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.

  • Article
  • Published:

Norepinephrine modulates calcium dynamics in cortical oligodendrocyte precursor cells promoting proliferation during arousal in mice

Abstract

Oligodendrocytes, the myelinating cells of the central nervous system (CNS), are generated from oligodendrocyte precursor cells (OPCs) that express neurotransmitter receptors. However, the mechanisms that affect OPC activity in vivo and the physiological roles of neurotransmitter signaling in OPCs are unclear. In this study, we generated a transgenic mouse line that expresses membrane-anchored GCaMP6s in OPCs and used longitudinal two-photon microscopy to monitor OPC calcium (Ca2+) dynamics in the cerebral cortex. OPCs exhibit focal and transient Ca2+ increases within their processes that are enhanced during locomotion-induced increases in arousal. The Ca2+ transients occur independently of excitatory neuron activity, rapidly decline when OPCs differentiate and are inhibited by anesthesia, sedative agents or noradrenergic receptor antagonists. Conditional knockout of α1A adrenergic receptors in OPCs suppresses spontaneous and locomotion-induced Ca2+ increases and reduces OPC proliferation. Our results demonstrate that OPCs are directly modulated by norepinephrine in vivo to enhance Ca2+ dynamics and promote population homeostasis.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: In vivo 2P imaging reveals that OPCs exhibit dynamic Ca2+ activity.
Fig. 2: OPC Ca2+ activity is significantly suppressed during anesthesia.
Fig. 3: Enforced locomotion enhances OPC Ca2+ activity in the mouse visual cortex.
Fig. 4: OPC Ca2+ activity is reduced when noradrenergic signaling is inhibited.
Fig. 5: OPC Ca2+ transients evoked by the α1 adrenergic receptor agonist are independent of synaptic activity in vitro.
Fig. 6: Deletion of Adra1a from OPCs eliminates alpha-adrenoceptor-mediated Ca2+ signaling.
Fig. 7: Spontaneous Ca2+ transients in OPCs progressively decline as they differentiate into myelinating oligodendrocytes.
Fig. 8: Selective deletion of Adra1a from OPCs reduces their proliferation.

Similar content being viewed by others

Data availability

The gene expression dataset in Extended Data Fig. 10c was deposited to the Gene Expression Omnibus with accession number GSE226635 and is available to the public immediately (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE226635). The mouse genome assembly (GRCm38/mm10) used in this study is available via the UCSC Genome Browser Gateway (https://genome.ucsc.edu/cgi-bin/hgGateway?db=mm10). All images and videos generated during this study are available from the corresponding author upon reasonable request.

Code availability

Code used for data acquisition and analysis in this study is available on GitHub (https://github.com/Bergles-lab/Lu-et-al-NN-2023).

