Visuomotor experience induces functional and structural plasticity of chandelier cells

  1. Department of Molecular Visual Plasticity, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
  2. Department of Axonal Signaling, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
  3. Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
  4. Department of Vision & Cognition, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
  5. Laboratory of Visual Brain Therapy, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de la Vision, Paris F-75012, France
  6. Department of Integrative Neurophysiology, Centre for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
  7. Department of Psychiatry, Academic Medical Center, University of Amsterdam. Amsterdam, The Netherlands
  8. Department of Cortical Structure & Function, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
  9. Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, the Netherlands

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, public reviews, and a response from the authors (if available).

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Brice Bathellier
    CNRS, Paris, France
  • Senior Editor
    Joshua Gold
    University of Pennsylvania, Philadelphia, United States of America

Reviewer #1 (Public Review):

Overall, the experiments are well-designed and the results of the study are exciting. We have one major concern, as well as a few minor comments that are detailed in the following.

Major:
1. The authors suggest that "Visuomotor experience induces functional and structural plasticity of chandelier cells". One puzzling thing here, however, is that mice constantly experience visuomotor coupling throughout life which is not different from experience in the virtual tunnel. Why do the authors think that the coupled experience in the VR induces stronger experience-dependent changes than the coupled experience in the home cage? Could this be a time-dependent effect (e.g. arousal levels could systematically decrease with the number of head-fixed VR sessions)? The control experiment here would be to have a group of mice that experience similar visual flow without coupling between movement and visual flow feedback. Either change would be experience-dependent of course, but having the "visuomotor experience dependent" in the title might be a bit strong given the lack of control for that. We would suggest changing the pitch of the manuscript to one of the conclusions the authors can make cleanly (e.g. Figure 4).

Minor:
2. "ChCs shape the communication hierarchy of cortical networks providing visual and contextual information." We are not sure what this means.

3. "respond to locomotion and visuomotor mismatch, indicating arousal-related activity" This is not clear. We think we understand what the authors mean but would suggest rephrasing.

4. 'based on morphological properties revealed that 87% (287/329) of labeled neurons were ChCs" Please specify the morphological properties used for the classification somewhere in the methods.

5. We may have missed this - in the patch clamp experiment (Fig.1 H-K), please add information about how many mice/slices these experiments were performed in.

6. "These findings suggest that the rabies-labeled L1-4 neurons providing monosynaptic input to ChCs are predominantly inhibitory neurons". We are not sure this conclusion is warranted given the sparse set of neurons labelled and the low number of cells recorded in the paired patch experiment. We would suggest properly testing (e.g. stain for GABA on the rabies data) or rephrasing.

7. Figure 2E. A direct comparison of dF/F across different cell types can be subject to a problematic interpretation. The transfer function from spikes to calcium can be different from cell type to cell type. Additionally, the two cell populations have been marked with different constructs (despite the fact that it's the same GECI) further reducing the reliability of dF/F comparisons. We would recommend using a different representation here that does not rely on a direct comparison of dF/F responses (e.g. like the "response strength" used in Figure 3B). Assuming calcium dynamics are different in ChCs and PyCs - this similarity in calcium response is likely a coincidence.

8. If ChCs are more strongly driven by locomotion and arousal, then it's a bit counterintuitive that at the beginning of the visual corridor when locomotion speed consistently increases, the activity of ChCs consistently decreases. This does not appear to be driven by suppression by visual stimuli as it is present also in the first and last 20cm of the tunnel where there are no visual stimuli. How do the authors explain this?

9. The authors mention that "ChC responses underwent sensory-evoked plasticity during the repeated visual exposure, even though the visual stimuli were different from those encountered during training in the virtual tunnel". How would this work? And would this mean all visual responses are reduced? What is special about the visual experience in the virtual tunnel? It does not inherently differ from visual experience in the home cage, given that the test stimuli (full field gratings) are different from both.

10. Just as a point to consider for future experiments: For the open-loop control experiments, the visual flow is constant (20cm/s) - ideally, this would be a replay of the running speed the mouse previously generated to match statistics.

11. We would recommend specifying the parameters used for neuropil correction in the methods section.

12. If we understand correctly, the F0 used for the dF/F calculation is different from that used for division. Why is this?

13. Authors compare neuronal responses using "baseline-corrected average". Please specify the parameters of the baseline correction (i.e. what is used as baseline here).

