Zusammenfassung
Aktuelle Forschungsergebnisse im Bereich neurodegenerativer Erkrankungen deuten vermehrt darauf hin, dass die Ernährung und damit assoziiert die Zusammensetzung des Darm-Mikrobioms einen entscheidenden Einfluss auf die Entstehung und den Verlauf verschiedenster Krankheiten haben. Die sogenannte Darm-Hirn Achse, oder präziser die Darm-Mikrobiom-Hirn Achse hat dadurch deutlich an Aufmerksamkeit gewonnen. Dabei kann der Darm das zentrale Nervensystem auf unterschiedliche Weisen beeinflussen, I) direkt durch bakterielle Bestandteile und Metaboliten von Bakterien, II) durch Manipulation der im Körper zirkulierenden Immunzellen, oder III) durch direkten Kontakt, z. B. über den N. vagus.
Fortschritte auf dem Gebiet der Molekularbiologie, wie das Next Generation Sequencing ermöglichen aufgrund ihres hohen Auflösungsvermögens die genaue Identifikation von Bakterien und die Kompositionen ganzer Mikrobiome. Dadurch ist es möglich, die Interaktionen zwischen dem intestinalen Mikrobiom, dem Metabolom und dem Darm- assoziierten Immunsystem detailliert zu erforschen.
In dieser Arbeit diskutieren wir den Einfluss des Mikrobioms, der Ernährung und den damit verbundenen Gesundheitszustand auf die Neuroregeneration. Der Fokus liegt dabei auf der Möglichkeit, wie dieses Wissen in Zukunft für therapeutische Zwecke genutzt werden kann.
Abstract
Recent advances in the field of neurodegenerative disorders point to a possible association between diet, gut microbiota composition and disease incidence. Hence, the so-called gut-brain axis, or more precisely the gut-microbiome-brain axis, has gained increasing attention. There are several ways in which gut content can impact the central nervous system, i. e. either I) directly via bacterial components and dietary metabolites that are systematically available, II) by intermediates, such as circulating immune cells or III) via direct neuronal connections, i. e. the vagus nerve.
New technologies for the identification of bacteria, like next generation sequencing, are enabling a higher resolution understanding of microbiota composition. Therefore, it is now possible to elucidate direct interactions between the gut microbiome, the metabolome, and the gut-associated immune system. In addition to these interactions and of equal importance are the interdependencies of gut metabolites with cells of the central nervous system. In this review, we discuss how the gut microbiome can promote neuronal regeneration or degeneration, depending on health status and diet, and how its modulation may be exploited for novel therapeutic applications.
Introduction
The gut and its microbiome
Colonization of the human gut by various bacterial species occurs initially during or shortly after birth. Already the method of birth, i. e. by caesarean section or naturally, is the key factor for the abundance (Toscano et al., 2017) and amount of gut bacteria (Huurre et al., 2008). In addition to the vaginal passage (Dominguez-Bello et al., 2010), another factor known to contribute to the origin of gut microbiota, is breast-feeding (Backhed et al., 2015). Also, the possibility of colonization in utero is currently being examined (Willyard, 2018), which is supported by the presence of bacterial DNA in the placenta (Aagaard et al., 2014).
In humans, the colon is the organ containing the highest number of microbial species (Sender et al., 2016). The majority of these microbes are members of three bacterial phyla, referred to as enterotypes, namely Firmicutes, Bacteroidetes, and Actinobacteria (Tap et al., 2009). Due to the enormous number and diversity of these gut-dwelling bacterial species, the human organism is capable of digesting several food-derived nutrients by taking advantage of bacterially-derived enzymes. Bacteroides thetaiotamicro, for example, produces a variety of enzymes that can degrade a range of carbohydrates (Xu et al., 2003). This symbiotic relationship that enables a broad range of nutritive sources has figured largely in human evolution. Perhaps the most prominent example involves mitochondria, bacteria that lost their cellular autonomy and became endosymbiotically-derived organelles (Stilling et al., 2014, Archibald, 2015, Raina et al., 2018).
The primary metabolites and end products of bacterial fermentation include short chain fatty acids (SCFA) (Salminen et al., 1998), micronutrients such as vitamins (Fresia Fernandez, 1987), and secondary bile acids (Ajouz et al., 2014). These microbial products diffuse passively, or are actively transported across gastrointestinal tract endothelia, where they become available for downstream organs via blood circulation (Conlon and Bird, 2014). However, this route is not only used by essentially beneficial metabolites, but also by potentially harmful products of pathogenic bacteria or pathobionts. However, during a state of healthy homeostasis, or eubiosis, these potentially harmful metabolites are less relevant, since non-pathogenic bacteria outnumber, and thus suppress the growth of pathogenic species (Kamada et al., 2012).
In addition to the influence of host genetics (Bonder et al., 2016), use of antibiotics (Dethlefsen et al., 2008), or immune defense (Wang et al., 2015), the contribution of diet is known to be a key regulator of microbial composition (Ley et al., 2008). Diet begins shaping development of the gut microbiome immediately following birth. For example, components of breast milk (as opposed to formula milk) already affect bacterial gut composition in newborns (Harmsen et al., 2000). During the first three years of life, this composition of bacterial communities evolves towards those found in adults. Furthermore, the composition of fecal microbiota differs between human populations of different origins, highlighting the influence of additional selective pressures, such as dietary habits, hygiene, and general lifestyle, i. e. exercise and smoking.Additionally, gut bacterial diversity increases, in a population-independent manner, during adolescence (Yatsunenko et al., 2012).
