Adult hippocampal neurogenesis buffers stress responses and depressive behaviour

Jason S Snyder, Amélie Soumier, Michelle Brewer, James Pickel, Heather A Cameron, Jason S Snyder, Amélie Soumier, Michelle Brewer, James Pickel, Heather A Cameron

Abstract

Glucocorticoids are released in response to stressful experiences and serve many beneficial homeostatic functions. However, dysregulation of glucocorticoids is associated with cognitive impairments and depressive illness. In the hippocampus, a brain region densely populated with receptors for stress hormones, stress and glucocorticoids strongly inhibit adult neurogenesis. Decreased neurogenesis has been implicated in the pathogenesis of anxiety and depression, but direct evidence for this role is lacking. Here we show that adult-born hippocampal neurons are required for normal expression of the endocrine and behavioural components of the stress response. Using either transgenic or radiation methods to inhibit adult neurogenesis specifically, we find that glucocorticoid levels are slower to recover after moderate stress and are less suppressed by dexamethasone in neurogenesis-deficient mice than intact mice, consistent with a role for the hippocampus in regulation of the hypothalamic-pituitary-adrenal (HPA) axis. Relative to controls, neurogenesis-deficient mice also showed increased food avoidance in a novel environment after acute stress, increased behavioural despair in the forced swim test, and decreased sucrose preference, a measure of anhedonia. These findings identify a small subset of neurons within the dentate gyrus that are critical for hippocampal negative control of the HPA axis and support a direct role for adult neurogenesis in depressive illness.