Reviewer #2 (Public Review):

Summary:
Seignette et al. investigated the potential roles of axo-axonic (chandelier) cells (ChCs) in a sensory system, namely visual processing. As introduced by the authors, the axo-axonic cell type has remained (and still is) somehow mysterious in its function. Seignette and colleagues leveraged the development of a transgenic mouse line selective for ChC, and applied a very wide range of techniques: transsynaptic rabies tracing, optogenetic input activation, in vitro electrophysiology, 2-photon recording in vivo, behavior and chemogenetic manipulations, to precisely determine the contribution of ChCs to the primary visual cortex network.

The main findings are 1) the identification of synaptic inputs to ChC, with a majority of local, deep layer principal neurons (PN), 2) the demonstration that ChC is strongly and synchronously activated by visual stimuli with low specificity in naive animals, 3) the recruitment of ChC by arousal/visuomotor mismatch, 4) the induction of functional and structural plasticity at the ChC-PN module, and, 5) the weak disinhibition of PNs induced by ChCs silencing. All these findings are strongly supported by experimental data and thoroughly compared to available evidence.

Strengths:
This article reports an impressive range of very demanding experiments, which were well executed and analyzed, and are presented in a very clear and balanced manner. Moreover, the manuscript is well-written throughout, making it appealing to future readers. It has also been a pleasure to review this article.

In sum, this is an impressive study and an excellent manuscript, that presents no major flaws.

Notably, this study is one of the first studies to report on the activities and potential roles of axo-axonic cells in an active, integrated brain process, beyond locomotion as reported and published in V1. This type of research was much awaited in the fields of interneuron and vision research.

Weaknesses:
There are no fundamental weaknesses; the latter mainly concern the presentation of the main results.

The main weakness may be that the different sections appear somehow disconnected conceptually.

Additionally, some parts deserve a more in-depth clarification/simplification of concepts and analytic methods for scientists outside the subfield of V1 research. Indeed, this paper will be of key interest to researchers of various backgrounds.

Reviewer #3 (Public Review):

Summary:
The authors set out to characterize the anatomical connectivity profile and the functional responses of chandelier cells (ChCs) in the mouse primary visual cortex. Using retrograde rabies tracing, optogenetics, and in vitro electrophysiology, they found that the primary source of input to ChCs are local layer 5 pyramidal cells, as well as long-range thalamic and cortical connections. ChCs provided input to local layer 2/3 pyramidal neurons, but did not receive reciprocal connections.

With two-photon calcium imaging recordings during passive viewing of drifting gratings, the authors showed that ChCs exhibit weakly selective visual responses, high correlations within their own population, and strong responses during periods of arousal (assessed by locomotion and pupil size). These results were replicated and extended in experiments with natural images and prediction of receptive field structure using a convolutional neural network.

Furthermore, the authors employed a learned visuomotor task in a virtual corridor to show that ChCs exhibit strong responses to mismatches between visual flow and locomotion, locomotion-related activation (similar to what was shown above), and visually-evoked suppression. They also showed the existence of two clusters of pyramidal neurons with functionally different responses - a cluster with "classically visual" responses and a cluster with locomotion- and mismatch-driven responses (the latter more correlated with ChCs). Comparing naive and trained mice, the authors found that visual responses of ChCs are suppressed following task learning, accompanied by a shortening of the axon initial segment (AIS) of pyramidal cells and an increase in the proportion of AIS contacted by ChCs. However, additional controls would be required to identify which component(s) of the experimental paradigm led to the functional and anatomical changes observed.

Finally, using a chemogenetic inactivation of ChCs, the authors propose weak connectivity to pyramidal cells (due to small effects in pyramidal cell activity). However, these results are not unequivocally supported, as the baseline activity of ChCs before inactivation is considerably lower, suggesting a potentially confounding homeostatic plasticity mechanism might already be operating.

Strengths:
The authors bring a comprehensive, state-of-the-art methodology to bear, including rabies tracing, in vivo two-photon calcium imaging, in vitro electrophysiology, optogenetics and chemogenetics, and deep neural networks. Their analyses and statistical tests are sound and for the most part, support their claims. Their results are in line with previous findings and extend them to the primary visual cortex.

Weaknesses:
- Some of the results (e.g. arousal-related responses) are not entirely surprising given that similar results exist in other cortical areas.

- Control analyses regarding locomotion patterns before and after learning the task (Figure 5), and additional control experiments to identify whether functional and anatomical changes following task learning were due to learning, repeated visual exposure, exposure to reward, or visuomotor experience would strengthen the claims made.

- The strength of the results of the chemogenetics experiment is impacted by the lower baseline activity of ChCs that express the KORD receptor. At present, it is not possible to exclude the presence of homeostatic plasticity in the network *before* the inactivation takes place.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation