Gut microbiome alterations in Alzheimer's disease

Nicholas M Vogt, Robert L Kerby, Kimberly A Dill-McFarland, Sandra J Harding, Andrew P Merluzzi, Sterling C Johnson, Cynthia M Carlsson, Sanjay Asthana, Henrik Zetterberg, Kaj Blennow, Barbara B Bendlin, Federico E Rey, Nicholas M Vogt, Robert L Kerby, Kimberly A Dill-McFarland, Sandra J Harding, Andrew P Merluzzi, Sterling C Johnson, Cynthia M Carlsson, Sanjay Asthana, Henrik Zetterberg, Kaj Blennow, Barbara B Bendlin, Federico E Rey

Abstract

Alzheimer's disease (AD) is the most common form of dementia. However, the etiopathogenesis of this devastating disease is not fully understood. Recent studies in rodents suggest that alterations in the gut microbiome may contribute to amyloid deposition, yet the microbial communities associated with AD have not been characterized in humans. Towards this end, we characterized the bacterial taxonomic composition of fecal samples from participants with and without a diagnosis of dementia due to AD. Our analyses revealed that the gut microbiome of AD participants has decreased microbial diversity and is compositionally distinct from control age- and sex-matched individuals. We identified phylum- through genus-wide differences in bacterial abundance including decreased Firmicutes, increased Bacteroidetes, and decreased Bifidobacterium in the microbiome of AD participants. Furthermore, we observed correlations between levels of differentially abundant genera and cerebrospinal fluid (CSF) biomarkers of AD. These findings add AD to the growing list of diseases associated with gut microbial alterations, as well as suggest that gut bacterial communities may be a target for therapeutic intervention.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Alzheimer’s disease is associated with alterations in gut microbiome composition. (A) Faith’s Phylogenetic Diversity is decreased in the microbiome of AD participants. *p < 0.05. (B) Non-metric multidimensional scaling (NMDS) plot of weighted UniFrac analysis of relative sample OTU composition. NMDS analysis was limited to two dimensions, with a stress measurement of 0.17. Each dot represents a scaled measure of the composition of a given participant, color- and shape-coded by cohort. (C) Differential abundance analysis identified 14 OTUs that were increased and 68 OTUs that were decreased in AD relative to Control participants (p < 0.05, FDR-corrected). Each point represents an OTU. Data plotted as log2 fold change; OTUs to the right of the zero line are more abundant and OTUs to the left of the zero line are less abundant in AD compared to Control groups. OTUs are organized on the y-axis according to the lowest taxonomic classification possible. (D) OTUs grouped at the phylum level and analyzed using Metastats show that AD participants have decreased abundance of Firmicutes and Actinobacteria, and increased abundance of Bacteroidetes compared to Control participants (p < 0.05, FDR-corrected). Tukey plots show median, IQR, and participant data points for phylum relative abundance.
Figure 2
Figure 2
Bacterial families and genera differentially represented in feces from AD participants compared to Control participants (p < 0.05, FDR-corrected). Tukey plots are colored by phylum and show median, IQR, and participant data points for genus or family relative abundance.
Figure 3
Figure 3
Bacterial taxa correlate with CSF biomarkers of AD pathology. 13 genera identified as differentially abundant in AD were correlated with CSF biomarkers of AD including the Aβ42/Aβ40 ratio (with lower CSF levels reflecting greater amyloid deposition in the brain), phosphorylated tau (p-tau), and the p-tau/Aβ42 ratio (which incorporates both facets of AD pathology). Correlations were calculated separately for all 40 participants (All), 31 non-demented participants (ND), and 9 AD participants (AD). In general, genera identified as more abundant in AD were associated with greater AD pathology, while genera identified as less abundant in AD were associated with less AD pathology. Genera are ordered from most to least abundant. Shape and color of ellipses represent strength of Spearman’s rank correlation coefficients (rho). Bolded ellipse borders represent significant correlations (two-sided, p < 0.05 uncorrected).