References

  1. Xiao, L. et al. Rapid production of new oligodendrocytes is required in the earliest stages of motor-skill learning. Nat. Neurosci. 19, 1210–1217 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hughes, E. G., Orthmann-Murphy, J. L., Langseth, A. J. & Bergles, D. E. Myelin remodeling through experience-dependent oligodendrogenesis in the adult somatosensory cortex. Nat. Neurosci. 21, 696–706 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chang, A., Nishiyama, A., Peterson, J., Prineas, J. & Trapp, B. D. NG2-positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions. J. Neurosci. 20, 6404–6412 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Orthmann-Murphy, J. et al. Remyelination alters the pattern of myelin in the cerebral cortex. eLife 9, e56621 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Buchanan, J. et al. Oligodendrocyte precursor cells ingest axons in the mouse neocortex. Proc. Natl Acad. Sci. USA 119, e2202580119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Falcão, A. M. et al. Disease-specific oligodendrocyte lineage cells arise in multiple sclerosis. Nat. Med. 24, 1837–1844 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Kirby, L. et al. Oligodendrocyte precursor cells present antigen and are cytotoxic targets in inflammatory demyelination. Nat. Commun. 10, 3887 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Auguste, Y. S. S. et al. Oligodendrocyte precursor cells engulf synapses during circuit remodeling in mice. Nat. Neurosci. 25, 1273–1278 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Xiao, Y., Petrucco, L., Hoodless, L. J., Portugues, R. & Czopka, T. Oligodendrocyte precursor cells sculpt the visual system by regulating axonal remodeling. Nat. Neurosci. 25, 280–284 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Djogo, T. et al. Adult NG2-glia are required for median eminence-mediated leptin sensing and body weight control. Cell Metab. 23, 797–810 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Larson, V. A., Zhang, Y. & Bergles, D. E. Electrophysiological properties of NG2+ cells: matching physiological studies with gene expression profiles. Brain Res. 1638, 138–160 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hrvatin, S. et al. Single-cell analysis of experience-dependent transcriptomic states in the mouse visual cortex. Nat. Neurosci. 21, 120–129 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Bergles, D. E., Roberts, J. D. B., Somogyi, P. & Jahr, C. E. Glutamatergic synapses on oligodendrocyte precursor cells in the hippocampus. Nature 405, 187–191 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Lin, S. & Bergles, D. E. Synaptic signaling between GABAergic interneurons and oligodendrocyte precursor cells in the hippocampus. Nat. Neurosci. 7, 24–32 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Biase, L. M. D., Nishiyama, A. & Bergles, D. E. Excitability and synaptic communication within the oligodendrocyte lineage. J. Neurosci. 30, 3600–3611 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Chen, T.-J. et al. In vivo regulation of oligodendrocyte precursor cell proliferation and differentiation by the AMPA-receptor subunit GluA2. Cell Rep. 25, 852–861 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Káradóttir, R., Cavelier, P., Bergersen, L. H. & Attwell, D. NMDA receptors are expressed in oligodendrocytes and activated in ischaemia. Nature 438, 1162–1166 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Khawaja, R. R. et al. GluA2 overexpression in oligodendrocyte progenitors promotes postinjury oligodendrocyte regeneration. Cell Rep. 35, 109147 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kougioumtzidou, E. et al. Signalling through AMPA receptors on oligodendrocyte precursors promotes myelination by enhancing oligodendrocyte survival. eLife 6, e28080 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Mei, F. et al. Identification of the kappa-opioid receptor as a therapeutic target for oligodendrocyte remyelination. J. Neurosci. 36, 7925–7935 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Deshmukh, V. A. et al. A regenerative approach to the treatment of multiple sclerosis. Nature 502, 327–332 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hughes, E. G., Kang, S. H., Fukaya, M. & Bergles, D. E. Oligodendrocyte progenitors balance growth with self-repulsion to achieve homeostasis in the adult brain. Nat. Neurosci. 16, 668–676 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Marisca, R. et al. Functionally distinct subgroups of oligodendrocyte precursor cells integrate neural activity and execute myelin formation. Nat. Neurosci. 23, 363–374 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rungta, R. L., Chaigneau, E., Osmanski, B.-F. & Charpak, S. Vascular compartmentalization of functional hyperemia from the synapse to the pia. Neuron 99, 362–375 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, T.-W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kang, S. H., Fukaya, M., Yang, J. K., Rothstein, J. D. & Bergles, D. E. NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 68, 668–681 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Thrane, A. S. et al. General anesthesia selectively disrupts astrocyte calcium signaling in the awake mouse cortex. Proc. Natl Acad. Sci. USA 109, 18974–18979 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Paukert, M. et al. Norepinephrine controls astroglial responsiveness to local circuit activity. Neuron 82, 1263–1270 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Silvestre, J. S. & Prous, J. Research on adverse drug events. I. Muscarinic M3 receptor binding affinity could predict the risk of antipsychotics to induce type 2 diabetes. Methods Find. Exp. Clin. Pharmacol. 27, 289 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Cohen, R. I. & Almazan, G. Norepinephrine-stimulated PI hydrolysis in oligodendrocytes is mediated by alpha 1A-adrenoceptors. Neuroreport 4, 1115–1118 (1993).

