Combination of gut microbiota and plasma amyloid-β as a potential index for identifying preclinical Alzheimer's disease: a cross-sectional analysis from the SILCODE study

Can Sheng, Kun Yang, Beiqi He, Wenying Du, Yanning Cai, Ying Han, Can Sheng, Kun Yang, Beiqi He, Wenying Du, Yanning Cai, Ying Han

Abstract

Background: Plasma amyloid-β (Aβ) may facilitate identification of individuals with brain amyloidosis. Gut microbial dysbiosis in Alzheimer's disease (AD) is increasingly being recognized. However, knowledge about alterations of gut microbiota in preclinical AD, as well as whether the combination of plasma Aβ and gut microbiota could identify preclinical AD, remains largely unknown.

Methods: This study recruited 34 Aβ-negative cognitively normal (CN-) participants, 32 Aβ-positive cognitively normal (CN+) participants, and 22 patients with cognitive impairment (CI), including 11 patients with mild cognitive impairment (MCI) and 11 AD dementia patients. All participants underwent neuropsychological assessments and fecal microbiota analysis through 16S ribosomal RNA (rRNA) Illumina Miseq sequencing technique. Meso Scale Discovery (MSD) kits were used to quantify the plasma Aβ40, Aβ42, and Aβ42/Aβ40 in CN- and CN+ participants. Using Spearman's correlation analysis, the associations of global standard uptake value rate (SUVR) with altered gut microbiota and plasma Aβ markers were separately evaluated. Furthermore, the discriminative power of the combination of gut microbiota and plasma Aβ markers for identifying CN+ individuals was investigated.

Results: Compared with the CN- group, the CN+ group showed significantly reduced plasma Aβ42 (p = 0.011) and Aβ42/Aβ40 (p = 0.003). The relative abundance of phylum Bacteroidetes was significantly enriched, whereas phylum Firmicutes and class Deltaproteobacteria were significantly decreased in CN+ individuals in comparison with that in CN- individuals. Particularly, the relative abundance of phylum Firmicutes and its corresponding SCFA-producing bacteria exhibited a progressive decline tendency from CN- to CN+ and CI. Besides, the global brain Aβ burden was negatively associated with the plasma Aβ42/Aβ40 (r = -0.298, p = 0.015), family Desulfovibrionaceae (r = -0.331, p = 0.007), genus Bilophila (r = -0.247, p = 0.046), and genus Faecalibacterium (r = -0.291, p = 0.018) for all CN participants. Finally, the combination of plasma Aβ markers, altered gut microbiota, and cognitive performance reached a relatively good discriminative power in identifying individuals with CN+ from CN- (AUC = 0.869, 95% CI 0.782 ~ 0.955).

Conclusions: This study provided the evidence that the gut microbial composition was altered in preclinical AD. The combination of plasma Aβ and gut microbiota may serve as a non-invasive, cost-effective diagnostic tool for early AD screening. Targeting the gut microbiota may be a novel therapeutic strategy for AD.

Trial registration: This study has been registered in ClinicalTrials.gov (NCT03370744, https://www.clinicaltrials.gov ) in November 15, 2017.