The effect of dietary change on the microbiome, as an important environmental factor, depends on the duration of change. Short-term alterations in dietary behavior can influence microbial composition without a large effect on the enterotypes. By contrast, sustained and long-lasting changes to diet are able to affect enterotype states (Wu et al., 2011).
In general, a typical human diet consists of three major components, i. e. carbohydrates, proteins, and fat, which also serve as different substrates for our gut bacteria. In the Western diet these substrates upon reaching the colon, mainly consist of dietary fibers, which comprise non-starch polysaccharides. Additional substrates found here include simple sugars, oligosaccharides, starch, proteins, and lipids (Conlon and Bird, 2014). Importantly, consuming a high amount of dietary fiber is beneficial for health, as it has been shown repeatedly to reduce risk of coronary heart disease (Liu et al., 1999) and diabetes (Montonen et al., 2003). Furthermore, organic acids, the resulting end products of carbohydrate catabolism, serve as additional energy sources for resident bacteria. It is also here where SCFA, in particular, exert a vast number of effects, especially by acting on the immune system once they have been absorbed by the gut epithelium (Conlon and Bird, 2014). However, the direct effects of SCFA on the central nervous system (CNS), need to be better understood.
Dysbiosis and disease
Symbiosis between the commensal microbiome and the innate and adaptive immune system has provided crucial developmental advantages in eukaryote evolution. Hence, it is not surprising, that a dysbalance of the microbiome composition, also denoted as dysbiosis, exerts detrimental effects on human health and immunity (Levy et al., 2017). A dysbiosis of the gut microbiome can be caused by internal or external factors, such as sleep deprivation, stress, use of antibiotics, or dietary components (Dethlefsen et al., 2008, Bailey et al., 2011, Devkota et al., 2012) and an increasing number of chronic disorders are associated with an altered microbial composition. For instance, high salt intake exacerbates disease activity in several – mainly autoimmune – diseases, mediated by an increase in pro-inflammatory immune cell subsets (Jorg et al., 2016).
The commensal microbiome essentially participates in regulating the immune tolerance (Weiner et al., 2011). As such, regulatory immune components such as regulatory T cells (Treg) are crucial for development of a sufficient immune tolerance, especially towards enteric microbiota (Sakaguchi et al., 2008). Pathobionts occur at low abundances in healthy individuals, but tend to expand in the diseased organism, as is the case for inflammatory bowel disease (IBD). IBD is characterized by chronic inflammation of the gastrointestinal tract (de Souza and Fiocchi, 2016), leading to a compositional shift in the commensal microbiome (Frank et al., 2007, Lupp et al., 2007, Butto et al., 2015).
Besides gastrointestinal and autoimmune diseases, a dysbiotic state is currently discussed in neurodegenerative disorders. An important microbiome-related metabolic pathway – the kynurenine pathway – for example, was shown to be associated with several neurodegenerative and neuroinflammatory diseases, and depression (Lombardi et al., 2018). For decades, the focus in chronic diseases, such as autoimmune and neurodegenerative diseases has been on the association with a genetic predisposition. However, more recently the direct impact of environmental factors like the microbiome composition has gained attention (Chen et al., 2016b). The microbiome involvement is further supported by animal studies, showing an attenuation or even the complete absence of neurologic disease, once the animals were kept under bacteria-free conditions (Wu et al., 2010, Berer et al., 2011). Human migration studies have demonstrated that multiple sclerosis (MS) incidence increases in subjects who move from low risk countries to countries with higher MS prevalence, usually countries far north of the equator (Gale and Martyn, 1995). This notion initially led to the theory of latitude, i. e. reduced sun exposure being the major risk factor, not considering dietary habits. How certain components of the diet may have an impact on neuroinflammation and – degeneration was recently shown for fatty acids, especially the differential roles of short versus long chain fatty acids (SCFA, LCFA). We could show, that administration of LCFA in the experimental model of MS worsened disease course via polarization towards T-helper (Th) 1 and Th17 cells, whereas the SCFA propionic acid diminished clinical symptoms due to an increase of intestine-derived Treg (Haghikia et al., 2015).

Microbiome metabolites differentially affect the immune and the central nervous system Short chain fatty acids (SCFA) are metabolites, originating from the fermentation of fiber-rich diet by the commensal gut bacteria. SCFA serve as an energy source for gut epithelia, but also pass from the gut lumen into blood circulation and interact with immune cells within the lamina propria cells of the submucosal nerve plexus. The impact of short chain fatty acids within the CNS is indirect in the case of a shifted immune cell balance towards Treg. Especially in diseases with a disrupted blood-brain-barrier, such as MS, changes in CNS-resident immune cells can directly affect the CNS. BA, butyric acid, PA, propionic acid, AA, acetic acid.
Additionally, the observed conversion to a rather anti-inflammatory environment by propionic acid treatment is accompanied with a decrease in demyelination and less axonal loss, which we observed during disease course in mice by Luxol Fast Blue and Bielschowsky silver staining (Haghikia et al., 2015). The question, if these neuroprotective effects are only mediated by reduced inflammation or via a local action of propionic acid on CNS cells, remains unanswered. A recent study has shown that Treg are able to directly increase neuronal remyelination and oligodendrocyte differentiation, thereby affecting remyelination processes (Dombrowski et al., 2017). In addition, anti-inflammatory cytokines like interleukin-10 are capable to trigger neuroregeneration (Chen et al., 2016a). There is cumulative evidence for the neuroprotective capacity of (mainly regulatory) immune-mediated processes, nevertheless, various findings point to a direct neuroprotective effect by commensal metabolic components. However, these components can also exert damaging effects on neurons, especially in high concentrations. In animal models investigating autism-like diseases, administration of high dose propionic acid (e. g. 500 mg/kg body weight), provoked autism-like behavior (Macfabe, 2012, Choi et al., 2018). This corresponds with findings within the disease propionic-acidemia, which is characterized by a reduced activity of the propionyl-CoA carboxylase, leading to an impaired metabolism of propionic acid, accompanied with propionic acid accumulation. This observed autism-like behavior is supposed to be triggered by an increased inhibitory GABA-ergic neurotransmission (Morland et al., 2018).
