Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Microbiota-dependent increase in δ-valerobetaine alters neuronal function and is responsible for age-related cognitive decline

Abstract

Understanding the physiological origins of age-related cognitive decline is of critical importance given the rising age of the world’s population1. Previous work in animal models has established a strong link between cognitive performance and the microbiota2,3,4,5, and it is known that the microbiome undergoes profound remodeling in older adults6. Despite growing evidence for the association between age-related cognitive decline and changes in the gut microbiome, the mechanisms underlying such interactions between the brain and the gut are poorly understood. Here, using fecal microbiota transplantation (FMT), we demonstrate that age-related remodeling of the gut microbiota leads to decline in cognitive function in mice and that this impairment can be rescued by transplantation of microbiota from young animals. Moreover, using a metabolomic approach, we found elevated concentrations of δ-valerobetaine, a gut microbiota-derived metabolite, in the blood and brain of aged mice and older adults. We then demonstrated that δ-valerobetaine is deleterious to learning and memory processes in mice. At the neuronal level, we showed that δ-valerobetaine modulates inhibitory synaptic transmission and neuronal network activity. Finally, we identified specific bacterial taxa that significantly correlate with δ-valerobetaine levels in the brain. Based on our findings, we propose that δ-valerobetaine contributes to microbiota-driven brain aging and that the associated mechanisms represent a promising target for countering age-related cognitive decline.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Microbiota transplant from young mice reinstates microbiota composition and rescues memory deficits in aged mice.
Fig. 2: δ-valerobetaine, an age-associated microbiota-derived metabolite, is modulated in serum and brain by microbiota transplantations.
Fig. 3: Specific bacterial taxa correlate with δ-valerobetaine levels in the brain.
Fig. 4: Injection of δ-valerobetaine recapitulates age-associated memory deficits and diminishes the effects of microbiota from young mice.

Similar content being viewed by others

Data availability

Microbiome profiling (16S rRNA-seq) raw data are accessible at the European Nucleotide Archive under accession number PRJEB46041. Data on TwinsUK twin participants are available to bona fide researchers under managed access due to governance and ethical constraints. Raw data should be requested via the website (http://twinsuk.ac.uk/resources-for-researchers/access-our-data/), and requests are reviewed by the TwinsUK Resource Executive Committee regularly. Other data that support the findings of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Code availability

R scripts used for microbiome profiling are available online at https://github.com/OmarMossad/Mossad_FMT_NatAging.

References

  1. Livingston, G. et al. Dementia prevention, intervention, and care. Lancet 390, 2673–2734 (2017).

    Article  PubMed  Google Scholar 

  2. Desbonnet, L., Clarke, G., Shanahan, F., Dinan, T. G. & Cryan, J. F. Microbiota is essential for social development in the mouse. Mol. Psychiatry 19, 146–148 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Hoban, A. E. et al. The microbiome regulates amygdala-dependent fear recall. Mol. Psychiatry 23, 1134–1144 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Savignac, H. M., Tramullas, M., Kiely, B., Dinan, T. G. & Cryan, J. F. Bifidobacteria modulate cognitive processes in an anxious mouse strain. Behav. Brain Res. 287, 59–72 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Magnusson, K. R. et al. Relationships between diet-related changes in the gut microbiome and cognitive flexibility. Neuroscience 300, 128–140 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Claesson, M. J. et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc. Natl Acad. Sci. USA 108, 4586–4591 (2011).

    Article  CAS  PubMed  Google Scholar 

  7. Koistinen, V. M. et al. Contribution of gut microbiota to metabolism of dietary glycine betaine in mice and in vitro colonic fermentation. Microbiome 7, 103 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Koeth, R. A. et al. γ-butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of l-carnitine to TMAO. Cell Metab. 20, 799–812 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Shin, S. Y. et al. An atlas of genetic influences on human blood metabolites. Nat. Genet. 46, 543–550 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Long, T. et al. Whole-genome sequencing identifies common-to-rare variants associated with human blood metabolites. Nat. Genet. 49, 568–578 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Vaadia, E. et al. Dynamics of neuronal interactions in monkey cortex in relation to behavioural events. Nature 373, 515–518 (1995).

    Article  CAS  PubMed  Google Scholar 

  12. Courtin, J. et al. Prefrontal parvalbumin interneurons shape neuronal activity to drive fear expression. Nature 505, 92–96 (2014).

