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.

Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice

Subjects

Abstract

As human lifespan increases, a greater fraction of the population is suffering from age-related cognitive impairments, making it important to elucidate a means to combat the effects of aging1,2. Here we report that exposure of an aged animal to young blood can counteract and reverse pre-existing effects of brain aging at the molecular, structural, functional and cognitive level. Genome-wide microarray analysis of heterochronic parabionts—in which circulatory systems of young and aged animals are connected—identified synaptic plasticity–related transcriptional changes in the hippocampus of aged mice. Dendritic spine density of mature neurons increased and synaptic plasticity improved in the hippocampus of aged heterochronic parabionts. At the cognitive level, systemic administration of young blood plasma into aged mice improved age-related cognitive impairments in both contextual fear conditioning and spatial learning and memory. Structural and cognitive enhancements elicited by exposure to young blood are mediated, in part, by activation of the cyclic AMP response element binding protein (Creb) in the aged hippocampus. Our data indicate that exposure of aged mice to young blood late in life is capable of rejuvenating synaptic plasticity and improving cognitive function.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Heterochronic parabiosis enhances dendritic spine number and synaptic plasticity in the aged hippocampus and elicits a plasticity-related expression profile.
Figure 2: Administration of young blood plasma improves hippocampal-dependent learning and memory in aged mice.
Figure 3: Creb mediates the enhancements in dendritic spine number and hippocampal-dependent learning and memory elicited by young blood in aged mice.

References

  1. Hebert, L.E., Scherr, P.A., Bienias, J.L., Bennett, D.A. & Evans, D.A. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch. Neurol. 60, 1119–1122 (2003).

    Article  PubMed  Google Scholar 

  2. Bishop, N.A., Lu, T. & Yankner, B.A. Neural mechanisms of ageing and cognitive decline. Nature 464, 529–535 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hedden, T. & Gabrieli, J.D. Insights into the ageing mind: a view from cognitive neuroscience. Nat. Rev. Neurosci. 5, 87–96 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Raz, N., Gunning-Dixon, F.M., Head, D., Dupuis, J.H. & Acker, J.D. Neuroanatomical correlates of cognitive aging: evidence from structural magnetic resonance imaging. Neuropsychology 12, 95–114 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Mattson, M.P. & Magnus, T. Ageing and neuronal vulnerability. Nat. Rev. Neurosci. 7, 278–294 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rapp, P.R. & Heindel, W.C. Memory systems in normal and pathological aging. Curr. Opin. Neurol. 7, 294–298 (1994).

    Article  CAS  PubMed  Google Scholar 

  7. Andrews-Hanna, J.R. et al. Disruption of large-scale brain systems in advanced aging. Neuron 56, 924–935 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Scheff, S.W., Price, D.A., Schmitt, F.A., DeKosky, S.T. & Mufson, E.J. Synaptic alterations in CA1 in mild Alzheimer disease and mild cognitive impairment. Neurology 68, 1501–1508 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Nicholson, D.A., Yoshida, R., Berry, R.W., Gallagher, M. & Geinisman, Y. Reduction in size of perforated postsynaptic densities in hippocampal axospinous synapses and age-related spatial learning impairments. J. Neurosci. 24, 7648–7653 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Smith, T.D., Adams, M.M., Gallagher, M., Morrison, J.H. & Rapp, P.R. Circuit-specific alterations in hippocampal synaptophysin immunoreactivity predict spatial learning impairment in aged rats. J. Neurosci. 20, 6587–6593 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Morrison, J.H. & Baxter, M.G. The ageing cortical synapse: hallmarks and implications for cognitive decline. Nat. Rev. Neurosci. 13, 240–250 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Villeda, S.A. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90–94 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pavlopoulos, E. et al. Molecular mechanism for age-related memory loss: the histone-binding protein RbAp48. Sci. Transl. Med. 5, 200ra115 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Conboy, I.M. et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433, 760–764 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Brack, A.S. et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 317, 807–810 (2007).

    CAS  PubMed  Google Scholar 

  16. Ruckh, J.M. et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 10, 96–103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Loffredo, F.S. et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell 153, 828–839 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Geinisman, Y., de Toledo-Morrell, L. & Morrell, F. Loss of perforated synapses in the dentate gyrus: morphological substrate of memory deficit in aged rats. Proc. Natl. Acad. Sci. USA 83, 3027–3031 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rosenzweig, E.S. & Barnes, C.A. Impact of aging on hippocampal function: plasticity, network dynamics, and cognition. Prog. Neurobiol. 69, 143–179 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Small, S.A., Schobel, S.A., Buxton, R.B., Witter, M.P. & Barnes, C.A. A pathophysiological framework of hippocampal dysfunction in ageing and disease. Nat. Rev. Neurosci. 12, 585–601 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Alberini, C.M. Transcription factors in long-term memory and synaptic plasticity. Physiol. Rev. 89, 121–145 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Bliss, T.V. & Collingridge, G.L. A synaptic model of memory: long-term potentiation in the hippocampus. Nature 361, 31–39 (1993).

