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.

Parvalbumin interneurons mediate neuronal circuitry–neurogenesis coupling in the adult hippocampus

Abstract

Using immunohistology, electron microscopy, electrophysiology and optogenetics, we found that proliferating adult mouse hippocampal neural precursors received immature GABAergic synaptic inputs from parvalbumin-expressing interneurons. Recently shown to suppress adult quiescent neural stem cell activation, parvalbumin interneuron activation promoted newborn neuronal progeny survival and development. Our results suggest a niche mechanism involving parvalbumin interneurons that couples local circuit activity to the diametric regulation of two critical early phases of adult hippocampal neurogenesis.

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: PV+ interneurons form immature synaptic inputs onto proliferating newborn progeny in the adult dentate gyrus.
Figure 2: Activation of PV+, but not SST+, interneurons in the dentate gyrus promotes survival of proliferative newborn progeny during early phases of adult hippocampal neurogenesis.
Figure 3: Suppression of PV+, but not SST+, interneuron activity in the adult dentate gyrus decreases survival of newborn progeny in the normal and enriched environment.

Similar content being viewed by others

References

  1. Sierra, A. et al. Cell Stem Cell 7, 483–495 (2010).

    Article  CAS  Google Scholar 

  2. Snyder, J.S. et al. J. Neurosci. 29, 14484–14495 (2009).

    Article  CAS  Google Scholar 

  3. Kronenberg, G. et al. J. Comp. Neurol. 467, 455–463 (2003).

    Article  Google Scholar 

  4. Mandyam, C.D., Harburg, G.C. & Eisch, A.J. Neuroscience 146, 108–122 (2007).

    Article  CAS  Google Scholar 

  5. Ge, S. et al. Nature 439, 589–593 (2006).

    Article  CAS  Google Scholar 

  6. Kaplan, M.S. & Bell, D.H. J. Neurosci. 4, 1429–1441 (1984).

    Article  CAS  Google Scholar 

  7. Tozuka, Y., Fukuda, S., Namba, T., Seki, T. & Hisatsune, T. Neuron 47, 803–815 (2005).

    Article  CAS  Google Scholar 

  8. Liu, X., Wang, Q., Haydar, T.F. & Bordey, A. Nat. Neurosci. 8, 1179–1187 (2005).

    Article  CAS  Google Scholar 

  9. Song, J. et al. Nature 489, 150–154 (2012).

    Article  CAS  Google Scholar 

  10. Markwardt, S.J., Dieni, C.V., Wadiche, J.I. & Overstreet-Wadiche, L. Nat. Neurosci. 14, 1407–1409 (2011).

    Article  CAS  Google Scholar 

  11. Gao, Z. et al. Nat. Neurosci. 12, 1090–1092 (2009).

    Article  CAS  Google Scholar 

  12. Kuwabara, T. et al. Nat. Neurosci. 12, 1097–1105 (2009).

    Article  CAS  Google Scholar 

  13. Ming, G.L. & Song, H. Neuron 70, 687–702 (2011).

    Article  CAS  Google Scholar 

  14. Mann, E.O. & Paulsen, O. Trends Neurosci. 30, 343–349 (2007).

    Article  CAS  Google Scholar 

  15. Deisseroth, K. et al. Neuron 42, 535–552 (2004).

    Article  CAS  Google Scholar 

  16. Chambers, R.A., Potenza, M.N., Hoffman, R.E. & Miranker, W. Neuropsychopharmacology 29, 747–758 (2004).

    Article  Google Scholar 

  17. Kim, W.R. et al. Eur. J. Neurosci. 29, 1408–1421 (2009).

    Article  Google Scholar 

  18. Sahay, A. et al. Nature 472, 466–470 (2011).

    Article  CAS  Google Scholar 

  19. Gu, Y. et al. Nat. Neurosci. 15, 1700–1706 (2013).

    Article  Google Scholar 

  20. Tashiro, A., Sandler, V.M., Toni, N., Zhao, C. & Gage, F.H. Nature 442, 929–933 (2006).

    Article  CAS  Google Scholar 

  21. van Praag, H. et al. Nature 415, 1030–1034 (2002).

