Microbiota-derived short chain fatty acids modulate microglia and promote Aβ plaque deposition

Alessio Vittorio Colombo, Rebecca Katie Sadler, Gemma Llovera, Vikramjeet Singh, Stefan Roth, Steffanie Heindl, Laura Sebastian Monasor, Aswin Verhoeven, Finn Peters, Samira Parhizkar, Frits Kamp, Mercedes Gomez de Aguero, Andrew J MacPherson, Edith Winkler, Jochen Herms, Corinne Benakis, Martin Dichgans, Harald Steiner, Martin Giera, Christian Haass, Sabina Tahirovic, Arthur Liesz, Alessio Vittorio Colombo, Rebecca Katie Sadler, Gemma Llovera, Vikramjeet Singh, Stefan Roth, Steffanie Heindl, Laura Sebastian Monasor, Aswin Verhoeven, Finn Peters, Samira Parhizkar, Frits Kamp, Mercedes Gomez de Aguero, Andrew J MacPherson, Edith Winkler, Jochen Herms, Corinne Benakis, Martin Dichgans, Harald Steiner, Martin Giera, Christian Haass, Sabina Tahirovic, Arthur Liesz

Abstract

Previous studies have identified a crucial role of the gut microbiome in modifying Alzheimer's disease (AD) progression. However, the mechanisms of microbiome-brain interaction in AD were so far unknown. Here, we identify microbiota-derived short chain fatty acids (SCFA) as microbial metabolites which promote Aβ deposition. Germ-free (GF) AD mice exhibit a substantially reduced Aβ plaque load and markedly reduced SCFA plasma concentrations; conversely, SCFA supplementation to GF AD mice increased the Aβ plaque load to levels of conventionally colonized (specific pathogen-free [SPF]) animals and SCFA supplementation to SPF mice even further exacerbated plaque load. This was accompanied by the pronounced alterations in microglial transcriptomic profile, including upregulation of ApoE. Despite increased microglial recruitment to Aβ plaques upon SCFA supplementation, microglia contained less intracellular Aβ. Taken together, our results demonstrate that microbiota-derived SCFA are critical mediators along the gut-brain axis which promote Aβ deposition likely via modulation of the microglial phenotype.

Keywords: alzheimer's disease; amyloid; immunology; inflammation; metabolites; microbiome; microglia; mouse; neuroinflammation; neuroscience.

Conflict of interest statement

AC, RS, GL, VS, SR, SH, LS, AV, FP, SP, FK, MG, EW, JH, CB, MD, HS, MG, CH, ST, AL No competing interests declared, AM Reviewing editor, eLife

© 2021, Colombo et al.

Figures

Figure 1.. Germ-free APPPS1 mice show reduced…
Figure 1.. Germ-free APPPS1 mice show reduced Alzheimer’s disease (AD) pathology.
(A) Representative images and analysis of brain cortices from 5 months old germ-free (GF), naturally recolonized (Rec) and conventionally colonized (specific pathogen-free [SPF]) APPPS1 mice immunostained for Aβ (clone 6E10). Quantification of parenchymal plaque load reveals significantly reduced Aβ burden in GF mice compared to the Rec and SPF groups. Values are expressed as percentages of amyloid plaque area and normalized to GF group (ANOVA; n(GF) = 9, n(Rec) = 6, n(SPF) = 9). (B) Representative results and quantification from Barnes maze behavioral analysis of 5 months old GF, Rec and SPF APPPS1 mice. Quantification of frequency of error zone entering in the Barnes maze reveals a better performance in GF mice compared to Rec and SPF mice (ANOVA; n(GF) = 6, n(Rec) = 8, n(SPF) = 6). (C) Sagittal overview image indicating the analysis ROIs in the frontal cortex (blue: Methoxy-X04-positive plaques) and representative image demonstrating segmentation of Methoxy-X04 fluorescence intensity into individual plaques. The images show maximal intensity projections. Individual plaques are labeled with different colors. (D) Frequency distribution of plaque radius in 5 months old GF and SPF APPPS1 mice (two-way ANOVA; n(GF) = 5, n(SPF) = 5). All data are derived from at least three individual experiments and presented as mean ± SEM.
