The ageing systemic milieu negatively regulates neurogenesis and cognitive function

Saul A Villeda, Jian Luo, Kira I Mosher, Bende Zou, Markus Britschgi, Gregor Bieri, Trisha M Stan, Nina Fainberg, Zhaoqing Ding, Alexander Eggel, Kurt M Lucin, Eva Czirr, Jeong-Soo Park, Sebastien Couillard-Després, Ludwig Aigner, Ge Li, Elaine R Peskind, Jeffrey A Kaye, Joseph F Quinn, Douglas R Galasko, Xinmin S Xie, Thomas A Rando, Tony Wyss-Coray, Saul A Villeda, Jian Luo, Kira I Mosher, Bende Zou, Markus Britschgi, Gregor Bieri, Trisha M Stan, Nina Fainberg, Zhaoqing Ding, Alexander Eggel, Kurt M Lucin, Eva Czirr, Jeong-Soo Park, Sebastien Couillard-Després, Ludwig Aigner, Ge Li, Elaine R Peskind, Jeffrey A Kaye, Joseph F Quinn, Douglas R Galasko, Xinmin S Xie, Thomas A Rando, Tony Wyss-Coray

Abstract

In the central nervous system, ageing results in a precipitous decline in adult neural stem/progenitor cells and neurogenesis, with concomitant impairments in cognitive functions. Interestingly, such impairments can be ameliorated through systemic perturbations such as exercise. Here, using heterochronic parabiosis we show that blood-borne factors present in the systemic milieu can inhibit or promote adult neurogenesis in an age-dependent fashion in mice. Accordingly, exposing a young mouse to an old systemic environment or to plasma from old mice decreased synaptic plasticity, and impaired contextual fear conditioning and spatial learning and memory. We identify chemokines--including CCL11 (also known as eotaxin)--the plasma levels of which correlate with reduced neurogenesis in heterochronic parabionts and aged mice, and the levels of which are increased in the plasma and cerebrospinal fluid of healthy ageing humans. Lastly, increasing peripheral CCL11 chemokine levels in vivo in young mice decreased adult neurogenesis and impaired learning and memory. Together our data indicate that the decline in neurogenesis and cognitive impairments observed during ageing can be in part attributed to changes in blood-borne factors.

Conflict of interest statement

Competing Interests Statement: The authors declare that they have no competing financial interests.

Figures

Figure 1. Heterochronic parabiosis alters neurogenesis in…
Figure 1. Heterochronic parabiosis alters neurogenesis in an age-dependent fashion
a, Schematic showing parabiotic pairings. b,e, Representative fields of Doublecortin (b) and BrdU (e) immunostaining of young (3–4 months; yellow) and old (18–20 months; gray) isochronic and heterochronic parabionts five weeks after parabiosis (arrowheads point to individual cells, scale bar: 100μm). c–f Quantification of neurogenesis (c,d) and proliferating cells (e,f) in the young (c,e; top) and old (d,f; bottom) DG after parabiosis. Data from 12 young isochronic, 10 young heterochronic, 6 old isochronic and 12 old heterochronic parabionts. g,h, Population spike amplitude (PSA) was recorded from DG of young parabionts. Representative electrophysiological profiles (g) and LTP levels (h) are shown for young heterochronic and isochronic parabionts. Data from 4–5 mice per group. All data represented as Mean + SEM; *P<0.05; **P<0.01 t-test.
Figure 2. Factors from an old systemic…
Figure 2. Factors from an old systemic environment decrease neurogenesis and impair learning and memory
a, Schematic of young (3–4 months) or old (18–22 months) plasma extraction and intravenous injection into young (3 months) adult mice. b, Representative field of Doublecortin immunostaining of young adult mice after plasma injection treatment four times over ten days (scale bar: 100μm). c, Quantification of neurogenesis in the young DG after plasma injection. Data from 8 young plasma and 7 old plasma injected mice. d,e Hippocampal learning and memory assessed by contextual fear conditioning (d) and RAWM (e) paradigms in young adult mice after young or old plasma injections nine times over 24 days. d, Percent freezing time 24 hours after training. Data from 8 mice per group. e, Number of entry arm errors prior to finding platform. Data from 12 mice per group. All data represented as Mean ± SEM; *P< 0.05; **P< 0.01; t-test (c,d), repeated measures ANOVA, Bonferroni post-hoc test (e).
Figure 3. Systemic chemokine levels increase during…
Figure 3. Systemic chemokine levels increase during aging and heterochronic parabiosis and correlate with decreased neurogenesis
a, Venn diagram of results from aging and parabiosis proteomic screens. Seventeen age-related plasma factors correlated strongest with decreased neurogenesis in gray, fifteen plasma factors increased between young isochronic and young heterochronic parabionts in red, and six factors elevated in both screens in brown intersection. Data from 5–6 animals per age group. b,c Changes in plasma concentrations of CCL11 with age (b) and young heterochronic parabionts pre- and post- parabiotic pairing (c). d,e Changes in plasma (d) and CSF (e) concentrations of CCL11 with age in healthy human subjects. All data represented as dot plots with mean; *P< 0.05; **P< 0.01; ***P< 0.001 t-test (c,e), ANOVA, Tukey’s post-hoc test (a,b), and Mann-Whitney U Test (d).
Figure 4. Systemic exposure to CCL11 inhibits…
Figure 4. Systemic exposure to CCL11 inhibits neurogenesis and impairs learning and memory
a, Schematic of young (3–4 months) mice injected intraperitoneally with CCL11 or vehicle, and in combination with anti-CCL11 neutralizing or isotype control antibody. b, Representative field of Dcx-positive cells for each treatment group (n = 6–10 mice) treated four times over ten days. (scale bar: 100μm). c, Quantification of neurogenesis in the DG after treatment. d, Schematic of young adult mice given unilateral stereotaxic injections of anti-CCL11 neutralizing or isotype control antibody followed by systemic injections with either recombinant CCL11 or PBS (vehicle). e, Representative field of Dcx-positive cells in adjacent sides of the DG for each treatment group (n = 3–11 mice) f, Quantification of neurogenesis in the DG after systemic and stereotaxic treatment. g,h, Learning and memory assessed by contextual fear conditioning (g) and RAWM (h) paradigms in young adult mice injected with CCL11 or vehicle every three days for five weeks (n = 12–16 mice per group). All data represented as Mean ± SEM; *P< 0.05; **P<0.01; ANOVA, Dunnet’s or Tukey’s post-hoc test (c,f); repeated measures ANOVA, Bonferroni post-hoc test (k).

