Summary

In Vivo Visualization of Spontaneous Activity in Neonatal Mouse Sensory Cortex at a Single-Neuron Resolution

Published: November 21, 2023
doi:

Summary

Primary sensory areas in the neocortex exhibit unique spontaneous activities during development. This article describes how to visualize individual neuron activities and primary sensory areas to analyze area-specific synchronous activities in neonatal mice in vivo.

Abstract

The mammalian brain undergoes dynamic developmental changes at both the cellular and circuit levels throughout prenatal and postnatal periods. Following the discovery of numerous genes contributing to these developmental changes, it is now known that neuronal activity also substantially modulates these processes. In the developing cerebral cortex, neurons exhibit synchronized activity patterns that are specialized to each primary sensory area. These patterns markedly differ from those observed in the mature cortex, emphasizing their role in regulating area-specific developmental processes. Deficiencies in neuronal activity during development can lead to various brain diseases. These findings highlight the need to examine the regulatory mechanisms underlying activity patterns in neuronal development. This paper summarizes a series of protocols to visualize primary sensory areas and neuronal activity in neonatal mice, to image the activity of individual neurons within the cortical subfields using two-photon microscopy in vivo, and to analyze subfield-related activity correlations. We show representative results of patchwork-like synchronous activity within individual barrels in the somatosensory cortex. We also discuss various potential applications and some limitations of this protocol.

Introduction

The cerebral cortex contains several sensory areas with distinct functions. The areas receive inputs originating from their corresponding sensory organs, mostly conveyed through the spinal cord or brainstem and relayed via the thalamus1,2. Notably, neurons in each primary sensory area exhibit uniquely synchronized activity during early developmental stages, which also originate from sensory organs or the lower nervous centers, but essentially differ from the activities observed in the mature cortex3.

In neonatal rodents, for example, the primary visual area (V1) displays wave-like activity, which originates in the retina (retinal wave) and propagates through the entire visual pathway while conserving retinotopy4. The primary auditory area (A1) exhibits synchronous activity organized in band-shaped subregions that correspond to the isofrequency bands in the mature brain. The activity emanates from the cochlea's inner hair cells5,6. The barrel cortex in the primary somatosensory area (S1) shows a patchwork-like activity pattern in which layer 4 neurons within individual barrels, namely, neurons responsive to individual whiskers, are synchronously activated7. Although proposed to originate from the trigeminal ganglion, the source of the activity remains unknown7. Consequently, neonatal activity patterns are specialized both within each primary sensory area and within intra-areal subfields. The simultaneous visualization of neuronal activity and structure of primary sensory areas may facilitate an inquiry into the contribution of these activity patterns to the development of sensory systems.

In this article, we summarized a series of protocols: (1) to visualize individual neuronal activities using sparse labeling of GCaMP and primary sensory areas using TCA-RFP mice that express red fluorescent protein in thalamocortical axons7, (2) to image single cell-level activity in neonatal mice using two-photon microscopy in vivo, and (3) to analyze the activity correlations within S1 barrel cortex. The representative results show patchwork-like synchronized activity within individual barrels of a postnatal day (P)6 mouse. Despite some limitations, this technique can be used for chronic imaging, wide-field imaging across multiple sensory areas, and various manipulation experiments. The multifaceted analysis of neuronal activity during development will enrich our comprehension of brain circuit formation mechanisms.

Protocol

All the experiments were conducted in accordance with the guidelines for animal experimentation of Kumamoto University and the National Institute of Genetics and approved by the animal experimentation committees. 1. In utero electroporation (IUE) Mate male TCA-RFP mice of ICR background with female wild-type ICR mice. Observe the vaginal plug to check for mating early morning of the next day. Observe the abdomen to check for pregnancy 2 weeks later. …

Representative Results

Figure 1 shows the representative results of layer 4 neuron activities in the barrel cortex of a P6 pup visualized using the present protocol. Two-photon images of the green channel (GCaMP) and red channel (TCA-RFP) were temporally averaged and shown in Figure 1A. Because TCA-RFP fluorescence was much weaker than GCaMP fluorescence, the GCaMP signal leaked into the red channel (Figure 1A1,A2). Fourteen ROIs were dra…

Discussion

Given that the spontaneous activities emerge from the sensory organ or lower nervous system and travel to the primary sensory area through a pathway equivalent to that of a mature nervous system3, it is crucial to define the primary sensory area and the location of imaged neurons within the area. In this protocol, we addressed this requirement by employing transgenic mice that visualize thalamocortical axons and the Supernova system that expresses GCaMP sparsely8. These tec…

Offenlegungen

The authors have nothing to disclose.

Acknowledgements

This work was supported by the Japan Society for the Promotion of Science Grants-in-Aid for Transformative Research Areas (B) (22H05092, 22H05094) and for Scientific Research Grants 20K06876, AMED under Grant Number 21wm0525015, the Takeda Science Foundation, the Naito Foundation, the Kato Memorial Bioscience Foundation, the Kowa Life Science Foundation, NIG-JOINT (24A2021) (to H.M.); and Japan Society for the Promotion of Science Grants-in-Aid for Scientific Research Grants 19K06887 and 22K06446, the Kodama Memorial Fund for Medical Research, the Uehara Memorial Foundation, the Kato Memorial Bioscience Foundation, and the Takeda Science Foundation (to N.N-T.). We thank Dr. Takuji Iwasato for the TCA-RFP mice.