Figure 1—figure supplement 1.. Sex-specific analysis of…
Figure 1—figure supplement 1.. Sex-specific analysis of amyloid plaque load.
Plaque load of animals depicted in Figure 1A was individually analyzed in male (A; n = 5 per group) and female (B; n = 4 per group) mice, revealing a similar trend for increased plaque load in specific pathogen-free (SPF) mice compared to germ-free (GF) mice. Values are expressed as percentages of amyloid plaque area and normalized to GF group (unpaired T-test; at least three individual experiments).
Figure 2.. Short chain fatty acids (SCFA)…
Figure 2.. Short chain fatty acids (SCFA) are mediators of Aβ plaque deposition.
(A) Plasma SCFA concentrations of acetate, butyrate, and propionate were quantified using GC/MS-based metabolomics analysis, showing an increase in specific pathogen-free (SPF) compared to germ-free (GF) mice (ANOVA; n(GF) = 4, n(Rec) = 5, n(SPF) = 4; two individual experiments). (B) Experimental plan for SCFA supplementation in GF mice. Four weeks old GF mice have been treated with SCFA in drinking water for 8 weeks. (C) Representative images from GF (control-treatment), Rec, and GF supplemented with SCFA mice showing a significant increase in Aβ plaque load upon SCFA administration (ANOVA; n(GF) = 7, n(Rec) = 9, n(GF+SCFA) = 6; three individual experiments). (D) SPF APPPS1 mice were supplemented with control or SCFA in drinking water for 4 weeks (from 8 to 12 weeks of age). Histological analysis revealed a significantly increased plaque load in SCFA-supplemented SPF mice compared to control treatment (unpaired T-test; n(Ctrl) = 4, n(SCFA) = 5; one individual experiment). All data in this figure are presented as mean ± SEM.
Figure 2—figure supplement 1.. Total short chain…
Figure 2—figure supplement 1.. Total short chain fatty acids (SCFA) concentrations.
Total plasma SCFA concentrations of acetate, butyrate, and propionate in specific pathogen-free (SPF), germ-free (GF), and GF+SCFA treated mice (ANOVA; n(GF) = 5, n(SPF) = 5, n(GF+SCFA) = 4; two individual experiments). Data are presented as mean ± SEM.
Figure 3.. Short chain fatty acids (SCFA)…
Figure 3.. Short chain fatty acids (SCFA) mildly increase amyloidogenic processing.
(A) Western blot analysis and (B) its densitometry quantification of 3 months old brain homogenates of control (Ctrl)- and SCFA-supplemented germ-free (GF) APPPS1 animals. The Aβ level is significantly increased in SCFA group in comparison to Ctrl, despite unaffected APP FL levels. APP CTFs show a decreased C83 (**) to C99 (*) ratio. We could not detect alterations in protein levels of secretases involved in APP processing (ADAM10, BACE1, and γ-secretase/PSEN1). m = ADAM10 mature form; im = ADAM10 immature form. Data represent mean ± SEM (unpaired T-test; n(Ctrl) = 3, n(SCFA) = 3). (C) Upper panel: γ-Secretase reconstituted into lipid vesicles was incubated at 37°C together with the C99-based substrate C100-His6 in the presence of increasing doses of a SCFA mixture (375 and 750 μM final concentration of total SCFA of an equimolar mixture of Na-acetate, Na-propionate, and Na-butyrate) for 24 hr. Production of Aβ was analyzed by immunoblotting. γ-Secretase inhibitor (GSI) L-685,458 (0.4 μM) was used as a negative control. No alterations in Aβ levels were detected in the presence of SCFA. Lower panel: Qualitative analysis of individual Aβ species via Tris‐Bicine‐Urea SDS‐PAGE reveals that SCFA treatment does not alter the ratio among the different Aβ species (Aβ37-38-40-42/43) suggesting no direct effects on modulation of γ-secretase cleavage. (D) Aggregation kinetics of monomeric Aβ40 recorded by the increase in fluorescence of Thioflavin T incubated with either 30 mM NaCl (Ctrl) or 30 mM SCFA mixture do not show any significant difference, suggesting that SCFA do not directly modify Aβ fibrillarization. Data points represent mean ± SD from three independent experiments.
