Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer's disease

Myles R Minter, Can Zhang, Vanessa Leone, Daina L Ringus, Xiaoqiong Zhang, Paul Oyler-Castrillo, Mark W Musch, Fan Liao, Joseph F Ward, David M Holtzman, Eugene B Chang, Rudolph E Tanzi, Sangram S Sisodia, Myles R Minter, Can Zhang, Vanessa Leone, Daina L Ringus, Xiaoqiong Zhang, Paul Oyler-Castrillo, Mark W Musch, Fan Liao, Joseph F Ward, David M Holtzman, Eugene B Chang, Rudolph E Tanzi, Sangram S Sisodia

Abstract

Severe amyloidosis and plaque-localized neuro-inflammation are key pathological features of Alzheimer's disease (AD). In addition to astrocyte and microglial reactivity, emerging evidence suggests a role of gut microbiota in regulating innate immunity and influencing brain function. Here, we examine the role of the host microbiome in regulating amyloidosis in the APPSWE/PS1ΔE9 mouse model of AD. We show that prolonged shifts in gut microbial composition and diversity induced by long-term broad-spectrum combinatorial antibiotic treatment regime decreases Aβ plaque deposition. We also show that levels of soluble Aβ are elevated and that levels of circulating cytokine and chemokine signatures are altered in this setting. Finally, we observe attenuated plaque-localised glial reactivity in these mice and significantly altered microglial morphology. These findings suggest the gut microbiota community diversity can regulate host innate immunity mechanisms that impact Aβ amyloidosis.