    CAS  PubMed  Google Scholar 

  33. Papay, R. et al. Localization of the mouse α1A-adrenergic receptor (AR) in the brain: α1AAR is expressed in neurons, GABAergic interneurons, and NG2 oligodendrocyte progenitors. J. Comp. Neurol. 497, 209–222 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Ye, L. et al. Ethanol abolishes vigilance-dependent astroglia network activation in mice by inhibiting norepinephrine release. Nat. Commun. 11, 6157 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. He, D. et al. lncRNA functional networks in oligodendrocytes reveal stage-specific myelination control by an lncOL1/Suz12 complex in the CNS. Neuron 93, 362–378 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Schain, A. J., Hill, R. A. & Grutzendler, J. Label-free in vivo imaging of myelinated axons in health and disease with spectral confocal reflectance microscopy. Nat. Med. 20, 443–449 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hill, R. A., Li, A. M. & Grutzendler, J. Lifelong cortical myelin plasticity and age-related degeneration in the live mammalian brain. Nat. Neurosci. 21, 683–695 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Barres, A., Lazar, M. A. & Raff, M. C. A novel role for thyroid hormone, glucocorticoids and retinoic acid in timing oligodendrocyte development. Development 120, 1097–1108 (1994).

    Article  CAS  PubMed  Google Scholar 

  39. Luttrell, L. M., Daaka, Y. & Lefkowitz, R. J. Regulation of tyrosine kinase cascades by G-protein-coupled receptors. Curr. Opin. Cell Biol. 11, 177–183 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Hamilton, N., Vayro, S., Wigley, R. & Butt, A. M. Axons and astrocytes release ATP and glutamate to evoke calcium signals in NG2-glia. Glia 58, 66–79 (2010).

    Article  PubMed  Google Scholar 

  41. Ge, W.-P. et al. Long-term potentiation of neuron-glia synapses mediated by Ca2+-permeable AMPA receptors. Science 312, 1533–1537 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Paez, P. M., Fulton, D. J., Spreur, V., Handley, V. & Campagnoni, A. T. Multiple kinase pathways regulate voltage-dependent Ca2+ influx and migration in oligodendrocyte precursor cells. J. Neurosci. 30, 6422–6433 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Harlow, D. E., Saul, K. E., Komuro, H. & Macklin, W. B. Myelin proteolipid protein complexes with αv integrin and AMPA receptors in vivo and regulates AMPA-dependent oligodendrocyte progenitor cell migration through the modulation of cell-surface GluR2 expression. J. Neurosci. 35, 12018–12032 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Maldonado, P. P., Vélez-Fort, M., Levavasseur, F. & Angulo, M. C. Oligodendrocyte precursor cells are accurate sensors of local K+ in mature gray matter. J. Neurosci. 33, 2432–2442 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Spitzer, S. O. et al. Oligodendrocyte progenitor cells become regionally diverse and heterogeneous with age. Neuron 101, 459–471 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Schwarz, L. A. & Luo, L. Organization of the locus coeruleus-norepinephrine system. Curr. Biol. 25, R1051–R1056 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Qian, L. et al. β2 adrenergic receptor activation induces microglial NADPH oxidase activation and dopaminergic neurotoxicity through an ERK-dependent/protein kinase A-independent pathway. Glia 57, 1600–1609 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Jhaveri, D. J. et al. Norepinephrine directly activates adult hippocampal precursors via β3-adrenergic receptors. J. Neurosci. 30, 2795–2806 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bojarskaite, L. et al. Astrocytic Ca2+ signaling is reduced during sleep and is involved in the regulation of slow wave sleep. Nat. Commun. 11, 3240 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Srinivasan, R. et al. Ca2+ signaling in astrocytes from Ip3r2−/− mice in brain slices and during startle responses in vivo. Nat. Neurosci. 18, 708–717 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Papay, R. et al. Mouse α1B-adrenergic receptor is expressed in neurons and NG2 oligodendrocytes. J. Comp. Neurol. 478, 1–10 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Agarwal, A. et al. Transient opening of the mitochondrial permeability transition pore induces microdomain calcium transients in astrocyte processes. Neuron 93, 587–605 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Aston-Jones, G. & Bloom, F. E. Activity of norepinephrine-containing locus coeruleus neurons in behaving rats anticipates fluctuations in the sleep-waking cycle. J. Neurosci. 1, 876–886 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Matsumoto, Y. et al. Differential proliferation rhythm of neural progenitor and oligodendrocyte precursor cells in the young adult hippocampus. PLoS ONE 6, e27628 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bellesi, M. et al. Effects of sleep and wake on oligodendrocytes and their precursors. J. Neurosci. 33, 14288–14300 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Noble, M., Murray, K., Stroobant, P., Waterfield, M. D. & Riddle, P. Platelet-derived growth factor promotes division and motility and inhibits premature differentiation of the oligodendrocyte/type-2 astrocyte progenitor ceil. Nature 333, 560–562 (1988).