Figure 4.. Short chain fatty acids (SCFA)…
Figure 4.. Short chain fatty acids (SCFA) modulate microglia.
(A) Morphological analysis of microglia shows an increase in the circularity index, indicating a more activated phenotype, in SCFA- compared to control-treated germ-free (GF) APPPS1 mice. Iba1 (red) has been used as microglial marker. For each group, each shade of color represents the microglia from a single mouse (U test; n(Ctrl) = 6, n(SCFA) = 5). (B) smFISH analysis of microglial cells (red, Cx3cr1 mRNA particles) surrounding amyloid plaques (green, anti-Aβ clone 2D8) shows an increased number of Cx3cr1-positive cells (>4 puncta) clustering around Aβ plaques in SCFA- compared to control-supplemented GF APPPS1 mice. DAPI (blue) was used as nuclear dye (U test; n(Ctrl) = 5, n(SCFA) = 5 mice, 5 FOV per mouse). (C) Experimental outline for APPPS1 brain homogenate injection into a WT mouse brain showing the injection site (red dot). (D) smFISH analysis of Cx3cr1-positive cells (>4 puncta) surrounding the APPPS1 brain homogenate injection site (analysis area relative to injection site for all brains) showing an enhanced recruitment of microglial cells in specific pathogen-free (SPF) versus GF WT mice. (E) APPPS1 brain homogenate injection induces higher microglial activation in SPF in comparison to GF WT mice as shown by the higher amount of Trem2 mRNA puncta (red) per Cx3cr1-positive (green) microglia. DAPI (blue) was used as nuclear dye. In D and E, each different shade of color represents the microglia from a single mouse (U test, n(GF) = 3, n(SPF) = 3; two individual experiments, 120 analyzed images (with multiple microglia) per mouse). (F) Microglial recruitment and Aβ plaque uptake were histologically quantified in SCFA- and control-supplemented SPF APPPS1 mice. Although significantly more CD68-positive (green) and Iba1-positive (red) microglial cells were located at Aβ plaques (white, anti-Aβ clone 3552), the number of Aβ-positive microglia was significantly reduced in the SCFA- compared to the control-treated group. Values are expressed as percentages and normalized to the control-treated group (unpaired T-test; n(Ctrl) = 4, n(SCFA) = 5; one individual experiment).
Figure 4—figure supplement 1.. Ex vivo amyloid…
Figure 4—figure supplement 1.. Ex vivo amyloid plaque clearance assay.
Primary WT microglia treated with short chain fatty acids (SCFA; 250 µM) do not show alteration in their phagocytic capacity toward Aβ plaques in comparison to the control-treated cells (Ctrl). CD68 (green) was used to visualize microglia. Amyloid plaques were visualized using both Thiazine red (ThR, red, fibrillar Aβ) and an anti-Aβ antibody (3552, white, total Aβ). DAPI (blue) was used as nuclear dye. Values are expressed as percentages of phagocytosed Aβ plaques normalized to the control-treated group and presented as mean ± SEM (unpaired T-test; n(Ctrl) = 3, n(SCFA) = 3; three independent experiments with two technical replicates per experiment).
Figure 5.. Increased ApoE expression marks microglial…
Figure 5.. Increased ApoE expression marks microglial activation upon short chain fatty acids (SCFA) supplementation.
(A) Heatmap of Nanostring transcriptomic analysis from control- and SCFA-supplemented germ-free (GF) APPPS1 mice at 3 months of age. Row values were scaled using unit variance scaling. Genes previously associated with microglial function have been annotated on the heatmap. Three mice per group have been analyzed. (B) Top 20 upregulated genes in SCFA- versus control-treated samples. Most of transcriptome hits have been previously associated with microglial activation. (C) Functional gene interaction network analysis using Ingenuity Pathway Analysis. Genes are colored based on fold-change values determined by RNA-Seq analysis, where red indicates an increase in SCFA- and green in control-treated animals. Network analysis revealed upregulation of the ApoE-TREM2 axis as one of the principal biological pathways upregulated by SCFA. (D) Representative sagittal brain section indicating location of the analyzed region of interest in the frontal cortex and representative image showing ApoE (green) distribution. Quantification of ApoE signal showed a SCFA-dependent increase of ApoE expression (unpaired T-test; n(Ctrl) = 5, n(SCFA ) = 5; three individual experiments). (E) Quantification of ApoE colocalization (absolute coverage area in mm2) with microglia and astrocytes in control- and SCFA-supplemented GF APPPS1 mice at 3 months of age (unpaired T-test; n(Ctrl) = 5, n(SCFA) = 5; two individual experiments). (F) Analysis of Methoxy-X04-stained brain sections showed a specific increase in plaques of smaller sizes (4–8 µm radius) in 3 months old SCFA- compared to Ctrl-supplemented GF APPPS1 mice (unpaired T-test per bin; n(Ctrl) = 5, n(SCFA) = 5; three individual experiments). Data represent mean ± SEM.