    Article  CAS  PubMed  Google Scholar 

  23. Frey, U. & Morris, R.G. Synaptic tagging and long-term potentiation. Nature 385, 533–536 (1997).

    Article  CAS  PubMed  Google Scholar 

  24. Jeong, H. et al. Sirt1 mediates neuroprotection from mutant huntingtin by activation of the TORC1 and CREB transcriptional pathway. Nat. Med. 18, 159–165 (2012).

    Article  CAS  Google Scholar 

  25. Merrill, D.A., Karim, R., Darraq, M., Chiba, A.A. & Tuszynski, M.H. Hippocampal cell genesis does not correlate with spatial learning ability in aged rats. J. Comp. Neurol. 459, 201–207 (2003).

    Article  PubMed  Google Scholar 

  26. Bizon, J.L. & Gallagher, M. Production of new cells in the rat dentate gyrus over the lifespan: relation to cognitive decline. Eur. J. Neurosci. 18, 215–219 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Drapeau, E. et al. Spatial memory performances of aged rats in the water maze predict levels of hippocampal neurogenesis. Proc. Natl. Acad. Sci. USA 100, 14385–14390 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Smolinsky, A.N. et al. Analysis of grooming behavior and its utility in studying animal stress, anxiety, and depression. in Mouse Models of Mood and Anxiety Disorders (ed. Gould, T.) 21–36 (Humana Press, NY, 2009).

    Chapter  Google Scholar 

  29. Gould, T.D. Mood and Anxiety Related Phenotypes in Mice: Characterization Using Behavioral Tests (Humana Press, New York, 2009).

  30. Luo, J. et al. Glia-dependent TGF-β signaling, acting independently of the TH17 pathway, is critical for initiation of murine autoimmune encephalomyelitis. J. Clin. Invest. 117, 3306–3315 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xie, X. & Smart, T.G. Modulation of long-term potentiation in rat hippocampal pyramidal neurons by zinc. Pflugers Arch. 427, 481–486 (1994).

    Article  CAS  PubMed  Google Scholar 

  32. Grimm, D. et al. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 82, 5887–5911 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xu, W. et al. Distinct neuronal coding schemes in memory revealed by selective erasure of fast synchronous synaptic transmission. Neuron 73, 990–1001 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Raber, J. et al. Irradiation enhances hippocampus-dependent cognition in mice deficient in extracellular superoxide dismutase. Hippocampus 21, 72–80 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Alamed, J., Wilcock, D.M., Diamond, D.M., Gordon, M.N. & Morgan, D. Two-day radial-arm water maze learning and memory task; robust resolution of amyloid-related memory deficits in transgenic mice. Nat. Protoc. 1, 1671–1679 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Eggel, K. Lucin and N. Woodling for critical review and advice, and D. Jing and F. Lee (Cornell University) for Golgi stain reagents. This work was funded by California Institute for Regenerative Medicine (CIRM) fellowships (K.E.P. and K.L.), a Netherlands Organization for Scientific Research (NWO) Rubicon fellowship (J.M.), a Child Health Research Institute fellowship (Stanford National Institutes of Health (NIH)/National Center for Research Resources CTSA-UL1-RR025744, J.M.C.), a Jane Coffin Childs fellowship (J.M.C.), National Science Foundation fellowships (K.I.M. and J.U.), a National Research Service Award fellowship (1F31-AG034045-01, S.A.V.), anonymous (T.W.-C.), Veterans Affairs (T.W.-C.), the National Institute on Aging (AG045034, AG03144, T.W.-C.), CIRM (T.W.-C.), the University of California San Francisco (UCSF) Program for Breakthrough Biomedical Research, the Sandler Foundation (S.A.V.), the UCSF Clinical and Translational Science Institute (UL1-TR000004, S.A.V.) and an NIH Director's Independence Award (DP5-OD12178, S.A.V.).

Author information

Authors and Affiliations

Authors

Contributions

S.A.V., K.E.P., J.M., J.M.C., K.I.M., J.L., L.K.S. and K.L. performed parabiosis. S.A.V., K.I.M., G.B. and D.B. performed and/or analyzed microarray. S.A.V., K.E.P., R.W. and E.G.W. performed histological studies. J.M. and D.A.S. performed Golgi studies. B.Z. and X.S.X. performed electrophysiological studies. S.A.V., K.E.P., J.M.C., J.L., L.K.S., G.B., K.L. and J.U. performed plasma cognitive studies. J.M.C. performed maintenance and stress studies. J.M.C. and S.A.V. performed the denaturation study. K.E.P. and G.B. generated viral constructs. K.E.P. performed viral studies. F.M.L. provided reagents. S.A.V. and T.W.-C. designed and supervised the study and wrote the manuscript.

Corresponding authors

Correspondence to Saul A Villeda or Tony Wyss-Coray.

Ethics declarations

Competing interests

T.W.-C. has formed a company that follows up on the work described here.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 and Supplementary Table 1 (PDF 3938 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Villeda, S., Plambeck, K., Middeldorp, J. et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat Med 20, 659–663 (2014). https://doi.org/10.1038/nm.3569

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3569

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing