Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota

T Harach, N Marungruang, N Duthilleul, V Cheatham, K D Mc Coy, G Frisoni, J J Neher, F Fåk, M Jucker, T Lasser, T Bolmont, T Harach, N Marungruang, N Duthilleul, V Cheatham, K D Mc Coy, G Frisoni, J J Neher, F Fåk, M Jucker, T Lasser, T Bolmont

Abstract

Alzheimer's disease is the most common form of dementia in the western world, however there is no cure available for this devastating neurodegenerative disorder. Despite clinical and experimental evidence implicating the intestinal microbiota in a number of brain disorders, its impact on Alzheimer's disease is not known. To this end we sequenced bacterial 16S rRNA from fecal samples of Aβ precursor protein (APP) transgenic mouse model and found a remarkable shift in the gut microbiota as compared to non-transgenic wild-type mice. Subsequently we generated germ-free APP transgenic mice and found a drastic reduction of cerebral Aβ amyloid pathology when compared to control mice with intestinal microbiota. Importantly, colonization of germ-free APP transgenic mice with microbiota from conventionally-raised APP transgenic mice increased cerebral Aβ pathology, while colonization with microbiota from wild-type mice was less effective in increasing cerebral Aβ levels. Our results indicate a microbial involvement in the development of Abeta amyloid pathology, and suggest that microbiota may contribute to the development of neurodegenerative diseases.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. Comparison of the gut microbiota…
Figure 1. Comparison of the gut microbiota between conventionally raised (CONVR-) APPPS1 and wild type (WT) mice analyzed with multiple t-tests together with the Holm-Sidak method to correct for multiple comparisons.
(a) Mean sequence relative abundance of gut microbial taxa at phylum level in CONVR-APPPS1 and WT mice, aged 1 month (n = 6 and n = 8 respectively), 3.5 months (n = 7 and n = 8) and 8 months (n = 6 and n = 7). Bacteroides, Firmicutes, Verrucomicrobia and Tenericutes were significantly different between 8 month-old APPPS1 and WT mice (p < 0.001), as well as Proteobacteria (p < 0.01) and Actinobacteria (p < 0.01). No significant differences were found in the younger mice. (b) At genus level, 4 microbial taxa with relative abundance >5% were significantly different between 8 month-old CONVR-APPPS1 and WT mice (***p < 0.001), while younger mice did not show any significant differences. (c) Rarefaction curves (α-diversity vs. sequencing effort) and weighted Unifrac PCoA plot to compare phylogenetic distance matrices of CONVR-APPPS1 and WT mice. Species richness was significantly increased in the 8 month-old APPPS1 mice (p < 0.05, observed species test in QIIME with FDR correction for multiple comparisons). Younger mice did not show significant differences in α-diversity. Clustering of CONVR-APPPS1 mice in the PCoA was significantly separated from the WT mice at 8 months of age (p < 0.001 for both the ANOSIM and the Adonis non-parametric test in QIIME). Experiments were performed three times, and data represent mean values of a representative experiment.
Figure 2. Reduction of Aβ levels in…
Figure 2. Reduction of Aβ levels in brain and blood of GF-APPPS1 transgenic mice.