Keywords: Alzheimer’s disease; Amyloid-β; Gut microbiota; Plasma; Preclinical.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Plasma Aβ levels for CN− and CN+ participants. Scatter plot presented mean with range. No significant difference was observed in plasma Aβ40 (A), whereas there were significant differences in plasma Aβ42 (B) and Aβ42/Aβ40 (C) between CN− and CN+. *p < 0.05; **p < 0.01. Aβ amyloid-β; CN−, amyloid-β-negative cognitively normal participants; CN+, amyloid-β-positive cognitively normal participants
Fig. 2
Fig. 2
The overall gut microbiota community for CN− and CN+ participants. A The Venn diagram illustrated the overlap of ASVs between CN− and CN+, with 1929 ASVs shared in both groups, and 147 ASVs unique for CN+. B The key ASVs in both groups. The abundance of ASV_19 and ASV_9 was significantly reduced in CN+ compared with that in CN−. C The alpha diversity of gut microbiota between CN− and CN+. Each bar graph represented the mean and standard deviation. There were no significant differences in Chao1, ACE, Simpson, and Shannon indexes between CN− and CN+. D The PCoA based on the distribution of ASVs. The structure of gut microbiota in the CN+ group was not significantly different from that in the CN− group. *p< 0.05; **p < 0.01. CN− amyloid-β-negative cognitively normal participants, CN+ amyloid-β-positive cognitively normal participants, ASV amplicon sequence variants, PCoA principal coordinates analysis, PERMANOVA permutational multivariate analysis of variance
Fig. 3
Fig. 3
The gut microbial compositions for CN− and CN+ participants. A The bacterial community in both groups at different taxonomic levels. Bar graphs indicated the relative abundance of phylum-level, class-level, order-level, and family-level taxa. B LEfSe analysis between CN− and CN+. As shown in the histogram of LDA scores for differentially abundant taxa, positive LDA scores indicated the enrichment of taxa in the CN+ group (green), and negative LDA scores indicated the enriched taxa in the CN− group (red). The LDA scores (log10) > 2 and p < 0.05 were listed. Cladogram indicated the phylogenetic distribution of gut bacteria. Colors represented different groups (CN−, red; CN+, green). Nodes with different colors represented important taxa in different groups. Yellow nodes suggested no significantly differential taxa between the two groups. C The heatmap showing the relative abundance and distribution of differentially abundant taxa identified by the LEfSe method. CN− amyloid-β-negative cognitively normal participants, CN+ amyloid-β-positive cognitively normal participants, LEfSe linear discriminant analysis (LDA) effect size
Fig. 4
Fig. 4
The association of plasma Aβ markers and gut microbiota with global brain Aβ burden. A There was negative correlation between global SUVR and plasma Aβ42/Aβ40. The family Desulfovibrionaceae (B), genus Bilophila (C), and genus Faecalibacterium (D) were negatively correlated with the global SUVR. Aβ amyloid-β, SUVR standard uptake value rate
Fig. 5
Fig. 5
ROCs for CN+ participants. A The discriminative power of individual plasma Aβ markers and the combined panel in identifying CN+ from CN−; B The discriminative power of each of the gut taxa and the combined panel in identifying CN+ from CN−; C The predicted values of the combined panels in identifying CN+ from CN−. Aβ, amyloid-β; CN−, amyloid-β-negative cognitively normal participants; CN+, amyloid-β-positive cognitively normal participants; panel 1, the combined plasma Aβ40, Aβ42, and Aβ42/ Aβ40; panel 2, the combined taxa 1, taxa 2, and taxa 3; panel 3, the combined clinical cognitive tests (including MoCA-B, AVLT-long delayed recall, AVLT-R, STT-A, STT-B, AFT, BNT); panel 4, the combined plasma Aβ markers, gut taxa, and cognitive tests