Figures

Figure 1. Composition of the gastrointestinal microbiome…
Figure 1. Composition of the gastrointestinal microbiome is altered in ABX-treated APPSWE/PS1ΔE9 mice.
(A) Q-PCR analysis of isolated DNA from cecal contents of 6 month old male APPSWE/PS1ΔE9 mice analysing 16s rRNA gene copy number (n = 9–10). (B) Q-PCR analysis of isolated DNA from fecal contents of 6 month old male APPSWE/PS1ΔE9 mice analysing 16s rRNA gene copy number (n = 9–10). To obtain a 16s rRNA gene copy number value, expression levels were normalized to both DNA concentration and an amplification standard curve of a 16s rRNA gene-containing plasmid with known copy number. Illumina® MiSeq based sequencing of the 16s rRNA gene from cecal and fecal contents of 6 month old male APPSWE/PS1ΔE9 mice was performed and a microbial (C) phylogenetic tree and (D) diversity histogram were generated based on quality-controlled OTU reads. Only families and genus’ with relative abundance >0.5% were included. (E) α-diversity analysis of the sequencing data using the Shannon index, reveals a significant decrease in microbial diversification in ABX-treated APPSWE/PS1ΔE9 mice (n = 9–10, *p = 0.0429, unpaired two-tailed Student’s t-test). Principal co-ordinate analysis of (F) un-weighted, accounting for presence of OTUs only, and (G) weighted β-diversity, accounting for both presence and relative abundance of OTUs, demonstrates distinct alterations in microbial diversity in ABX-treated APPSWE/PS1ΔE9 mice (n = 9–10 mice, each with cecal and fecal sequencing). The percentage of data variance explained by each IPCA is displayed. Data are displayed as X/Y scatter or mean ± SEM. See Supplementary Figure 2–3, statistical Table 1 and Supplementary table 2 for additional information.
Figure 2. The circulating inflammatory mediator profile…
Figure 2. The circulating inflammatory mediator profile is altered in ABX-treated APPSWE/PS1ΔE9 mice.
(A) Immunoblot-based array of inflammatory mediators in isolated serum from vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice (n = 10, pooled sera). (B) Densitometry of select inflammatory mediators from the aforementioned array. Expression levels in ABX-treated mice are expressed in arbitrary units relative to vehicle control (dashed line). Data are displayed as mean alone. See Supplementary Figures 4 and 5 for additional information.
Figure 3. Amyloidosis is altered in ABX-treated…
Figure 3. Amyloidosis is altered in ABX-treated male APPSWE/PS1ΔE9 mice.
(A) Representative immunohistochemical images of Aβ plaque deposition in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice using anti-Aβ mAb 3D6. Each staining run was performed using sections from 12 month old APPSWE/PS1ΔE9 mice as a positive control and no primary antibody negative controls. (B) Plaque burden quantification in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice using particle analysis of 3D6+ve immunofluorescence (n = 10, *p = 0.0169, unpaired two-tailed Student’s t-test). 3D6+ve area was averaged from 4 sections/mouse (240 μm apart) and expressed relative to total cortical and hippocampal area. (C) Representative x60 magnification z-stack maximum projection images of 3D6+ve Aβ plaques from vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice. (D) Quantification of plaque area using threshold-limiting immunofluorescence detection (n = 10, *p = 0.02, unpaired two-tailed Student’s t-test). (E) MSD Mesoscale® analysis of TFA-soluble (TBS-insoluble) Aβ1-40 and Aβ1-42 levels in combined cortical and hippocampal tissue of vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice using anti-Aβ mAb 4G8 (n = 10, *p = 0.0491, unpaired two-tailed Student’s t-test). (F) MSD Mesoscale® analysis of TBS-soluble Aβ1-40 and Aβ1-42 levels in combined cortical and hippocampal tissue of vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice using anti-Aβ mAb 4G8 (n = 10, *p < 0.05, unpaired two-tailed Student’s t-test). (G) Quantification of the Aβ1-40:Aβ1-42 ratio from the TBS-soluble MSD Mesoscale® data in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice (n = 10, *p = 0.014, unpaired two-tailed Student’s t-test). Data are displayed as mean ± SEM. See Supplementary Figure 6–9 and statistical table 1 for additional information.
Figure 4. Plaque-localized gliosis is influenced by…
Figure 4. Plaque-localized gliosis is influenced by ABX treatment in male APPSWE/PS1ΔE9 mice.
(A) Representative x60 magnification z-stack maximum projection images of IBA-1+ve Aβ plaque-localized microglia, co-stained with DAPI, in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice. (B) Quantification of plaque-localized IBA-1+ve microglial number in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice (n = 8, ***p < 0.001, unpaired two-tailed Student’s t-test). (C) Plaque-localized IBA+ve microglial number expressed relative to 3D6+ve Aβ plaque area in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice (n = 8). (D) Representative 3D IMARIS-based reconstructions of x60 magnification z-stack images (0.8 μm apart) depicting plaque-localized IBA-1+ve microglial morphology in in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice. Microglial branching, terminal points and Aβ plaque surfaces can be seen. The (E) length and (F) number of microglial dendritic branches alongside (G) projection terminal points were quantified (n = 4, *p = 0.0237, **p < 0.01, unpaired two-tailed Student’s t-test). (H) Representative x60 magnification z-stack maximum projection images of GFAP+ve Aβ plaque-localized astrocytes, co-stained with DAPI, in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice. (I) Quantification of plaque-localized GFAP+ve astrocyte number in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice (n = 8, **p = 0.001, unpaired two-tailed Student’s t-test). (J) Plaque-localized GFAP+ve astrocyte number expressed relative to 3D6+ve Aβ plaque area in vehicle control and ABX-treated male 6 month old APPSWE/PS1ΔE9 mice (n = 8). Data are displayed as mean ± SEM. See statistical table 1 for additional information.

