nezu et al., 2017). Consistent with this, across several mouse models for AD, infection with Porphyromonas gingivalis exacerbated AD pathology resulting in an increase in neuroinflammation, neurodegeneration and amyloidosis (Dominy et al., 2019; Ilievski et al., 2018; Ishida et al., 2017). This evidence linking bacterial and viral infection to AD may be related to some studies proposing that native function of Aβ is an antimicrobial peptide involved innate immune defense (Eimer et al., 2018; Kumar et al., 2016; Moir et al., 2018). Whether the gut microbiome may play a role in regulating host responses to AD-associated infections remains poorly understood.
Emerging Studies of the Gut Microbiome in Other NDs
ALS is a fatal neurological disease accompanied with the progressive degeneration of motor neurons in the brain and spinal cord. A few studies have compared the gut microbial composition in patients with ALS and healthy controls and highlighted certain species. In an investigation of 37 patients with ALS and 29 healthy familial controls, metagenomic sequencing of gut microbiome revealed a significantly distinct microbial composition compared with healthy controls, among which the abundance of Anaerostipes hadrus, Bacteroidales bacterium, Bifidobacterium pseudocatenulatum, are marginally increased whereas Clostridium leptum and Escherichia coli are marginally decreased in ALS-patients compared to non-affected controls (Blacher et al., 2019). In addition, reduced levels of nicotinamide (NAM), correlated with decreased abundance of Bifidobacterium pseudocatenulatum, was observed in the serum and CSF of patients with ALS by metabolomic analysis (Blacher et al., 2019). Another study of 6 ALS-patients highlights the reduced ratio of Firmicutes/Bacteroidetes in fecal microbiota of ALS-patients, accompanied with increased abundance of Dorea and decreased abundance of Oscillibacter, Anaerostipes and Lachnospiraceae (Fang et al., 2016). By contrast, a study involving 25 ALS-patients did not find significant differences in the gut microbial composition between ALS patients and healthy controls, even though an increase of the overall number of microbial species was observed in ALS patients compared with healthy control (Brenner et al., 2018). The contradictory results from different studies may be due at least in part to the limited power of the studies, which highlights the need for systematic investigation of the microbiota in large cohorts of ALS patients and controls.
In addition to independent studies reporting correlations between microbiota composition and ALS, one study reported a role for the microbiota in protecting against ALS symptoms in the Sod1 transgenic mouse model. Depletion of gut microbiota by broad-spectrum antibiotics exacerbated the motor deficits in Sod1 mice, as evaluated in the rotarod test and wire-hanging test (Blacher et al., 2019). In line with this, Sod1 mice reared as germ-free exhibited higher mortality rates than those raised as conventionally colonized. This contrasts findings from mouse models of PD and AD, where absence of the microbiota abrogates pathology and behavioral abnormalities. 16S rRNA sequencing and shotgun metagenomic sequencing revealed that Sod1 mice exhibited reduced abundance of A. muciniphila compared to wildtype littermates. Colonizing antibiotic-treated Sod1 mice with A. muciniphila not only extended lifespan but also ameliorated the brain atrophy and motor deficits of the mice. Consistent with the association of reduced nicotinamide with ALS incidence in a companion clinical study, increased levels of nicotinamide were observed in the sera of Sod1 mice administered A. muciniphila, and administering nicotinamide to Sod1 mice recapitulated the protective effect of A. muciniphila (Blacher et al., 2019). Overall, this study suggests that gut microbes can regulate motor and neurophysiological endophenotypes of ALS by altering the systemic bioavailability of select metabolites. In another study of the C9orf72 mouse model of ALS and FTD, researchers similarly identified a role for microbiome alterations in regulating the severity of motor impairments and lifespan (Burberry et al., 2020). The microbiome alterations correlated with increases in pro-inflammatory cytokines and activated microglia in the spinal cord, suggesting a neuroimmune-mediated path. Altogether, the findings contribute to growing interest in uncovering functions for previously uncharacterized metabolites that are regulated by the gut microbiome.
Similar links between the microbiota and HD are just beginning to be explored. HD is a progressive neurological disorder caused by a trinucleotide expansion in the huntingtin gene, which causes death of neurons in various brain regions, leading to chorea and abnormal alterations in behavior, emotion and cognition. Severe gliosis was observed in the postmortem brains of individuals with HD (Singhrao et al., 1999). Given that the microbiota can affect the neuronal activity and glial function in the brain, studying the involvement of microbiota in HD can improve our understanding of whether alterations in environment factors can modulate risk for HD. While no studies to date have sequenced the human gut microbiome from HD patients, one study reported the presence of various fungal and bacterial species in the striatum of HD patients, including Candida, Pseudomonas, Acinetobacter, and Burkholderia (Alonso et al.,2019).. While the study suggests that microbial infection may be associated with HD, the technical contamination of brain tissue needs to be excluded. Another study compared the gut microbiota in the R6/1 mouse model of HD with wildtype littermates and reported sexual dimorphism in the bacterial signature of R6/1 mice. An increase in Bacteroidales, Lactobacillales and a decrease in Clostridiales were observed in male R6/1 mice, while an increase in Coriobacteriales, Erysipelotrichales, Bacteroidales, Burkholderiale and a decrease in Clostridiales were observed in females. In addition, male R6/1 mice show higher microbial diversity compared to both female R6/1 mice and wildtype littermates (Kong et al., 2018). Whether these microbiota alterations contribute to symptoms of the R6/1 model remain unclear. However, in the BACHD model, mice reared as germ-free exhibited reduced levels of myelin-related proteins and decreased numbers of mature oligodendrocytes in the prefrontal cortex compared to mice reared conventionally-colonized (Radulescu et al., 2019). This generally aligns with findings from the Sod1 model, where absence of the microbiota exacerbates endophenotypes of disease.
There are as yet only a few studies of the microbiome in other NDs such as MSA. A study analyzing 15 fecal samples from Chinese MSA patients found increased Roseburia hominis, Akkermansia muciniphila, Alistipes onderdonkii, Streptococcus parasanguinis, and Staphylococcus xylosus, and decreased Bacteroides coprocola, Megamonas funiformis, Bifidobacterium pseudocatenulatum, Clostridium nexile, B. plebeius, and Granulicatella adiacens relative to healthy controls (Wan et al., 2019). Another study sequencing 17 fecal samples of Malaysian MSA patients identified increased abundance of Bacteroides and reduced abundance of Paraprevotella (Tan et al., 2018). For LBD, there is as yet no study examining the microbiome. Additional research is warranted to replicate and extend interrogations into the microbiota in ALS, HD MSA and LBD, among other NDs.
Comparing Microbiomes Across NDs
Some NDs share common risk factors and symptomatology, raising the question of whether there are any shared features of the microbiome across different NDs. Decreases in Lachnospiraceae have been commonly reported in aging, PD and ALS, as well as Crohn’s disease (Gevers et al., 2014). Lachnospiraceae suppresses colonic inflammation in experimental colitis models and there are negative correlations between Lachnospiraceae and pro-inflammatory intestinal microbes. Moreover, Lachnospiraceae are a major producer of short chain fatty acids (SCFAs) which elicit widespread effects on host metabolism, immunity and neurophysiology. SCFAs have been shown to activate GPR109 on myeloid cells, which in turn induces regulatory T cell polarization to attenuate intestinal inflammation (Singh et al., 2014). Such interactions between select microbiota alterations, microbiome-dependent metabolites and immunity could be relevant to links between infection, inflammation and NDs such as AD and PD. Additionally, the SCFA propionate was reported to reduce oxidative stress via GPR41 in cells of the blood brain barrier (Hoyles et al., 2018), further warranting future investigation into the impact of SCFAs on NDs.
Another notable bacterial species associated with NDs is Akkermansia, which exhibits increased abundance in both PD and AD, but decreased abundance in aging and the ALS mouse model. Administration of Akkermansia ameliorates motor impairment in the Sod1 ALS mouse model via select microbial metabolites like nicotinamide (Blacher et al., 2019). A previous study has found that administration Akkermansia with Parabacteroides to epilepsy mouse models elevated levels of GABA relative to glutamate levels in the hippocampus (Olson et al., 2018). While the signaling pathways and the downstream effects of metabolites deriving from Akkermansia are still elusive, these observations suggest that there may be distinctive effects of Akkermansia in different NDs. Nevertheless, the reported increased abundance of Akkermansia in PD/AD could well be a result or confounding effect, rather than a modifier of PD/AD pathogenesis. Therefore, further studies are needed to discern any causal links between microbial alterations and the onset or development of NDs.
Microbiome-Related Interventions for NDs
Despite our limited mechanistic understanding of how the microbiota may predispose to symptoms of NDs, efforts to manipulate the microbiota through transplantation and probiotic treatment highlight the potential for microbial amelioration of ND-related pathology and behavior in laboratory animals. Transplantation of conventional microbiota into MPTP mice ameliorated motor deficits, increased dopaminergic neurons in the substantial nigra and reduced numbers of activated microglia and astrocytes (Sun et al., 2018). Similarly, AD mouse models exhibited improved learning and memory behavior, reductions in brain Aβ and tau aggregation, decreased neuroinflammation, and attenuated synaptic dysfunction after microbiota transplantation (Kim et al., 2020; Sun, J. et al., 2019). In the ADLPAPT mouse line, which expresses the five mutations in APP and PS1 found in the 5xFAD model in addition to the P301L mutation in tau, daily fecal microbiota transplant (FMT) from wildtype mice into ADLPAPT mice over 4 months resulted in improved spatial short-term memory and long-term memory (Kim et al., 2020). In addition, measuring the total plaque area of the frontal cortex and hippocampus revealed a decrease in Aβ plaque burden in treated mice compared to control ADLP mice. Furthermore, FMT decreased tau aggregates, reduced the number of activated microglia and attenuated gliosis. In the APP/PS1 mouse model, daily FMTs over 4 weeks also resulted in improved cognition as indicated by better performance in the Morris water maze and object recognition test (Sun, J. et al., 2019). Aβ deposition and hyperphosphorylated tau in the hippocampus were reduced in the treated mice compared to controls. Targeting select bacteria for treatment has also revealed beneficial effects in ND animal models. Administration of probiotic bacterium Bacillus subtilis to transgenic C. elegans expressing human alpha-synuclein increased clearance of alpha-synuclein, ultimately leading to a reversal of aggregation (Goya et al., 2020). Another series of experiments examining the effects probiotic treatment used the 3xTg mouse model which express single mutation in each of the human transgenes for APP, PS1 and tau. Chronic treatment of 3xTg mice with a cocktail of Lactobacillus and Bifidobacteria (SLAB51) promoted working memory, increased cortical thickness and decreased amyloid burden in the brain (Bonfili et al., 2017). Furthermore, bacterial treatment activated sirtuin-1 dependent pathways to promote antioxidative responses (Bonfili et al., 2018), improved glucose reuptake in the brain, and reduced levels of hyperphosphorylated tau (Bonfili et al., 2019). Similarly, APP/PS1 mice treated with Clostridium butyricum had reduced microglial activation and decreased signs of neurodegeneration (Sun et al., 2020). These studies provide proof-of-concept for microbiome interventions in animal models. However, there is as yet no evidence that such approaches would be safe or effective for clinical NDs.