References

    1. Jia J, Wei C, Chen S, Li F, Tang Y, Qin W, et al. The cost of Alzheimer's disease in China and re-estimation of costs worldwide. Alzheimers Dement. 2018;14:483–491.
    1. Jack CR, Jr, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA research framework: toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018;14:535–562.
    1. Wang X, Huang W, Su L, Xing Y, Jessen F, Sun Y, et al. Neuroimaging advances regarding subjective cognitive decline in preclinical Alzheimer's disease. Mol Neurodegener. 2020;15:55.
    1. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer's disease. Lancet. 2016;388:505–517.
    1. Cryan JF, O'Riordan KJ, Sandhu K, Peterson V, Dinan TG. The gut microbiome in neurological disorders. Lancet Neurol. 2020;19:179–194.
    1. Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The microbiota-gut-brain axis. Physiol Rev. 2019;99:1877–2013.
    1. Sherwin E, Dinan TG, Cryan JF. Recent developments in understanding the role of the gut microbiota in brain health and disease. Ann N Y Acad Sci. 2018;1420:5–25.
    1. Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N, Festari C, 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.
    1. Abraham D, Feher J, Scuderi GL, Szabo D, Dobolyi A, Cservenak M, et al. Exercise and probiotics attenuate the development of Alzheimer's disease in transgenic mice: role of microbiome. Exp Gerontol. 2019;115:122–131.
    1. Bonfili L, Cecarini V, Berardi S, Scarpona S, Suchodolski JS, Nasuti C, et al. Microbiota modulation counteracts Alzheimer's disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci Rep. 2017;7:2426.
    1. Liu P, Wu L, Peng G, Han Y, Tang R, Ge J, et al. Altered microbiomes distinguish Alzheimer's disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav Immun. 2019;80:633–643.
    1. Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, et al. Gut microbiome alterations in Alzheimer's disease. Sci Rep. 2017;7:13537.
    1. Li B, He Y, Ma J, Huang P, Du J, Cao L, et al. Mild cognitive impairment has similar alterations as Alzheimer's disease in gut microbiota. Alzheimers Dement. 2019;15:1357–1366.
    1. Sheng C, Lin L, Lin H, Wang X, Han Y, Liu SL. Altered gut microbiota in adults with subjective cognitive decline: the SILCODE study. J Alzheimer's Dis. 2021;82:513–26.
    1. Chouraki V, Beiser A, Younkin L, Preis SR, Weinstein G, Hansson O, et al. Plasma amyloid-β and risk of Alzheimer's disease in the Framingham Heart Study. Alzheimers Dement. 2015;11:249–57.e1.
    1. Hanon O, Vidal JS, Lehmann S, Bombois S, Allinquant B, Tréluyer JM, et al. Plasma amyloid levels within the Alzheimer's process and correlations with central biomarkers. Alzheimers Dement. 2018;14:858–868.
    1. Song F, Poljak A, Valenzuela M, Mayeux R, Smythe GA, Sachdev PS. Meta-analysis of plasma amyloid-β levels in Alzheimer's disease. J Alzheimer's Dis. 2011;26:365–375.
    1. Lim YY, Maruff P, Kaneko N, Doecke J, Fowler C, Villemagne VL, et al. Plasma amyloid-β biomarker associated with cognitive decline in preclinical Alzheimer's disease. J Alzheimer's Dis. 2020;77:1057–1065.
    1. Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Doré V, et al. High performance plasma amyloid-β biomarkers for Alzheimer's disease. Nature. 2018;554:249–254.
    1. Pérez-Grijalba V, Arbizu J, Romero J, Prieto E, Pesini P, Sarasa L, et al. Plasma Aβ42/40 ratio alone or combined with FDG-PET can accurately predict amyloid-PET positivity: a cross-sectional analysis from the AB255 Study. Alzheimers Res Ther. 2019;11:96.
    1. Li X, Wang X, Su L, Hu X, Han Y. Sino Longitudinal Study on Cognitive Decline (SILCODE): protocol for a Chinese longitudinal observational study to develop risk prediction models of conversion to mild cognitive impairment in individuals with subjective cognitive decline. BMJ Open. 2019;9:e028188.
    1. Bondi MW, Edmonds EC, Jak AJ, Clark LR, Delano-Wood L, McDonald CR, et al. Neuropsychological criteria for mild cognitive impairment improves diagnostic precision, biomarker associations, and progression rates. J Alzheimer's Dis. 2014;42:275–289.
    1. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Jr, Kawas CH, 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.
    1. Zhao Q, Lv Y, Zhou Y, Hong Z, Guo Q. Short-term delayed recall of auditory verbal learning test is equivalent to long-term delayed recall for identifying amnestic mild cognitive impairment. PLoS One. 2012;7:e51157.
    1. Zhao Q, Guo Q, Li F, Zhou Y, Wang B, Hong Z. The Shape Trail Test: application of a new variant of the Trail making test. PLoS One. 2013;8:e57333.
    1. Guo QJL, Hong Z, Lv C. A specific phenomenon of animal fluency test in chinese elderly. Chin Mental Health J. 2007;21:622–625.
    1. Guo QHZ, Shi W, Sun Y, Lv C. Boston naming test in Chinese elderly, patient with mild cognitive impairment and Alzheimer's dementia. Chin Mental Health J. 2006;20:81–84.
    1. Chen KL, Xu Y, Chu AQ, Ding D, Liang XN, Nasreddine ZS, et al. Validation of the Chinese version of montreal cognitive assessment basic for screening mild cognitive impairment. J Am Geriatr Soc. 2016;64:e285–ee90.
    1. Qian Y, Yang X, Xu S, Wu C, Song Y, Qin N, et al. Alteration of the fecal microbiota in Chinese patients with Parkinson’s disease. Brain Behav Immun. 2018;70:194–202.
    1. Rognes T, Flouri T, Nichols B, Quince C, Mahé F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4:e2584.
    1. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460–2461.
    1. Chen K, Roontiva A, Thiyyagura P, Lee W, Liu X, Ayutyanont N, et al. Improved power for characterizing longitudinal amyloid-β PET changes and evaluating amyloid-modifying treatments with a cerebral white matter reference region. J Nucl Med. 2015;56:560–566.
    1. Li TR, Wu Y, Jiang JJ, Lin H, Han CL, Jiang JH, et al. Radiomics analysis of magnetic resonance imaging facilitates the identification of preclinical Alzheimer's Disease: An Exploratory study. Front Cell Dev Biol. 2020;8:605734.
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44:837–845.
    1. Lövheim H, Elgh F, Johansson A, Zetterberg H, Blennow K, Hallmans G, et al. Plasma concentrations of free amyloid β cannot predict the development of Alzheimer's disease. Alzheimers Dement. 2017;13:778–782.
    1. Rembach A, Faux NG, Watt AD, Pertile KK, Rumble RL, Trounson BO, et al. Changes in plasma amyloid beta in a longitudinal study of aging and Alzheimer's disease. Alzheimers Dement. 2014;10:53–61.
    1. Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, et al. CSF and blood biomarkers for the diagnosis of Alzheimer's disease: a systematic review and meta-analysis. Lancet Neurol. 2016;15:673–684.
    1. Verberk IMW, Thijssen E, Koelewijn J, Mauroo K, Vanbrabant J, de Wilde A, et al. Combination of plasma amyloid beta((1-42/1-40)) and glial fibrillary acidic protein strongly associates with cerebral amyloid pathology. Alzheimers Res Ther. 2020;12:118.
    1. Bhat MI, Kapila R. Dietary metabolites derived from gut microbiota: critical modulators of epigenetic changes in mammals. Nutr Rev. 2017;75:374–389.
    1. Kumar H, Lund R, Laiho A, Lundelin K, Ley RE, Isolauri E, et al. Gut microbiota as an epigenetic regulator: pilot study based on whole-genome methylation analysis. mBio. 2014;5:e02113–14.
    1. Megur A, Baltriukienė D, Bukelskienė V, Burokas A. The microbiota-gut-brain axis and Alzheimer's disease: neuroinflammation is to blame? Nutrients. 2020;13:37.
    1. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012;336:1268–1273.
    1. Goyal D, Ali SA, Singh RK. Emerging role of gut microbiota in modulation of neuroinflammation and neurodegeneration with emphasis on Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry. 2021;106:110112.
    1. Welcome MO. Gut microbiota disorder, gut epithelial and blood-brain barrier dysfunctions in etiopathogenesis of dementia: molecular mechanisms and signaling pathways. Neuromolecular Med. 2019;21:205–226.
    1. Zhang L, Wang Y, Xiayu X, Shi C, Chen W, Song N, et al. Altered gut microbiota in a mouse model of Alzheimer's disease. J Alzheimer's Dis. 2017;60:1241–1257.
    1. Canfora EE, Jocken JW, Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol. 2015;11:577–591.
    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.
    1. Sanna S, van Zuydam NR, Mahajan A, Kurilshikov A, Vich Vila A, Võsa U, et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet. 2019;51:600–605.
    1. Silva YP, Bernardi A, Frozza RL. The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol (Lausanne) 2020;11:25.
    1. Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014;6:263ra158.
    1. Allin KH, Tremaroli V, Caesar R, Jensen BAH, Damgaard MTF, Bahl MI, et al. Aberrant intestinal microbiota in individuals with prediabetes. Diabetologia. 2018;61:810–820.
    1. Zhuang ZQ, Shen LL, Li WW, Fu X, Zeng F, Gui L, et al. Gut Microbiota is altered in patients with Alzheimer's disease. J Alzheimer's Dis. 2018;63:1337–1346.
    1. Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev. 2014;38:996–1047.
    1. Zhan X, Stamova B, Sharp FR. Lipopolysaccharide associates with amyloid plaques, neurons and oligodendrocytes in Alzheimer's disease brain: a review. Front Aging Neurosci. 2018;10:42.
    1. Lukiw WJ. Bacteroides fragilis lipopolysaccharide and inflammatory signaling in Alzheimer's disease. Front Microbiol. 2016;7:1544.
    1. Waite DW, Chuvochina M, Pelikan C, Parks DH, Yilmaz P, Wagner M, et al. Proposal to reclassify the proteobacterial classes Deltaproteobacteria and Oligoflexia, and the phylum Thermodesulfobacteria into four phyla reflecting major functional capabilities. Int J Syst Evol Microbiol. 2020;70:5972–6016.
    1. Attene-Ramos MS, Wagner ED, Plewa MJ, Gaskins HR. Evidence that hydrogen sulfide is a genotoxic agent. Mol Cancer Res. 2006;4:9–14.
    1. Sheng C, Sun Y, Wang M, Wang X, Liu Y, Pang D, et al. Combining visual rating scales for medial temporal lobe atrophy and posterior atrophy to identify amnestic mild cognitive impairment from cognitively normal older adults: evidence based on two cohorts. J Alzheimer's Dis. 2020;77:323–337.
    1. Harper L, Fumagalli GG, Barkhof F, Scheltens P, O'Brien JT, Bouwman F, et al. MRI visual rating scales in the diagnosis of dementia: evaluation in 184 post-mortem confirmed cases. Brain. 2016;139:1211–1225.
    1. Liu YX, Qin Y, Chen T, Lu M, Qian X, Guo X, et al. A practical guide to amplicon and metagenomic analysis of microbiome data. Protein Cell. 2021;12:315–30.

Source: PubMed

3
Subscribe