Figure 5—figure supplement 1.. Effect of individual…
Figure 5—figure supplement 1.. Effect of individual short chain fatty acids (SCFA) on microglial polarization.
Primary microglial cells isolated from specific pathogen-free (SPF) WT mice were cultured and treated with the individual SCFA acetate, butyrate, and propionate, the combined SCFA or the respective control (NaCl). Effect of SCFA on microglial polarization was tested by Nanostring analysis of the microglial transcriptome as in Figure 5 (n = 3 samples per group; data is combined from two individual experiments comparing combined SCFA with Ctrl and individual SCFA to Ctrl, respectively). (A) Heatmap of significantly regulated genes between control and combined SCFA treatment. Row values were scaled using unit variance scaling. (B) Venn diagram for common genes that were significantly regulated (in comparison to the respective salt control) for the individual and combined SCFA treatments. (C) Heatmap for scores of biological processes from gene ontology (GO) analysis (combined scores of p-value and z-score per GO term). Color code indicates min to max values per GO term, and gray color indicates not regulated pathway.

References

    1. Abdel-Haq R, Schlachetzki JCM, Glass CK, Mazmanian SK. Microbiome–microglia connections via the gut–brain axis. Journal of Experimental Medicine. 2019;216:41–59. doi: 10.1084/jem.20180794.
    1. Attar A, Liu T, Chan WT, Hayes J, Nejad M, Lei K, Bitan G. A shortened barnes maze protocol reveals memory deficits at 4-months of age in the triple-transgenic mouse model of Alzheimer's disease. PLOS ONE. 2013;8:e80355. doi: 10.1371/journal.pone.0080355.
    1. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut Microbiota as an environmental factor that regulates fat storage. PNAS. 2004;101:15718–15723. doi: 10.1073/pnas.0407076101.
    1. Benakis C, Martin-Gallausiaux C, Trezzi JP, Melton P, Liesz A, Wilmes P. The microbiome-gut-brain Axis in acute and chronic brain diseases. Current Opinion in Neurobiology. 2020;61:1–9. doi: 10.1016/j.conb.2019.11.009.
    1. Bohlen CJ, Bennett FC, Tucker AF, Collins HY, Mulinyawe SB, Barres BA. Diverse requirements for microglial survival, specification, and function revealed by Defined-Medium cultures. Neuron. 2017;94:759–773. doi: 10.1016/j.neuron.2017.04.043.
    1. Brandscheid C, Schuck F, Reinhardt S, Schäfer KH, Pietrzik CU, Grimm M, Hartmann T, Schwiertz A, Endres K. Altered gut microbiome composition and tryptic activity of the 5xfad alzheimer's Mouse Model. Journal of Alzheimer's Disease. 2017;56:775–788. doi: 10.3233/JAD-160926.
    1. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, Koeglsperger T, Dake B, Wu PM, Doykan CE, Fanek Z, Liu L, Chen Z, Rothstein JD, Ransohoff RM, Gygi SP, Antel JP, Weiner HL. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nature Neuroscience. 2014;17:131–143. doi: 10.1038/nn.3599.
    1. Colombo A, Dinkel L, Müller SA, Sebastian Monasor L, Schifferer M, Cantuti-Castelvetri L, König J, Vidatic L, Bremova-Ertl T, Lieberman AP, Hecimovic S, Simons M, Lichtenthaler SF, Strupp M, Schneider SA, Tahirovic S. Loss of NPC1 enhances phagocytic uptake and impairs lipid trafficking in microglia. Nature Communications. 2021;12:1158. doi: 10.1038/s41467-021-21428-5.