Figures

Figure 1
Figure 1
GFAP-TK mice show specific inhibition of adult neurogenesis. a) Confocal image of endogenous GFAP and transgenic TK expression in a radial precursor cell in the dentate gyrus (arrow). b) Confocal photographs of valganciclovir-treated mice show GFAP+ astrocytes in the hilus and molecular layer in both genotypes, despite strong TK expression in all GFAP-expressing cells in TK mice. c) Valganciclovir treatment did not affect numbers of GFAP+ astrocytes (genotype effect F1,20=1.7, P=0.2; valganciclovir effect F1,20=1.0, P=0.3; interaction F1,20=1.5, P=0.2; n=6/group), confirming the expectation that valganciclovir does not kill astrocytes, which are postmitotic in the adult. d) Confocal photographs of dentate gyrus doublecortin (DCX) immunostaining in mice treated with valganciclovir (v-WT and v-TK) for 4 weeks. DCX+ young neurons are abundant in v-WT mice but absent in v-TK mice. e) The number of BrdU+/DCX+ young neurons in was unaltered in v-WT mice but reduced by 99% in v-TK mice (genotype effect F1,20=20, P=0.0002; valganciclovir effect F1,20=19, P=0.0003; interaction F1,20=40, P<0.0001; *P<0.001 vs. untreated TK and v-WT; n=6/group). Inset shows example of 1-day-old BrdU+/DCX+ neuron (arrow). Error bars show s.e.m.; scale bars 10 μm in a, e and 100 μm in b, d. MOL, molecular layer; GCL, granule cell layer; HIL, hilus.
Figure 2
Figure 2
The glucocorticoid response to stress is increased in neurogenesis-deficient mice. a) Restraint, a moderate psychogenic stressor, resulted in higher corticosterone in neurogenesis-deficient v-TK mice than in v-WT mice 30 min after the end of stress. b) The effect of restraint was still observed after repeated exposure to stress (for both days: genotype effect F1,65>5, P<0.05; time effect F2,65>24, P<0.001; *P<0.05 post hoc vs. v-WT; n=6-17/group/timepoint). c) In untreated control mice, corticosterone levels 30 min after restraint stress were not different between WT and TK, indicating that altered glucocorticoid response to stress is not a non-specific effect of transgene expression (t30=0.1, P=0.95; n=16/group). d) Valganciclovir-treated v-TK mice show impaired dexamethasone suppression of corticosterone in response to restraint (*t13=2.5, P=0.03; n=7-8/group). Error bars show s.e.m.
Figure 3
Figure 3
Increased stress response is not due to reduced neurogenesis in the subventricular zone. a) Increased corticosterone response 30 min after restraint was confirmed in mice in which neurogenesis was reduced by irradiation (irradiation effect F1,50=2.0, P=0.16; time effect F2,50=5.1, P=0.01; irradiation × time interaction F2,50=5.7, P=0.006; *P<0.01 post hoc; n=4-6/group/time point). Green squares at the 30 min time point indicate corticosterone levels in irradiated mice that showed unaffected neurogenesis in the subventricular zone (SVZ). b) Confocal images of BrdU+ and DCX+ cells in the dentate gyrus; neurogenesis was reduced in all irradiated mice. c) Confocal images of BrdU+ and DCX+ cells illustrating sparing of neurogenesis in the SVZ. Scale bars 100 μm. LV, lateral ventricle; MOL, molecular layer; GCL, granule cell layer; HIL, hilus.
Figure 4
Figure 4
Mice lacking neurogenesis show increased anxiety/depression-like behaviors. a) In the novelty-suppressed feeding (NSF) test, v-TK mice showed increased latency to feed in a novel environment following restraint stress but not under control conditions (genotype effect F1,75=5.9, P=0.02; stress effect F1,75=1.9, P=0.17; genotype × stress interaction F1,75=4.8, P=0.03; *P<0.05 vs. v-TK control and *P<0.01 vs. v-WT stressed; n=13-25/group). b) Cumulative distribution of feeding latencies for the NSF test (log-rank test; *P<0.05 vs. all other groups). c) Neurogenesis-deficient v-TK mice became immobile faster in the forced swim test. Restraint stress reduced the latency to immobility in v-WT mice but did not affect v-TK mice (genotype effect F1,88=1.1, P=0.3; stress effect F1,88=2.2, P=0.14; genotype × stress interaction F1,88=2.6, P=0.11; *T46=2.1, P<0.05 vs. v-WT stressed; v-TK control vs. v-TK stressed T42=0.1, P=0.9; n=22-26/group). d) Under control conditions, the total time spent immobile was greater in v-TK mice than in v-WT mice. Restraint stress significantly increased total immobility in v-WT mice but had no effect on v-TK mice (genotype effect F1,88=0.3, P=0.6; stress effect F1,88=4.2, P=0.04; genotype × stress interaction F1,88=9.1, P=0.003; *P<0.05 vs. control v-TK, *P<0.001 vs. stressed v-WT, stressed v-WT vs. stressed v-TK P>0.05; n=22-26/group). e) Neurogenesis-deficient v-TK mice showed reduced preference for sucrose in an acute test, compared to v-WT mice, under both control and restraint conditions (genotype effect F1,20=11.2, P<0.01; stress effect F1,20=3.1, P=0.09; genotype × stress interaction F1,20=0.2, P=0.7; n=4-8/group). f) Sucrose preference remained lower in v-TK mice compared to v-WT mice during the subsequent dark cycle (genotype effect F1,25=6.8, P=0.01; stress effect F1,25=0.8, P=0.4; genotype × stress interaction F1,25=0.5, P=0.5; n=4-10/group). Error bars show s.e.m.