References

    1. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016;8:595–608. doi: 10.15252/emmm.201606210.
    1. Hill JM, et al. Pathogenic microbes, the microbiome, and Alzheimer’s disease (AD) Front. Aging. Neurosci. 2014;6:1–5.
    1. Zhao Y, Dua P, Lukiw WJ. Microbial sources of amyloid and relevance to amyloidogenesis and Alzheimer’s disease (AD) J. Alzheimers Dis. Parkinsonism. 2015;5:177.
    1. Bhattacharjee S, Lukiw WJ. Alzheimer’s disease and the microbiome. Front. Cell. Neurosci. 2013;7:1–4. doi: 10.3389/fncel.2013.00153.
    1. Ghaisas S, Maher J, Kanthasamy A. Gut microbiome in health and disease: Linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol. Ther. 2016;158:52–62. doi: 10.1016/j.pharmthera.2015.11.012.
    1. Chow J, Lee SM, Shen Y, Khosravi A, Mazmanian SK. Host-bacterial symbiosis in health and disease. Adv. Immunol. 2010;107:243–274. doi: 10.1016/B978-0-12-381300-8.00008-3.
    1. Koenig JE, et al. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl. Acad. Sci. USA. 2011;108(Suppl 1):4578–4585. doi: 10.1073/pnas.1000081107.
    1. Human Microbiome Project Consortium Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214. doi: 10.1038/nature11234.
    1. Claesson MJ, et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl. Acad. Sci. USA. 2011;108(Suppl 1):4586–4591. doi: 10.1073/pnas.1000097107.
    1. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148:1258–1270. doi: 10.1016/j.cell.2012.01.035.
    1. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017;20:145–155. doi: 10.1038/nn.4476.
    1. Scheperjans F, et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov. Disord. 2015;30:350–358. doi: 10.1002/mds.26069.
    1. Keshavarzian A, et al. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015;30:1351–1360. doi: 10.1002/mds.26307.
    1. Cattaneo A, et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging. 2017;49:60–68. doi: 10.1016/j.neurobiolaging.2016.08.019.
    1. Minter MR, et al. Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer’s disease. Sci. Rep. 2016;6:1–12. doi: 10.1038/srep30028.
    1. Harach T, et al. Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci. Rep. 2017;7:41802. doi: 10.1038/srep41802.
    1. Morris MC, et al. MIND diet associated with reduced incidence of Alzheimer’s disease. Alzheimers Dement. 2015;11:1007–1014. doi: 10.1016/j.jalz.2014.11.009.
    1. Langille MGI, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat. Biotechnol. 2013;31:814–821. doi: 10.1038/nbt.2676.
    1. Blennow K, Zetterberg H. The past and the future of Alzheimer’s disease CSFbiomarkers - a journey toward validated biochemical tests covering the whole spectrum of molecular events. Front. Neurosci. 2015;9:1–8. doi: 10.3389/fnins.2015.00345.
    1. Hellwig K, et al. Neurogranin and YKL-40: independent markers of synaptic degeneration and neuroinflammation in Alzheimer’s disease. Alz. Res. Therapy. 2015;7:74. doi: 10.1186/s13195-015-0161-y.
    1. Craig-Schapiro R, et al. YKL-40: a novel prognostic fluid biomarker for preclinical Alzheimer’s disease. Biol. Psychiatry. 2010;68:903–912. doi: 10.1016/j.biopsych.2010.08.025.
    1. Rosén C, et al. Increased Levels of Chitotriosidase and YKL-40 in Cerebrospinal Fluid from Patients with Alzheimer’s disease. Dement. Geriatr. Cogn. Dis. Extra. 2014;4:297–304. doi: 10.1159/000362164.
    1. Pistollato F, et al. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr. Rev. 2016;74:624–634. doi: 10.1093/nutrit/nuw023.
    1. Turnbaugh PJ, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–484. doi: 10.1038/nature07540.
    1. Larsen N, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE. 2010;5:1–10.
    1. Dicksved J, et al. Molecular analysis of the gut microbiota of identical twins with Crohn’s disease. ISME J. 2008;2:716–727. doi: 10.1038/ismej.2008.37.