    Article  CAS  PubMed  Google Scholar 

  57. Cooper, G. M. The eukaryotic cell cycle. In The Cell: A Molecular Approach 2nd edn (Sinauer Associates, 2000).

  58. Bellesi, M. et al. Myelin modifications after chronic sleep loss in adolescent mice. Sleep 41, zsy034 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  59. McKenzie, I. A. et al. Motor skill learning requires active central myelination. Science 346, 318–322 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Grant, S. J., Aston-Jones, G. & Redmond, D. E. Responses of primate locus coeruleus neurons to simple and complex sensory stimuli. Brain Res. Bull. 21, 401–410 (1988).

    Article  CAS  PubMed  Google Scholar 

  61. Wang, Y. et al. Accurate quantification of astrocyte and neurotransmitter fluorescence dynamics for single-cell and population-level physiology. Nat. Neurosci. 22, 1936–1944 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Dubbs, A., Guevara, J. & Yuste, R. moco: fast motion correction for calcium imaging. Front. Neuroinform. 10, 6 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Kalman, R. E. A new approach to linear filtering and prediction problems. J. Basic Eng. 82, 35–45 (1960).

    Article  Google Scholar 

  64. Kang, S. H. et al. Degeneration and impaired regeneration of gray matter oligodendrocytes in amyotrophic lateral sclerosis. Nat. Neurosci. 16, 571–579 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wolff, C. et al. Multi-view light-sheet imaging and tracking with the MaMuT software reveals the cell lineage of a direct developing arthropod limb. eLife 7, e34410 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank our colleagues for their support. T. Babola, Y. Wang and G. Yu provided helpful suggestions for analyzing OPC Ca2+ activity. C. Call provided assistance in SCoRe microscopy. D. G. Caro, R. Catenacci and M. Smith provided assistance in OPC live-cell imaging. N. Ye and A. E. Bush helped with mouse husbandry. M. Pucak and A. E. Bush provided assistance with daily operation and maintenance of the microscopes essential to this study. We appreciate the generosity of the SciDraw community, especially L. Petrucco (https://doi.org/10.5281/zenodo.3925903, used in Fig. 1a) and E. Tyler and L. Kravitz (https://doi.org/10.5281/zenodo.3925975, used in Fig. 3a). This study was supported by grants from the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation, the National Institute of Neurological Disorders and Stroke (R01 NS041435) and the National Institute on Aging (R01 AG072305). E.T.H. is supported by the National Science Foundation (2019278189).

Author information

Authors and Affiliations

Authors

Contributions

T.-Y.L. and D.E.B. designed the research and experiments. T.-Y.L. performed and analyzed all the experiments. P.H. contributed to the analysis of the longitudinal OPC Ca2+ activity and helped to perform the OPC proliferation and fate-mapping experiments in vivo. E.T.H. constructed the enforced locomotion rig. A.A. designed and generated the Rosa26-lsl-mGCaMP6s and Rosa26-lsl-GCaMP6s transgenic mouse lines. R.K. performed the bulk RNA sequencing analysis. P.A.C. provided instruments for live-cell imaging and feedback to the manuscript. T.-Y.L., P.H., A.A. and D.E.B. co-wrote and edited the manuscript.