    Article  PubMed  Google Scholar 

  13. Prezioso, M. et al. Spike-timing-dependent plasticity learning of coincidence detection with passively integrated memristive circuits. Nat. Commun. 9, 5311 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dejean, C. et al. Prefrontal neuronal assemblies temporally control fear behaviour. Nature 535, 420–424 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Yang, Y. & Mailman, R. B. Strategic neuronal encoding in medial prefrontal cortex of spatial working memory in the T-maze. Behav. Brain Res. 343, 50–60 (2018).

    Article  PubMed  Google Scholar 

  16. Morici, J. F., Bekinschtein, P. & Weisstaub, N. V. Medial prefrontal cortex role in recognition memory in rodents. Behav. Brain Res. 292, 241–251 (2015).

    Article  PubMed  Google Scholar 

  17. Kells, P. A., Gautam, S. H., Fakhraei, L., Li, J. & Shew, W. L. Strong neuron-to-body coupling implies weak neuron-to-neuron coupling in motor cortex. Nat. Commun. 10, 1575 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Okun, M. et al. Diverse coupling of neurons to populations in sensory cortex. Nature 521, 511–515 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sweeney, Y. & Clopath, C. Population coupling predicts the plasticity of stimulus responses in cortical circuits. eLife 9, e56053 (2020).

  20. Erny, D. et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 18, 965–977 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Blank, T., Goldmann, T. & Prinz, M. Microglia fuel the learning brain. Trends Immunol. 35, 139–140 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wu, Y., Dissing-Olesen, L., MacVicar, B. A. & Stevens, B. Microglia: dynamic mediators of synapse development and plasticity. Trends Immunol. 36, 605–613 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Boehme, M. et al. Microbiota from young mice counteracts selective age-associated behavioral deficits. Nat Aging 1, 666–676 (2021).

    Article  Google Scholar 

  25. Dagher, N. N. et al. Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3×Tg-AD mice. J. Neuroinflammation 12, 139 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Lawton, M. P. et al. Health, valuation of life, and the wish to live. Gerontologist 39, 406–416 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Gazzaley, A., Cooney, J. W., Rissman, J. & D’Esposito, M. Top–down suppression deficit underlies working memory impairment in normal aging. Nat. Neurosci. 8, 1298–1300 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Grady, C. L. et al. Age-related reductions in human recognition memory due to impaired encoding. Science 269, 218–221 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Jermakowicz, W. J. & Casagrande, V. A. Neural networks a century after Cajal. Brain Res. Rev. 55, 264–284 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Cohen, M. R. & Kohn, A. Measuring and interpreting neuronal correlations. Nat. Neurosci. 14, 811–819 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Averbeck, B. B., Latham, P. E. & Pouget, A. Neural correlations, population coding and computation. Nat. Rev. Neurosci. 7, 358–366 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Rolls, E. T., Treves, A. & Tovee, M. J. The representational capacity of the distributed encoding of information provided by populations of neurons in primate temporal visual cortex. Exp. Brain Res. 114, 149–162 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Shew, W. L., Yang, H., Yu, S., Roy, R. & Plenz, D. Information capacity and transmission are maximized in balanced cortical networks with neuronal avalanches. J. Neurosci. 31, 55–63 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Goossens, T., Vercammen, C., Wouters, J. & van Wieringen, A. Aging affects neural synchronization to speech-related acoustic modulations. Front. Aging Neurosci. 8, 133 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Biagi, E. et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE 5, e10667 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Langille, M. G. et al. Microbial shifts in the aging mouse gut. Microbiome 2, 50 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Rath, S., Rud, T., Pieper, D. H. & Vital, M. Potential TMA-producing bacteria are ubiquitously found in Mammalia. Front. Microbiol. 10, 2966 (2019).

    Article  PubMed  Google Scholar 

  38. Karkkainen, O. et al. Whole grain intake associated molecule 5-aminovaleric acid betaine decreases β-oxidation of fatty acids in mouse cardiomyocytes. Sci. Rep. 8, 13036 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Mezo, C. et al. Different effects of constitutive and induced microbiota modulation on microglia in a mouse model of Alzheimer’s disease. Acta Neuropathol. Commun. 8, 119 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Spowart-Manning, L. & van der Staay, F. J. The T-maze continuous alternation task for assessing the effects of putative cognition enhancers in the mouse. Behav. Brain Res. 151, 37–46 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Evans, A. M., DeHaven, C. D., Barrett, T., Mitchell, M. & Milgram, E. Integrated, nontargeted ultrahigh performance liquid chromatography/electrospray ionization tandem mass spectrometry platform for the identification and relative quantification of the small-molecule complement of biological systems. Anal. Chem. 81, 6656–6667 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Rath, S., Heidrich, B., Pieper, D. H. & Vital, M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 5, 54 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Takahashi, S., Tomita, J., Nishioka, K., Hisada, T. & Nishijima, M. Development of a prokaryotic universal primer for simultaneous analysis of bacteria and archaea using next-generation sequencing. PLoS ONE 9, e105592 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Schliep, K. P. phangorn: phylogenetic analysis in R. Bioinformatics 27, 592–593 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Foster, Z. S. L., Sharpton, T. J. & Grünwald, N. J. Metacoder: an R package for visualization and manipulation of community taxonomic diversity data. PLoS Comput. Biol. 13, e1005404 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  47. McMurdie, P. J. & Holmes, S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 8, e61217 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

T.B., O.M., D.E., T.L., E.N. and M.P. were supported by the DFG (SFB/TRR167 ‘NeuroMac’). M.P. was further supported by the Sobek Foundation, the Ernst-Jung Foundation, the DFG (SFB 992, SFB1160, Reinhart Koselleck grant, Gottfried Wilhelm Leibniz prize), the Alzheimer Forschung Initiative e.V. (AFI) and the Ministry of Science, Research and Arts, Baden-Wuerttemberg (Sonderlinie ‘Neuroinflammation’). This study was supported by the DFG under Germany’s Excellence Strategy (CIBSS, EXC-2189, project ID-390939984). We are grateful to M. Gomez De Agüero and S.C. Ganal-Vonarburg (Macpherson laboratory, Bern, Switzerland) for providing us with GF mice. TwinsUK is funded by the Wellcome Trust, the Medical Research Council, the European Union, the National Institute for Health Research-funded BioResource, the Clinical Research Facility and the Biomedical Research Centre based at Guy’s and St. Thomas’ NHS Foundation Trust in partnership with King’s College London. We are grateful to Plexxikon for providing the CSF1R inhibitor PLX5622. We thank A. Dumas for proofreading the article.

Author information

Authors and Affiliations

Authors

Contributions

O.M. conducted the majority of experiments and data analysis; E.N., T.L. and J.M.B. performed targeted metabolomics; S.W. and M.V. performed microbiome profiling; S.F. and J.F.S. performed electrophysiological recordings; D.S. and P.S. contributed to the FMT experimental paradigm; J.F.S., D.E., T.L., B.S., A.S., M.B. and M.P. designed experiments and interpreted results; T.B. initiated and conceived the project and designed experiments; O.M. and T.B. wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Thomas Blank.

Ethics declarations

Competing interests

T.B. is an author on a patent application, which is pending. All authors declare no other competing interests.

Additional information

Peer review information Nature Aging thanks Sven Pettersson, Pieter Dorrestein and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Effects of antibiotics and fecal microbiota transplantation.

(a) Gating on DAPI- Syto9+ live bacteria of fecal samples from SPF and ABX-treated mice. Representative plot for the flow cytometry gating. (b) Percentage of live Gram+ and Gram- bacteria and (c) Quantification of live bacteria per mg fecal sample. (d) NMDS plot of Beta diversity (Bray-Curtis distance matrix) of microbiota in fecal pellets. Samples collected from start and end of the experiments are shown as triangles and circles, respectively. (e) Abundance of cecal microbiota composition profiles at the phylum level (each color represents one bacterial phylum). (f, g) Alpha diversity in cecum measured by (f) Shannon and (g) Simpson indices. Box plots; centre = median, upper and lower “hinges” correspond to the first and third quartiles (the 25th and 75th percentiles), upper whisker extends from the hinge to the highest value that is within 1.5 * IQR of the hinge, and lower whisker extends from the hinge to the lowest value within 1.5 * IQR of the hinge, where IQR is the inter-quartile range, or distance between the first and third quartiles. Statistical analysis with (b, c) Mann–Whitney U test (two-tailed) (f, g) Wilcoxon test followed by Holm post-hoc test (two-tailed) (*p < 0.05, **p < 0.01, ***p < 0.001, ns = not significant). Exact p-values are reported in the source data.