Other microbiome-based interventions that aim to enhance the efficacy of existing treatments for NDs may be more tractable for clinical translation. With the finding that select bacteria metabolize L-dopa, a common treatment for PD (Maini Rekdal et al., 2019; van de Steeg et al., 2018; van Kessel et al., 2019), there is increased interest in whether inhibiting or reducing microbial metabolism of L-dopa could promote drug efficacy in PD. Similar approaches of modulating microbial metabolism of existing medications may be relevant more broadly to neurology, as select microbes from the gut microbiome have been reported to interact with a variety of common medications for neurological diseases (Cryan et al., 2020; Maini Rekdal et al., 2019). In addition, a wealth of evidence suggests that adherence to the Mediterranean diet reduces susceptibility to AD (Berti et al., 2018; Rainey-Smith et al., 2018), but whether the microbiome may mediate the effects of the diet on host physiology remains unclear. Indeed, the microbiome was altered in patients with mild cognitive impairment after treatment with the modified Mediterranean-ketogenic diet (Nagpal et al., 2019), and the microbiome was reported to mediate the neuromodulatory effects of the ketogenic diet in mouse models of refractory epilepsy (Olson et al., 2018). While diet-related interventions are being pursued for protecting against cognitive impairment in AD, further epidemiological research on potential relationships between diet, microbiome and risk for NDs is warranted.
Future Outlooks
Overall, studies in animal models provide intriguing evidence that perturbing the microbiota can modify risk for developing pathological and behavioral endophenotypes of PD, AD, ALS and HD (Table 3). In various genetic models of PD and AD, complete lack of or depletion of the microbiota yields animals with reduced amyloid pathology and improved motor or cognitive behavior. While only a few such studies have been conducted in mouse models of ALS and HD, they commonly report that the absence or depletion of the microbiota exacerbates neuropathological and behavioral symptoms of disease. While these studies provide intriguing proof of concept that the microbiome as a whole is important for host physiological responses related to NDs, whether more subtle alterations in complex microbial communities could predispose to disease remains largely unexplored. Intriguingly, microbiota transplant and bacterial treatments are capable of reducing symptoms, suggesting that modifying the gut microbiota in animal models of NDs can ameliorate neuroinflammatory, neuropathological and/or behavioral abnormalities triggered by genetic risk. These findings raise the question of whether there are cross-cutting features and shared influences of the gut microbiota across subsets of ND models (Figure 1). Microbial regulation of peripheral immune and neuroimmune function may be a point of convergence across the disorders that warrants future study. Direct effects of microbial products or microbially-regulated biochemicals on protein misfolding is another area for continued investigation.
Table 3:
Effects of microbiome manipulations in animal models for NDs
Parkinson’s disease: Microbial effects on pathology and behavior in animal models Subject Perturbation Sample Test Result Reference Pink1−/− mice Oral administration of Citrobacter rodentium n=5-72 males and females Grip strength test
Pole test
Open field test
Flow cytometry for brain infiltrated immune subsets
Histology for dopaminergic neurons in the striatum Impaired motor ability shown in Citrobacter rodentium treated Pink1−/− mice
Decreased density of dopaminergic axonal varicosities and enhanced CD8+ T cells in the brain of Citrobacter rodentium treated Pink1−/− mice (Matheoud et al., 2019) Thyl-αSyn mice Administration of microbiota from PD patients n=3-6 males Beam traversal test
Pole descent test
Nasal adhesive removal test Hindlimb clasping reflexes
Beam traversal test
Pole descent test
Nasal adhesive removal test
Hindlimb clasping reflex PD-derived gut microbiota promotes motor impairments (Sampson et al., 2016) Thyl-αSyn
mice GF vs SPF n=4-6 males Western blot and immunofluorescence staining for α-Syn aggregation Representative 3D reconstructions of Iba1-stained microglia in the caudoputamen Reduced locomotor deficits, α-syn accumulation and decreased activation of microglia in GF Thy1-αSyn mice (Sampson et al., 2016) Thyl-αSyn
mice Monocolonization of curli-sufficient vs curli-deficient E. coli n=7-9 males ELISA and immunofluorescence staining for α-Syn aggregation in the brain and gut Immunofluorescence staining for TH neurons, microglia activation in the brain Beam traversal
Pole descent test
Nasal adhesive removal test
Hindlimb clasping reflex
Wire hang test Increased locomotor deficits, α-syn accumulation and enhanced activation of microglia in Thy1-αSyn mice colonized with curli-sufficient Ecoli (Sampson et al., 2020) Aged Fischer 344 rats Oral administration of curli-sufficient E. coli vs curli-deficient E. coli n=9-13
males Histology for astrocytes, α-syn aggregation
Serum cytokine profiling Increased a-syn accumulation and enhanced activation of astrocytes in rats with curli-sufficient E. coli (Chen et al., 2016) C. elegans expressing human α-syn Curli-sufficient vs curli-deficient E. coli feeding n=15 Sex unknown Immunofluorescence staining for α-syn aggregation Increased α-syn accumulation in C. elegans feed with curli-sufficient E. coli (Chen et al., 2016) Thyl-αSyn mice Drinking water containing 10 μg/ml LPS 12 consecutive days treatment n is unknown males and females Hindlimb clasping reflex Mildly exacerbated motor impairments in LPS treated Thy1-αSyn mice (Gorecki et al., 2019) MPTP mouse model
Rotenone mouse model Probiotic cocktail containing Lactobacillus rhamnosus GG, Bifidobacterium animalis lactis and Lactobacillus acidophilus
4 weeks treatment before animal model establishment n=3-4 males Cylinder test
Beam traversal test
Challenge beam test
Stride length test
Histology for dopaminergic neurons and glial activation in the striatum and substantial nigra Mitigated behavioral impairments, ameliorated dopaminergic neuronal loss and gliosis in probiotics treated
MPTP and rotenone mouse model (Srivastav et al., 2019) 6-OHDA rat model Antibiotic (neomycin, 2 mg/mL; vancomycin 0.2 mg/mL; bacitracin, 0.5 mg/mL; pimaricin 1.2 μg/mL) treatment for 14 days before animal model establishment n=4-12 males Cylinder test
Forepaw adjusted steps test
Amphetamine-induced rotation
Histology for TH neuron loss in striatum and substantial nigra Q-PCR for expression of proinflammatory cytokines in striatum Attenuated motor deficits, decreased dopaminergic neuron loss and lower expression of proinflammatory cytokines in antibiotic treated 6-OHDA rat model (Koutzoumis et al., 2020) MPTP mouse model Fecal microbiota of transplantation after animal model establishment n=15 males Pole test
Traction test
Histology for dopaminergic neuron loss and glial activation in the substantial nigra Western blot for striatal TH expression Attenuated motor deficits, decreased dopaminergic neuron loss and glial activation in the MPTP mouse model transplanted fecal microbiota from wildtype littermates (Sun et al., 2018) C. elegans model of synucleinop athy Bacillus subtilis probiotic strain PXN21 feeding n=25 sex unknown Histology for αSyn aggregation reduced αSyn aggregation in the C. elegans feed with Bacillus subtilis (Goya et al., 2020) MPTP mouse model Oral administration of Proteus mirabilis n=9-12 males Rotarod test
Open field test
Histology for dopaminergic neuron loss and glial activation in the substantial nigra Histology for TH positive axons loss in the striatum Exacerbated motor deficits, loss of dopaminergic neurons and glial activation in MPTP mouse model received Proteus mirabilis (Choi et al., 2018) MPTP mouse model antibiotic (ampicillin 1 g/L, neomycin sulfate 1 g/L, metronidazole 1 g/L) treatment before animal model establishment n=10 males Histology for dopaminergic neuron loss in the substantial nigra Attenuated dopaminergic neuron loss in the antibiotic treated MPTP mouse model (Pu et al., 2019) Alzheimer’s disease: Microbial effects on pathology and behavior in mice Subject Perturbation Sample Test Result Reference APP/PS1 mice GF vs CONV n=5-8 males and females Histopathology for Aβ in brain
Immunostaining for microglia
Western blot for Aβ ELISA for Aβ38, Aβ40, Aβ42, and cytokines For GF-APP/PS1 mice: lower Aβ deposition in cortex and hippocampus, Aβ levels in western blot and Aβ42 levels in ELISA
reduction in Iba-1 positive microglial leading to reduction in cortical neuroinflammation
For CONV-APP/PS1 mice: increase in IL-1β, INF-χ, IL-2, IL-5 (Harach et al., 2017) APP/PS1 mice Antibiotics (gentamycin 1mg/mL, vancomycin 0.5mg/mL, metronidazole 2mg/mL, neomycin 0.5mg/mL, ampicillin 1mg/mL, kanamycin 3mg/mL, colistin 6000U/mL, cefaperazone 1mg/mL) for 1 week n=5-14 males Flow cytometry for immune cells Immunohistochemistry for Aβ, Iba-1, GFAP ELISA for Aβ Cytokine/chemokine array For ABX treated mice: reduced Aβ deposition in cortex and hippocampus
reduction in Aβ plaque localized microglial and astrocytes leading to reduction in neuroinflammation elevation in FOXP3+ Tregs upregulation of CCL11, IL-1β, IL-2, IL-3, SCF and downregulation of IL-6 (Minter et al., 2017) ADLP mice Fecal microbiota transplant daily for 4 months n=14-16 sex unknown Behavioral tests: Y-maze, contextual fear conditioning, open field
