Abnormal gut microbiota composition contributes to cognitive dysfunction in SAMP8 mice

Gaofeng Zhan, Ning Yang, Shan Li, Niannian Huang, Xi Fang, Jie Zhang, Bin Zhu, Ling Yang, Chun Yang, Ailin Luo, Gaofeng Zhan, Ning Yang, Shan Li, Niannian Huang, Xi Fang, Jie Zhang, Bin Zhu, Ling Yang, Chun Yang, Ailin Luo

Abstract

Alzheimer's disease is characterized by cognitive dysfunction and aging is an important predisposing factor; however, the pathological and therapeutic mechanisms are not fully understood. Recently, the role of gut microbiota in Alzheimer's disease has received increasing attention. The cognitive function in senescence-accelerated mouse prone 8 (SAMP8) mice was significantly decreased and the Chao 1 and Shannon indices, principal coordinates analysis, and principal component analysis results were notably abnormal compared with that of those in senescence-accelerated mouse resistant 1 (SAMR1) mice. Moreover, 27 gut bacteria at six phylogenetic levels differed between SAMP8 and SAMR1 mice. In a separate study, we transplanted fecal bacteria from SAMP8 or SAMR1 mice into pseudo germ-free mice. Interestingly, the pseudo germ-free mice had significantly lower cognitive function prior to transplant. Pseudo germ-free mice that received fecal bacteria transplants from SAMR1 mice but not from SAMP8 mice showed improvements in behavior and in α-diversity and β-diversity indices. In total, 14 bacteria at six phylogenetic levels were significantly altered by the gut microbiota transplant. These results suggest that cognitive dysfunction in SAMP8 mice is associated with abnormal composition of the gut microbiota. Thus, improving abnormal gut microbiota may provide an alternative treatment for cognitive dysfunction and Alzheimer's disease.

Keywords: Alzheimer’s disease; SAMP8; cognitive dysfunction; gut microbiota; pseudo germ-free mice.

Conflict of interest statement

CONFLICTS OF INTEREST: CY received research support from B. Braun. Other authors declared no conflicts of interest.