(ad) Levels of Aβ38, Aβ40, Aβ42 and Aβ42/Aβ40 ratio measured by ELISA in 3.5 and 8 month-old conventionally-raised (CONVR-APPPS1) and germ-free APPPS1 mice (GF-APPPS1). Levels of Aβ assessed by western blot in 3.5 month-old (n = 5) (e) and in 8 month-old mice (n = 6). Full blots are shown in Supplementary Fig. 6(f). Plasmatic levels of soluble Aβ40 (g), Aβ42 (h) and ratio Aβ42/Aβ40 (i) by ELISA. Experiments were performed three times, and data represent mean values of a representative experiment. Data represent mean ± SEM. Statistical differences between germ free and conventionally-raised mice: *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 3. Reduced amyloid load in GF-APPPS1…
Figure 3. Reduced amyloid load in GF-APPPS1 transgenic mice.
(a) Thioflavin T-stained brain sections encompassing the cortex and hippocampus of a 3.5 and 8 month-old CONVR-APPPS1 and age-matched GF-APPPS1 mice (scale bar: 150 um, all panels have the same magnification). (b) Quantification of amyloid load on Thioflavin-stained sections demonstrates a significant reduction of cerebral amyloid plaques in young and aged GF-APPPS1 mice (n = 5 and n = 6 respectively) in both cortex and hippocampus, as compared to CONVR-APPPS1 mice (n = 5 and n = 6 respectively). Statistical differences between GF- and CONVR-APPPS1 mice: *p < 0.05, **p < 0.01, ***p < 0.001. Shown are mean ± SEM.
Figure 4. Colonization of 4 month-old GF-APPPS1…
Figure 4. Colonization of 4 month-old GF-APPPS1 mice with the microbiota from aged CONVR-APPPS1 (COLOAD-APPPS1) and CONVR-wild type mice (COLOWT-APPPS1).
After 8 weeks of colonization, levels of cerebral soluble Aβ38 (a), Aβ40 (b), Aβ42 (c) and ratio Aβ42/Aβ40 (d) were evaluated by ELISA in (6 month-old) GF-APPPS1, COLOWT-APPPS1, COLOAD-APPPS1 and CONVR-APPPS1 mice. Levels of Aβ assessed by western blot in 6 month-old GF-APPPS1 and COLOWT-APPPS1 mice (n = 6) (e) and in 6 month-old COLOAD-APPPS1, COLOWT-APPPS1 animals and CONVR-APPPS1 (n = 4) (f) Full blots are shown in Supplementary Fig. 7. Plasmatic levels of soluble Aβ40 (g), Aβ42 (h) and ratio Aβ42/Aβ40 (i) by ELISA. Description of used genotypes (j). Experiments were performed three times, and data represent mean values of a representative experiment. Data represent mean ± SEM. Statistical differences between germ free and conventionally-raised mice: *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 5. Comparison of the gut microbiota…
Figure 5. Comparison of the gut microbiota between COLOWT-APPPS1 and COLOAD-APPPS1 mice at 6 weeks after colonization.
(a) Mean sequence relative abundance of gut microbial taxa at phylum level in COLOWT-APPPS1 and COLOAD-APPPS1 mice (n = 6 and n = 6 respectively). No significant differences were observed at this time point. (b) At genus level, the 4 bacterial taxa that differed in the aged CONVR-APPPS1 and CONVR-WT mice showed similar trends regarding Akkermansia abundance in the colonized mice, but did not reach statistical significance. Experiments were performed three times, and data represent mean values of a representative experiment. Data represent mean ± SEM. Rarefaction curves (α-diversity vs. sequencing effort) and weighted Unifrac PCoA plot to compare phylogenetic distance matrices of (c) COLOWT-APPPS1 and (d) COLOAD APPPS1 mice at day 1, day 4 and week 2, 4 and 6 of colonization. Significant differences in α-diversity were found between COLOAD-APPPS1 and COLOWT-APPPS1 mice at Day 1 (p < 0.05) as well as between COLOAD-APPPS1 at Day 1 and Day 4, Week 2, 4 and 6 (all p < 0.05).