ELISA for Aβ
Immunohistochemistry for Aβ, tau, GFAP For FMT treated mice: better performance on behavioral tests reduction in Aβ deposition in frontal cortex and hippocampus reduction in cortex Aβ40 levels reduction in tau aggregates in hippocampus
reduction in activated microglia and astrocytes in frontal cortex (Kim et al., 2020) APP/PS1 mice Fecal microbiota transplant daily for 4 weeks n=4-6 males Behavioral tests: Morris Water Maze, object recognition test ELISA for Aβ40 and Aβ42
Western blot for tau Immunostaining for PSD-95, CD11b and COX-2
NMR for SCFAs For FMT treated mice: better performance on behavioral tests compared to controls
reduction in Aβ deposition in cortex and hippocampus and in Aβ40/Aβ42 levels
reduction in tau phosphorylation increase in PSD-95 staining
reduction in COX-2 and CD11b levels increase in butyrate levels (Sun, J. et al., 2019) APP/PS1 mice Oral administration of Clostridium butyricum for 4 weeks n=20 sex unknown Behavioral tests: Morris Water Maze, object recognition test
Histology for Aβ and CD11b
ELISA for Aβ42
Butyrate assay Treated mice performed better on behavioral tests compared to controls
Decrease of Aβ levels in the brain for treated mice
Reduction of activated microglial in treated mice
Reduction of IL-1β and TNF-α in treated mice
Higher levels of butyrate in treated mice (Sun et al., 2020) 3xTg mice SLAB51 probiotic treatment n=48-64 males Behavioral test: open field, novel object recognition and elevated maze
GC for SCFAs
ELISA for cytokines
Histology and immunostaining for Aβ Western blot for tau For SLAB51 treated mice: better performance on behavioral tests
Reduction in Aβ deposition in hippocampus
Reduction in tau phosphorylation
Reduction in oxidative stress (Bonfili et al., 2017; Bonfili et al., 2018; Bonfili et al., 2019) APPPS1-21 mice Antibiotics (kanamycin 4 mg/ml, gentamicin 0.35 mg/ml, colistin 8,500 U/ml, metronidazole 2.15 mg/ml, vancomycin 0.45 mg/ml) from P14-P21 n=6-10 males and females Immunofluorescence for Ab and microglia activation Antibiotics treatment reduce Aβ pathology and activation of microglia only in the male brain but not female (Dodiya et al., 2019) Amyotrophic lateral sclerosis: Microbial effects on pathology and behavior in mice Subject Perturbation Sample Test Result Reference SOD1G93A mice 2% sodium butyrate in water n=5-10 sex unknown ALS progression (body weight loss, lifespan)
Intestinal tight junction (western blot and immunofluorescence staining for ZO-1) Delayed ALS progression Increased the intestinal tight junction (Zhang, Y.G. et al., 2017) SOD1G93A mice Repeated oral administration of Akkermansia muciniphila into antibiotic pre-treated Sod1 mice at 6-day intervals for a total of 15 treatments. n= 5-61 males and females lifespan analysis rotarod locomotor test
Hanging-wire grip test
Histological staining for spinal cord
T2 relaxation time Extended lifespan
Ameliorated locomotor deficits
Increased number of motor neurons in spinal cord
Reduced brain atrophy (Blacher et al., 2019) Huntington’s disease: Microbial effects on pathology and behavior in mice Subject Perturbation Sample Test Result Reference BACHD mice GF vs SPF n=4-12 males and females Transmission electron microscopy for myelination
Q-PCR and western blot for myelin based protein Reduced levels of myelin-related proteins and decreased numbers of mature oligodendrocytes in the prefrontal cortex of BACHD mice reared in GF condition (Radulescu et al., 2019) Figure 1. Proposed pathways for microbial modulation of neurodegenerative diseases.
The gut microbiome is increasingly implicated in modifying risk for NDs. While precise mechanisms are unknown, a few pathways have been proposed. The gut microbiome is critical for conditioning peripheral immune homeostasis and autoimmune responses, which could influence the manifestation of NDs (far left). In addition, the gut microbiome is important for promoting intestinal colonization resistance and protecting against pathogens, which could influence the likelihood of host exposure to infection and inflammation, as risk factors for subsets of NDs (center left). The gut microbiome regulates a vast repertoire of biochemicals, some of which may interact directly with amyloids to promote aggregation in neurons and propagation from the periphery to the brain (center right). The gut microbiome modulates biochemicals that modulate neuroimmune development and function, including the activity of brain microglia, which could impact the manifestation of subsets of NDs (far right).
While an increasing number of studies continue to link the microbiome to the modulation of ND symptoms in animal models, the most formidable challenge will be in assessing the relevance of these preclinical studies to the complex and varied manifestations of human NDs. Expanded epidemiological and translational research on the microbiome and NDs is needed to identify associations between the microbiome and various NDs. These studies will need to account for the many influences of diet, medical co-morbidity and environmental exposures, among other factors, to variation in the human microbiome. The findings from these clinical studies will be paramount to guiding hypothesis generation and experimental design toward ensuring that basic studies of the microbiome in NDs will ultimately be relevant to the human conditions. The future holds tremendous opportunity to uncover the influences of the gut microbiota on risk for symptoms of NDs, with the potential to reveal fundamental interactions across the microbiota-gut-brain axis and inform new approaches for identifying and treating NDs.
Acknowledgements
P.F. and S.A.K. are supported by funding from the Chan Zuckerberg Initiative Ben Barres Career Acceleration Award. K.G.J. is supported by Ruth L. Kirschstein National Research Service Award GM007185 from the National Institutes of Health. E.Y.H. is a New York Stem Cell Foundation—Robertson Investigator and is supported by a Packard Fellowship in Science and Engineering and Chan Zuckerberg Initiative Ben Barres Career Acceleration Award.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Abbott KN, Arnott CK, Westbrook RF, and Tran DMD (2019). The effect of high fat, high sugar, and combined high fat-high sugar diets on spatial learning and memory in rodents: A meta-analysis. Neurosci Biobehav Rev. 107, 399–421. Published online 2019/08/28 DOI: 10.1016/j.neubiorev.2019.08.010. [DOI] [PubMed] [Google Scholar]
- Aho VTE, Pereira PAB, Voutilainen S, Paulin L, Pekkonen E, Auvinen P, and Scheperjans F (2019). Gut microbiota in Parkinson’s disease: Temporal stability and relations to disease progression. EBioMedicine. 44, 691–707. Published online 2019/06/22 DOI: 10.1016/j.ebiom.2019.05.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alonso R, Pisa D, and Carrasco L (2019). Brain Microbiota in Huntington’s Disease Patients. Front Microbiol. 10, 2622 Published online 2019/12/05 DOI: 10.3389/fmicb.2019.02622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- An R, Wilms E, Masclee AAM, Smidt H, Zoetendal EG, and Jonkers D (2018). Age-dependent changes in GI physiology and microbiota: time to reconsider? Gut. 67(12), 2213–2222. Published online 2018/09/09 DOI: 10.1136/gutjnl-2017-315542. [DOI] [PubMed] [Google Scholar]
- Barcena C, Valdes-Mas R, Mayoral P, Garabaya C, Durand S, Rodriguez F, Fernandez-Garcia MT, Salazar N, Nogacka AM, Garatachea N, et al. (2019). Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat Med. 25(8), 1234–1242. Published online 2019/07/25 DOI: 10.1038/s41591-019-0504-5. [DOI] [PubMed] [Google Scholar]
- Bedarf JR, Hildebrand F, Coelho LP, Sunagawa S, Bahram M, Goeser F, Bork P, and Wullner U (2017). Functional implications of microbial and viral gut metagenome changes in early stage L-DOPA-naive Parkinson’s disease patients. Genome Med. 9(1), 39 Published online 2017/04/30 DOI: 10.1186/s13073-017-0428-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bennett JP Jr., Keeney PM, and Brohawn DG (2019). RNA Sequencing Reveals Small and Variable Contributions of Infectious Agents to Transcriptomes of Postmortem Nervous Tissues From Amyotrophic Lateral Sclerosis, Alzheimer’s Disease and Parkinson’s Disease Subjects, and Increased Expression of Genes From Disease-Activated Microglia. Front Neurosci. 13, 235 Published online 2019/04/16 DOI: 10.3389/fnins.2019.00235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berti V, Walters M, Sterling J, Quinn CG, Logue M, Andrews R, Matthews DC, Osorio RS, Pupi A, Vallabhajosula S, et al. (2018). Mediterranean diet and 3-year Alzheimer brain biomarker changes in middle-aged adults. Neurology. 90(20), e1789–e1798. Published online 2018/04/15 DOI: 10.1212/WNL.0000000000005527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beydoun MA, Beydoun HA, Elbejjani M, Dore GA, and Zonderman AB (2018). Helicobacter pylori seropositivity and its association with incident all-cause and Alzheimer’s disease dementia in large national surveys. Alzheimers Dement. 14(9), 1148–1158. Published online 2018/09/12 DOI: 10.1016/j.jalz.2018.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biagi E, Franceschi C, Rampelli S, Severgnini M, Ostan R, Turroni S, Consolandi C, Quercia S, Scurti M, Monti D, et al. (2016). Gut Microbiota and Extreme Longevity. Curr Biol. 26(11), 1480–1485. Published online 2016/05/18 DOI: 10.1016/j.cub.2016.04.016. [DOI] [PubMed] [Google Scholar]