Apart from secondary neurodegeneration as a result of inflammation, in patients with Parkinson’s disease, a dysbiosed microbiota has been observed, that is characterized by a reduced abundance of Prevotellaceae. Furthermore, changes in Parkinson’s patients’ microbiome also correlate with disease progression (Minato et al., 2017): the relative abundance of Enterobacteriaceae is associated with motor dysfunction (Scheperjans et al., 2015). Additionally, psychiatric diseases such as depression and autism spectrum disorders (ASD) have also been linked to alterations of the gut microbiome (Kang et al., 2013, Jiang et al., 2015). By high-throughput pyrosequencing of bacterial genomic DNA extracted from fecal samples of patients with major depressive disorder, an increase in the levels of Bacterioidetes, Proteobacteria, and Actinobacteria was found, while the level of Firmicutes was decreased in comparison to healthy controls. Furthermore, the severity of depressive symptoms is inversely correlated with a reduction of Faecalibacterium. ASD have also been linked to a less diverse gut microbiome and to alterations in different bacterial genera, namely reduced abundances of Prevotella, Coprococcus, and unclassified Veillonellaceae (Kang et al., 2013), as well as to gastrointestinal disruption that correlates with disease severity (Adams et al., 2011). Therefore, the effect of microbiota transfer therapy has been investigated in a clinical trial. After a 2-week antibiotic treatment followed by bowel cleanse and a subsequent performance of fecal microbiota transplantation, patients displayed a reduction of gastrointestinal symptoms, accompanied by a significant improvement in behavioral ASD symptoms (Kang et al., 2017). Although this example promises hope for fighting other diseases that have been associated with an altered microbiome, it also needs to be assessed carefully. Since most of the microbiome sequencing studies lack a temporal and causal relationship between microbial alterations and disease initiation, the use of probiotics and fecal transplant therapies is still a matter of debate. It is not yet understood, if depleting disease-associated pathogens, by probiotics or fecal transplants, will have any positive effect on these diseases (Khoruts and Sadowsky, 2016).
In contrast to the aforementioned correlations between an altered gut microbiome and neurologic diseases, a healthy diet is associated with lower risk of first clinical diagnosis of central nervous system demyelination; a healthy diet defined as rich in vegetables, legumes, fish, poultry, and eggs (Black et al., 2018).
Interactions between gut and the brain
The term gut-brain axis has emerged from observing the direct effects of gut metabolites on the CNS and accordingly, the interaction between the gut and the enteric nervous system is well worked out. Classically, the interaction has long been understood as a “ONE WAY” process concerning the regulation of gastrointestinal functions by the brain. The sympathetic and parasympathetic nervous system modulates gut function, e. g. motility or secretion of several components into the gut lumen (Rhee et al., 2009). However, the impact is reciprocal, and this facet has only recently gained attention (Mayer et al., 2014). Besides the beneficial effects of gut-derived SCFA on the CNS via the immune system, other bacterial products have long been known to directly manipulate neurons. Botulinum and tetanus neurotoxins are bacterial toxins, which directly exert severe damage in neurons. Botulinum toxin blocks synaptic vesicle fusion and the release of neurotransmitters (Rossetto et al., 2014), whereas tetanus toxin is internalized into signaling endosomes and retrogradely transported to the neuronal soma, where it blocks neurotransmission (Calvo et al., 2012, Yang and Chiu, 2017). The effects of the many bacterial metabolites on neurons remain to be explored, yet scarce evidence suggests that some could affect CNS neurons up to the level of the dopaminergic reward system (Diaz Heijtz et al., 2011).
These recent findings on the direct effects of gut metabolites on the CNS have initiated a paradigm shift also in drug development programs targeting (De Vadder et al., 2014, Stilling et al., 2016, Hoyles et al., 2018) Parkinson’s, Alzheimer’s and MS diseases (Berer et al., 2011, Hill et al., 2014, Sampson et al., 2016). The molecular mechanisms by which SCFA are able to directly influence cellular processes have also been analyzed in cancer research (Augenlicht et al., 2002, Matthews et al., 2012). These mechanisms are mediated either by receptor activation or by epigenetic modulation. SCFA activate the orphan G-protein coupled receptors (GPR) 41 and 43, also known as free fatty acid receptor (FFAR) 2 and 3 (Brown et al., 2003) among other effects, modulating the induction of immune regulatory mechanisms such as Treg differentiation (Smith et al., 2013). Due to their small molecular size, SCFA have also been shown to directly inhibit epigenetic modifiers like the histone deacetylases (HDAC) of class I and II (Candido et al., 1978, Davie, 2003, Harrison and Dexter, 2013). For instance, the key players active in maintaining homeostasis of lysine acetylation are histone acetyltransferases (HATs) and HDACs. HATs catalyze the transfer of an acetyl-group from acetyl-CoA onto lysine residues of histone proteins. This process leads to a relaxation of nuclear chromatin structure. By contrast, HDACs remove acetyl groups from lysine residues, causing chromatin condensation. Changing the conformation of the chromatin framework either increases (relaxation) or decreases (condensation) transcriptional processes (Chuang et al., 2009). HDAC inhibitors promote chromatin relaxation and thus translational activation. Since acetylation is not exclusively limited to chromatin but also in various proteins as post-translational modifications, HDAC inhibitors may exert varying effects on cellular processes. These effects include modulation of protein expression and function, mitochondrial behavior, intracellular transport, and metabolic processes (Kazantsev and Thompson, 2008). Protein post-translational modifications are discussed as important drivers of neurodegenerative diseases, since they could serve as a link for the gap between environmental factors and genetic disease susceptibility. An imbalance of HAT and HDAC activity is considered to favor neurodegenerative conditions (Chuang et al., 2009). Assuming that SCFA manipulate neurons by HDAC inhibition, our daily diet may have a greater impact on the development of, not just autoimmune diseases, but also neurodegenerative disorders. Beneficial effects of chromatin remodeling have already been shown in an Alzheimer’s disease model, using the HDAC inhibitor sodium 4-phenylbutyrate (Ricobaraza et al., 2009). Although there is sparse evidence that SCFA induce direct neuroregenerative effects, an altered HDAC activity has already been proven to participate in processes including neuronal damage. For instance, nuclear export of HDAC1 induces axonal damage that leads to to neuronal death (Kim et al., 2010). Hence SCFA, like known HDAC I and II inhibitors, may prove to have therapeutic potential.