Corresponding author

Correspondence to Dwight E. Bergles.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Kaylene Young and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Expressing membrane-anchored GCaMP6s (mGCaMP6s) in OPCs using Rosa26-lsl-mGCaMP6s knockin transgenic mice.

a, The design of the Rosa26-lsl-mGCaMP6s and Rosa26-lsl-GCaMP6s knockin transgenic mice. MARCKS: the N-terminal myristoylation sequence of myristoylated alanine-rich C-kinase substrate. b, Representative confocal images from 3 independent experiments showing the expression of mGCaMP6s (anti-GFP) in the cortical OPCs (anti-NG2) following immunohistochemistry 4 weeks after tamoxifen injection. c, Quantification of b (n = 3 mice). Error bars represent SEM. d, Representative confocal images from 3 independent experiments showing the expression of cytosolic GCaMP6s (anti-GFP) in the cortical OPCs (anti-NG2) following immunohistochemistry 4 weeks after tamoxifen injection. e, Quantification of d (n = 3 mice). Error bars represent s.e.m. f,g, Representative confocal images from 3 independent experiments showing single mGCaMP6s- (f) and GCaMP6s-expressing (g) OPCs with magnified views of their distal processes (yellow arrowheads) in the dotted squares, respectively. Note the lack of cytosolic GCaMP6s expression in the processes.

Extended Data Fig. 2 Basic properties of OPC Ca2+ events detected by mGCaMP6s and GCaMP6s.

a, Dot plots comparing the average (avg.) event frequency (# of events/min), active area, frequency normalized to active area, event area, amplitude, duration (the time between 50% onset time point – 50% offset time point), rise time (onset duration from 10% to 90% of the peak amplitude) and decay time (offset duration from 90% to 10% of the peak amplitude) between mGCaMP6s- and GCaMP6s-expressing OPCs. Each data dot represents an OPC. n = 6 OPCs from 6 mice. Black filled circles and error bars represent mean ± s.e.m. For data passed the Shapiro-Wilk normality test at the 0.05 level, Student’s t-test (two-sided) was further performed. For data that did not pass the Shapiro-Wilk normality test at the 0.05 level, including avg. event area and avg. rise time, Mann-Whitney test (two-sided) was performed instead to determine if the mean values between mGCaMP6s- and GCaMP6s-expressing OPCs are different or not. b, Quantification of the event origins from mGCaMP6s- and GCaMP6s-expressing OPCs (n = 6 OPCs from 6 mice). Student’s t-test, two-sided.

Extended Data Fig. 3 Propagation of OPC Ca2+ transients is independent of site of event origin, event amplitude, and somatic Ca2+ activity.

a, Average percentage of stationary (having an overall propagation score < 10 µm) and propagating (having an overall propagation score ≥ 10 µm) OPC membrane Ca2+ events (n = 6 OPCs from 6 mice). Black filled circles and error bars represent mean ± s.e.m. Student’s t-test, two-sided. b, A plot of the distance between event origin and soma (Origin from soma) versus overall propagation score. R: Pearson’s r. n = 1,100 propagating events from 6 mice. c, Plotting event amplitude against event overall propagation score. R: Pearson’s r. n = 1,100 propagating events from 6 mice. d, Directions of event propagation 10 s before and after the onset of a soma event. Event travelling direction was determined by total voxels travelled away from soma minus total voxels travelled toward soma. n = 911 propagating events from 6 mice. e, ΔF/F traces of the process events (thin gray lines) peaked 10 s before and after soma event onset in mGCaMP6s-expressing and GCaMP6s-expressing OPCs, respectively. Mean ΔF/F (solid blue and brown lines) is the average ΔF/F of 165 events in the mGCaMP6s-expressing mice (6 cells from 6 mice), and 509 events in the GCaMP6s-expressing mice (6 cells from 6 mice). Shuffled mean (dotted purple lines) is the average value after shuffling ΔF/F values of each event. Shaded areas indicate standard deviation.