- Blacher E, Bashiardes S, Shapiro H, Rothschild D, Mor U, Dori-Bachash M, Kleimeyer C, Moresi C, Harnik Y, Zur M, et al. (2019). Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature. 572(7770), 474–480. Published online 2019/07/23 DOI: 10.1038/s41586-019-1443-5. [DOI] [PubMed] [Google Scholar]
- Bloch A, Probst A, Bissig H, Adams H, and Tolnay M (2006). Alpha-synuclein pathology of the spinal and peripheral autonomic nervous system in neurologically unimpaired elderly subjects. Neuropathol Appl Neurobiol. 32(3), 284–295. Published online 2006/04/28 DOI: 10.1111/j.1365-2990.2006.00727.x. [DOI] [PubMed] [Google Scholar]
- Bonaz B, Bazin T, and Pellissier S (2018). The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis. Front Neurosci. 12, 49 DOI: 10.3389/fnins.2018.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonfili L, Cecarini V, Berardi S, Scarpona S, Suchodolski JS, Nasuti C, Fiorini D, Boarelli MC, Rossi G, and Eleuteri AM (2017). Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci Rep. 7(1), 2426 Published online 2017/05/27 DOI: 10.1038/s41598-017-02587-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonfili L, Cecarini V, Cuccioloni M, Angeletti M, Berardi S, Scarpona S, Rossi G, and Eleuteri AM (2018). SLAB51 Probiotic Formulation Activates SIRT1 Pathway Promoting Antioxidant and Neuroprotective Effects in an AD Mouse Model. Mol Neurobiol. 55(10), 7987–8000. Published online 2018/03/02 DOI: 10.1007/s12035-018-0973-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonfili L, Cecarini V, Gogoi O, Berardi S, Scarpona S, Angeletti M, Rossi G, and Eleuteri AM (2019). Gut microbiota manipulation through probiotics oral administration restores glucose homeostasis in a mouse model of Alzheimer’s disease. Neurobiol Aging. Published online 2019/12/10 DOI: 10.1016/j.neurobiolaging.2019.11.004. [DOI] [PubMed] [Google Scholar]
- Borghammer P, and Van Den Berge N (2019). Brain-First versus Gut-First Parkinson’s Disease: A Hypothesis. J Parkinsons Dis. 9(s2), S281–S295. DOI: 10.3233/JPD-191721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braak H, and Braak E (2000). Pathoanatomy of Parkinson’s disease. J Neurol. 247 Suppl 2, II3–10. Published online 2000/09/19 DOI: 10.1007/PL00007758. [DOI] [PubMed] [Google Scholar]
- Braak H, de Vos RA, Bohl J, and Del Tredici K (2006). Gastric alpha-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci Lett. 396(1), 67–72. Published online 2005/12/07 DOI: 10.1016/j.neulet.2005.11.012. [DOI] [PubMed] [Google Scholar]
- Braak H, Del Tredici K, Bratzke H, Hamm-Clement J, Sandmann-Keil D, and Rub U (2002). Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson’s disease (preclinical and clinical stages). J Neurol. 249 Suppl 3, III/ 1–5. Published online 2003/01/17 DOI: 10.1007/s00415-002-1301-4. [DOI] [PubMed] [Google Scholar]
- Braak H, Rub U, Sandmann-Keil D, Gai WP, de Vos RA, Jansen Steur EN, Arai K, and Braak E (2000). Parkinson’s disease: affection of brain stem nuclei controlling premotor and motor neurons of the somatomotor system. Acta Neuropathol. 99(5), 489–495. Published online 2000/05/11 DOI: 10.1007/s004010051150. [DOI] [PubMed] [Google Scholar]
- Brandscheid C, Schuck F, Reinhardt S, Schafer KH, Pietrzik CU, Grimm M, Hartmann T, Schwiertz A, and Endres K (2017). Altered Gut Microbiome Composition and Tryptic Activity of the 5xFAD Alzheimer’s Mouse Model. J Alzheimers Dis. 56(2), 775–788. Published online 2016/12/31 DOI: 10.3233/JAD-160926. [DOI] [PubMed] [Google Scholar]
- Brenner D, Hiergeist A, Adis C, Mayer B, Gessner A, Ludolph AC, and Weishaupt JH (2018). The fecal microbiome of ALS patients. Neurobiol Aging. 61, 132–137. Published online 2017/10/25 DOI: 10.1016/j.neurobiolaging.2017.09.023. [DOI] [PubMed] [Google Scholar]
- Bullain S, and Doody R (2020). What works and what does not work in Alzheimer’s disease? From interventions on risk factors to anti-amyloid trials. J Neurochem. Published online 2020/04/12 DOI: 10.1111/jnc.15023. [DOI] [PubMed] [Google Scholar]
- Burberry A, Wells MF, Limone F, Couto A, Smith KS, Keaney J, Gillet G, van Gastel N, Wang JY, Pietilainen O, et al. (2020). C9orf72 suppresses systemic and neural inflammation induced by gut bacteria. Nature. DOI: 10.1038/s41586-020-2288-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cersosimo MG, and Benarroch EE (2012). Pathological correlates of gastrointestinal dysfunction in Parkinson’s disease. Neurobiol Dis. 46(3), 559–564. Published online 2011/11/04 DOI: 10.1016/j.nbd.2011.10.014. [DOI] [PubMed] [Google Scholar]
- Challis C, Hori A, Sampson TR, Yoo BB, Challis RC, Hamilton AM, Mazmanian SK, Volpicelli-Daley LA, and Gradinaru V (2020). Gut-seeded alpha-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat Neurosci. 23(3), 327–336. Published online 2020/02/19 DOI: 10.1038/s41593-020-0589-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen SG, Stribinskis V, Rane MJ, Demuth DR, Gozal E, Roberts AM, Jagadapillai R, Liu R, Choe K, Shivakumar B, et al. (2016). Exposure to the Functional Bacterial Amyloid Protein Curli Enhances Alpha-Synuclein Aggregation in Aged Fischer 344 Rats and Caenorhabditis elegans. Sci Rep. 6, 34477 DOI: 10.1038/srep34477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi JG, Kim N, Ju IG, Eo H, Lim SM, Jang SE, Kim DH, and Oh MS (2018). Oral administration of Proteus mirabilis damages dopaminergic neurons and motor functions in mice. Sci Rep. 8(1), 1275 Published online 2018/01/21 DOI: 10.1038/s41598-018-19646-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chorell E, Andersson E, Evans ML, Jain N, Gotheson A, Aden J, Chapman MR, Almqvist F, and Wittung-Stafshede P (2015). Bacterial Chaperones CsgE and CsgC Differentially Modulate Human alpha-Synuclein Amyloid Formation via Transient Contacts. PLoS One. 10(10), e0140194 Published online 2015/10/16 DOI: 10.1371/journal.pone.0140194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Claesson MJ, Cusack S, O’Sullivan O, Greene-Diniz R, de Weerd H, Flannery E, Marchesi JR, Falush D, Dinan T, Fitzgerald G, et al. (2011). Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci U S A. 108 Suppl 1, 4586–4591. Published online 2010/06/24 DOI: 10.1073/pnas.1000097107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clark RI, Salazar A, Yamada R, Fitz-Gibbon S, Morselli M, Alcaraz J, Rana A, Rera M, Pellegrini M, Ja WW, et al. (2015). Distinct Shifts in Microbiota Composition during Drosophila Aging Impair Intestinal Function and Drive Mortality. Cell Rep. 12(10), 1656–1667. Published online 2015/09/01 DOI: 10.1016/j.celrep.2015.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cox LM, Schafer MJ, Sohn J, Vincentini J, Weiner HL, Ginsberg SD, and Blaser MJ (2019). Calorie restriction slows age-related microbiota changes in an Alzheimer’s disease model in female mice. Sci Rep. 9(1), 17904 Published online 2019/12/01 DOI: 10.1038/s41598-019-54187-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cryan JF, O’Riordan KJ, Sandhu K, Peterson V, and Dinan TG (2020). The gut microbiome in neurological disorders. Lancet Neurol. 19(2), 179–194. Published online 2019/11/23 DOI: 10.1016/S1474-4422(19)30356-4. [DOI] [PubMed] [Google Scholar]
- Dill-McFarland KA, Tang ZZ, Kemis JH, Kerby RL, Chen G, Palloni A, Sorenson T, Rey FE, and Herd P (2019). Close social relationships correlate with human gut microbiota composition. Sci Rep. 9(1), 703 Published online 2019/01/27 DOI: 10.1038/s41598-018-37298-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dodiya HB, Forsyth CB, Voigt RM, Engen PA, Patel J, Shaikh M, Green SJ, Naqib A, Roy A, Kordower JH, et al. (2020). Chronic stress-induced gut dysfunction exacerbates Parkinson’s disease phenotype and pathology in a rotenone-induced mouse model of Parkinson’s disease. Neurobiol Dis. 135, 104352 Published online 2018/12/24 DOI: 10.1016/j.nbd.2018.12.012. [DOI] [PubMed] [Google Scholar]
- Dodiya HB, Kuntz T, Shaik SM, Baufeld C, Leibowitz J, Zhang X, Gottel N, Zhang X, Butovsky O, Gilbert JA, et al. (2019). Sex-specific effects of microbiome perturbations on cerebral Abeta amyloidosis and microglia phenotypes. J Exp Med. 216(7), 1542–1560. Published online 2019/05/18 DOI: 10.1084/jem.20182386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dominy SS, Lynch C, Ermini F, Benedyk M, Marczyk A, Konradi A, Nguyen M, Haditsch U, Raha D, Griffin C, et al. (2019). Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv. 5(1), eaau3333 Published online 2019/02/13 DOI: 10.1126/sciadv.aau3333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donato V, Ayala FR, Cogliati S, Bauman C, Costa JG, Lenini C, and Grau R (2017). Bacillus subtilis biofilm extends Caenorhabditis elegans longevity through downregulation of the insulin-like signalling pathway. Nat Commun. 8, 14332 Published online 2017/01/31 DOI: 10.1038/ncomms14332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drolet RE, Cannon JR, Montero L, and Greenamyre JT (2009). Chronic rotenone exposure reproduces Parkinson’s disease gastrointestinal neuropathology. Neurobiol Dis. 36(1), 96–102. Published online 2009/07/15 DOI: 10.1016/j.nbd.2009.06.017. [DOI] [PubMed] [Google Scholar]