    Article  CAS  Google Scholar 

  22. Ge, S., Yang, C.H., Hsu, K.S., Ming, G.L. & Song, H. Neuron 54, 559–566 (2007).

    Article  CAS  Google Scholar 

  23. Kempermann, G., Kuhn, H.G. & Gage, F.H. Nature 386, 493–495 (1997).

    Article  CAS  Google Scholar 

  24. Atasoy, D., Aponte, Y., Su, H.H. & Sternson, S.M. J. Neurosci. 28, 7025–7030 (2008).

    Article  CAS  Google Scholar 

  25. Sohal, V.S., Zhang, F., Yizhar, O. & Deisseroth, K. Nature 459, 698–702 (2009).

    Article  CAS  Google Scholar 

  26. Cardin, J.A. et al. Nature 459, 663–667 (2009).

    Article  CAS  Google Scholar 

  27. Kim, J.Y. et al. Neuron 63, 761–773 (2009).

    Article  CAS  Google Scholar 

  28. Duan, X. et al. Cell 130, 1146–1158 (2007).

    Article  CAS  Google Scholar 

  29. Toni, N. et al. Nat. Neurosci. 11, 901–907 (2008).

    Article  CAS  Google Scholar 

  30. Toni, N. et al. Nat. Neurosci. 10, 727–734 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Song and Ming laboratories for discussion, L. Tsai and K. Deisseroth for initial help with optogenetics, and Q. Hussaini, Y. Cai and L. Liu for technical support. The electron microscopy images were acquired at the electron microscopy facility of the University of Lausanne. This work was supported by grants from the US National Institutes of Health (NS047344, ES021957) and The Brain & Behavior Research Foundation to H.S., from the US National Institutes of Health (NS048271, HD069184), the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation, The Brain & Behavior Research Foundation and Maryland Stem Cell Research Fund to G.M., from the Swiss National Science Foundation (PP00A-119026/1) to N.T., from The Brain & Behavior Research Foundationand Maryland Stem Cell Research Fund to K.M.C., by postdoctoral fellowships from Maryland Stem Cell Research Fund to J. Song, Z.W. and C.Z., from the Fondation Leenaards to J.M., and by a pre-doctoral fellowship from The Children's Tumor Foundation to G.J.S.

Author information

Authors and Affiliations

Authors

Contributions

J. Song led and contributed to all aspects of the study. J.M. and N.T. performed electron microscopy analyses. J. Sun, Z.W., G.J.S., D.H., C.Z., H.D. and K.M.C. contributed to tool development and data collection and analyses. G.M. and H.S. supervised the project and wrote the manuscript.

Corresponding authors

Correspondence to Nicolas Toni, Guo-li Ming or Hongjun Song.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Immunohistological characterization of newborn progeny during early proliferative phases of adult hippocampal neurogenesis.

(a) A schematic diagram of the current view of progenitor subtypes and their marker expression during early phases of adult hippocampal neurogenesis. (b-d) Time course analysis of marker expression by retrovirally-labelled precursors in the adult dentate gyrus. Engineered onco-retroviruses expressing GFP were stereotaxcially injected into the adult mouse dentate gyrus. Shown in (b) are sample confocal images of staining for GFP, MCM2, Tbr2, DCX and DAPI. Scale bar: 10 mm. Shown in (c-d) are quantifications of different precursor subtypes among all GFP+ cells at 2, 4 and 7 days post injection (dpi). Numbers associated with plots indicates numbers of animal examined. The same set of animals was used for (c and d). Values represent mean ± s.e.m.

Supplementary Figure 2 Electrophysiological characterization of newborn progeny during early proliferative phases of adult hippocampal neurogenesis.

(a) Sample whole-cell voltage-clamp (Vm = -65 mV) recording traces of a GFP+ cell in the slice acutely prepared from injected animals at 4 dpi in response to puff of GABA (10 mM) in the presence or absence of bicuculline (100 mM). (b) Sample confocal images of staining of GFP, synapsin I (a synaptic vesicle protein), GAD67 (a GABAergic neuron marker), and DAPI. Scale bar: 5 mm. Arrows point to GAD67+synapsin+ puncta. Please see Supplementary Movie 1 for 3D reconstruction and detail. (c) Sample whole-cell voltage-clamp recording traces from a GFP+ cell in the slice acutely prepared from injected animals at 4 dpi. Note a lack of spontaneous synaptic current (SSC) and evoked postsynaptic current (PSC) in response to low frequency field stimulation (0.1 Hz) of the dentate granule cell layer. (d) Sample whole-cell voltage-clamp recording traces of a GFP+ cell at 4 dpi in the acute slice in response to 5 Hz stimulation of dentate granule cell layer in the absence and presence of bicuculline (100 μM).