Potentially indirect neuroprotective effects of SCFA include the secretion of anti-inflammatory cytokines such as interleukin-10 from Tregs, and interleukin-4 from Th2 cells. These cytokines have been shown to protect damaged neurons and synapse formation after brain injury (Siffrin et al., 2010, Chen et al., 2016a, Vogelaar et al., 2018).
The presence and composition of our commensal microbiome not only participates in normal gastrointestinal function, but may also influence development of the brain, its function and the occurrence of its diseases. This is important, not only considering maintenance of a healthy diet in order to promote proper brain functioning, but most importantly for expecting mothers, who by maintaining a balanced diet can encourage a child’s normal brain development during pregnancy. It was demonstrated in mice that a maternal diet high in fat negatively impacts the offspring’s social behavior, caused by a reduction of oxytocin-immunoreactive neurons and a reduction of long term potentiation in dopaminergic neurons within the ventral tegmental area (Buffington et al., 2016). By contrast, a diet containing omega-3 polyunsaturated fatty acids has beneficial impacts on neurodevelopment by influencing the hypothalamic-pituitary-adrenal (HPA) axis. Since the HPA axis mainly determines stress reactivity, these metabolic components prevent depressive-like behaviors due to better stress resistance (Pusceddu et al., 2015). Hence microbiome manipulation, either by diet or by specific prebiotics, may open new therapeutic avenues for treating various systemic diseases including neurodegeneration.
About the authors

2010–2013: Bachelor of Science in Biology, Faculty of Biology and Biotechnology, Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Germany
2013–2015: Master of Science in Biology with focus on neurobiology, Faculty of Medicine, Neuroimmunology, Ruhr-University Bochum, Germany
since 2015: PhD study in Biology with focus on the direct effects of gut metabolites (i. e. short chain fatty acids) on neurons in the context of Multiple Sclerosis, Faculty of Medicine, Neuroimmunology, Ruhr-University Bochum, Germany

Aiden Haghikia hat sein Medizinstudium an der Ruhr-Universität Bochum abgeschlossen, wo er im Rahmen des neurowissenschaftlichen Graduierten Kolleg am Lehrstuhl für Neuroanatomie (Prof. Rolf Dermietzel) seine Promotionsarbeit zur funktionellen Charakterisierung von astrozytären Gap Junctions durchführte. Nach seiner Facharztausbildung in der Neurologischen Klinik der Ruhr-Universität Bochum im St. Josef-Hospital (Leitung Prof. Gold) arbeitete er als Postdoc im Labor von Prof. Lars Fugger an der University of Oxford (Neuroimmunogenetik). Er hält eine Universitätsprofessur für Translationale Neuroimmunologie inne und ist seit 2017 leitender Oberarzt der Neurologischen Klinik, Leiter der MS-Ambulanz/neuroimmunologischen Forschung in Bochum.