References

    1. Arends Y. M., Duyckaerts C., Rozemuller J. M., Eikelenboom P. & Hauw J. J. Microglia, amyloid and dementia in alzheimer disease. A correlative study. Neurobiology of aging 21, 39–47 (2000).
    1. Beach T. G., Walker R. & McGeer E. G. Patterns of gliosis in Alzheimer’s disease and aging cerebrum. Glia 2, 420–436, doi: 10.1002/glia.440020605 (1989).
    1. Delacourte A. General and dramatic glial reaction in Alzheimer brains. Neurology 40, 33–37 (1990).
    1. Cagnin A. et al. In-vivo measurement of activated microglia in dementia. Lancet 358, 461–467, doi: 10.1016/S0140-6736(01)05625-2 (2001).
    1. Yasuno F. et al. Increased binding of peripheral benzodiazepine receptor in mild cognitive impairment-dementia converters measured by positron emission tomography with [(1)(1)C]DAA1106. Psychiatry research 203, 67–74, doi: 10.1016/j.pscychresns.2011.08.013 (2012).
    1. Griciuc A. et al. Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron 78, 631–643, doi: 10.1016/j.neuron.2013.04.014 (2013).
    1. Guerreiro R. et al. TREM2 variants in Alzheimer’s disease. The New England journal of medicine 368, 117–127, doi: 10.1056/NEJMoa1211851 (2013).
    1. Jonsson T. et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. The New England journal of medicine 368, 107–116, doi: 10.1056/NEJMoa1211103 (2013).
    1. Lambert J. C. et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nature genetics 41, 1094–1099, doi: 10.1038/ng.439 (2009).
    1. Naj A. C. et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nature genetics 43, 436–441, doi: 10.1038/ng.801 (2011).
    1. Naj A. C. et al. Effects of multiple genetic loci on age at onset in late-onset Alzheimer disease: a genome-wide association study. JAMA neurology 71, 1394–1404, doi: 10.1001/jamaneurol.2014.1491 (2014).
    1. Minter M. R., Taylor J. M. & Crack P. J. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. Journal of neurochemistry 136, 457–474, doi: 10.1111/jnc.13411 (2016).
    1. Chakrabarty P. et al. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron 85, 519–533, doi: 10.1016/j.neuron.2014.11.020 (2015).
    1. Guillot-Sestier M. V. et al. Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology. Neuron 85, 534–548, doi: 10.1016/j.neuron.2014.12.068 (2015).
    1. He P. et al. Deletion of tumor necrosis factor death receptor inhibits amyloid beta generation and prevents learning and memory deficits in Alzheimer’s mice. The Journal of cell biology 178, 829–841, doi: 10.1083/jcb.200705042 (2007).
    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. Vom Berg J. et al. Inhibition of IL-12/IL-23 signaling reduces Alzheimer’s disease-like pathology and cognitive decline. Nature medicine 18, 1812–1819, doi: 10.1038/nm.2965 (2012).
    1. Baruch K. et al. Aging. Aging-induced type I interferon response at the choroid plexus negatively affects brain function. Science 346, 89–93, doi: 10.1126/science.1252945 (2014).
    1. Taylor J. M. et al. Type-1 interferon signaling mediates neuro-inflammatory events in models of Alzheimer’s disease. Neurobiology of aging 35, 1012–1023, doi: 10.1016/j.neurobiolaging.2013.10.089 (2014).
    1. Baruch K. et al. Breaking immune tolerance by targeting Foxp3(+) regulatory T cells mitigates Alzheimer’s disease pathology. Nature communications 6, 7967, doi: 10.1038/ncomms8967 (2015).
    1. Gyoneva S. et al. Systemic inflammation regulates microglial responses to tissue damage in vivo. Glia 62, 1345–1360, doi: 10.1002/glia.22686 (2014).
    1. Sherwin E., Rea K., Dinan T. G. & Cryan J. F. A gut (microbiome) feeling about the brain. Current opinion in gastroenterology 32, 96–102, doi: 10.1097/MOG.0000000000000244 (2016).
    1. Kim H. R. et al. Delivery modes and neonatal EEG: spatial pattern analysis. Early human development 75, 35–53 (2003).
    1. Juarez I., Gratton A. & Flores G. Ontogeny of altered dendritic morphology in the rat prefrontal cortex, hippocampus, and nucleus accumbens following Cesarean delivery and birth anoxia. The Journal of comparative neurology 507, 1734–1747, doi: 10.1002/cne.21651 (2008).
    1. Bashir M. E., Louie S., Shi H. N. & Nagler-Anderson C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. Journal of immunology 172, 6978–6987 (2004).
    1. Hormann N. et al. Gut microbial colonization orchestrates TLR2 expression, signaling and epithelial proliferation in the small intestinal mucosa. PloS one 9, e113080, doi: 10.1371/journal.pone.0113080 (2014).
    1. Wang Y. et al. An intestinal commensal symbiosis factor controls neuroinflammation via TLR2-mediated CD39 signalling. Nature communications 5, 4432, doi: 10.1038/ncomms5432 (2014).
    1. Erny D. et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nature neuroscience 18, 965–977, doi: 10.1038/nn.4030 (2015).