Supplementary Figure 3 Synaptic inputs onto proliferating neural progeny from PV+ interneurons.

Shown are sample confocal images of GFP+ newborn progeny at 4 dpi with immunostaining of synapsin, PV and a proliferation marker MCM2, and DAPI. Scale bars: 20 mm (left panel) and 10 mm (right two panels).

Supplementary Figure 4 Immuno-EM analyses of synapses and close appositions between PV+ axon terminals and newborn progeny in the adult dentate gyrus.

Shown in (a-f) are steps to identify synaptic contact between a labelled PV+ axon terminal and labelled newborn progeny as shown in Fig. 1c. Locations of the newborn progeny (NP) were identified at the light microscopic level (a) and followed through to the electron microscopic level (b-f). Electron micrographs at increasing magnifications (b-d) show the position of the PV+ axon terminal in relation to three of the newborn progeny (NPs 1-3). The high magnification view of the PV+ axon terminal forming a symmetrical synaptic contact (arrowheads) with the labelled newborn progeny is shown in (d) and Fig. 1c. The morphology of the NP1 cell body is lacking from (b-d), but is shown in (e) with the PV+ axon closely apposed. To further describe the relationship between the PV+ axon and NPs 1 and 2, they have been reconstructed in 3D shown in (f). Various contacts between PV+ axons and newborn progeny in the adult dentate gyrus are shown in (g-l). Shown in (g-j) are two examples of PV+ axons closely apposing the tips of filopodia extending from labelled newborn progeny (NPs 4 and 5), at low and high magnifications. Shown in (k-l), PV+ axon segments (arrows in k) are seen both in the vicinity of a labelled newborn progeny (NP6) and closely apposing its principal dendritic extension (arrowheads in l). Note also the presence of cytoskeleton fibers within the dendritic extension (d) of the newborn progeny. Shown in (m-p) are processes to identify synaptic contact between a labelled PV+ axonal terminal and an unlabelled mature granule neuron (MN) as shown in Fig. 1e. The location of the mature neuron (arrow) was identified at the light microscopic level (m) and followed through to the electron microscopic level (n-p). Electron micrographs at increasing magnifications (n-p) show the position of the PV+ axon terminal in relation to the mature neuron. The high magnification view of the PV+ axon terminal forming a symmetrical synaptic contact (arrowheads) with the mature neuron is shown in (p) and Fig. 1e. Newborn progeny are coloured in green, PV+ axons in red and mature granule neurons in blue. Scale bars: a, 10 μm; b, 2 μm; c, 0.5 μm; d, 0.2 μm; e, 2 μm; f, 2 μm; g, 0.2 μm; h, 0.1 μm; i, 0.5 μm; j, 0.2 μm; k, 2 μm; l, 0.5 μm; m, 10 μm; n, 2 μm; o, 1 μm; p, 0.2 μm.

Supplementary Figure 5 Characterization of PV+ neuron synaptic inputs onto newborn progeny with optogenetic tools.

(a-b) Targeting of PV+ neurons and newborn progeny with engineered AAV and onco-retrovirus, respectively. Shown in (a) is a schematic diagram of the experimental design. Engineered AAV with Cre-dependent expression of ChR2-YFP, NpHR-YFP or Arch-YFP was stereotaxically injected into the dentate gyrus of 5 week-old PV-Cre mice and retrovirus expressing RFP was stereotaxically injected 4 weeks later. Electrophysiological recordings of RFP+ cells were carried out in slices acutely prepared from injected animals at 3-5 dpi. Shown in (b) are sample confocal images of ChR2-YFP and RFP in the dentate gyrus from AAV and retroviral injected PV-Cre animals. Note the wide spread of YFP+ fibers surrounding RFP+ cells. Scale bars: 50 mm (left) and 10 mm (right). (c) Effective light-induced manipulation of PV+ neuron firing in slices acutely prepared from AAV injected animals. Shown are sample traces of whole-cell recording of a ChR2-YFP+ neuron under current-clamp mode upon light-stimulation (472 nm blue light at 8 Hz, 5 ms).