Materials

20× objective lens (water immersion)
250 mL Vacuum Filter/Storage Bottle System Corning 431096
4%-paraformaldehyde phosphate buffer solution (4% PFA) Nacalai 09154-85
Acrylic resin (UNIFAST II) GC N/A
Agarose Sigma A9793
Aspirator tube assembly Drummond 2-040-000
CaCl2•2H2O Nacalai 06731-05
Electroporator BEX GEB14
Eye drop (Scopisol) Senju Pharmaceutical N/A
Fluorescence stereo microscope Leica M165FC
Glucose Nacalai 16806-25
Heating pad Muromachi Kikai FHC-HPS
HEPES Gibco 15630-080
Isoflurane Pfizer N/A
KCl Nacalai 28514-75
MgSO4•7H2O Wako 131-00405
Micropipette puller Narishige PC-100
Multiphoton laser Spectra-Physics Mai Tai eHP DeepSee
Multiphoton microscope Zeiss LSM 7MP
NaCl Nacalai 31320-05
Non-woven fabric (Kimwipe) Kimberly Clark S-200
Phosphate buffered saline (PBS) Nacalai 27575-31
Plasmid: CAG-loxP-STOP-loxP-GCaMP6s-ires-tTA-WPRE Addgene pK175
Plasmid: TRE-nCre Addgene pK031
Precision calibrated micropipets Drummond 2-000-050
Razor blade Feather FA-10
Rimadyl (50 mg/mL Carprofen) Zoetis JP N/A
Round cover glass, 3-mm-diameter  Matsunami CS01078
Saline Otsuka 035175315
Sodium pentobarbital Nacalai 26427-72
Stage for imaging living pup (two single-axis translation stage for XY positioning, two-axis goniometer, base plate, adjustable pillar for z positioning) ThorLabs LT1/M, GN2/M, BM2060/M, MLP01/M
TCA-RFP mouse N/A N/A Mizuno et al., 2018a
Tissue adhesive (Vetbond) 3M 1469SB
Titanium bar Endo Scientific Instrument N/A Custom made (Mizuno et al., 2018b)
Titanium bar fixing plate N/A Custom made (Mizuno et al., 2018b)
Trypan blue Sigma T8154
Tweezers with platinum plate electrode, 5 mm diameter BEX CUY650P5
Wild-type ICR mouse Nihon SLC Slc:ICR

Referenzen

  1. Rao, M. S., Mizuno, H. Elucidating mechanisms of neuronal circuit formation in layer 4 of the somatosensory cortex via intravital imaging. Neuroscience Research. 167, 47-53 (2021).
  2. Iwasato, T., Erzurumlu, R. S. Development of tactile sensory circuits in the CNS. Current Opinion in Neurobiology. 53, 66-75 (2018).
  3. Martini, F. J., Guillamón-Vivancos, T., Moreno-Juan, V., Valdeolmillos, M., López-Bendito, G. Spontaneous activity in developing thalamic and cortical sensory networks. Neuron. 109 (16), 2519-2534 (2021).
  4. Ackman, J. B., Burbridge, T. J., Crair, M. C. Retinal waves coordinate patterned activity throughout the developing visual system. Nature. 490 (7419), 219-225 (2012).
  5. Tritsch, N. X., Yi, E., Gale, J. E., Glowatzki, E., Bergles, D. E. The origin of spontaneous activity in the developing auditory system. Nature. 450 (7166), 50-55 (2007).
  6. Babola, T. A., et al. Homeostatic control of spontaneous activity in the developing auditory system. Neuron. 99 (3), 511-524.e5 (2018).
  7. Mizuno, H., et al. Patchwork-type spontaneous activity in neonatal barrel cortex layer 4 transmitted via thalamocortical projections. Cell Reports. 22 (1), 123-135 (2018).
  8. Mizuno, H., et al. NMDAR-regulated dynamics of layer 4 neuronal dendrites during thalamocortical reorganization in neonates. Neuron. 82 (2), 365-379 (2014).
  9. Tabata, H., Nakajima, K. Efficient in utero gene transfer system to the developing mouse brain using electroporation: visualization of neuronal migration in the developing cortex. Neurowissenschaften. 103 (4), 865-872 (2001).
  10. Fukuchi-Shimogori, T., Grove, E. A. Neocortex patterning by the secreted signaling molecule FGF8. Science. 294 (5544), 1071-1074 (2001).
  11. Saito, T., Nakatsuji, N. Efficient gene transfer into the embryonic mouse brain using in vivo electroporation. Entwicklungsbiologie. 240 (1), 237-246 (2001).
  12. Holtmaat, A., et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial window. Nature Protocols. 4 (8), 1128-1144 (2009).
  13. Mizuno, H., Nakazawa, S., Iwasato, T. In vivo two-photon imaging of cortical neurons in neonatal mice. Journal of Visualized Experiments. 140, e58340 (2018).
  14. Egashira, T., et al. In vivo two-photon calcium imaging of cortical neurons in neonatal mice. STAR Protocols. 4 (2), 102245 (2023).
  15. Cantu, D. A., et al. EZcalcium: Open-source toolbox for analysis of calcium imaging data. Frontiers in Neural Circuits. 14, 25 (2020).
  16. Maruoka, H., et al. Lattice system of functionally distinct cell types in the neocortex. Science. 358 (6363), 610-615 (2017).
  17. Antón-Bolaños, N., et al. Prenatal activity from thalamic neurons governs the emergence of functional cortical maps in mice. Science. 364 (6444), 987-990 (2019).
  18. Guillamón-Vivancos, T., et al. Input-dependent segregation of visual and somatosensory circuits in the mouse superior colliculus. Science. 377 (6608), 845-850 (2022).
  19. Cardin, J. A., Crair, M. C., Higley, M. J. Mesoscopic imaging: Shining a wide light on large-scale neural dynamics. Neuron. 108 (1), 33-43 (2020).
  20. Chen, J. L., Voigt, F. F., Javadzadeh, M., Krueppel, R., Helmchen, F. Long-range population dynamics of anatomically defined neocortical networks. eLife. 5, e14679 (2016).
  21. Ota, K., et al. cell-resolution, contiguous-wide two-photon imaging to reveal functional network architectures across multi-modal cortical areas. Neuron. 109 (11), 1810-1824 (2021).
  22. Zariwala, H. A., et al. A Cre-dependent GCaMP3 reporter mouse for neuronal imaging in vivo. The Journal of Neuroscience. 32 (9), 3131-3141 (2012).
  23. Murakami, T., Matsui, T., Uemura, M., Ohki, K. Modular strategy for development of the hierarchical visual network in mice. Nature. 608 (7923), 578-585 (2022).
  24. Pnevmatikakis, E. A., et al. Simultaneous denoising, deconvolution, and demixing of Calcium imaging data. Neuron. 89 (2), 285-299 (2016).
  25. Shemesh, O. A., et al. Precision calcium imaging of dense neural populations via a cell-body-targeted calcium indicator. Neuron. 107 (3), 470-486 (2020).
  26. Giovannucci, A., et al. CaImAn an open source tool for scalable calcium imaging data analysis. Elife. 8, e38173 (2019).
  27. Pachitariu, M., et al. Suite2p: beyond 10,000 neurons with standard two-photon microscopy. BioRxiv. , (2017).
  28. Sitdikova, G., et al. Isoflurane suppresses early cortical activity. Annals of Clinical and Translational Neurology. 1 (1), 15-26 (2014).
  29. Marques-Smith, A., et al. A Transient translaminar GABAergic interneuron circuit connects thalamocortical recipient layers in neonatal somatosensory cortex. Neuron. 89 (3), 536-549 (2016).
  30. Tuncdemir, S. N., et al. Early somatostatin interneuron connectivity mediates the maturation of deep layer cortical circuits. Neuron. 89 (3), 521-535 (2016).
  31. Nakazawa, S., Yoshimura, Y., Takagi, M., Mizuno, H., Iwasato, T. Developmental phase transitions in spatial organization of spontaneous activity in postnatal barrel cortex layer 4. The Journal of Neuroscience. 40 (40), 7637-7650 (2020).
  32. Yu, Y. -. C., et al. Preferential electrical coupling regulates neocortical lineage-dependent microcircuit assembly. Nature. 486 (7401), 113-117 (2012).
  33. Siegel, F., Heimel, J. A., Peters, J., Lohmann, C. Peripheral and central inputs shape network dynamics in the developing visual cortex in vivo. Current Biology. 22 (3), 253-258 (2012).
  34. Nakagawa, N., Hosoya, T. Slow dynamics in microcolumnar gap junction network of developing neocortical pyramidal neurons. Neurowissenschaften. 406, 554-554 (2019).
  35. Valiullina, F., et al. Developmental changes in electrophysiological properties and a transition from electrical to chemical coupling between excitatory layer 4 neurons in the rat barrel cortex. Frontiers in Neural Circuits. 10, 1 (2016).
  36. Avitan, L., et al. Spontaneous and evoked activity patterns diverge over development. Elife. 10, e61942 (2021).
  37. Mölter, J., Avitan, L., Goodhill, G. J. Detecting neural assemblies in calcium imaging data. BMC Biology. 16 (1), 143 (2018).
  38. Nakazawa, S., Mizuno, H., Iwasato, T. Differential dynamics of cortical neuron dendritic trees revealed by long-term in vivo imaging in neonates. Nature Communications. 9 (1), 3106 (2018).
This article has been published
Video Coming Soon
Keep me updated:

.

Diesen Artikel zitieren
Nakagawa-Tamagawa, N., Egashira, T., Rao, M. S., Mizuno, H. In Vivo Visualization of Spontaneous Activity in Neonatal Mouse Sensory Cortex at a Single-Neuron Resolution. J. Vis. Exp. (201), e65899, doi:10.3791/65899 (2023).

View Video