2010–2013: Bachelor of Science in Biology, Faculty of Biology and Biotechnology, Developmental Neurobiology, Ruhr-University Bochum, Germany
2013–2015: Master of Science in Biology with focus on neurobiology, Faculty of Medicine, Neuroimmunology, Ruhr-University Bochum, Germany
since 2015: PhD study in Biology with focus on neuroregeneration and neurodegeneration in Multiple Sclerosis, Faculty of Medicine, Neuroimmunology, Ruhr-University Bochum, Germany
References
Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J (2014) The placenta harbors a unique microbiome. Science translational medicine 6:237ra265.10.1126/scitranslmed.3008599Search in Google Scholar PubMed PubMed Central
Adams JB, Johansen LJ, Powell LD, Quig D, Rubin RA (2011) Gastrointestinal flora and gastrointestinal status in children with autism--comparisons to typical children and correlation with autism severity. BMC gastroenterology 11:22.10.1186/1471-230X-11-22Search in Google Scholar PubMed PubMed Central
Ajouz H, Mukherji D, Shamseddine A (2014) Secondary bile acids: an underrecognized cause of colon cancer. World journal of surgical oncology 12:164.10.1186/1477-7819-12-164Search in Google Scholar PubMed PubMed Central
Archibald JM (2015) Endosymbiosis and Eukaryotic Cell Evolution. Current biology: CB 25:R911–921.10.1016/j.cub.2015.07.055Search in Google Scholar PubMed
Augenlicht LH, Mariadason JM, Wilson A, Arango D, Yang W, Heerdt BG, Velcich A (2002) Short chain fatty acids and colon cancer. The Journal of nutrition 132:3804S–3808S.10.1093/jn/132.12.3804SSearch in Google Scholar PubMed
Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y, Xia Y, Xie H, Zhong H, Khan MT, Zhang J, Li J, Xiao L, Al-Aama J, Zhang D, Lee YS, Kotowska D, Colding C, Tremaroli V, Yin Y, Bergman S, Xu X, Madsen L, Kristiansen K, Dahlgren J, Wang J (2015) Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell host & microbe 17:690–703.10.1016/j.chom.2015.04.004Search in Google Scholar PubMed
Bailey MT, Dowd SE, Galley JD, Hufnagle AR, Allen RG, Lyte M (2011) Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation. Brain, behavior, and immunity 25:397–407.10.1016/j.bbi.2010.10.023Search in Google Scholar PubMed PubMed Central
Berer K, Mues M, Koutrolos M, Rasbi ZA, Boziki M, Johner C, Wekerle H, Krishnamoorthy G (2011) Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479:538–541.10.1038/nature10554Search in Google Scholar PubMed
Black LJ, Rowley C, Sherriff J, Pereira G, Ponsonby AL, Lucas RM, Ausimmune Investigator G (2018) A healthy dietary pattern associates with a lower risk of a first clinical diagnosis of central nervous system demyelination. Multiple sclerosis 1352458518793524.10.1177/1352458518793524Search in Google Scholar PubMed
Bonder MJ, Kurilshikov A, Tigchelaar EF, Mujagic Z, Imhann F, Vila AV, Deelen P, Vatanen T, Schirmer M, Smeekens SP, Zhernakova DV, Jankipersadsing SA, Jaeger M, Oosting M, Cenit MC, Masclee AA, Swertz MA, Li Y, Kumar V, Joosten L, Harmsen H, Weersma RK, Franke L, Hofker MH, Xavier RJ, Jonkers D, Netea MG, Wijmenga C, Fu J, Zhernakova A (2016) The effect of host genetics on the gut microbiome. Nature genetics 48:1407–1412.10.1038/ng.3663Search in Google Scholar PubMed
Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ, Pike NB, Strum JC, Steplewski KM, Murdock PR, Holder JC, Marshall FH, Szekeres PG, Wilson S, Ignar DM, Foord SM, Wise A, Dowell SJ (2003) The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. The Journal of biological chemistry 278:11312–11319.10.1074/jbc.M211609200Search in Google Scholar
Buffington SA, Di Prisco GV, Auchtung TA, Ajami NJ, Petrosino JF, Costa-Mattioli M (2016) Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 165:1762–1775.10.1016/j.cell.2016.06.001Search in Google Scholar
Butto LF, Schaubeck M, Haller D (2015) Mechanisms of Microbe-Host Interaction in Crohn’s Disease: Dysbiosis vs. Pathobiont Selection. Frontiers in immunology 6:555.10.3389/fimmu.2015.00555Search in Google Scholar
Calvo AC, Olivan S, Manzano R, Zaragoza P, Aguilera J, Osta R (2012) Fragment C of tetanus toxin: new insights into its neuronal signaling pathway. International journal of molecular sciences 13:6883–6901.10.3390/ijms13066883Search in Google Scholar
Candido EP, Reeves R, Davie JR (1978) Sodium butyrate inhibits histone deacetylation in cultured cells. Cell 14:105–113.10.1016/0092-8674(78)90305-7Search in Google Scholar
Chen H, Lin W, Zhang Y, Lin L, Chen J, Zeng Y, Zheng M, Zhuang Z, Du H, Chen R, Liu N (2016a) IL-10 Promotes Neurite Outgrowth and Synapse Formation in Cultured Cortical Neurons after the Oxygen-Glucose Deprivation via JAK1/STAT3 Pathway. Scientific reports 6:30459.10.1038/srep30459Search in Google Scholar PubMed PubMed Central
Chen J, Chia N, Kalari KR, Yao JZ, Novotna M, Paz Soldan MM, Luckey DH, Marietta EV, Jeraldo PR, Chen X, Weinshenker BG, Rodriguez M, Kantarci OH, Nelson H, Murray JA, Mangalam AK (2016b) Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Scientific reports 6:28484.10.1038/srep28484Search in Google Scholar PubMed PubMed Central
Choi J, Lee S, Won J, Jin Y, Hong Y, Hur TY, Kim JH, Lee SR, Hong Y (2018) Pathophysiological and neurobehavioral characteristics of a propionic acid- mediated autism-like rat model. PloS one 13:e0192925.Search in Google Scholar