References

    1. van Praag H, Shubert T, Zhao C, Gage FH. Exercise enhances learning and hippocampal neurogenesis in aged mice. J Neurosci. 2005;25 (38):8680–8685.
    1. Gage FH. Mammalian neural stem cells. Science. 2000;287 (5457):1433–1438.
    1. Alvarez-Buylla A, Lim DA. For the long run: maintaining germinal niches in the adult brain. Neuron. 2004;41 (5):683–686.
    1. Zhao C, Deng W, Gage FH. Mechanisms and functional implications of adult neurogenesis. Cell. 2008;132 (4):645–660.
    1. van Praag H, et al. Functional neurogenesis in the adult hippocampus. Nature. 2002;415 (6875):1030–1034.
    1. Deng W, Aimone JB, Gage FH. New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat Rev Neurosci. 11(5):339–350.
    1. Clelland CD, et al. A functional role for adult hippocampal neurogenesis in spatial pattern separation. Science. 2009;325 (5937):210–213.
    1. Zhang CL, Zou Y, He W, Gage FH, Evans RM. A role for adult TLX-positive neural stem cells in learning and behaviour. Nature. 2008;451 (7181):1004–1007.
    1. Saxe MD, et al. Ablation of hippocampal neurogenesis impairs contextual fear conditioning and synaptic plasticity in the dentate gyrus. Proc Natl Acad Sci U S A. 2006;103 (46):17501–17506.
    1. Shen Q, et al. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science. 2004;304 (5675):1338–1340.
    1. Carpentier PA, Palmer TD. Immune influence on adult neural stem cell regulation and function. Neuron. 2009;64 (1):79–92.
    1. Renault VM, et al. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell. 2009;5 (5):527–539.
    1. Molofsky AV, et al. Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature. 2006;443 (7110):448–452.
    1. Lie DC, et al. Wnt signalling regulates adult hippocampal neurogenesis. Nature. 2005;437 (7063):1370–1375.
    1. Lucin KM, Wyss-Coray T. Immune activation in brain aging and neurodegeneration: too much or too little? Neuron. 2009;64 (1):110–122.
    1. Rapp PR, Heindel WC. Memory systems in normal and pathological aging. Curr Opin Neurol. 1994;7 (4):294–298.
    1. Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007;10 (12):1538–1543.
    1. Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the hippocampus. Nature. 1993;361 (6407):31–39.
    1. Conboy IM, et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005;433 (7027):760–764.
    1. Couillard-Despres S, et al. In vivo optical imaging of neurogenesis: watching new neurons in the intact brain. Mol Imaging. 2008;7 (1):28–34.
    1. Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003;302 (5651):1760–1765.
    1. Moriyama M, et al. Complement receptor 2 is expressed in neural progenitor cells and regulates adult hippocampal neurogenesis. J Neurosci. 31(11):3981–3989.
    1. Fumagalli M, d’Adda di Fagagna F. SASPense and DDRama in cancer and ageing. Nat Cell Biol. 2009;11 (8):921–923.
    1. Luo J, et al. Glia-dependent TGF-beta signaling, acting independently of the TH17 pathway, is critical for initiation of murine autoimmune encephalomyelitis. J Clin Invest. 2007;117 (11):3306–3315.
    1. Xie X, Smart TG. Modulation of long-term potentiation in rat hippocampal pyramidal neurons by zinc. Pflugers Arch. 1994;427 (5–6):481–486.
    1. Alamed J, Wilcock DM, Diamond DM, Gordon MN, Morgan D. Two-day radial-arm water maze learning and memory task; robust resolution of amyloid-related memory deficits in transgenic mice. Nat Protoc. 2006;1 (4):1671–1679.
    1. Raber J, et al. Irradiation enhances hippocampus-dependent cognition in mice deficient in extracellular superoxide dismutase. Hippocampus. 21(1):72–80.
    1. Zhang J, et al. CSF multianalyte profile distinguishes Alzheimer and Parkinson diseases. Am J Clin Pathol. 2008;129 (4):526–529.
    1. Li G, et al. Cerebrospinal fluid concentration of brain-derived neurotrophic factor and cognitive function in non-demented subjects. PLoS One. 2009;4 (5):e5424.
    1. Couillard-Despres S, et al. Human in vitro reporter model of neuronal development and early differentiation processes. BMC Neurosci. 2008;9:31.
    1. Buckwalter MS, et al. Chronically increased transforming growth factor-beta1 strongly inhibits hippocampal neurogenesis in aged mice. Am J Pathol. 2006;169 (1):154–164.

Source: PubMed

Подписаться