- Eimer WA, Vijaya Kumar DK, Navalpur Shanmugam NK, Rodriguez AS, Mitchell T, Washicosky KJ, Gyorgy B, Breakefield XO, Tanzi RE, and Moir RD (2018). Alzheimer’s Disease-Associated beta-Amyloid Is Rapidly Seeded by Herpesviridae to Protect against Brain Infection. Neuron. 99(1), 56–63 e53 Published online 2018/07/13 DOI: 10.1016/j.neuron.2018.06.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engen PA, Dodiya HB, Naqib A, Forsyth CB, Green SJ, Voigt RM, Kordower JH, Mutlu EA, Shannon KM, and Keshavarzian A (2017). The Potential Role of Gut-Derived Inflammation in Multiple System Atrophy. J Parkinsons Dis. 7(2), 331–346. DOI: 10.3233/JPD-160991. [DOI] [PubMed] [Google Scholar]
- Ezzat K, Pernemalm M, Palsson S, Roberts TC, Jarver P, Dondalska A, Bestas B, Sobkowiak MJ, Levanen B, Skold M, et al. (2019). The viral protein corona directs viral pathogenesis and amyloid aggregation. Nat Commun. 10(1), 2331 Published online 2019/05/28 DOI: 10.1038/s41467-019-10192-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fang X, Wang X, Yang S, Meng F, Wang X, Wei H, and Chen T (2016). Evaluation of the Microbial Diversity in Amyotrophic Lateral Sclerosis Using High-Throughput Sequencing. Front Microbiol. 7, 1479 Published online 2016/10/06 DOI: 10.3389/fmicb.2016.01479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fasano A, Visanji NP, Liu LW, Lang AE, and Pfeiffer RF (2015). Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 14(6), 625–639. Published online 2015/05/20 DOI: 10.1016/S1474-4422(15)00007-1. [DOI] [PubMed] [Google Scholar]
- Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, and Pace NR (2007). Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 104(34), 13780–13785. Published online 2007/08/19 DOI: 10.1073/pnas.0706625104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fransen F, van Beek AA, Borghuis T, Aidy SE, Hugenholtz F, van der Gaast-de Jongh C, Savelkoul HFJ, De Jonge MI, Boekschoten MV, Smidt H, et al. (2017). Aged Gut Microbiota Contributes to Systemical Inflammaging after Transfer to Germ-Free Mice. Front Immunol. 8, 1385 Published online 2017/11/23 DOI: 10.3389/fimmu.2017.01385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frohlich EE, Farzi A, Mayerhofer R, Reichmann F, Jacan A, Wagner B, Zinser E, Bordag N, Magnes C, Frohlich E, et al. (2016). Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain Behav Immun. 56, 140–155. Published online 2016/03/01 DOI: 10.1016/j.bbi.2016.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fung TC, Olson CA, and Hsiao EY (2017). Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci. 20(2), 145–155. Published online 2017/01/17 DOI: 10.1038/nn.4476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, Schwager E, Knights D, Song SJ, Yassour M, et al. (2014). The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 15(3), 382–392. Published online 2014/03/19 DOI: 10.1016/j.chom.2014.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giau VV, Wu SY, Jamerlan A, An SSA, Kim SY, and Hulme J (2018). Gut Microbiota and Their Neuroinflammatory Implications in Alzheimer’s Disease. Nutrients. 10(11). Published online 2018/11/18 DOI: 10.3390/nu10111765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gordon HA, Bruckner-Kardoss E, and Wostmann BS (1966). Aging in germ-free mice: life tables and lesions observed at natural death. J Gerontol. 21(3), 380–387. Published online 1966/07/01 DOI: 10.1093/geronj/21.3.380. [DOI] [PubMed] [Google Scholar]
- Gorecki AM, Preskey L, Bakeberg MC, Kenna JE, Gildenhuys C, MacDougall G, Dunlop SA, Mastaglia FL, Akkari PA, Koengten F, et al. (2019). Altered Gut Microbiome in Parkinson’s Disease and the Influence of Lipopolysaccharide in a Human alpha-Synuclein Over-Expressing Mouse Model. Front Neurosci. 13, 839 Published online 2019/08/24 DOI: 10.3389/fnins.2019.00839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goya ME, Xue F, Sampedro-Torres-Quevedo C, Arnaouteli S, Riquelme-Dominguez L, Romanowski A, Brydon J, Ball KL, Stanley-Wall NR, and Doitsidou M (2020). Probiotic Bacillus subtilis Protects against alpha-Synuclein Aggregation in C. elegans. Cell Rep. 30(2), 367–380 e367 Published online 2020/01/16 DOI: 10.1016/j.celrep.2019.12.078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grathwohl S, Quansah E, Maroof N, Steiner JA, Spycher L, Benmansour F, Duran-Pacheco G, Siebourg-Polster J, Oroszlan-Szovik K, Remy H, et al. (2019). Experimental colitis drives enteric alpha-synuclein accumulation and Parkinson-like brain pathology. 505164 DOI: 10.1101/505164%JbioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gray MT, Munoz DG, Gray DA, Schlossmacher MG, and Woulfe JM (2014). Alpha-synuclein in the appendiceal mucosa of neurologically intact subjects. Mov Disord. 29(8), 991–998. Published online 2013/12/20 DOI: 10.1002/mds.25779. [DOI] [PubMed] [Google Scholar]
- Gubert C, Kong G, Renoir T, and Hannan AJ (2020). Exercise, diet and stress as modulators of gut microbiota: Implications for neurodegenerative diseases. Neurobiol Dis. 134, 104621 Published online 2019/10/20 DOI: 10.1016/j.nbd.2019.104621. [DOI] [PubMed] [Google Scholar]
- Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD, Frisoni G, Neher JJ, Fak F, Jucker M, Lasser T, et al. (2017). Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci Rep. 7, 41802 Published online 2017/02/09 DOI: 10.1038/srep41802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hasegawa S, Goto S, Tsuji H, Okuno T, Asahara T, Nomoto K, Shibata A, Fujisawa Y, Minato T, Okamoto A, et al. (2015). Intestinal Dysbiosis and Lowered Serum Lipopolysaccharide-Binding Protein in Parkinson’s Disease. PLoS One. 10(11), e0142164 Published online 2015/11/06 DOI: 10.1371/journal.pone.0142164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawkes CH, Del Tredici K, and Braak H (2007). Parkinson’s disease: a dual-hit hypothesis. Neuropathol Appl Neurobiol. 33(6), 599–614. Published online 2007/10/27 DOI: 10.1111/j.1365-2990.2007.00874.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heintz-Buschart A, Pandey U, Wicke T, Sixel-Doring F, Janzen A, Sittig-Wiegand E, Trenkwalder C, Oertel WH, Mollenhauer B, and Wilmes P (2018). The nasal and gut microbiome in Parkinson’s disease and idiopathic rapid eye movement sleep behavior disorder. Mov Disord. 33(1), 88–98. DOI: 10.1002/mds.27105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hill-Burns EM, Debelius JW, Morton JT, Wissemann WT, Lewis MR, Wallen ZD, Peddada SD, Factor SA, Molho E, Zabetian CP, et al. (2017). Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov Disord. 32(5), 739–749. Published online 2017/02/15 DOI: 10.1002/mds.26942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holmqvist S, Chutna O, Bousset L, Aldrin-Kirk P, Li W, Bjorklund T, Wang ZY, Roybon L, Melki R, and Li JY (2014). Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 128(6), 805–820. Published online 2014/10/10 DOI: 10.1007/s00401-014-1343-6. [DOI] [PubMed] [Google Scholar]
- Hopfner F, Kunstner A, Muller SH, Kunzel S, Zeuner KE, Margraf NG, Deuschl G, Baines JF, and Kuhlenbaumer G (2017). Gut microbiota in Parkinson disease in a northern German cohort. Brain Res. 1667, 41–45. Published online 2017/05/17 DOI: 10.1016/j.brainres.2017.04.019. [DOI] [PubMed] [Google Scholar]
- Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, and McArthur S (2018). Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier. Microbiome. 6(1), 55 Published online 2018/03/23 DOI: 10.1186/s40168-018-0439-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hui KY, Fernandez-Hernandez H, Hu J, Schaffner A, Pankratz N, Hsu NY, Chuang LS, Carmi S, Villaverde N, Li X, et al. (2018). Functional variants in the LRRK2 gene confer shared effects on risk for Crohn’s disease and Parkinson’s disease. Sci Transl Med. 10(423). Published online 2018/01/13 DOI: 10.1126/scitranslmed.aai7795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iatsenko I, Boquete JP, and Lemaitre B (2018). Microbiota-Derived Lactate Activates Production of Reactive Oxygen Species by the Intestinal NADPH Oxidase Nox and Shortens Drosophila Lifespan. Immunity. 49(5), 929–942 e925 Published online 2018/11/18 DOI: 10.1016/j.immuni.2018.09.017. [DOI] [PubMed] [Google Scholar]
- Ilievski V, Zuchowska PK, Green SJ, Toth PT, Ragozzino ME, Le K, Aljewari HW, O’Brien-Simpson NM, Reynolds EC, and Watanabe K (2018). Chronic oral application of a periodontal pathogen results in brain inflammation, neurodegeneration and amyloid beta production in wild type mice. PLoS One. 13(10), e0204941 Published online 2018/10/04 DOI: 10.1371/journal.pone.0204941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishida N, Ishihara Y, Ishida K, Tada H, Funaki-Kato Y, Hagiwara M, Ferdous T, Abdullah M, Mitani A, Michikawa M, et al. (2017). Periodontitis induced by bacterial infection exacerbates features of Alzheimer’s disease in transgenic mice. NPJ Aging Mech Dis. 3, 15 Published online 2017/11/15 DOI: 10.1038/s41514-017-0015-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Itzhaki RF (2017). Herpes simplex virus type 1 and Alzheimer’s disease: possible mechanisms and signposts. FASEB J. 31(8), 3216–3226. Published online 2017/08/03 DOI: 10.1096/fj.201700360. [DOI] [PubMed] [Google Scholar]