    1. Cramer JV, Benakis C, Liesz A. T cells in the post-ischemic brain: troopers or paramedics? Journal of Neuroimmunology. 2019;326:33–37. doi: 10.1016/j.jneuroim.2018.11.006.
    1. Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain axis. Physiological Reviews. 2019;99:1877–2013. doi: 10.1152/physrev.00018.2018.
    1. Cryan JF, O'Mahony SM. The microbiome-gut-brain Axis: from bowel to behavior. Neurogastroenterology & Motility. 2011;23:187–192. doi: 10.1111/j.1365-2982.2010.01664.x.
    1. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nature Reviews Gastroenterology & Hepatology. 2019;16:461–478. doi: 10.1038/s41575-019-0157-3.
    1. Daria A, Colombo A, Llovera G, Hampel H, Willem M, Liesz A, Haass C, Tahirovic S. Young microglia restore amyloid plaque clearance of aged microglia. The EMBO Journal. 2017;36:583–603. doi: 10.15252/embj.201694591.
    1. Davie JR. Inhibition of histone deacetylase activity by butyrate. The Journal of Nutrition. 2003;133:2485S–2493. doi: 10.1093/jn/133.7.2485S.
    1. Dodiya HB, Kuntz T, Shaik SM, Baufeld C, Leibowitz J, Zhang X, Gottel N, Zhang X, Butovsky O, Gilbert JA, Sisodia SS. Sex-specific effects of microbiome perturbations on cerebral aβ amyloidosis and microglia phenotypes. Journal of Experimental Medicine. 2019;216:1542–1560. doi: 10.1084/jem.20182386.
    1. Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermöhlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I, Prinz M. Host Microbiota constantly control maturation and function of microglia in the CNS. Nature Neuroscience. 2015;18:965–977. doi: 10.1038/nn.4030.
    1. Guillot-Sestier MV, Doty KR, Town T. Innate immunity fights Alzheimer's Disease. Trends in Neurosciences. 2015;38:674–681. doi: 10.1016/j.tins.2015.08.008.
    1. Haghikia A, Jörg 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, Thöne J, Demir S, Müller DN, Gold R, Linker RA. Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine. Immunity. 2015;43:817–829. doi: 10.1016/j.immuni.2015.09.007.
    1. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer’s disease. Journal of Cell Biology. 2018;217:459–472. doi: 10.1083/jcb.201709069.
    1. Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD, Frisoni G, Neher JJ, Fåk F, Jucker M, Lasser T, Bolmont T. Reduction of abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut Microbiota. Scientific Reports. 2017;7:41802. doi: 10.1038/srep41802.
    1. Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, McCormick BA. Alzheimer's Disease Microbiome Is Associated with Dysregulation of the Anti-Inflammatory P-Glycoprotein Pathway. mBio. 2019;10:e00632-19. doi: 10.1128/mBio.00632-19.
    1. Hefendehl JK, Wegenast-Braun BM, Liebig C, Eicke D, Milford D, Calhoun ME, Kohsaka S, Eichner M, Jucker M. Long-term in vivo imaging of β-amyloid plaque appearance and growth in a mouse model of cerebral β-amyloidosis. Journal of Neuroscience. 2011;31:624–629. doi: 10.1523/JNEUROSCI.5147-10.2011.
    1. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, Gelpi E, Halle A, Korte M, Latz E, Golenbock DT. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493:674–678. doi: 10.1038/nature11729.
    1. Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nature Reviews Immunology. 2014;14:463–477. doi: 10.1038/nri3705.
    1. Ho L, Ono K, Tsuji M, Mazzola P, Singh R, Pasinetti GM. Protective roles of intestinal Microbiota derived short chain fatty acids in Alzheimer's disease-type beta-amyloid neuropathological mechanisms. Expert Review of Neurotherapeutics. 2018;18:83–90. doi: 10.1080/14737175.2018.1400909.
    1. Holtzman DM, Herz J, Bu G. Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in alzheimer disease. Cold Spring Harbor Perspectives in Medicine. 2012;2:a006312. doi: 10.1101/cshperspect.a006312.