    1. Jankowsky J. L. et al. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Human molecular genetics 13, 159–170, doi: 10.1093/hmg/ddh019 (2004).
    1. Jankowsky J. L. et al. Co-expression of multiple transgenes in mouse CNS: a comparison of strategies. Biomolecular engineering 17, 157–165 (2001).
    1. Lathrop S. K. et al. Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254, doi: 10.1038/nature10434 (2011).
    1. Sun J. et al. Pancreatic beta-Cells Limit Autoimmune Diabetes via an Immunoregulatory Antimicrobial Peptide Expressed under the Influence of the Gut Microbiota. Immunity 43, 304–317, doi: 10.1016/j.immuni.2015.07.013 (2015).
    1. Zhang Z. et al. Peripheral Lymphoid Volume Expansion and Maintenance Are Controlled by Gut Microbiota via RALDH(+) Dendritic Cells. Immunity 44, 330–342, doi: 10.1016/j.immuni.2016.01.004 (2016).
    1. Johnson-Wood K. et al. Amyloid precursor protein processing and A beta42 deposition in a transgenic mouse model of Alzheimer disease. Proceedings of the National Academy of Sciences of the United States of America 94, 1550–1555 (1997).
    1. Garai K., Verghese P. B., Baban B., Holtzman D. M. & Frieden C. The binding of apolipoprotein E to oligomers and fibrils of amyloid-beta alters the kinetics of amyloid aggregation. Biochemistry 53, 6323–6331, doi: 10.1021/bi5008172 (2014).
    1. Liao F. et al. Anti-ApoE antibody given after plaque onset decreases Abeta accumulation and improves brain function in a mouse model of Abeta amyloidosis. The Journal of neuroscience: the official journal of the Society for Neuroscience 34, 7281–7292, doi: 10.1523/JNEUROSCI.0646-14.2014 (2014).
    1. Liao F. et al. 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 3, 70, doi: 10.1186/s40478-015-0250-y (2015).
    1. Villeda S. A. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90–94, doi: 10.1038/nature10357 (2011).
    1. Lalli M. A. et al. Whole-genome sequencing suggests a chemokine gene cluster that modifies age at onset in familial Alzheimer’s disease. Molecular psychiatry 20, 1294–1300, doi: 10.1038/mp.2015.131 (2015).
    1. Erickson M. A., Morofuji Y., Owen J. B. & Banks W. A. Rapid transport of CCL11 across the blood-brain barrier: regional variation and importance of blood cells. The Journal of pharmacology and experimental therapeutics 349, 497–507, doi: 10.1124/jpet.114.213074 (2014).
    1. Song M. et al. TLR4 mutation reduces microglial activation, increases Abeta deposits and exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. Journal of neuroinflammation 8, 92, doi: 10.1186/1742-2094-8-92 (2011).
    1. Stefka A. T. et al. Commensal bacteria protect against food allergen sensitization. Proceedings of the National Academy of Sciences of the United States of America 111, 13145–13150, doi: 10.1073/pnas.1412008111 (2014).
    1. Hagihara H., Toyama K., Yamasaki N. & Miyakawa T. Dissection of hippocampal dentate gyrus from adult mouse. J Vis Exp, doi: 10.3791/1543 (2009).
    1. Wang Y. et al. 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis. The ISME journal 3, 944–954, doi: 10.1038/ismej.2009.37 (2009).
    1. Leone V. et al. Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism. Cell host & microbe 17, 681–689, doi: 10.1016/j.chom.2015.03.006 (2015).
    1. Caporaso J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nature methods 7, 335–336, doi: 10.1038/nmeth.f.303 (2010).
    1. McDonald D. et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. The ISME journal 6, 610–618, doi: 10.1038/ismej.2011.139 (2012).
    1. Caporaso J. G. et al. PyNAST: a flexible tool for aligning sequences to a template alignment. Bioinformatics 26, 266–267, doi: 10.1093/bioinformatics/btp636 (2010).
    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. Applied and environmental microbiology 73, 5261–5267, doi: 10.1128/AEM.00062-07 (2007).
    1. Price M. N., Dehal P. S. & Arkin A. P. FastTree 2–approximately maximum-likelihood trees for large alignments. PloS one 5, e9490, doi: 10.1371/journal.pone.0009490 (2010).
    1. Lozupone C. & Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Applied and environmental microbiology 71, 8228–8235, doi: 10.1128/AEM.71.12.8228-8235.2005 (2005).
    1. Wagner S. L. et al. Soluble gamma-secretase modulators selectively inhibit the production of Abeta42 and augment the production of multiple carboxy-truncated Abeta species. Biochemistry, doi: 10.1021/bi401537v (2014).
    1. Choi S. H. et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature, doi: 10.1038/nature13800 (2014).
    1. Zhang X. et al. Near-infrared fluorescence molecular imaging of amyloid beta species and monitoring therapy in animal models of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America 112, 9734–9739, doi: 10.1073/pnas.1505420112 (2015).

Source: PubMed

3
Tilaa