Figures

Figure 1
Figure 1
MWMT for SAMR1 and SAMP8 mice. (A) The schedule for MWMT and fecal sample collection. Seven days after accommodation, SAMR1 and SAMP8 mice were scheduled for MWMT and probe trial on days 1–6. On day 7, fecal samples were collected for 16S rRNA gene sequencing and fecal bacteria transplant. (B) Body weight (T-test, P > 0.05). (C) The trace graph of SAMR1 and SAMP8 mice in MWMT. (D) Escape latency (two-way ANOVA; Time: F4,20 = 6.062, P < 0.01; Group: F1,5 = 8.802, P < 0.05; Interaction: F4,20 = 3.476, P < 0.05). (E) Escape path length (two-way ANOVA. Time: F4,20 = 11.32, P < 0.001; Group: F1,5 = 4.834, P = 0.0792; Interaction: F4,20 = 3.271, P < 0.05). (F) Platform crossing (t-test, P < 0.05). (G) Time spent in each quadrant (t-test, P < 0.05). ANOVA: analysis of variance; MWMT: Morris water maze test. Data are shown as mean ± SEM (n = 6). *P < 0.05, **P < 0.01.
Figure 2
Figure 2
Differential profiles of the gut microbiota between SAMR1 and SAMP8 mice. (A) Heat map of differential levels of bacteria between the groups. (B) Chao 1 index (t-test, P < 0.01). (C) Shannon index (t-test, P < 0.05). (D) Simpson index (t-test, P > 0.05). (E) PCoA analysis of gut bacteria data (Bray–Curtis dissimilarity). (F) PCA analysis of gut bacteria data. α-diversity data are shown as mean ± SEM (n = 10). PCA: principal component analysis; PCoA: principal coordinates analysis. *P < 0.05, **P < 0.01.
Figure 3
Figure 3
Differential levels of the gut bacteria between SAMR1 and SAMP8 mice. (A) Phylum Deferribacteres (t-test, P < 0.05). (B) Class Deferribacteres (t-test, P < 0.05). (C) Class Deltaproteobacteria (t-test, P < 0.001). (D) Order Deferribacterales (t-test, P < 0.05). (E) Order Desulfovibrionales (t-test, P < 0.001). (F) Order Mollicutes RF9 (t-test, P < 0.01). (G) Family Clostridiales vadinBB60 group (t-test, P < 0.05). (H) Family Desulfovibrionaceae (t-test, P < 0.001). (I) Family Christensenellaceae (t-test, P < 0.001). (J) Family Family XIII (t-test, P < 0.001). (K) Family Deferribacteraceae (t-test, P < 0.001). (L) Family Ruminococcaceae (t-test, P < 0.001). (M) Genus Mucispirillum (t-test, P < 0.05). (N) Genus Serratia (t-test, P < 0.05). (O) Genus Family XIII AD3011 group (t-test, P < 0.001). (P) Genus Christensenellaceae R-7 group (t-test, P < 0.01). (Q) Genus Subdoligranulum (t-test, P < 0.05). (R) Genus Desulfovibrio (t-test, P < 0.001). (S) Genus Ruminiclostridium 9 (t-test, P < 0.01). (T) Genus Coprococcus 1 (t-test, P < 0.05). (U) Genus Ruminococcaceae UCG 004 (t-test, P < 0.05). (V) Genus Lachnospiraceae NK4A136 group (t-test, P < 0.05). (W) Genus Lachnospiraceae oscillibacter (t-test, P < 0.001). (X) Species Desulfovibrio sp. UNSW3caefatS (t-test, P < 0.001). (Y) Species uncultured Deferribacteraceae bacterium (t-test, P < 0.05). (Z) Species uncultured Bacteroidales bacterium (t-test, P < 0.001). (AA) Species Unidentified (t-test, P < 0.05).
Figure 4
Figure 4
Effects of SAMR1 and SAMP8 mice fecal microbiota transplant on behavior in pseudo germ-free mice. (A) Schedule of fecal microbiota transplant on behavioral testing in pseudo germ-free mice. Mice were first treated by administering large doses of antibiotic solution for 14 consecutive days. Thereafter, mice were orally treated with fetal microbiota of SAMR1 and SAMP8 mice. MWMT was performed on days 29–33, and the probe trial was performed on day 34. On day 35, fecal samples were collected for 16S rRNA gene sequencing testing. (B) Body weight (Two-way ANOVA; Time: F2,14 = 25.59, P < 0.001; Group: F3,21 = 0.1132, P = 0.9514; Interaction: F6,42 = 0.6019, P = 0.7272.). (C) The trace graph of mice in MWMT. (D) Escape latency (two-way ANOVA; Time: F4,28 = 17.23, P < 0.001; Group: F3,21 = 3.27, P < 0.05; Interaction: F12,84 = 0.9568, P = 0.4961). (E) Escape path length (two-way ANOVA; Time: F4,28 = 32.04, P < 0.001; Group: F3,21 = 8.668, P < 0.001; Interaction: F12,84 = 0.7349, P = 0.7137.). (F) Platform crossing (one-way ANOVA; F3,28 = 8.745, P < 0.001.). (G) Time spent in each quadrant (one-way ANOVA; F3,28 = 9.133, P < 0.001). ANOVA: analysis of variance; MWMT: Morris water maze test. Data are shown as mean ± SEM (n = 8). *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 5
Figure 5
α-diversity and β-diversity of fecal microbiota transplant in pseudo germ-free mice. (A) Chao 1 index (One-way ANOVA; F3,24 = 21.28, P < 0.001). (B) Observed species index (One-way ANOVA; F3,24 = 17.29, P < 0.001). (C) PD whole tree (One-way ANOVA; F3,24 = 16.76, P < 0.001). (D) Shannon index (One-way ANOVA; F3,24 = 50.19, P < 0.001). Data are shown as mean ± SEM (n = 7). *P < 0.05, **P < 0.01, ***P < 0.001. (E) PCoA analysis of gut bacteria data (Bray–Curtis dissimilarity). (F) NMDS analysis of gut bacteria data. NMDS: non-metric multi-dimensional scaling; PCoA: principal coordinates analysis.
Figure 6
Figure 6
Effects of fecal microbiota transplant on levels of gut microbiota in pseudo germ-free mice. (A) Phylum Firmicutes (One-way ANOVA; F3,24 = 34.61, P < 0.001). (B) Class Betaproteobacteria (One-way ANOVA; F3,24 = 37.98, P < 0.001). (C) Order Bacteroidales (One-way ANOVA; F3,24 = 17.77, P < 0.001). (D) Order Burkholderiales (One-way ANOVA; F3,24 = 41.43, P < 0.001). (E) Order Clostridiales (One-way ANOVA; F3,24 = 36.92, P < 0.001). (F) Family Alcaligenaceae (One-way ANOVA; F3,24 = 34.95, P < 0.001). (G) Family Lachnospiraceae (One-way ANOVA; F3,24 = 40.75, P < 0.001). (H) Family Prevotellaceae (One-way ANOVA; F3,24 = 26.69, P < 0.001). (I) Genus Lachnospiraceae NK4A136 group (One-way ANOVA; F3,24 = 30.02, P < 0.001). (J) Genus Moryella (Fisher’s exact test; P < 0.001). (K) Genus Parasutterella (One-way ANOVA; F3,24 = 37.64, P < 0.001). (L) Genus Peptococcus (Fisher’s exact test; P < 0.001). (M) Genus Prevotellaceae NK3B31 group (Fisher’s exact test; P < 0.05). (N) Species Lachnospiraceae bacterium 615 (Fisher’s exact test; P < 0.05). ANOVA: analysis of variance