    1. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, Lemere CA, Selkoe DJ, Stevens B. Complement and microglia mediate early synapse loss in alzheimer mouse models. Science. 2016;352:712–716. doi: 10.1126/science.aad8373.
    1. Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI. Molecular analysis of commensal host-microbial relationships in the intestine. Science. 2001;291:881–884. doi: 10.1126/science.291.5505.881.
    1. Hoving LR, Heijink M, van Harmelen V, van Dijk KW, Giera M. GC-MS analysis of Short-Chain fatty acids in feces, cecum content, and blood samples. Methods in Molecular Biology. 2018;1730:247–256. doi: 10.1007/978-1-4939-7592-1_17.
    1. Huuskonen J, Suuronen T, Nuutinen T, Kyrylenko S, Salminen A. Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids. British Journal of Pharmacology. 2004;141:874–880. doi: 10.1038/sj.bjp.0705682.
    1. Janakiraman M, Krishnamoorthy G. Emerging role of diet and Microbiota interactions in neuroinflammation. Frontiers in Immunology. 2018;9:2067. doi: 10.3389/fimmu.2018.02067.
    1. Layden BT, Angueira AR, Brodsky M, Durai V, Lowe WL. Short chain fatty acids and their receptors: new metabolic targets. Translational Research. 2013;161:131–140. doi: 10.1016/j.trsl.2012.10.007.
    1. Lewcock JW, Schlepckow K, Di Paolo G, Tahirovic S, Monroe KM, Haass C. Emerging microglia biology defines novel therapeutic approaches for alzheimer's Disease. Neuron. 2020;108:801–821. doi: 10.1016/j.neuron.2020.09.029.
    1. Li Z, Yi CX, Katiraei S, Kooijman S, Zhou E, Chung CK, Gao Y, van den Heuvel JK, Meijer OC, Berbée JFP, Heijink M, Giera M, Willems van Dijk K, Groen AK, Rensen PCN, Wang Y. Butyrate reduces appetite and activates Brown adipose tissue via the gut-brain neural circuit. Gut. 2018;67:1269–1279. doi: 10.1136/gutjnl-2017-314050.
    1. Liao F, Zhang TJ, Jiang H, Lefton KB, Robinson GO, Vassar R, Sullivan PM, Holtzman DM. Murine versus human apolipoprotein E4: differential facilitation of and co-localization in cerebral amyloid angiopathy and amyloid plaques in APP transgenic mouse models. Acta Neuropathologica Communications. 2015;3:70. doi: 10.1186/s40478-015-0250-y.
    1. Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, Giese T, Veltkamp R. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nature Medicine. 2009;15:192–199. doi: 10.1038/nm.1927.
    1. Lin HV, Frassetto A, Kowalik EJ, Nawrocki AR, Lu MM, Kosinski JR, Hubert JA, Szeto D, Yao X, Forrest G, Marsh DJ. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLOS ONE. 2012;7:e35240. doi: 10.1371/journal.pone.0035240.
    1. Long JM, Holtzman DM. Alzheimer disease: an update on pathobiology and treatment strategies. Cell. 2019;179:312–339. doi: 10.1016/j.cell.2019.09.001.
    1. Marizzoni M, Cattaneo A, Mirabelli P, Festari C, Lopizzo N, Nicolosi V, Mombelli E, Mazzelli M, Luongo D, Naviglio D, Coppola L, Salvatore M, Frisoni GB. Short-Chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in Alzheimer's Disease. Journal of Alzheimer's Disease. 2020;78:683–697. doi: 10.3233/JAD-200306.
    1. Metsalu T, Vilo J. ClustVis: a web tool for visualizing clustering of multivariate data using principal component analysis and heatmap. Nucleic Acids Research. 2015;43:W566–W570. doi: 10.1093/nar/gkv468.
    1. Minter MR, Zhang C, Leone V, Ringus DL, Zhang X, Oyler-Castrillo P, Musch MW, Liao F, Ward JF, Holtzman DM, Chang EB, Tanzi RE, Sisodia SS. Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of alzheimer's disease. Scientific Reports. 2016;6:30028. doi: 10.1038/srep30028.