- Johnson ME, Stecher B, Labrie V, Brundin L, and Brundin P (2019). Triggers, Facilitators, and Aggravators: Redefining Parkinson’s Disease Pathogenesis. Trends Neurosci. 42(1), 4–13. Published online 2018/10/22 DOI: 10.1016/j.tins.2018.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson ME, Stringer A, and Bobrovskaya L (2018). Rotenone induces gastrointestinal pathology and microbiota alterations in a rat model of Parkinson’s disease. Neurotoxicology. 65, 174–185. Published online 2018/02/23 DOI: 10.1016/j.neuro.2018.02.013. [DOI] [PubMed] [Google Scholar]
- Kelly LP, Carvey PM, Keshavarzian A, Shannon KM, Shaikh M, Bakay RA, and Kordower JH (2014). Progression of intestinal permeability changes and alpha-synuclein expression in a mouse model of Parkinson’s disease. Mov Disord. 29(8), 999–1009. Published online 2014/06/06 DOI: 10.1002/mds.25736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keshavarzian A, Green SJ, Engen PA, Voigt RM, Naqib A, Forsyth CB, Mutlu E, and Shannon KM (2015). Colonic bacterial composition in Parkinson’s disease. Mov Disord. 30(10), 1351–1360. Published online 2015/07/17 DOI: 10.1002/mds.26307. [DOI] [PubMed] [Google Scholar]
- Killinger BA, Madaj Z, Sikora JW, Rey N, Haas AJ, Vepa Y, Lindqvist D, Chen H, Thomas PM, Brundin P, et al. (2018). The vermiform appendix impacts the risk of developing Parkinson’s disease. Sci Transl Med. 10(465). Published online 2018/11/02 DOI: 10.1126/scitranslmed.aar5280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim MS, Kim Y, Choi H, Kim W, Park S, Lee D, Kim DK, Kim HJ, Choi H, Hyun DW, et al. (2020). Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 69(2), 283–294. Published online 2019/09/01 DOI: 10.1136/gutjnl-2018-317431. [DOI] [PubMed] [Google Scholar]
- Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, Lee JH, Kim WR, Kook M, Foss CA, et al. (2019). Transneuronal Propagation of Pathologic alpha-Synuclein from the Gut to the Brain Models Parkinson’s Disease. Neuron. 103(4), 627–641 e627 Published online 2019/07/01 DOI: 10.1016/j.neuron.2019.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong G, Cao KL, Judd LM, Li S, Renoir T, and Hannan AJ (2018). Microbiome profiling reveals gut dysbiosis in a transgenic mouse model of Huntington’s disease. Neurobiol Dis. 104268 Published online 2018/09/09 DOI: 10.1016/j.nbd.2018.09.001. [DOI] [PubMed] [Google Scholar]
- Koutzoumis DN, Vergara M, Pino J, Buddendorff J, Khoshbouei H, Mandel RJ, and Torres GE (2020). Alterations of the gut microbiota with antibiotics protects dopamine neuron loss and improve motor deficits in a pharmacological rodent model of Parkinson’s disease. Exp Neurol. 325, 113159 Published online 2019/12/18 DOI: 10.1016/j.expneurol.2019.113159. [DOI] [PubMed] [Google Scholar]
- Kowalski K, and Mulak A (2019). Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J Neurogastroenterol Motil. 25(1), 48–60. Published online 2019/01/17 DOI: 10.5056/jnm18087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar DK, Choi SH, Washicosky KJ, Eimer WA, Tucker S, Ghofrani J, Lefkowitz A, McColl G, Goldstein LE, Tanzi RE, et al. (2016). Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med. 8(340), 340ra372 Published online 2016/05/27 DOI: 10.1126/scitranslmed.aaf1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kundu P, Blacher E, Elinav E, and Pettersson S (2017). Our Gut Microbiome: The Evolving Inner Self. Cell. 171(7), 1481–1493. Published online 2017/12/16 DOI: 10.1016/j.cell.2017.11.024. [DOI] [PubMed] [Google Scholar]
- Lai F, Jiang R, Xie W, Liu X, Tang Y, Xiao H, Gao J, Jia Y, and Bai Q (2018). Intestinal Pathology and Gut Microbiota Alterations in a Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Mouse Model of Parkinson’s Disease. Neurochem Res. 43(10), 1986–1999. Published online 2018/09/02 DOI: 10.1007/s11064-018-2620-x. [DOI] [PubMed] [Google Scholar]
- Langille MG, Meehan CJ, Koenig JE, Dhanani AS, Rose RA, Howlett SE, and Beiko RG (2014). Microbial shifts in the aging mouse gut. Microbiome. 2(1), 50 Published online 2014/12/19 DOI: 10.1186/s40168-014-0050-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lautenschlager S, Gill P, Luo ZX, Fagan MJ, and Rayfield EJ (2017). Morphological evolution of the mammalian jaw adductor complex. Biol Rev Camb Philos Soc. 92(4), 1910–1940. Published online 2016/11/24 DOI: 10.1111/brv.12314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee WJ, and Hase K (2014). Gut microbiota-generated metabolites in animal health and disease. Nat Chem Biol. 10(6), 416–424. Published online 2014/05/20 DOI: 10.1038/nchembio.1535. [DOI] [PubMed] [Google Scholar]
- Li F, Wang P, Chen Z, Sui X, Xie X, and Zhang J (2019). Alteration of the fecal microbiota in North Eastern Han Chinese population with sporadic Parkinson’s disease. Neurosci Lett. 707, 134297 Published online 2019/06/15 DOI: 10.1016/j.neulet.2019.134297. [DOI] [PubMed] [Google Scholar]
- Li W, Wu X, Hu X, Wang T, Liang S, Duan Y, Jin F, and Qin B (2017). Structural changes of gut microbiota in Parkinson’s disease and its correlation with clinical features. Sci China Life Sci. 60(11), 1223–1233. Published online 2017/05/26 DOI: 10.1007/s11427-016-9001-4. [DOI] [PubMed] [Google Scholar]
- Lin A, Zheng W, He Y, Tang W, Wei X, He R, Huang W, Su Y, Huang Y, Zhou H, et al. (2018). Gut microbiota in patients with Parkinson’s disease in southern China. Parkinsonism Relat Disord. 53, 82–88. Published online 2018/05/20 DOI: 10.1016/j.parkreldis.2018.05.007. [DOI] [PubMed] [Google Scholar]
- Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, and Knight R (2012). Diversity, stability and resilience of the human gut microbiota. Nature. 489(7415), 220–230. Published online 2012/09/14 DOI: 10.1038/nature11550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luczynski P, McVey Neufeld KA, Oriach CS, Clarke G, Dinan TG, and Cryan JF (2016). Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int J Neuropsychopharmacol. 19(8). Published online 2016/02/26 DOI: 10.1093/ijnp/pyw020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maini Rekdal V, Bess EN, Bisanz JE, Turnbaugh PJ, and Balskus EP (2019). Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science. 364(6445). Published online 2019/06/15 DOI: 10.1126/science.aau6323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matheoud D, Cannon T, Voisin A, Penttinen AM, Ramet L, Fahmy AM, Ducrot C, Laplante A, Bourque MJ, Zhu L, et al. (2019). Intestinal infection triggers Parkinson’s disease-like symptoms in Pink1(−/−) mice. Nature. 571(7766), 565–569. Published online 2019/07/19 DOI: 10.1038/s41586-019-1405-y. [DOI] [PubMed] [Google Scholar]
- Menegon A, Board PG, Blackburn AC, Mellick GD, and Le Couteur DG (1998). Parkinson’s disease, pesticides, and glutathione transferase polymorphisms. Lancet. 352(9137), 1344–1346. Published online 1998/11/05 DOI: 10.1016/s0140-6736(98)03453-9. [DOI] [PubMed] [Google Scholar]
- Minato T, Maeda T, Fujisawa Y, Tsuji H, Nomoto K, Ohno K, and Hirayama M (2017). Progression of Parkinson’s disease is associated with gut dysbiosis: Two-year follow-up study. PLoS One. 12(11), e0187307 DOI: 10.1371/journal.pone.0187307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Minter MR, Hinterleitner R, Meisel M, Zhang C, Leone V, Zhang X, Oyler-Castrillo P, Zhang X, Musch MW, Shen X, et al. (2017). Antibiotic-induced perturbations in microbial diversity during postnatal development alters amyloid pathology in an aged APPSWE/PS1DeltaE9 murine model of Alzheimer’s disease. Sci Rep. 7(1), 10411 Published online 2017/09/07 DOI: 10.1038/s41598-017-11047-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miraglia F, Ricci A, Rota L, and Colla E (2018). Subcellular localization of alpha-synuclein aggregates and their interaction with membranes. Neural Regen Res. 13(7), 1136–1144. Published online 2018/07/22 DOI: 10.4103/1673-5374.235013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moir RD, Lathe R, and Tanzi RE (2018). The antimicrobial protection hypothesis of Alzheimer’s disease. Alzheimers Dement. 14(12), 1602–1614. Published online 2018/10/14 DOI: 10.1016/j.jalz.2018.06.3040. [DOI] [PubMed] [Google Scholar]
- Nagpal R, Neth BJ, Wang S, Craft S, and Yadav H (2019). Modified Mediterranean-ketogenic diet modulates gut microbiome and short-chain fatty acids in association with Alzheimer’s disease markers in subjects with mild cognitive impairment. EBioMedicine. 47, 529–542. Published online 2019/09/04 DOI: 10.1016/j.ebiom.2019.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nezu A, Kubota T, Maruyama S, Nagata M, Nohno K, Morozumi T, and Yoshie H (2017). Expression of neprilysin in periodontitis-affected gingival tissues. Arch Oral Biol. 79, 35–41. Published online 2017/03/13 DOI: 10.1016/j.archoralbio.2017.03.003. [DOI] [PubMed] [Google Scholar]
- Noorian AR, Rha J, Annerino DM, Bernhard D, Taylor GM, and Greene JG (2012). Alpha-synuclein transgenic mice display age-related slowing of gastrointestinal motility associated with transgene expression in the vagal system. Neurobiol Dis. 48(1), 9–19. Published online 2012/06/23 DOI: 10.1016/j.nbd.2012.06.005. [DOI] [PubMed] [Google Scholar]
- O’Toole PW, and Jeffery IB (2015). Gut microbiota and aging. Science. 350(6265), 1214–1215. Published online 2016/01/20 DOI: 10.1126/science.aac8469. [DOI] [PubMed] [Google Scholar]