Chuang DM, Leng Y, Marinova Z, Kim HJ, Chiu CT (2009) Multiple roles of HDAC inhibition in neurodegenerative conditions. Trends in neurosciences 32:591–601.10.1016/j.tins.2009.06.002Search in Google Scholar PubMed PubMed Central
Conlon MA, Bird AR (2014) The impact of diet and lifestyle on gut microbiota and human health. Nutrients 7:17–44.10.3390/nu7010017Search in Google Scholar PubMed PubMed Central
Davie JR (2003) Inhibition of histone deacetylase activity by butyrate. The Journal of nutrition 133:2485S-2493S.10.1093/jn/133.7.2485SSearch in Google Scholar PubMed
de Souza HS, Fiocchi C (2016) Immunopathogenesis of IBD: current state of the art. Nature reviews Gastroenterology & hepatology 13:13–27.10.1038/nrgastro.2015.186Search in Google Scholar
De Vadder F, Kovatcheva-Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, Backhed F, Mithieux G (2014) Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 156:84–96.10.1016/j.cell.2013.12.016Search in Google Scholar
Dethlefsen L, Huse S, Sogin ML, Relman DA (2008) The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS biology 6:e280.10.1371/journal.pbio.0060280Search in Google Scholar
Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, Nadimpalli A, Antonopoulos DA, Jabri B, Chang EB (2012) Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/-mice. Nature 487:104–108.10.1038/nature11225Search in Google Scholar
Diaz Heijtz R, Wang S, Anuar F, Qian Y, Bjorkholm B, Samuelsson A, Hibberd ML, Forssberg H, Pettersson S (2011) Normal gut microbiota modulates brain development and behavior. Proceedings of the National Academy of Sciences of the United States of America 108:3047–3052.10.1073/pnas.1010529108Search in Google Scholar
Dombrowski Y, O’Hagan T, Dittmer M, Penalva R, Mayoral SR, Bankhead P, Fleville S, Eleftheriadis G, Zhao C, Naughton M, Hassan R, Moffat J, Falconer J, Boyd A, Hamilton P, Allen IV, Kissenpfennig A, Moynagh PN, Evergren E, Perbal B, Williams AC, Ingram RJ, Chan JR, Franklin RJM, Fitzgerald DC (2017) Regulatory T cells promote myelin regeneration in the central nervous system. Nature neuroscience 20:674–680.10.1038/nn.4528Search in Google Scholar
Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R (2010) Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proceedings of the National Academy of Sciences of the United States of America 107:11971–11975.10.1073/pnas.1002601107Search in Google Scholar
Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR (2007) Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proceedings of the National Academy of Sciences of the United States of America 104:13780–13785.10.1073/pnas.0706625104Search in Google Scholar
Fresia Fernandez MDC (1987) Vitamin K composition of anaerobic gut bacteria. FEMS Microbiology Letters 41:175–180.10.1111/j.1574-6968.1987.tb02191.xSearch in Google Scholar
Gale CR, Martyn CN (1995) Migrant studies in multiple sclerosis. Progress in neurobiology 47:425–448.10.1016/0301-0082(95)80008-VSearch in Google Scholar
Haghikia A, Jorg S, Duscha A, Berg J, Manzel A, Waschbisch A, Hammer A, Lee DH, May C, Wilck N, Balogh A, Ostermann AI, Schebb NH, Akkad DA, Grohme DA, Kleinewietfeld M, Kempa S, Thone J, Demir S, Muller DN, Gold R, Linker RA (2015) Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine. Immunity 43:817–829.10.1016/j.immuni.2015.09.007Search in Google Scholar PubMed
Harmsen HJ, Wildeboer-Veloo AC, Raangs GC, Wagendorp AA, Klijn N, Bindels JG, Welling GW (2000) Analysis of intestinal flora development in breast-fed and formula-fed infants by using molecular identification and detection methods. Journal of pediatric gastroenterology and nutrition 30:61–67.10.1097/00005176-200001000-00019Search in Google Scholar PubMed
Harrison IF, Dexter DT (2013) Epigenetic targeting of histone deacetylase: therapeutic potential in Parkinson’s disease? Pharmacology & therapeutics 140:34–52.10.1016/j.pharmthera.2013.05.010Search in Google Scholar PubMed
Hill JM, Clement C, Pogue AI, Bhattacharjee S, Zhao Y, Lukiw WJ (2014) Pathogenic microbes, the microbiome, and Alzheimer’s disease (AD). Frontiers in aging neuroscience 6:127.Search in Google Scholar
Hoyles L, Snelling T, Umlai UK, Nicholson JK, Carding SR, Glen RC, McArthur S (2018) Microbiome-host systems interactions: protective effects of propionate upon the blood-brain barrier. Microbiome 6:55.10.1186/s40168-018-0439-ySearch in Google Scholar PubMed PubMed Central
Huurre A, Kalliomaki M, Rautava S, Rinne M, Salminen S, Isolauri E (2008) Mode of delivery – effects on gut microbiota and humoral immunity. Neonatology 93:236–240.10.1159/000111102Search in Google Scholar PubMed
Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J, Li L, Ruan B (2015) Altered fecal microbiota composition in patients with major depressive disorder. Brain, behavior, and immunity 48:186–194.10.1016/j.bbi.2015.03.016Search in Google Scholar PubMed
Jorg S, Kissel J, Manzel A, Kleinewietfeld M, Haghikia A, Gold R, Muller DN, Linker RA (2016) High salt drives Th17 responses in experimental autoimmune encephalomyelitis without impacting myeloid dendritic cells. Experimental neurology 279:212–222.10.1016/j.expneurol.2016.03.010Search in Google Scholar PubMed
Kamada N, Kim YG, Sham HP, Vallance BA, Puente JL, Martens EC, Nunez G (2012) Regulated virulence controls the ability of a pathogen to compete with the gut microbiota. Science 336:1325–1329.10.1126/science.1222195Search in Google Scholar PubMed PubMed Central