    1. Minter MR, Hinterleitner R, Meisel M, Zhang C, Leone V, Zhang X, Oyler-Castrillo P, Zhang X, Musch MW, Shen X, Jabri B, Chang EB, Tanzi RE, Sisodia SS. Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged APPSWE/PS1ΔE9 murine model of Alzheimer's disease. Scientific Reports. 2017;7:10411. doi: 10.1038/s41598-017-11047-w.
    1. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut Microbiota and their impact on human metabolism. Gut Microbes. 2016;7:189–200. doi: 10.1080/19490976.2015.1134082.
    1. Moschen I, Bröer A, Galić S, Lang F, Bröer S. Significance of short chain fatty acid transport by members of the monocarboxylate transporter family (MCT) Neurochemical Research. 2012;37:2562–2568. doi: 10.1007/s11064-012-0857-3.
    1. Nagpal R, Neth BJ, Wang S, Craft S, Yadav H. 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. 2019;47:529–542. doi: 10.1016/j.ebiom.2019.08.032.
    1. Parhizkar S, Arzberger T, Brendel M, Kleinberger G, Deussing M, Focke C, Nuscher B, Xiong M, Ghasemigharagoz A, Katzmarski N, Krasemann S, Lichtenthaler SF, Müller SA, Colombo A, Monasor LS, Tahirovic S, Herms J, Willem M, Pettkus N, Butovsky O, Bartenstein P, Edbauer D, Rominger A, Ertürk A, Grathwohl SA, Neher JJ, Holtzman DM, Meyer-Luehmann M, Haass C. Loss of TREM2 function increases amyloid seeding but reduces plaque-associated ApoE. Nature Neuroscience. 2019;22:191–204. doi: 10.1038/s41593-018-0296-9.
    1. Peters F, Salihoglu H, Rodrigues E, Herzog E, Blume T, Filser S, Dorostkar M, Shimshek DR, Brose N, Neumann U, Herms J. BACE1 inhibition more effectively suppresses initiation than progression of β-amyloid pathology. Acta Neuropathologica. 2018;135:695–710. doi: 10.1007/s00401-017-1804-9.
    1. Radde R, Bolmont T, Kaeser SA, Coomaraswamy J, Lindau D, Stoltze L, Calhoun ME, Jäggi F, Wolburg H, Gengler S, Haass C, Ghetti B, Czech C, Hölscher C, Mathews PM, Jucker M. Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Reports. 2006;7:940–946. doi: 10.1038/sj.embor.7400784.
    1. Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, Mazmanian SK. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human Microbiota. Science. 2011;332:974–977. doi: 10.1126/science.1206095.
    1. Sadler R, Singh V, Benakis C, Garzetti D, Brea D, Stecher B, Anrather J, Liesz A. Microbiota differences between commercial breeders impacts the post-stroke immune response. Brain, Behavior, and Immunity. 2017;66:23–30. doi: 10.1016/j.bbi.2017.03.011.
    1. Sadler R, Cramer JV, Heindl S, Kostidis S, Betz D, Zuurbier KR, Northoff BH, Heijink M, Goldberg MP, Plautz EJ, Roth S, Malik R, Dichgans M, Holdt LM, Benakis C, Giera M, Stowe AM, Liesz A. Short-Chain Fatty Acids Improve Poststroke Recovery via Immunological Mechanisms. The Journal of Neuroscience. 2020;40:1162–1173. doi: 10.1523/JNEUROSCI.1359-19.2019.
    1. Sala Frigerio C, Wolfs L, Fattorelli N, Thrupp N, Voytyuk I, Schmidt I, Mancuso R, Chen WT, Woodbury ME, Srivastava G, Möller T, Hudry E, Das S, Saido T, Karran E, Hyman B, Perry VH, Fiers M, De Strooper B. The major risk factors for alzheimer's Disease: Age, Sex, and Genes Modulate the Microglia Response to Aβ Plaques. Cell Reports. 2019;27:1293–1306. doi: 10.1016/j.celrep.2019.03.099.
    1. 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. Gut Microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's Disease. Cell. 2016;167:1469–1480. doi: 10.1016/j.cell.2016.11.018.
    1. Sarlus H, Heneka MT. Microglia in Alzheimer’s disease. Journal of Clinical Investigation. 2017;127:3240–3249. doi: 10.1172/JCI90606.
    1. Sebastian Monasor L, Müller SA, Colombo AV, Tanrioever G, König J, Roth S, Liesz A, Berghofer A, Piechotta A, Prestel M, Saito T, Saido TC, Herms J, Willem M, Haass C, Lichtenthaler SF, Tahirovic S. Fibrillar aβ triggers microglial proteome alterations and dysfunction in alzheimer mouse models. eLife. 2020;9:e54083. doi: 10.7554/eLife.54083.
    1. Shi Y, Holtzman DM. Interplay between innate immunity and alzheimer disease: apoe and TREM2 in the spotlight. Nature Reviews Immunology. 2018;18:759–772. doi: 10.1038/s41577-018-0051-1.
    1. Shirotani K, Tomioka M, Kremmer E, Haass C, Steiner H. Pathological activity of familial alzheimer's disease-associated mutant presenilin can be executed by six different gamma-secretase complexes. Neurobiology of Disease. 2007;27:102–107. doi: 10.1016/j.nbd.2007.04.011.
    1. Singh V, Roth S, Llovera G, Sadler R, Garzetti D, Stecher B, Dichgans M, Liesz A. Microbiota dysbiosis controls the neuroinflammatory response after stroke. Journal of Neuroscience. 2016;36:7428–7440. doi: 10.1523/JNEUROSCI.1114-16.2016.
    1. Singh V, Sadler R, Heindl S, Llovera G, Roth S, Benakis C, Liesz A. The gut microbiome primes a cerebroprotective immune response after stroke. Journal of Cerebral Blood Flow & Metabolism. 2018;38:1293–1298. doi: 10.1177/0271678X18780130.
    1. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic treg cell homeostasis. Science. 2013;341:569–573. doi: 10.1126/science.1241165.
    1. Spangenberg E, Severson PL, Hohsfield LA, Crapser J, Zhang J, Burton EA, Zhang Y, Spevak W, Lin J, Phan NY, Habets G, Rymar A, Tsang G, Walters J, Nespi M, Singh P, Broome S, Ibrahim P, Zhang C, Bollag G, West BL, Green KN. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer's disease model. Nature Communications. 2019;10:3758. doi: 10.1038/s41467-019-11674-z.
    1. Tremlett H, Bauer KC, Appel-Cresswell S, Finlay BB, Waubant E. The gut microbiome in human neurological disease: a review. Annals of Neurology. 2017;81:369–382. doi: 10.1002/ana.24901.
    1. van de Wouw M, Boehme M, Lyte JM, Wiley N, Strain C, O'Sullivan O, Clarke G, Stanton C, Dinan TG, Cryan JF. Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain-gut Axis alterations. The Journal of Physiology. 2018;596:4923–4944. doi: 10.1113/JP276431.
    1. Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, Vieira-Saecker A, Schwartz S, Santarelli F, Kummer MP, Griep A, Gelpi E, Beilharz M, Riedel D, Golenbock DT, Geyer M, Walter J, Latz E, Heneka MT. Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer's disease. Nature. 2017;552:355–361. doi: 10.1038/nature25158.
    1. Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, Carlsson CM, Asthana S, Zetterberg H, Blennow K, Bendlin BB, Rey FE. Gut microbiome alterations in Alzheimer's disease. Scientific Reports. 2017;7:13537. doi: 10.1038/s41598-017-13601-y.
    1. Winkler E, Kamp F, Scheuring J, Ebke A, Fukumori A, Steiner H. Generation of alzheimer Disease-associated amyloid β42/43 peptide by γ-Secretase can be inhibited directly by modulation of membrane thickness. Journal of Biological Chemistry. 2012;287:21326–21334. doi: 10.1074/jbc.M112.356659.
    1. Yamasaki A, Eimer S, Okochi M, Smialowska A, Kaether C, Baumeister R, Haass C, Steiner H. The GxGD motif of presenilin contributes to catalytic function and substrate identification of gamma-secretase. Journal of Neuroscience. 2006;26:3821–3828. doi: 10.1523/JNEUROSCI.5354-05.2006.

Source: PubMed

3
Tilaa