- Obata F, Fons CO, and Gould AP (2018). Early-life exposure to low-dose oxidants can increase longevity via microbiome remodelling in Drosophila. Nat Commun. 9(1), 975 Published online 2018/03/09 DOI: 10.1038/s41467-018-03070-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, Abe F, and Osawa R (2016). Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 16, 90 Published online 2016/05/26 DOI: 10.1186/s12866-016-0708-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olsen A, Jonsson A, and Normark S (1989). Fibronectin binding mediated by a novel class of surface organelles on Escherichia coli. Nature. 338(6217), 652–655. Published online 1989/04/20 DOI: 10.1038/338652a0. [DOI] [PubMed] [Google Scholar]
- Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, and Hsiao EY (2018). The Gut Microbiota Mediates the Anti-Seizure Effects of the Ketogenic Diet. Cell. 173(7), 1728–1741 e1713. Published online 2018/05/29 DOI: 10.1016/j.cell.2018.04.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palacios N, Hughes KC, Cereda E, Schwarzschild MA, and Ascherio A (2018). Appendectomy and risk of Parkinson’s disease in two large prospective cohorts of men and women. Mov Disord. 33(9), 1492–1496. Published online 2018/09/16 DOI: 10.1002/mds.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan-Montojo F, Anichtchik O, Dening Y, Knels L, Pursche S, Jung R, Jackson S, Gille G, Spillantini MG, Reichmann H, et al. (2010). Progression of Parkinson’s disease pathology is reproduced by intragastric administration of rotenone in mice. PLoS One. 5(1), e8762 Published online 2010/01/26 DOI: 10.1371/journal.pone.0008762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan-Montojo F, Schwarz M, Winkler C, Arnhold M, O’Sullivan GA, Pal A, Said J, Marsico G, Verbavatz JM, Rodrigo-Angulo M, et al. (2012). Environmental toxins trigger PD-like progression via increased alpha-synuclein release from enteric neurons in mice. Sci Rep. 2, 898 Published online 2012/12/04 DOI: 10.1038/srep00898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng C, Gathagan RJ, Covell DJ, Medellin C, Stieber A, Robinson JL, Zhang B, Pitkin RM, Olufemi MF, Luk KC, et al. (2018). Cellular milieu imparts distinct pathological alpha-synuclein strains in alpha-synucleinopathies. Nature. 557(7706), 558–563. Published online 2018/05/11 DOI: 10.1038/s41586-018-0104-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez-Pardo P, Dodiya HB, Engen PA, Forsyth CB, Huschens AM, Shaikh M, Voigt RM, Naqib A, Green SJ, Kordower JH, et al. (2019). Role of TLR4 in the gut-brain axis in Parkinson’s disease: a translational study from men to mice. Gut. 68(5), 829–843. Published online 2018/12/17 DOI: 10.1136/gutjnl-2018-316844. [DOI] [PubMed] [Google Scholar]
- Perez-Pardo P, Dodiya HB, Engen PA, Naqib A, Forsyth CB, Green SJ, Garssen J, Keshavarzian A, and Kraneveld AD (2018). Gut bacterial composition in a mouse model of Parkinson’s disease. Benef Microbes. 9(5), 799–814. Published online 2018/08/14 DOI: 10.3920/BM2017.0202. [DOI] [PubMed] [Google Scholar]
- Peter I, Dubinsky M, Bressman S, Park A, Lu C, Chen N, and Wang A (2018). Anti-Tumor Necrosis Factor Therapy and Incidence of Parkinson Disease Among Patients With Inflammatory Bowel Disease. JAMA Neurol. 75(8), 939–946. Published online 2018/05/02 DOI: 10.1001/jamaneurol.2018.0605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrov VA, Saltykova IV, Zhukova IA, Alifirova VM, Zhukova NG, Dorofeeva YB, Tyakht AV, Kovarsky BA, Alekseev DG, Kostryukova ES, et al. (2017). Analysis of Gut Microbiota in Patients with Parkinson’s Disease. Bull Exp Biol Med. 162(6), 734–737. Published online 2017/04/22 DOI: 10.1007/s10517-017-3700-7. [DOI] [PubMed] [Google Scholar]
- Pfeiffer RF (2011). Gastrointestinal dysfunction in Parkinson’s disease. Parkinsonism Relat Disord. 17(1), 10–15. Published online 2010/09/11 DOI: 10.1016/j.parkreldis.2010.08.003. [DOI] [PubMed] [Google Scholar]
- Pu Y, Chang L, Qu Y, Wang S, Zhang K, and Hashimoto K (2019). Antibiotic-induced microbiome depletion protects against MPTP-induced dopaminergic neurotoxicity in the brain. Aging (Albany NY). 11(17), 6915–6929. Published online 2019/09/04 DOI: 10.18632/aging.102221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qian Y, Yang X, Xu S, Wu C, Song Y, Qin N, Chen SD, and Xiao Q (2018). Alteration of the fecal microbiota in Chinese patients with Parkinson’s disease. Brain Behav Immun. 70, 194–202. Published online 2018/03/05 DOI: 10.1016/j.bbi.2018.02.016. [DOI] [PubMed] [Google Scholar]
- Radulescu CI, Garcia-Miralles M, Sidik H, Bardile CF, Yusof N, Lee HU, Ho EXP, Chu CW, Layton E, Low D, et al. (2019). Manipulation of microbiota reveals altered callosal myelination and white matter plasticity in a model of Huntington disease. Neurobiol Dis. 127, 65–75. Published online 2019/02/26 DOI: 10.1016/j.nbd.2019.02.011. [DOI] [PubMed] [Google Scholar]
- Rainey-Smith SR, Gu Y, Gardener SL, Doecke JD, Villemagne VL, Brown BM, Taddei K, Laws SM, Sohrabi HR, Weinborn M, et al. (2018). Mediterranean diet adherence and rate of cerebral Abeta-amyloid accumulation: Data from the Australian Imaging, Biomarkers and Lifestyle Study of Ageing. Transl Psychiatry. 8(1), 238 Published online 2018/10/31 DOI: 10.1038/s41398-018-0293-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rapsinski GJ, Wynosky-Dolfi MA, Oppong GO, Tursi SA, Wilson RP, Brodsky IE, and Tukel C (2015). Toll-like receptor 2 and NLRP3 cooperate to recognize a functional bacterial amyloid, curli. Infect Immun. 83(2), 693–701. DOI: 10.1128/IAI.02370-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Readhead B, Haure-Mirande JV, Funk CC, Richards MA, Shannon P, Haroutunian V, Sano M, Liang WS, Beckmann ND, Price ND, et al. (2018). Multiscale Analysis of Independent Alzheimer’s Cohorts Finds Disruption of Molecular, Genetic, and Clinical Networks by Human Herpesvirus. Neuron. 99(1), 64–82 e67 Published online 2018/06/26 DOI: 10.1016/j.neuron.2018.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren T, Gao Y, Qiu Y, Jiang S, Zhang Q, Zhang J, Wang L, Zhang Y, Wang L, and Nie K (2020). Gut Microbiota Altered in Mild Cognitive Impairment Compared With Normal Cognition in Sporadic Parkinson’s Disease. Front Neurol. 11, 137 Published online 2020/03/13 DOI: 10.3389/fneur.2020.00137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampson TR, Challis C, Jain N, Moiseyenko A, Ladinsky MS, Shastri GG, Thron T, Needham BD, Horvath I, Debelius JW, et al. (2020). A gut bacterial amyloid promotes alpha-synuclein aggregation and motor impairment in mice. Elife. 9 Published online 2020/02/12 DOI: 10.7554/eLife.53111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V, et al. (2016). Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell. 167(6), 1469–1480 e1412 Published online 2016/12/03 DOI: 10.1016/j.cell.2016.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, Haapaniemi E, Kaakkola S, Eerola-Rautio J, Pohja M, et al. (2015). Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 30(3), 350–358. Published online 2014/12/06 DOI: 10.1002/mds.26069. [DOI] [PubMed] [Google Scholar]
- Schirmer M, Garner A, Vlamakis H, and Xavier RJ (2019). Microbial genes and pathways in inflammatory bowel disease. Nat Rev Microbiol. 17(8), 497–511. Published online 2019/06/30 DOI: 10.1038/s41579-019-0213-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shannon KM, Keshavarzian A, Mutlu E, Dodiya HB, Daian D, Jaglin JA, and Kordower JH (2012). Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov Disord. 27(6), 709–715. Published online 2011/07/19 DOI: 10.1002/mds.23838. [DOI] [PubMed] [Google Scholar]
- Sherwin E, Dinan TG, and Cryan JF (2018). Recent developments in understanding the role of the gut microbiota in brain health and disease. Ann N Y Acad Sci. 1420(1), 5–25. Published online 2017/08/03 DOI: 10.1111/nyas.13416. [DOI] [PubMed] [Google Scholar]
- Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH, et al. (2014). Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 40(1), 128–139. Published online 2014/01/15 DOI: 10.1016/j.immuni.2013.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singhrao SK, Neal JW, Morgan BP, and Gasque P (1999). Increased complement biosynthesis by microglia and complement activation on neurons in Huntington’s disease. Exp Neurol. 159(2), 362–376. Published online 1999/10/03 DOI: 10.1006/exnr.1999.7170. [DOI] [PubMed] [Google Scholar]
- Smith P, Willemsen D, Popkes M, Metge F, Gandiwa E, Reichard M, and Valenzano DR (2017). Regulation of life span by the gut microbiota in the short-lived African turquoise killifish. Elife. 6 Published online 2017/08/23 DOI: 10.7554/eLife.27014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sovran B, Hugenholtz F, Elderman M, Van Beek AA, Graversen K, Huijskes M, Boekschoten MV, Savelkoul HFJ, De Vos P, Dekker J, et al. (2019). Age-associated Impairment of the Mucus Barrier Function is Associated with Profound Changes in Microbiota and Immunity. Sci Rep. 9(1), 1437 Published online 2019/02/07 DOI: 10.1038/s41598-018-35228-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srivastav S, Neupane S, Bhurtel S, Katila N, Maharjan S, Choi H, Hong JT, and Choi DY (2019). Probiotics mixture increases butyrate, and subsequently rescues the nigral dopaminergic neurons from MPTP and rotenone-induced neurotoxicity. J Nutr Biochem. 69, 73–86. Published online 2019/05/08 DOI: 10.1016/j.jnutbio.2019.03.021. [DOI] [PubMed] [Google Scholar]
