Elsevier

Neurobiology of Aging

Volume 31, Issue 11, November 2010, Pages 1937-1949
Neurobiology of Aging

Hippocampal gene expression patterns underlying the enhancement of memory by running in aged mice

https://doi.org/10.1016/j.neurobiolaging.2008.10.016Get rights and content

Abstract

Physical activity preserves cognition in the aging brain, but the mechanisms remain obscure. In order to identify candidate genes and pathways responsible for the preservation of cognitive function by exercise, we trained mice that had been exposed to lifelong running or sedentary lifestyle for 16 months in the hippocampus-dependent water maze. After water maze training, we analyzed the expression of 24,000 genes in the hippocampus using Illumina bead microarray. Runners show greater activation of genes associated with synaptic plasticity and mitochondrial function, and also exhibit significant downregulation of genes associated with oxidative stress and lipid metabolism. Running also modified the effects of learning on the expression of genes involved in cell excitability, energy metabolism, and insulin, MAP kinase and Wnt signaling. These results suggest that the enhancement of cognitive function by lifelong exercise is associated with an altered transcriptional profile following learning.

Introduction

The aging brain retains its responsiveness to the environment. Studies have shown that environmental enrichment can influence a number of factors relevant to brain aging, including synaptic plasticity and neuronal morphology (van Praag et al., 2000). Enriched environments typically afford increased opportunities for social and cognitive stimulation, as well as physical activity. Among these components of the enrichment paradigm, physical activity in the form of wheel running has emerged as a potent modulator of central neuroplasticity. Wheel running preserves learning and memory, and enhances adult neurogenesis in the hippocampus of aging mice (van Praag et al., 2005). These effects of voluntary wheel running on learning and memory are associated with changes in genes expression. For example, learning (Cavallaro et al., 2002) and environmental complexity (Rampon et al., 2000) alter gene transcription in young animals. Pathways influenced by these experiences include cell-survival associated genes and genes involved in synaptic plasticity. The gene expression profile to learning has also been shown to differ between young and aged animals (Rowe et al., 2007).

The hippocampus plays a critical role in learning and memory, and is susceptible to dysfunction and degeneration in aging and Alzheimer's disease (Mattson and Magnus, 2006). Several studies have now shown that running in a wheel influences gene transcription in the hippocampus of young adult mice and rats (Molteni et al., 2002, Tong et al., 2001, Hunsberger et al., 2007). The transcripts upregulated by running overlap considerably with those influenced by enrichment and learning. In addition, the pathways influenced by running oppose the effects of aging at multiple levels. However, it remains unknown whether exercise influences the transcriptome in aging animals. Moreover, it is uncertain what changes in gene transcription might underlie the established ability of exercise to preserve learning and memory in aging animals (van Praag et al., 2005).

In these studies, we characterized the effects of lifelong running on learning-induced transcriptional modifications in aged animals. Under basal, unstimulated conditions, the differences between runners and controls were restricted to a small pool of genes. In contrast, when we compare the regulation of gene expression by learning across runners and sedentary animals, a large number of gene transcripts are differentially regulated. Running enhanced hippocampus-dependent learning in aged mice, and the enhancement of spatial memory was accompanied by differential expression of genes important for learning and synaptic plasticity, as well as genes involved in energy metabolism, and insulin and Wnt signaling. These studies suggest that aged runners differentially express certain gene transcripts in the hippocampus, and may recruit an entirely different transcriptional profile following learning.

Section snippets

Animals and activity monitoring

Animal care and experimental procedures followed NIH guidelines and were approved by the National Institute on Aging Animal Care and Use Committee. Male C57BL/6 mice were housed individually with (n = 24) or without (n = 18) running wheels (Campden Instruments), beginning at 2–4 months of age. The number of wheel turns was collected automatically with custom software, and the room was maintained on a 12 h light/dark schedule with food and water available ad libitum.

Experimental design

After 16 months in their

Running enhances hippocampus-dependent learning in aged mice

All mice housed with running wheels ran extensively (mean distance = 1.54 ± 0.32 km/day) and there was a significant decrease in running activity with age (Pearson's correlation, time × distance; r2 = 0.76, p = 0.0005). In concurrence with previous reports (van Praag et al., 2005) running was associated with reduced escape latency in the maze (F1,48 = 6.81, p = 0.02; Fig. 1A). Runners also took a more direct path to the platform (F1,48 = 5.76, p = 0.003; Fig. 1B). In the probe trial, animals that had engaged in

Discussion

Running enhances hippocampus-dependent learning in aged mice. Improvements in learning were not attributable to changes in a small number of genes, but instead involved changes in the number and diversity of the molecular pathways influenced by water maze training. Runners recruited a more varied set of transcriptional categories following learning. These categories included transcripts involved in dendritic and synaptic plasticity, as well as genes implicated in learning and memory. Mice

Acknowledgements

This research was supported by the Intramural Research Program of the National Institute on Aging. We would like to thank Xiangru Xu for valuable discussions. The authors have no actual or potential conflicts of interest.

References (31)

  • M.A. Aberg et al.

    Peripheral infusion of IGF-I selectively induces neurogenesis in the adult rat hippocampus

    J. Neurosci.

    (2000)
  • S. Cavallaro et al.

    Memory-specific temporal profiles of gene expression in the hippocampus

    Proc. Natl. Acad. Sci. U.S.A.

    (2002)
  • C.H. Hillman et al.

    Be smart, exercise your heart: exercise effects on brain and cognition

    Nat. Rev. Neurosci.

    (2008)
  • S. Hoyer et al.

    Long-term abnormalities in brain glucose/energy metabolism after inhibition of the neuronal insulin receptor: implication of tau-protein

    J. Neural Transm. Suppl.

    (2007)
  • J.G. Hunsberger et al.

    Antidepressant actions of the exercise-regulated gene VGF

    Nat. Med.

    (2007)
  • Cited by (120)

    • Metabolic regulation of aging and age-related disease

      2020, Ageing Research Reviews
      Citation Excerpt :

      These studies included mice sacrificed under basal conditions, after swimming in the water maze, or after hidden platform training (Stranahan et al., 2010). Side-by-side comparison of the two studies revealed limited overlap between the genes and pathways differentially activated at baseline in the hippocampus of aged runners, relative to the transcriptional effects of caloric restriction in the hippocampus of aged mice from the AGEMAP study (Xu et al., 2007; Stranahan et al., 2010; Fig. 2). The lack of overlap between the two datasets could reflect distinct metabolic programs associated with energy expenditure in runners, relative to cellular adaptations in response to reduced energy intake in mice on CR.

    View all citing articles on Scopus
    View full text