References

    1. Harris SE, Riggio V, Evenden L, Gilchrist T, McCafferty S, Murphy L, Wrobel N, Taylor AM, Corley J, Pattie A, Cox SR, Martin-Ruiz C, Prendergast J, et al.. Age-related gene expression changes, and transcriptome wide association study of physical and cognitive aging traits, in the Lothian Birth Cohort 1936. Aging (Albany NY). 2017; 9:2489–503.
    1. Bories C, Arsenault D, Lemire M, Tremblay C, De Koninck Y, Calon F. Transgenic autoinhibition of p21-activated kinase exacerbates synaptic impairments and fronto-dependent behavioral deficits in an animal model of Alzheimer’s disease. Aging (Albany NY). 2017; 9:1386–403.
    1. Villemagne VL, Masters CL. Alzheimer disease: the landscape of ageing--insights from AD imaging markers. Nat Rev Neurol. 2014; 10:678–79. 10.1038/nrneurol.2014.214
    1. Kang J, Lu J, Zhang X. Metabolomics-based promising candidate biomarkers and pathways in Alzheimer’s disease. Pharmazie. 2015; 70:277–82.
    1. Huang JL, Qin MC, Zhou Y, Xu ZH, Yang SM, Zhang F, Zhong J, Liang MK, Chen B, Zhang WY, Wu DP, Zhong ZG. Comprehensive analysis of differentially expressed profiles of Alzheimer’s disease associated circular RNAs in an Alzheimer’s disease mouse model. Aging (Albany NY). 2018; 10:253–65.
    1. Currais A, Goldberg J, Farrokhi C, Chang M, Prior M, Dargusch R, Daugherty D, Armando A, Quehenberger O, Maher P, Schubert D. A comprehensive multiomics approach toward understanding the relationship between aging and dementia. Aging (Albany NY). 2015; 7:937–52. 10.18632/aging.100838
    1. O’Toole PW, Jeffery IB. Gut microbiota and aging. Science. 2015; 350:1214–15. 10.1126/science.aac8469
    1. Petra AI, Panagiotidou S, Hatziagelaki E, Stewart JM, Conti P, Theoharides TC. Gut-microbiota-brain axis and its effect on neuropsychiatric disorders with suspected immune dysregulation. Clin Ther. 2015; 37:984–95. 10.1016/j.clinthera.2015.04.002
    1. Bienenstock J, Kunze W, Forsythe P. Microbiota and the gut-brain axis. Nutr Rev. 2015. (Suppl 1); 73:28–31. 10.1093/nutrit/nuv019
    1. Yip LY, Chan EC. Investigation of host-gut microbiota modulation of therapeutic outcome. Drug Metab Dispos. 2015; 43:1619–31. 10.1124/dmd.115.063750
    1. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest. 2015; 125:926–38. 10.1172/JCI76304
    1. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015; 28:203–09.
    1. Yang C, Fujita Y, Ren Q, Ma M, Dong C, Hashimoto K. Bifidobacterium in the gut microbiota confer resilience to chronic social defeat stress in mice. Sci Rep. 2017; 7:45942. 10.1038/srep45942
    1. Yang C, Qu Y, Fujita Y, Ren Q, Ma M, Dong C, Hashimoto K. Possible role of the gut microbiota-brain axis in the antidepressant effects of (R)-ketamine in a social defeat stress model. Transl Psychiatry. 2017; 7:1294. 10.1038/s41398-017-0031-4
    1. Mancuso C, Santangelo R. Alzheimer’s disease and gut microbiota modifications: the long way between preclinical studies and clinical evidence. Pharmacol Res. 2018; 129:329–36. 10.1016/j.phrs.2017.12.009
    1. Leblhuber F, Egger M, Schuetz B, Fuchs D. Commentary: effect of probiotic supplementation on cognitive function and metabolic status in Alzheimer’s disease: a randomized, double-blind and controlled trial. Front Aging Neurosci. 2018; 10:54. 10.3389/fnagi.2018.00054
    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. Sci Rep. 2017; 7:41802. 10.1038/srep41802
    1. Jiang C, Li G, Huang P, Liu Z, Zhao B. The gut microbiota and Alzheimer’s disease. J Alzheimers Dis. 2017; 58:1–15. 10.3233/JAD-161141
    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 Rev Neurother. 2018; 18:83–90. 10.1080/14737175.2018.1400909
    1. Cattaneo A, Cattane N, Galluzzi S, Provasi S, Lopizzo N, Festari C, Ferrari C, Guerra UP, Paghera B, Muscio C, Bianchetti A, Volta GD, Turla M, et al., and INDIA-FBP Group. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol Aging. 2017; 49:60–68. 10.1016/j.neurobiolaging.2016.08.019