- Stebegg M, Silva-Cayetano A, Innocentin S, Jenkins TP, Cantacessi C, Gilbert C, and Linterman MA (2019). Heterochronic faecal transplantation boosts gut germinal centres in aged mice. Nat Commun. 10(1), 2443 Published online 2019/06/06 DOI: 10.1038/s41467-019-10430-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun BL, Li WW, Wang J, Xu YL, Sun HL, Tian DY, Wang YJ, and Yao XQ (2019). Gut Microbiota Alteration and Its Time Course in a Tauopathy Mouse Model. J Alzheimers Dis. 70(2), 399–412. Published online 2019/06/10 DOI: 10.3233/JAD-181220. [DOI] [PubMed] [Google Scholar]
- Sun J, Xu J, Ling Y, Wang F, Gong T, Yang C, Ye S, Ye K, Wei D, Song Z, et al. (2019). Fecal microbiota transplantation alleviated Alzheimer’s disease-like pathogenesis in APP/PS1 transgenic mice. Transl Psychiatry. 9(1), 189 Published online 2019/08/07 DOI: 10.1038/s41398-019-0525-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun J, Xu J, Yang B, Chen K, Kong Y, Fang N, Gong T, Wang F, Ling Z, and Liu J (2020). Effect of Clostridium butyricum against Microglia-Mediated Neuroinflammation in Alzheimer’s Disease via Regulating Gut Microbiota and Metabolites Butyrate. Mol Nutr Food Res. 64(2), e1900636 Published online 2019/12/14 DOI: 10.1002/mnfr.201900636. [DOI] [PubMed] [Google Scholar]
- Sun MF, Zhu YL, Zhou ZL, Jia XB, Xu YD, Yang Q, Cui C, and Shen YQ (2018). Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF-alpha signaling pathway. Brain Behav Immun. 70, 48–60. Published online 2018/02/23 DOI: 10.1016/j.bbi.2018.02.005. [DOI] [PubMed] [Google Scholar]
- Svensson E, Horvath-Puho E, Stokholm MG, Sorensen HT, Henderson VW, and Borghammer P (2016). Appendectomy and risk of Parkinson’s disease: A nationwide cohort study with more than 10 years of follow-up. Mov Disord. 31(12), 1918–1922. Published online 2016/09/14 DOI: 10.1002/mds.26761. [DOI] [PubMed] [Google Scholar]
- Svensson E, Horvath-Puho E, Thomsen RW, Djurhuus JC, Pedersen L, Borghammer P, and Sorensen HT (2015). Vagotomy and subsequent risk of Parkinson’s disease. Ann Neurol. 78(4), 522–529. Published online 2015/06/03 DOI: 10.1002/ana.24448. [DOI] [PubMed] [Google Scholar]
- Tan AH, Chong CW, Song SL, Teh CSJ, Yap IKS, Loke MF, Tan YQ, Yong HS, Mahadeva S, Lang AE, et al. (2018). Altered gut microbiome and metabolome in patients with multiple system atrophy. Mov Disord. 33(1), 174–176. Published online 2017/10/31 DOI: 10.1002/mds.27203. [DOI] [PubMed] [Google Scholar]
- Tavassoly O, Sade D, Bera S, Shaham-Niv S, Vocadlo DJ, and Gazit E (2018). Quinolinic Acid Amyloid-like Fibrillar Assemblies Seed alpha-Synuclein Aggregation. J Mol Biol. 430(20), 3847–3862. Published online 2018/08/12 DOI: 10.1016/j.jmb.2018.08.002. [DOI] [PubMed] [Google Scholar]
- Thevaranjan N, Puchta A, Schulz C, Naidoo A, Szamosi JC, Verschoor CP, Loukov D, Schenck LP, Jury J, Foley KP, et al. (2017). Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe. 21(4), 455–466 e454. Published online 2017/04/14 DOI: 10.1016/j.chom.2017.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ticinesi A, Milani C, Lauretani F, Nouvenne A, Mancabelli L, Lugli GA, Turroni F, Duranti S, Mangifesta M, Viappiani A, et al. (2017). Gut microbiota composition is associated with polypharmacy in elderly hospitalized patients. Sci Rep. 7(1), 11102 Published online 2017/09/13 DOI: 10.1038/s41598-017-10734-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uemura N, Yagi H, Uemura MT, Hatanaka Y, Yamakado H, and Takahashi R (2018). Inoculation of alpha-synuclein preformed fibrils into the mouse gastrointestinal tract induces Lewy body-like aggregates in the brainstem via the vagus nerve. Mol Neurodegener. 13(1), 21 Published online 2018/05/13 DOI: 10.1186/s13024-018-0257-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Unger MM, Spiegel J, Dillmann KU, Grundmann D, Philippeit H, Burmann J, Fassbender K, Schwiertz A, and Schafer KH (2016). Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat Disord. 32, 66–72. Published online 2016/11/05 DOI: 10.1016/j.parkreldis.2016.08.019. [DOI] [PubMed] [Google Scholar]
- van de Steeg E, Schuren FHJ, Obach RS, van Woudenbergh C, Walker GS, Heerikhuisen M, Nooijen IHG, and Vaes WHJ (2018). An Ex Vivo Fermentation Screening Platform to Study Drug Metabolism by Human Gut Microbiota. Drug Metab Dispos. 46(11), 1596–1607. Published online 2018/08/31 DOI: 10.1124/dmd.118.081026. [DOI] [PubMed] [Google Scholar]
- van Kessel SP, Frye AK, El-Gendy AO, Castejon M, Keshavarzian A, van Dijk G, and El Aidy S (2019). Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat Commun. 10(1), 310 Published online 2019/01/20 DOI: 10.1038/s41467-019-08294-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vidal-Martinez G, Chin B, Camarillo C, Herrera GV, Yang B, Sarosiek I, and Perez RG (2020). A Pilot Microbiota Study in Parkinson’s Disease Patients versus Control Subjects, and Effects of FTY720 and FTY720-Mitoxy Therapies in Parkinsonian and Multiple System Atrophy Mouse Models. J Parkinsons Dis. 10(1), 185–192. Published online 2019/09/29 DOI: 10.3233/JPD-191693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villumsen M, Aznar S, Pakkenberg B, Jess T, and Brudek T (2019). Inflammatory bowel disease increases the risk of Parkinson’s disease: a Danish nationwide cohort study 1977-2014. Gut. 68(1), 18–24. Published online 2018/05/23 DOI: 10.1136/gutjnl-2017-315666. [DOI] [PubMed] [Google Scholar]
- Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, Carlsson CM, Asthana S, Zetterberg H, Blennow K, et al. (2017). Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 7(1), 13537 Published online 2017/10/21 DOI: 10.1038/s41598-017-13601-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Voigt RM, Forsyth CB, Green SJ, Mutlu E, Engen P, Vitaterna MH, Turek FW, and Keshavarzian A (2014). Circadian disorganization alters intestinal microbiota. PLoS One. 9(5), e97500 DOI: 10.1371/journal.pone.0097500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wakabayashi K, Takahashi H, Takeda S, Ohama E, and Ikuta F (1988). Parkinson’s disease: the presence of Lewy bodies in Auerbach’s and Meissner’s plexuses. Acta Neuropathol. 76(3), 217–221. Published online 1988/01/01 DOI: 10.1007/bf00687767. [DOI] [PubMed] [Google Scholar]
- Wan L, Zhou X, Wang C, Chen Z, Peng H, Hou X, Peng Y, Wang P, Li T, Yuan H, et al. (2019). Alterations of the Gut Microbiota in Multiple System Atrophy Patients. Front Neurosci. 13, 1102 Published online 2019/11/05 DOI: 10.3389/fnins.2019.01102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang L, Magen I, Yuan PQ, Subramaniam SR, Richter F, Chesselet MF, and Tache Y (2012). Mice overexpressing wild-type human alpha-synuclein display alterations in colonic myenteric ganglia and defecation. Neurogastroenterol Motil. 24(9), e425–436. Published online 2012/07/12 DOI: 10.1111/j.1365-2982.2012.01974.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang XL, Zeng J, Yang Y, Xiong Y, Zhang ZH, Qiu M, Yan X, Sun XY, Tuo QZ, Liu R, et al. (2015). Helicobacter pylori filtrate induces Alzheimer-like tau hyperphosphorylation by activating glycogen synthase kinase-3beta. J Alzheimers Dis. 43(1), 153–165. Published online 2014/08/01 DOI: 10.3233/JAD-140198. [DOI] [PubMed] [Google Scholar]
- Weimers P, Halfvarson J, Sachs MC, Saunders-Pullman R, Ludvigsson JF, Peter I, Burisch J, and Olen O (2019). Inflammatory Bowel Disease and Parkinson’s Disease: A Nationwide Swedish Cohort Study. Inflamm Bowel Dis. 25(1), 111–123. Published online 2018/05/23 DOI: 10.1093/ibd/izy190. [DOI] [PubMed] [Google Scholar]
- Yang X, Qian Y, Xu S, Song Y, and Xiao Q (2017). Longitudinal Analysis of Fecal Microbiome and Pathologic Processes in a Rotenone Induced Mice Model of Parkinson’s Disease. Front Aging Neurosci. 9, 441 Published online 2018/01/24 DOI: 10.3389/fnagi.2017.00441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yilmaz R, Bayram E, Ulukan C, Altinok MK, and Akbostanci MC (2017). Appendectomy History is not Related to Parkinson’s Disease. J Parkinsons Dis. 7(2), 347–352. Published online 2017/04/08 DOI: 10.3233/JPD-171071. [DOI] [PubMed] [Google Scholar]
- Zhai CD, Zheng JJ, An BC, Huang HF, and Tan ZC (2019). Intestinal microbiota composition in patients with amyotrophic lateral sclerosis: establishment of bacterial and archaeal communities analyses. Chin Med J (Engl). 132(15), 1815–1822. Published online 2019/07/16 DOI: 10.1097/CM9.0000000000000351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Wang Y, Xiayu X, Shi C, Chen W, Song N, Fu X, Zhou R, Xu YF, Huang L, et al. (2017). Altered Gut Microbiota in a Mouse Model of Alzheimer’s Disease. J Alzheimers Dis. 60(4), 1241–1257. Published online 2017/10/19 DOI: 10.3233/JAD-170020. [DOI] [PubMed] [Google Scholar]
- Zhang YG, Wu S, Yi J, Xia Y, Jin D, Zhou J, and Sun J (2017). Target Intestinal Microbiota to Alleviate Disease Progression in Amyotrophic Lateral Sclerosis. Clin Ther. 39(2), 322–336. Published online 2017/01/29 DOI: 10.1016/j.clinthera.2016.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhuang ZQ, Shen LL, Li WW, Fu X, Zeng F, Gui L, Lu Y, Cai M, Zhu C, Tan YL, et al. (2018). Gut Microbiota is Altered in Patients with Alzheimer’s Disease. J Alzheimers Dis. 63(4), 1337–1346. Published online 2018/05/16 DOI: 10.3233/JAD-180176. [DOI] [PubMed] [Google Scholar]
RetroSearch is an open source project built by @garambo
| Open a GitHub Issue
Search and Browse the WWW like it's 1997 | Search results from DuckDuckGo
HTML:
3.2
| Encoding:
UTF-8
| Version:
0.7.3