Kang DW, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, Khoruts A, Geis E, Maldonado J, McDonough-Means S, Pollard EL, Roux S, Sadowsky MJ, Lipson KS, Sullivan MB, Caporaso JG, Krajmalnik-Brown R (2017) Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome 5:10.10.1186/s40168-016-0225-7Search in Google Scholar PubMed PubMed Central
Kang DW, Park JG, Ilhan ZE, Wallstrom G, Labaer J, Adams JB, Krajmalnik-Brown R (2013) Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. PloS one 8:e68322.10.1371/journal.pone.0068322Search in Google Scholar PubMed PubMed Central
Kazantsev AG, Thompson LM (2008) Therapeutic application of histone deacetylase inhibitors for central nervous system disorders. Nature reviews Drug discovery 7:854–868.10.1038/nrd2681Search in Google Scholar PubMed
Khoruts A, Sadowsky MJ (2016) Understanding the mechanisms of faecal microbiota transplantation. Nature reviews Gastroenterology & hepatology 13:508–516.10.1038/nrgastro.2016.98Search in Google Scholar PubMed PubMed Central
Kim JY, Shen S, Dietz K, He Y, Howell O, Reynolds R, Casaccia P (2010) HDAC1 nuclear export induced by pathological conditions is essential for the onset of axonal damage. Nature neuroscience 13:180–189.10.1038/nn.2471Search in Google Scholar PubMed PubMed Central
Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E (2017) Dysbiosis and the immune system. Nature reviews Immunology 17:219–232.10.1038/nri.2017.7Search in Google Scholar PubMed
Ley RE, Hamady M, Lozupone C, Turnbaugh PJ, Ramey RR, Bircher JS, Schlegel ML, Tucker TA, Schrenzel MD, Knight R, Gordon JI (2008) Evolution of mammals and their gut microbes. Science 320:1647–1651.10.1126/science.1155725Search in Google Scholar PubMed PubMed Central
Liu S, Stampfer MJ, Hu FB, Giovannucci E, Rimm E, Manson JE, Hennekens CH, Willett WC (1999) Whole-grain consumption and risk of coronary heart disease: results from the Nurses’ Health Study. The American journal of clinical nutrition 70:412–419.10.1093/ajcn/70.3.412Search in Google Scholar PubMed
Lombardi VC, De Meirleir KL, Subramanian K, Nourani SM, Dagda RK, Delaney SL, Palotas A (2018) Nutritional modulation of the intestinal microbiota; future opportunities for the prevention and treatment of neuroimmune and neuroinflammatory disease. The Journal of nutritional biochemistry 61:1–16.10.1016/j.jnutbio.2018.04.004Search in Google Scholar PubMed PubMed Central
Lupp C, Robertson ML, Wickham ME, Sekirov I, Champion OL, Gaynor EC, Finlay BB (2007) Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell host & microbe 2:119–129.10.1016/j.chom.2007.06.010Search in Google Scholar PubMed
Macfabe DF (2012) Short-chain fatty acid fermentation products of the gut microbiome: implications in autism spectrum disorders. Microbial ecology in health and disease 23.10.3402/mehd.v23i0.19260Search in Google Scholar PubMed PubMed Central
Matthews GM, Howarth GS, Butler RN (2012) Short-chain fatty acids induce apoptosis in colon cancer cells associated with changes to intracellular redox state and glucose metabolism. Chemotherapy 58:102–109.10.1159/000335672Search in Google Scholar PubMed
Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K (2014) Gut microbes and the brain: paradigm shift in neuroscience. The Journal of neuroscience: the official journal of the Society for Neuroscience 34:15490–15496.10.1523/JNEUROSCI.3299-14.2014Search in Google Scholar PubMed PubMed Central
Minato T, Maeda T, Fujisawa Y, Tsuji H, Nomoto K, Ohno K, Hirayama M (2017) Progression of Parkinson’s disease is associated with gut dysbiosis: Two-year follow-up study. PloS one 12:e0187307.10.1371/journal.pone.0187307Search in Google Scholar PubMed PubMed Central
Montonen J, Knekt P, Jarvinen R, Aromaa A, Reunanen A (2003) Whole-grain and fiber intake and the incidence of type 2 diabetes. The American journal of clinical nutrition 77:622–629.10.1093/ajcn/77.3.622Search in Google Scholar PubMed
Morland C, Froland AS, Pettersen MN, Storm-Mathisen J, Gundersen V, Rise F, Hassel B (2018) Propionate enters GABAergic neurons, inhibits GABA transaminase, causes GABA accumulation and lethargy in a model of propionic acidemia. The Biochemical journal 475:749–758.10.1042/BCJ20170814Search in Google Scholar PubMed
Pusceddu MM, El Aidy S, Crispie F, O’Sullivan O, Cotter P, Stanton C, Kelly P, Cryan JF, Dinan TG (2015) N-3 Polyunsaturated Fatty Acids (PUFAs) Reverse the Impact of Early-Life Stress on the Gut Microbiota. PloS one 10:e0139721.Search in Google Scholar
Raina JB, Eme L, Pollock FJ, Spang A, Archibald JM, Williams TA (2018) Symbiosis in the microbial world: from ecology to genome evolution. Biology open 7.10.1242/bio.032524Search in Google Scholar PubMed PubMed Central
Rhee SH, Pothoulakis C, Mayer EA (2009) Principles and clinical implications of the brain-gut-enteric microbiota axis. Nature reviews Gastroenterology & hepatology 6:306–314.10.1038/nrgastro.2009.35Search in Google Scholar PubMed PubMed Central
Ricobaraza A, Cuadrado-Tejedor M, Perez-Mediavilla A, Frechilla D, Del Rio J, Garcia-Osta A (2009) Phenylbutyrate ameliorates cognitive deficit and reduces tau pathology in an Alzheimer’s disease mouse model. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology 34:1721–1732.10.1038/npp.2008.229Search in Google Scholar PubMed