    1. DuPont AW, DuPont HL. The intestinal microbiota and chronic disorders of the gut. Nat. Rev. Gastroenterol. Hepatol. 2011;8:523–531. doi: 10.1038/nrgastro.2011.133.
    1. Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012;13:701–712. doi: 10.1038/nrn3346.
    1. Schwiertz A, et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity. 2009;18:190–195. doi: 10.1038/oby.2009.167.
    1. Ley RE, et al. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA. 2005;102:11070–11075. doi: 10.1073/pnas.0504978102.
    1. Rawlings AM, et al. Diabetes in midlife and cognitive change over 20 years: a cohort study. Ann. Intern. Med. 2014;161:785–793. doi: 10.7326/M14-0737.
    1. Ott A, et al. Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology. 1999;53:1937–1942. doi: 10.1212/WNL.53.9.1937.
    1. la Monte de SM, Wands JR. Review of insulin and insulin-like growth factor expression, signaling, and malfunction in the central nervous system: relevance to Alzheimer’s disease. J. Alzheimers Dis. 2005;7:45–61. doi: 10.3233/JAD-2005-7106.
    1. Willette AA, et al. Association of insulin resistance with cerebral glucose uptake in late middle-aged adults at risk for Alzheimer disease. JAMA Neurol. 2015;72:1013–1020. doi: 10.1001/jamaneurol.2015.0613.
    1. Willette AA, et al. Insulin resistance predicts brain amyloid deposition in late middle-aged adults. Alzheimers Dement. 2015;11:504–510. doi: 10.1016/j.jalz.2014.03.011.
    1. Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol. Rev. 2014;38:996–1047. doi: 10.1111/1574-6976.12075.
    1. Cani PD, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–1772. doi: 10.2337/db06-1491.
    1. Asti A, Gioglio L. Can a bacterial endotoxin be a key factor in the kinetics of amyloid fibril formation? J. Alzheimers Dis. 2014;39:169–179.
    1. Sheng JG, et al. Lipopolysaccharide-induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid beta peptide in APPswe transgenic mice. Neurobiol. Dis. 2003;14:133–145. doi: 10.1016/S0969-9961(03)00069-X.
    1. Kahn MS, et al. Prolonged elevation in hippocampal Aβ and cognitive deficits following repeated endotoxin exposure in the mouse. Behav. Brain. Res. 2012;229:176–184. doi: 10.1016/j.bbr.2012.01.010.
    1. Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J. Neurosci. 2005;25:8843–8853. doi: 10.1523/JNEUROSCI.2868-05.2005.
    1. Sy M, et al. Inflammation induced by infection potentiates tau pathological features in transgenic mice. Am. J. Pathol. 2011;178:2811–2822. doi: 10.1016/j.ajpath.2011.02.012.
    1. Man AL, et al. Age-associated modifications of intestinal permeability and innate immunity in human small intestine. Clin. Sci. 2015;129:515–527. doi: 10.1042/CS20150046.
    1. Stehle JR, et al. Lipopolysaccharide-binding protein, a surrogate marker of microbial translocation, is associated with physical function in healthy older adults. J. Gerontol. Ser. A-Biol. Sci. Med. Sci. 2012;67:1212–1218. doi: 10.1093/gerona/gls178.
    1. Zhan X, et al. Gram-negative bacterial molecules associate with Alzheimer disease pathology. Neurology. 2016;87:2324–2332. doi: 10.1212/WNL.0000000000003391.
    1. O’Callaghan A, van Sinderen D. Bifidobacteria and their role as members of the human gut microbiota. Front. Microbiol. 2016;7:1–23.
    1. Arboleya S, Watkins C, Stanton C, Ross RP. Gut bifidobacteria populations in human health and aging. Front. Microbiol. 2016;7:1–9. doi: 10.3389/fmicb.2016.01204.
    1. Underwood MA, German JB, Lebrilla CB, Mills DA. Bifidobacterium longum subspecies infantis: champion colonizer of the infant gut. Pediatr. Res. 2015;77:229–235.
    1. Wang Z, et al. The role of bifidobacteria in gut barrier function after thermal injury in rats. J. Trauma. 2006;61:650–657. doi: 10.1097/01.ta.0000196574.70614.27.