Supplementary Figure 6 Activation of PV+ neurons at 8 Hz reduces cell death of newborn progeny.

Shown are sample confocal images of staining of Iba-1 (a microglia marker), EdU and DAPI (a). Note engulfing of EdU+ and EdU- pyknotic nuclei by Iba-1+ microglia. Scale bars: 10 mm. Also shown are percentages of EdU+ cells that were surrounded by Iba-1+ microglia (EdU+Iba-1#; b) and total number of EdU- cells that were surrounded by Iba-1+ microglia (EdU-Iba-1#; c) at 4 dpi. Numbers associated with bar graphs indicates numbers of animal examined. Values represent mean ± s.e.m. (**: P < 0.01; Student's t-test).

Supplementary Figure 7 Effect of optogenetic activation of PV+ or SST+ interneurons during early proliferative phases of adult hippocampal neurogenesis in vivo.

(a) Increased NeuroD+ newborn progeny upon PV+ neuron activation. Shown on the left are sample confocal images of NeuroD immunostaining, EdU and DAPI. Arrows point to EdU+NeuroD+ newborn progeny. Scale bar: 50 mm. Shown on the right is a summary of quantification of EdU+NeuroD+ cells under different conditions. Numbers associated with bar graphs indicates numbers of animal examined. Values represent mean ± s.e.m. (*: P < 0.05; Student's t-test). (b-c) Quantifications of percentages of EdU+ cells that were also MCM2+ (proliferating neural progeny; b), or were DCX+ (newborn neuronal progeny; c) at 4 dpi. Values represent mean ± s.e.m. (P > 0.10; Student's t-test). The same groups of animals as in Figs. 2c-d were examined.

Supplementary Figure 8 Effect of suppressing PV+, or SST+, interneuron activation during early phases of adult hippocampal neurogenesis in vivo.

(a) A schematic diagram of experimental design for Figs. 3a-e. EdU (41.1 mg/kg body weight) was i.p. injected 4 times 2.5 hours apart at day 0 and continuous yellow light (593 nm; constant or no light sham control) was delivered 8 hrs per day between day 1 and day 4. Animals were processed for analysis on day 1 or day 4. (b) Sample confocal images of NpHR-YFP and RFP in the dentate gyrus from AAV and retroviral injected PV-Cre animals. Note the wide spread of YFP+ fibers surrounding RFP+ cells. Scale bars: 50 mm (left) and 10 mm (right). (c) Effective light-induced suppression of PV+ neuron firing in slices acutely prepared from AAV injected animals. Shown are sample traces of whole-cell recordings of NpHR- or Arch-expressing YFP+ neurons under current-clamp mode upon light stimulation (593 nm yellow light; constant). A current pulse of 100 pA current was delivered to YFP+ neurons, and the number of action potentials was quantified with or without yellow light on. Values represent mean ± s.e.m. (n = 6 cells; **: P < 0.01; Student's t-test). (d-e) Quantification of percentages of EdU+ cells that were MCM2+ (proliferating neural progeny; d), or were DCX+ (newborn neuronal progeny; e) at 4 dpi. The same groups of animals as in Figs. 3a-b were examined. Numbers associated with bar graphs indicates numbers of animal examined. Values represent mean ± s.e.m. (P > 0.10; Student's t-test). (f-h) Quantification of percentages of EdU+ cells that were MCM2+ (proliferating neural progeny, f), or were DCX+ (newborn neuronal progeny, g), and stereological quantification of EdU+NeuroD+ cells (h) at 4 dpi. The same groups of animals as in Fig. 3e were analyzed. Values represent mean ± s.e.m. (**: P < 0.01; two-way ANOVA).

Supplementary Figure 9 Optogenetic manipulations do not appear to affect properties of mature dentate granule neurons in the adult dentate gyrus.

Mature dentate granule neurons were recorded in acutely prepared slices after optogenetic stimulation (AAV-ChR2 expression in PV-Cre mice) or suppression (AAV-Arch expression in PV-Cre mice) paradigm (as in Fig. 2a and Supplementary Fig. 8a). (a) Shown are sample whole-cell voltage-clamp recording traces of GABAergic SSCs (left panel) and summaries of frequency and amplitude of GABAergic SSCs recorded (right two panels). (b) Same as in (a), except that glutamatergic SSCs were examined. Numbers associated with bar graphs indicate numbers of cells examined. Values represent mean ± s.e.m. No significant differences were found (P > 0.1; Student's t-test).

Supplementary Figure 10 PV+ neuron activity regulates dendritic growth of newborn neurons during adult hippocampal neurogenesis.

(a) Presence of GABAergic synaptic inputs onto newborn neurons at 7 dpi from PV+ neurons. Same as in Fig. 1g, except that GFP+ newborn progeny at 4 and 7 dpi were examined and summaries for percentages of GFP+ cells recorded that exhibited PSCs (left), the mean amplitudes of PSCs (middle) and the PSC induction rate (right) are shown. The same data for 4 dpi as in Fig. 1g are shown for comparison. (b-f) Optogenetic manipulation of PV+ neuron activity affects dendritic development of newborn neurons. Shown in (b) is a schematic diagram of experimental procedure. Shown in (c) are sample projected confocal images of immunostaining for GFP and RFP. Scale bars: 20 mm. Also shown are summaries of total dendritic length (d) and branch numbers (e). Each dot represents data from one individual neuron (**: P < 0.01; *: P < 0.05; two sample Kolmogorov–Smirnov test). Shown in (f) is a Sholl analysis for dendritic complexity. Values represent mean ± s.e.m. (**: P < 0.01; two sample Kolmogorov–Smirnov test).

Supplementary Figure 11 Models of activity-dependent diametric regulation of adult hippocampal neurogenesis processes and two critical periods of activity-dependent survival of newborn neuronal progeny.

(a) Diametric regulation of two sequential proliferative processes of adult hippocampal neurogenesis by PV+ neuron activity. Shown on the left is an illustration of the dentate circuitry where entorhinal cortical inputs activate dentate granule neurons, which in turn activate PV+ interneurons. mGC: mature granule cell; IN: interneuron; RGL: radial glia-like precursor; NP: newborn progeny. Shown on the right is a model of diametric regulation of quiescent neural stem cell activation and survival and maturation of their proliferative neuronal progeny: during heightened activity within dentate gyrus (top panel), activation of PV+ neurons promotes the survival and maturation of proliferating neuronal progenitor and inhibits quiescent neural stem cell activation; conversely, when the activity in the dentate gyrus is low (bottom panel), decreased PV+ neuron activity suppresses the survival of proliferating neuronal progenitors and simultaneously promotes expansion of the quiescent neural stem cell pool via symmetric cell division. (b) Two critical periods of activity-dependent regulation of progeny survival during adult hippocampal neurogenesis. The first phase occurs during GABAergic synaptic integration of proliferative newborn progeny involving PV+ local interneurons. The second phase occurs during glutamatergic synaptic integration of post-mitotic newborn neurons via a glutamate-mediated mechanism.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 and Supplementary Table 1 (PDF 9794 kb)

Close association between GFP+ newborn progeny and GAD67+Synapsin I+ synaptic puncta in the adult dentate gyrus.

Adult mice injected with onco-retroviruses to express GFP in proliferating progenitors in the adult SGZ. Shown is a surface-rendered reconstruction of a series of confocal images of the dentate gyrus (90 x 90 x 30 mm) for immunostaining of GFP (green), GAD67 (blue) and synapsin I (red) at 4 dpi. (AVI 14969 kb)

Close association between GFP+ newborn progeny and PV+Synapsin I+ synaptic puncta in the adult dentate gyrus.

Adult mice were injected with onco-retroviruses to express GFP in proliferating progenitors in the adult SGZ. Shown is a surface-rendered reconstruction of a series of confocal images of the dentate gyrus (90 x 90 x 30 mm) for immunostaining of GFP (green), PV (blue) and synapsin I (red) at 4 dpi. (AVI 23452 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Song, J., Sun, J., Moss, J. et al. Parvalbumin interneurons mediate neuronal circuitry–neurogenesis coupling in the adult hippocampus. Nat Neurosci 16, 1728–1730 (2013). https://doi.org/10.1038/nn.3572

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3572

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