Source data

Extended Data Fig. 2 Age-associated microbiota alterations on genus level.

(a,b) Differentially abundant taxa in (a) aged vs young animals, and (b) oFMTiAged vs yFMTiAged animals with Padj<0.05. Each dot represents one ASV. Genus classification is depicted on y-axis, and log2FoldChange on x-axis. Taxa with unclassified genus were filtered out. Color represents different phyla.

Extended Data Fig. 3 δ-Valerobetaine enhances inhibitory postsynaptic currents and population synchronization in the medial prefrontal cortex.

(a) Dose-dependent increase in δ-valerobetainein the brain represented by area under the curve (AUC) of the mass-spec peaks.Error bars represent SEM. (b) T-maze (c,d) Familiarization (c) and Testing sessions (d) with a 6 h gap for the Novel object recognition test (NOR) (n=5). Statistical analysis with one-way ANOVA compared to the control group (0 mg/kg) followed by Bonferroni post-hoctest (*p < 0.05, **p < 0.01, ***p < 0.001). (e) Examples of spontaneous excitatory postsynaptic currents (EPSCs, left) and spontaneous inhibitory postsynaptic currents (IPSCs, right) of prefrontal pyramidal cells under control conditions (black) and after pre-incubation with 10 µM δ-valerobetaine (green). Data arefrom patch-clamp recordings in acute brain slices. (f) δ-valerobetaine increased the frequency (p=0.006, unpairedt-test) but not the amplitude of spontaneous IPSCs (p=0.174, unpaired t-test) while EPSC properties were unaltered (p=0.089 and p=0.641 for frequency andamplitude, respectively). n=10 control and 9 δ-valerobetaine-treated neurons. (a-d,f) Error bars represent SEM (g) Single unit recordings from them PFC to assess the effect of δ-valerobetaine on population activity in vivo. Example traces show a shortsegment of the summed population firing rate from all units of one recordingsession (black, binned in 100 ms time epochs, see Methods) and the binned spikerate of one neuron (bottom) before (left) and after (right) δ-valerobetaineinjection. Note that the unit showed increased coupling to the populationactivity upon δ-valerobetaine injection, indicating enhanced synchronization inthe prefrontal network. (h) Quantification of spike rates and population coupling of all neurons revealedno effect of δ-valerobetaine on average spike frequency but a significant increase in population coupling of the recorded cells (control: n=77 units from 4 mice, δ-valerobetaine: n=56 units from 4, Welch’s tests). Black bars indicatemeans. Statistical analysis (f,h) with Mann–Whitney U test (two-tailed)(**p < 0.01, ns = not significant).

Source data

Extended Data Fig. 4 Beneficial cognitive effect of young FMT is independent of microglia.

(a) Schematic diagram of the experimental setting: as in (Fig.1a) with the addition of the mice receiving either control AIN-76A standard chow (NC) or supplemented with PLX5622 (PLX, 1200 mg/kg) to deplete CSF1R-dependent cells, including microglia. (b) Representative immunofluorescence images of Iba-1+ (red), DAPI (blue) in cortex and hippocampus of PLX5622-treated mice upon sacrifice after behavioral testing. Scale bar, 150μm. (c) Quantification of Iba1+ cell density in cortex and hippocampus as one ROI upon PLX5622 treatment. Error bars represent SEM. (d)T-maze (e,f) Familiarization (e) and Testing sessions (f) with a 6 h gap for Novel object recognition test (NOR) (young (NC), aged (NC), aged (PLX), yFMTiAged (NC), yFMTiAged (PLX), oFMTiAged; n=4, 7, 8, 7, 8, 7, respectively). (a-f) Data represent two independent experiments. Errorbars represent SEM. Statistical analysis with (c-f) one-way ANOVA followed by Tukey post-hoc test (*p < 0.05, **p < 0.01, ***p < 0.001, ns = not significant). Exact p-values are reported in the source data.

Source data

Extended Data Fig. 5 Graphical abstract.

Heterochronic microbiota transplantation ameliorates the age-related cognitive decline through modulating the levels of the microbiota-dependent metabolite, δ-valerobetaine.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1 and 2.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mossad, O., Nent, E., Woltemate, S. et al. Microbiota-dependent increase in δ-valerobetaine alters neuronal function and is responsible for age-related cognitive decline. Nat Aging 1, 1127–1136 (2021). https://doi.org/10.1038/s43587-021-00141-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00141-4

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research