    1. Griffiths EA, et al. In vivo effects of bifidobacteria and lactoferrin on gut endotoxin concentration and mucosal immunity in Balb/c mice. Dig. Dis. Sci. 2004;49:579–589. doi: 10.1023/B:.
    1. Romond M-B, et al. Does the intestinal bifidobacterial colonisation affect bacterial translocation? Anaerobe. 2008;14:43–48. doi: 10.1016/j.anaerobe.2007.09.003.
    1. Akbari E, et al. Effect of probiotic supplementation on cognitive function and metabolic status in Alzheimer’s disease: a randomized, double-blind and controlled trial. Front. Aging. Neurosci. 2016;8:1–8. doi: 10.3389/fnagi.2016.00256.
    1. Shen L, Liu L, Ji H-F. Alzheimer’s disease histological and behavioral manifestations in transgenic mice correlate with specific gut microbiome state. J. Alzheimers Dis. 2017;56:385–390. doi: 10.3233/JAD-160884.
    1. Nguyen TLA, Vieira-Silva S, Liston A, Raes J. How informative is the mouse for human gut microbiota research? Dis. Model. Mech. 2015;8:1–16. doi: 10.1242/dmm.017400.
    1. Hansen RA, et al. Efficacy and safety of donepezil, galantamine, and rivastigmine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. Clin. Interv. Aging. 2008;3:211–225.
    1. Sager MA, Hermann B, La Rue A. Middle-aged children of persons with Alzheimer’s disease: APOE genotypes and cognitive function in the Wisconsin Registry for Alzheimer’s Prevention. J. Geriatr. Psychiatry Neurol. 2005;18:245–249. doi: 10.1177/0891988705281882.
    1. Johnson SC, et al. The effect of TOMM40 poly-T length on gray matter volume and cognition in middle-aged persons with APOE ε3/ε3 genotype. Alzheimers Dement. 2011;7:456–465. doi: 10.1016/j.jalz.2010.11.012.
    1. McKhann GM, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–269. doi: 10.1016/j.jalz.2011.03.005.
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand. J. Gastroenterol. 1997;32:920–924. doi: 10.3109/00365529709011203.
    1. Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl. Environ. Microbiol. 2013;79:5112–5120. doi: 10.1128/AEM.01043-13.
    1. Schloss PD, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 2009;75:7537–7541. doi: 10.1128/AEM.01541-09.
    1. Westcott SL, Schloss PD. OptiClust, an improved method for assigning amplicon-based sequence data to operational taxonomic units. mSphere. 2017;2:1–11. doi: 10.1128/mSphereDirect.00073-17.
    1. Bokulich NA, et al. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat. Methods. 2013;10:57–59. doi: 10.1038/nmeth.2276.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq. 2. Genome Biol. 2014;15:31–21. doi: 10.1186/s13059-014-0550-8.
    1. McMurdie PJ, Holmes S. Waste not, want not: why rarefying microbiome data is inadmissible. PLoS Comput. Biol. 2014;10:1–12. doi: 10.1371/journal.pcbi.1003531.
    1. White JR, Nagarajan N, Pop M. Statistical methods for detecting differentially abundant features in clinical metagenomic samples. PLoS Comput. Biol. 2009;5:1–11. doi: 10.1371/journal.pcbi.1000352.
    1. Segata N, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(R60):1–18.
    1. Lewczuk P, Lelental N, Spitzer P, Maler JM, Kornhuber J. Amyloid-β 42/40 cerebrospinal fluid concentration ratio in the diagnostics of Alzheimer’s disease: validation of two novel assays. J. Alzheimers Dis. 2015;43:183–191.
    1. Racine AM, et al. Associations between performance on an abbreviated CogState battery, other measures of cognitive function, and biomarkers in people at risk for Alzheimer’s sisease. J. Alzheimers Dis. 2016;54:1395–1408. doi: 10.3233/JAD-160528.
    1. Racine AM, et al. Association of longitudinal white matter degeneration and cerebrospinal fluid biomarkers of neurodegeneration, inflammation and Alzheimer’s disease in late-middle-aged adults. Brain Imaging Behav. 2017;59:306.

Source: PubMed

3
Subskrybuj