Rossetto O, Pirazzini M, Montecucco C (2014) Botulinum neurotoxins: genetic, structural and mechanistic insights. Nature reviews Microbiology 12:535–549.10.1038/nrmicro3295Search in Google Scholar PubMed
Sakaguchi S, Yamaguchi T, Nomura T, Ono M (2008) Regulatory T cells and immune tolerance. Cell 133:775–787.10.1016/j.cell.2008.05.009Search in Google Scholar PubMed
Salminen S, Bouley C, Boutron-Ruault MC, Cummings JH, Franck A, Gibson GR, Isolauri E, Moreau MC, Roberfroid M, Rowland I (1998) Functional food science and gastrointestinal physiology and function. The British journal of nutrition 80 Suppl 1:S147–171.10.1079/BJN19980108Search in Google Scholar
Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V, Chesselet MF, Keshavarzian A, Shannon KM, Krajmalnik-Brown R, Wittung-Stafshede P, Knight R, Mazmanian SK (2016) Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 167:1469–1480 e1412.10.1016/j.cell.2016.11.018Search in Google Scholar PubMed PubMed Central
Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, Haapaniemi E, Kaakkola S, Eerola-Rautio J, Pohja M, Kinnunen E, Murros K, Auvinen P (2015) Gut microbiota are related to Parkinson’s disease and clinical phenotype. Movement disorders: official journal of the Movement Disorder Society 30:350–358.10.1002/mds.26069Search in Google Scholar PubMed
Sender R, Fuchs S, Milo R (2016) Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS biology 14:e1002533.10.1371/journal.pbio.1002533Search in Google Scholar PubMed PubMed Central
Siffrin V, Radbruch H, Glumm R, Niesner R, Paterka M, Herz J, Leuenberger T, Lehmann SM, Luenstedt S, Rinnenthal JL, Laube G, Luche H, Lehnardt S, Fehling HJ, Griesbeck O, Zipp F (2010) In vivo imaging of partially reversible th17 cell- induced neuronal dysfunction in the course of encephalomyelitis. Immunity 33:424–436.10.1016/j.immuni.2010.08.018Search in Google Scholar PubMed
Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341:569–573.10.1126/science.1241165Search in Google Scholar PubMed PubMed Central
Stilling RM, Bordenstein SR, Dinan TG, Cryan JF (2014) Friends with social benefits: host-microbe interactions as a driver of brain evolution and development? Frontiers in cellular and infection microbiology 4:147.10.3389/fcimb.2014.00147Search in Google Scholar PubMed PubMed Central
Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF (2016) The neuropharmacology of butyrate: The bread and butter of the microbiota-gut- brain axis? Neurochemistry international 99:110–132.10.1016/j.neuint.2016.06.011Search in Google Scholar PubMed
Tap J, Mondot S, Levenez F, Pelletier E, Caron C, Furet JP, Ugarte E, Munoz-Tamayo R, Paslier DL, Nalin R, Dore J, Leclerc M (2009) Towards the human intestinal microbiota phylogenetic core. Environmental microbiology 11:2574–2584.10.1111/j.1462-2920.2009.01982.xSearch in Google Scholar PubMed
Toscano M, De Grandi R, Peroni DG, Grossi E, Facchin V, Comberiati P, Drago L (2017) Impact of delivery mode on the colostrum microbiota composition. BMC microbiology 17:205.10.1186/s12866-017-1109-0Search in Google Scholar PubMed PubMed Central
Vogelaar CF, Mandal S, Lerch S, Birkner K, Birkenstock J, Buhler U, Schnatz A, Raine CS, Bittner S, Vogt J, Kipnis J, Nitsch R, Zipp F (2018) Fast direct neuronal signaling via the IL-4 receptor as therapeutic target in neuroinflammation. Science translational medicine 10.10.1126/scitranslmed.aao2304Search in Google Scholar PubMed
Wang S, Charbonnier LM, Noval Rivas M, Georgiev P, Li N, Gerber G, Bry L, Chatila TA (2015) MyD88 Adaptor-Dependent Microbial Sensing by Regulatory T Cells Promotes Mucosal Tolerance and Enforces Commensalism. Immunity 43:289–303.10.1016/j.immuni.2015.06.014Search in Google Scholar PubMed PubMed Central
Weiner HL, da Cunha AP, Quintana F, Wu H (2011) Oral tolerance. Immunological reviews 241:241–259.10.1111/j.1600-065X.2011.01017.xSearch in Google Scholar PubMed PubMed Central
Willyard C (2018) Could baby’s first bacteria take root before birth? Nature 553:264–266.10.1038/d41586-018-00664-8Search in Google Scholar PubMed
Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, Sinha R, Gilroy E, Gupta K, Baldassano R, Nessel L, Li H, Bushman FD, Lewis JD (2011) Linking long-term dietary patterns with gut microbial enterotypes. Science 334:105–108.10.1126/science.1208344Search in Google Scholar PubMed PubMed Central
Wu HJ, Ivanov, II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C, Mathis D (2010) Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32:815–827.10.1016/j.immuni.2010.06.001Search in Google Scholar PubMed PubMed Central
Xu J, Bjursell MK, Himrod J, Deng S, Carmichael LK, Chiang HC, Hooper LV, Gordon JI (2003) A genomic view of the human-Bacteroides thetaiotaomicron symbiosis. Science 299:2074–2076.10.1126/science.1080029Search in Google Scholar PubMed
Yang NJ, Chiu IM (2017) Bacterial Signaling to the Nervous System through Toxins and Metabolites. Journal of molecular biology 429:587–605.10.1016/j.jmb.2016.12.023Search in Google Scholar PubMed PubMed Central
Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI (2012) Human gut microbiome viewed across age and geography. Nature 486:222–227.10.1038/nature11053Search in Google Scholar PubMed PubMed Central
© 2018 Walter de Gruyter GmbH, Berlin/Boston