References

    1. Holsboer F, Ising M. Stress hormone regulation: biological role and translation into therapy. Annual review of psychology. 2010;61:81–109. C101–111.
    1. McEwen BS. Physiology and neurobiology of stress and adaptation: central role of the brain. Physiol Rev. 2007;87:873–904.
    1. Mirescu C, Gould E. Stress and adult neurogenesis. Hippocampus. 2006;16:233–238.
    1. Sapolsky RM. Is impaired neurogenesis relevant to the affective symptoms of depression? Biol Psychiatry. 2004;56:137–139.
    1. Drew MR, Hen R. Adult hippocampal neurogenesis as target for the treatment of depression. CNS & neurological disorders drug targets. 2007;6:205–218.
    1. Roozendaal B, et al. Memory retrieval impairment induced by hippocampal CA3 lesions is blocked by adrenocortical suppression. Nat Neurosci. 2001;4:1169–1171.
    1. Jankord R, Herman JP. Limbic regulation of hypothalamo-pituitary-adrenocortical function during acute and chronic stress. Ann N Y Acad Sci. 2008;1148:64–73.
    1. Garcia AD, Doan NB, Imura T, Bush TG, Sofroniew MV. GFAP-expressing progenitors are the principal source of constitutive neurogenesis in adult mouse forebrain. Nat Neurosci. 2004;7:1233–1241.
    1. Seri B, Garcia-Verdugo JM, McEwen BS, Alvarez-Buylla A. Astrocytes give rise to new neurons in the adult mammalian hippocampus. J Neurosci. 2001;21:7153–7160.
    1. Borrelli E, Heyman RA, Arias C, Sawchenko PE, Evans RM. Transgenic mice with inducible dwarfism. Nature. 1989;339:538–541.
    1. Burns KA, Murphy B, Danzer SC, Kuan C-Y. Developmental and post-injury cortical gliogenesis: a genetic fate-mapping study with Nestin-CreER mice. Glia. 2009;57:1115–1129.
    1. Santarelli L, et al. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science. 2003;301:805–809.
    1. Furay AR, Bruestle AE, Herman JP. The Role of the Forebrain Glucocorticoid Receptor in Acute and Chronic Stress. Endocrinology. 2008
    1. Wojtowicz JM. Irradiation as an experimental tool in studies of adult neurogenesis. Hippocampus. 2006;16:261–266.
    1. Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A. Subventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell. 1999;97:703–716.
    1. Airan RD, et al. High-speed imaging reveals neurophysiological links to behavior in an animal model of depression. Science. 2007;317:819–823.
    1. Surget A, et al. Antidepressants recruit new neurons to improve stress response regulation. Mol Psychiatry. 2011
    1. David DJ, et al. Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression. Neuron. 2009;62:479–493.
    1. Anacker C, et al. Antidepressants increase human hippocampal neurogenesis by activating the glucocorticoid receptor. Molecular Psychiatry. 2011
    1. Ridder S, et al. Mice with genetically altered glucocorticoid receptor expression show altered sensitivity for stress-induced depressive reactions. J Neurosci. 2005;25:6243–6250.
    1. Castagne V, Moser P, Porsolt RD. Behavioral Assessment of Antidepressant Activity in Rodents. 2009.
    1. Revest J-M, et al. Adult hippocampal neurogenesis is involved in anxiety-related behaviors. Mol Psychiatry. 2009;14:959–967.
    1. Treadway MT, Zald DH. Reconsidering anhedonia in depression: lessons from translational neuroscience. Neurosci Biobehav Rev. 2011;35:537–555.
    1. Barkus C, et al. Hippocampal NMDA receptors and anxiety: at the interface between cognition and emotion. Eur J Pharmacol. 2010;626:49–56.
    1. Duman CH. Models of depression. Vitam Horm. 2010;82:1–21.
    1. Sandi C, Pinelo-Nava MT. Stress and memory: behavioral effects and neurobiological mechanisms. Neural plasticity. 2007;2007:78970.
    1. Oomen CA, et al. Early maternal deprivation affects dentate gyrus structure and emotional learning in adult female rats. Psychopharmacology (Berl) 2010
    1. Delaney CL, Brenner M, Messing A. Conditional ablation of cerebellar astrocytes in postnatal transgenic mice. J Neurosci. 1996;16:6908–6918.
    1. Snyder J, et al. Adult-Born Hippocampal Neurons Are More Numerous, Faster Maturing, and More Involved in Behavior in Rats than in Mice. J Neurosci. 2009;29:14484–14495.
    1. Brown TS, Murphy HM. Factors affecting sucrose preference behavior in rats with hippocampal lesions. Physiol Behav. 1973;11:833–844.

Source: PubMed

3
Suscribir