References

    1. Hardy J. & Selkoe D. J. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356, doi: 10.1126/science.1072994 (2002).
    1. Belkaid Y. & Hand T. W. Role of the microbiota in immunity and inflammation. Cell 157, 121–141, doi: 10.1016/j.cell.2014.03.011 (2014).
    1. Hooper L. V., Littman D. R. & Macpherson A. J. Interactions between the microbiota and the immune system. Science 336, 1268–1273, doi: 10.1126/science.1223490 (2012).
    1. O’Toole P. W. & Jeffery I. B. Gut microbiota and aging. Science 350, 1214–1215, doi: 10.1126/science.aac8469 (2015).
    1. Scheperjans F. et al.. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord, doi: 10.1002/mds.26069 (2014).
    1. Collins S. M. & Bercik P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology 136, 2003–2014, doi: 10.1053/j.gastro.2009.01.075 (2009).
    1. Cryan J. F. & O’Mahony S. M. The microbiome-gut-brain axis: from bowel to behavior. Neurogastroenterology and motility: the official journal of the European Gastrointestinal Motility Society 23, 187–192, doi: 10.1111/j.1365-2982.2010.01664.x (2011).
    1. Dinan T. G. & Cryan J. F. Regulation of the stress response by the gut microbiota: implications for psychoneuroendocrinology. Psychoneuroendocrinology 37, 1369–1378, doi: 10.1016/j.psyneuen.2012.03.007 (2012).
    1. Forsythe P., Sudo N., Dinan T., Taylor V. H. & Bienenstock J. Mood and gut feelings. Brain, behavior, and immunity 24, 9–16, doi: 10.1016/j.bbi.2009.05.058 (2010).
    1. Hsiao E. Y. et al.. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 155, 1451–1463, doi: 10.1016/j.cell.2013.11.024 (2013).
    1. Berer K. et al.. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541, doi: 10.1038/nature10554 (2011).
    1. Cattaneo A. et al.. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiology of aging 49, 60–68, doi: 10.1016/j.neurobiolaging.2016.08.019 (2016).
    1. Minter M. R. et al.. Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer’s disease. Scientific reports 6, 30028, doi: 10.1038/srep30028 (2016).
    1. Radde R. et al.. Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep 7, 940–946, doi: 10.1038/sj.embor.7400784 (2006).
    1. Rabot S. et al.. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB journal: official publication of the Federation of American Societies for Experimental Biology 24, 4948–4959, doi: 10.1096/fj.10-164921 (2010).
    1. Fernandez-Gamba A., Leal M. C., Morelli L. & Castano E. M. Insulin-degrading enzyme: structure-function relationship and its possible roles in health and disease. Current pharmaceutical design 15, 3644–3655 (2009).
    1. Miners J. S., Barua N., Kehoe P. G., Gill S. & Love S. Abeta-degrading enzymes: potential for treatment of Alzheimer disease. Journal of neuropathology and experimental neurology 70, 944–959, doi: 10.1097/NEN.0b013e3182345e46 (2011).
    1. Bhattacharjee S. & Lukiw W. J. Alzheimer’s disease and the microbiome. Front Cell Neurosci 7, 153, doi: 10.3389/fncel.2013.00153 (2013).
    1. Collins S. M., Surette M. & Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol 10, 735–742, doi: 10.1038/nrmicro2876 (2012).
    1. Nicholson J. K. et al.. Host-gut microbiota metabolic interactions. Science 336, 1262–1267, doi: 10.1126/science.1223813 (2012).
    1. Ericsson A. C. et al.. Effects of vendor and genetic background on the composition of the fecal microbiota of inbred mice. PloS one 10, e0116704, doi: 10.1371/journal.pone.0116704 (2015).
    1. Reardon S. A mouse’s house may ruin experiments. Nature 530, 264, doi: 10.1038/nature.2016.19335 (2016).
    1. Couturier-Maillard A. et al.. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. The Journal of clinical investigation 123, 700–711, doi: 10.1172/JCI62236 (2013).
    1. Everard A. et al.. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA 110, 9066–9071, doi: 10.1073/pnas.1219451110 (2013).
    1. Haan M. N. Therapy Insight: type 2 diabetes mellitus and the risk of late-onset Alzheimer’s disease. Nature clinical practice. Neurology 2, 159–166, doi: 10.1038/ncpneuro0124 (2006).
    1. Ishibashi K., Onishi A., Fujiwara Y., Ishiwata K. & Ishii K. Plasma Glucose Levels Affect Cerebral 18F-FDG Distribution in Cognitively Normal Subjects With Diabetes. Clinical nuclear medicine, doi: 10.1097/RLU.0000000000001147 (2016).
    1. Pimplikar S. W. Neuroinflammation in Alzheimer’s disease: from pathogenesis to a therapeutic target. Journal of clinical immunology 34 Suppl 1, S64–69, doi: 10.1007/s10875-014-0032-5 (2014).
    1. Tang Y. et al.. Early inflammation-associated factors blunt sterol regulatory element-binding proteins-1-mediated lipogenesis in high-fat diet-fed APP/PSEN1dE9 mouse model of Alzheimer’s disease. Journal of neurochemistry, doi: 10.1111/jnc.13437 (2015).
    1. Kumar D. K. et al.. Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Science translational medicine 8, 340ra372, doi: 10.1126/scitranslmed.aaf1059 (2016).
    1. Dolpady J. et al.. Oral Probiotic VSL#3 Prevents Autoimmune Diabetes by Modulating Microbiota and Promoting Indoleamine 2,3-Dioxygenase-Enriched Tolerogenic Intestinal Environment. Journal of diabetes research 2016, 7569431, doi: 10.1155/2016/7569431 (2016).
    1. Lavasani S. et al.. A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PloS one 5, e9009, doi: 10.1371/journal.pone.0009009 (2010).
    1. Schippa S. et al.. A distinctive ‘microbial signature’ in celiac pediatric patients. BMC microbiology 10, 175, doi: 10.1186/1471-2180-10-175 (2010).
    1. Shen X. J. et al.. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut microbes 1, 138–147, doi: 10.4161/gmic.1.3.12360 (2010).
    1. Matziouridou C., Marungruang N., Nguyen T. D., Nyman M. & Fak F. Lingonberries reduce atherosclerosis in Apoe-/- mice in association with altered gut microbiota composition and improved lipid profile. Molecular nutrition & food research, doi: 10.1002/mnfr.201500738 (2016).
    1. Anand R., Gill K. D. & Mahdi A. A. Therapeutics of Alzheimer’s disease: Past, present and future. Neuropharmacology 76 Pt A, 27–50, doi: 10.1016/j.neuropharm.2013.07.004 (2014).
    1. Erny D. et al.. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18, 965–977, doi: 10.1038/nn.4030 (2015).
    1. Heneka M. T. et al.. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493, 674–678, doi: 10.1038/nature11729 (2013).
    1. Vijay-Kumar M. et al.. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 328, 228–231, doi: 10.1126/science.1179721 (2010).
    1. Klindworth A. et al.. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res 41, e1, doi: 10.1093/nar/gks808 (2013).
    1. Caporaso J. G. et al.. QIIME allows analysis of high-throughput community sequencing data. Nat Methods 7, 335–336, doi: 10.1038/nmeth.f.303 (2010).
    1. Edgar R. C. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461, doi: 10.1093/bioinformatics/btq461 (2010).
    1. DeSantis T. Z. et al.. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol 72, 5069–5072, doi: 10.1128/AEM.03006-05 (2006).
    1. Wang Q., Garrity G. M., Tiedje J. M. & Cole J. R. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol 73, 5261–5267, doi: 10.1128/AEM.00062-07 (2007).
    1. Klipper-Aurbach Y. et al.. Mathematical formulae for the prediction of the residual beta cell function during the first two years of disease in children and adolescents with insulin-dependent diabetes mellitus. Medical hypotheses 45, 486–490 (1995).
    1. Segata N. et al.. Metagenomic biomarker discovery and explanation. Genome Biol 12, R60, doi: 10.1186/gb-2011-12-6-r60 (2011).

Source: PubMed

3
Sottoscrivi