    1. Nadal A, Quesada I, Tudurí E, Nogueiras R, Alonso-Magdalena P. Endocrine-disrupting chemicals and the regulation of energy balance. Nat Rev Endocrinol. 2017; 13:536–46. 10.1038/nrendo.2017.51
    1. Bermon S, Petriz B, Kajėnienė A, Prestes J, Castell L, Franco OL. The microbiota: an exercise immunology perspective. Exerc Immunol Rev. 2015; 21:70–79.
    1. Huang KY, Liang S, Yu ML, Fu SP, Chen X, Lu SF. A systematic review and meta-analysis of acupuncture for improving learning and memory ability in animals. BMC Complement Altern Med. 2016; 16:297. 10.1186/s12906-016-1298-3
    1. Sawano E, Iwatani K, Tominaga-Yoshino K, Ogura A, Tashiro T. Reduction in NPY-positive neurons and dysregulation of excitability in young senescence-accelerated mouse prone 8 (SAMP8) hippocampus precede the onset of cognitive impairment. J Neurochem. 2015; 135:287–300. 10.1111/jnc.13274
    1. Dong W, Guo W, Zheng X, Wang F, Chen Y, Zhang W, Shi H. Electroacupuncture improves cognitive deficits associated with AMPK activation in SAMP8 mice. Metab Brain Dis. 2015; 30:777–84. 10.1007/s11011-014-9641-1
    1. Torres-Fuentes C, Schellekens H, Dinan TG, Cryan JF. The microbiota-gut-brain axis in obesity. Lancet Gastroenterol Hepatol. 2017; 2:747–56. 10.1016/S2468-1253(17)30147-4
    1. Cerdó T, Ruíz A, Suárez A, Campoy C. Probiotic, prebiotic, and brain development. Nutrients. 2017; 9:1247. 10.3390/nu9111247
    1. Wang J, Ye F, Cheng X, Zhang X, Liu F, Liu G, Ni M, Qiao S, Zhou W, Zhang Y. The effects of lw-afc on intestinal microbiome in senescence-accelerated mouse prone 8 strain, a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2016; 53:907–19. 10.3233/JAD-160138
    1. Farzi A, Fröhlich EE, Holzer P. Gut microbiota and the neuroendocrine system. Neurotherapeutics. 2018; 15:5–22. 10.1007/s13311-017-0600-5
    1. Parker A, Lawson MA, Vaux L, Pin C. Host-microbe interaction in the gastrointestinal tract. Environ Microbiol. 2017. 10.1111/1462-2920.13926
    1. Hernández-Chirlaque C, Aranda CJ, Ocón B, Capitán-Cañadas F, Ortega-González M, Carrero JJ, Suárez MD, Zarzuelo A, Sánchez de Medina F, Martínez-Augustin O. Germ-free and antibiotic-treated mice are highly susceptible to epithelial injury in DSS colitis. J Crohn’s Colitis. 2016; 10:1324–35. 10.1093/ecco-jcc/jjw096
    1. Hoffman JD, Parikh I, Green SJ, Chlipala G, Mohney RP, Keaton M, Bauer B, Hartz AM, Lin AL. Age drives distortion of brain metabolic, vascular and cognitive functions, and the gut microbiome. Front Aging Neurosci. 2017; 9:298. 10.3389/fnagi.2017.00298
    1. Sanguinetti E, Collado MC, Marrachelli VG, Monleon D, Selma-Royo M, Pardo-Tendero MM, Burchielli S, Iozzo P. Microbiome-metabolome signatures in mice genetically prone to develop dementia, fed a normal or fatty diet. Sci Rep. 2018; 8:4907. 10.1038/s41598-018-23261-1
    1. Nagai F, Morotomi M, Sakon H, Tanaka R. Parasutterella excrementihominis gen. nov., sp. nov., a member of the family Alcaligenaceae isolated from human faeces. Int J Syst Evol Microbiol. 2009; 59:1793–97. 10.1099/ijs.0.002519-0
    1. Vorhees CV, Williams MT. Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nat Protoc. 2006; 1:848–58. 10.1038/nprot.2006.116
    1. Ge X, Zhao W, Ding C, Tian H, Xu L, Wang H, Ni L, Jiang J, Gong J, Zhu W, Zhu M, Li N. Potential role of fecal microbiota from patients with slow transit constipation in the regulation of gastrointestinal motility. Sci Rep. 2017; 7:441. 10.1038/s41598-017-00612-y
    1. Sun H, Wang N, Cang Z, Zhu C, Zhao L, Nie X, Cheng J, Xia F, Zhai H, Lu Y. Modulation of microbiota-gut-brain axis by berberine resulting in improved metabolic status in high-fat diet-fed rats. Obes Facts. 2016; 9:365–78. 10.1159/000449507

Source: PubMed

3
구독하다