Extended Data Fig. 4 Activation of visual cortex by visual stimulation with light does not alter OPC Ca2+ events in vivo.

a, Schematic illustration of the experiment setup. A customized 3D-printed objective shield was used to prevent LED light from entering the objective. The bottom part of the objective shield is not depicted in the illustration in order to display the cranial window. See Methods for details. b, Schematic illustration of the experiment design. Baseline OPC Ca2+ activity was recorded for 60 seconds (s) followed by 3 brief LED stimulations 30 s apart that lasted for 0.1 s each. An infrared (IR) camera was on throughout the experiment to observe mouse behaviors during image acquisition. c, Representative heat maps showing the ΔF/F value and duration of OPC Ca2+ events sorted according to the time of event onset. OPC Ca2+ events that occurred during 10 s of quiescence or 10 s after LED stimulation were overlaid onto a single frame (Maximum projection), respectively. d, Averaging the OPC Ca2+ activity during 20 s of quiescence (gray) and around LED stimulation (blue) suggests that LED stimulation does not influence OPC Ca2+ activity in vivo. Shaded areas represent standard deviation. n = 4 mice. e, Quantification of OPC Ca2+ event frequency, area, duration and amplitude during 10 s of quiescence and 10 s post LED stimulation. n = 8 randomly-selected quiescent periods and 10 LED trials in 4 mice (color-coded). Black filled circles and error bars represent mean ± s.e.m. Student’s t-test, two-sided.

Extended Data Fig. 5 Exposure to carrier (DMSO) does not significantly alter OPC Ca2+ activity.

Quantification of OPC Ca2+ event frequency (# of events/min), area, duration, and amplitude before (Baseline) and 20 minutes after DMSO injection (DMSO). Black filled circles and error bars represent mean ± s.e.m. n = 5 OPCs from five mice each. Paired sample Student’s t-test, two-sided.

Extended Data Fig. 6 α1A adrenergic receptors mRNA is enriched in cortical OPCs relative to pre-myelinating oligodendrocytes.

a, Representative confocal images of an adult B6 visual cortex hybridized with probes recognizing the OPC marker, Pdgfra (green) and Adra1a (red) mRNA. DAPI (blue) stains cell nuclei. Adra1a mRNA is found around Pdgfra+ nuclei, suggesting that cortical OPCs express ADRA1A (n = 19 cells, 3 mice). b, Representative confocal images of an adult B6 visual cortex hybridized with probes recognizing the pre-myelinating oligodendrocyte marker, lncOL1 (green), and Adra1a (red) mRNA. DAPI stains cell nuclei (n = 6 cells, 2 mice). c, Quantification of a and b. Black filled circles and error bars represent mean ± s.e.m.

Extended Data Fig. 7 An example of an mGCaMP6s-expressing OPC undergoing cell death.

The mGCaMP6s-expressing OPC is highlighted in yellow. Note the round-shaped and intensely bright cell body (red arrowhead) as well as the fragmented processes on Day 9. We could not identify any Ca2+ events in the fragmented processes. Representative data from 2 independent experiments.

Extended Data Fig. 8 Local myelin profiles do not change around stable OPCs.

The mGCaMP6s-expressing OPC (highlighted in green) was followed for 16 days and local myelin profile was recorded by SCoRE microscopy concurrently. Local myelin profile remained unchanged from Day 0 (green) to Day 16 (magenta). Representative data from 4 independent experiments that yielded similar results.

Extended Data Fig. 9 Myelinating oligodendrocytes exhibit Ca2+ events in only a select few myelin sheaths.

a, Local calcium events detected (randomly pseudocolored by AQuA) in the same imaging plane where the traced OPC (Fig. 7f, highlighted in blue) became undetectable on Day 0. We did not observe persistent or enhanced Ca2+ events that can be attributed to the pre-myelinating OPC during this stage of maturation. Representative data from 4 independent experiments that yielded similar results. b, Representative confocal images from 3 independent experiments showing the expression of mGCaMP6s (anti-GFP) in the cortical myelinating oligodendrocytes (anti-MBP) using oligodendrocyte-specific and tamoxifen-inducible Cre transgenic line, Mobp-iCreER. c, The magnified views of the dotted squares in b. d, Schematic illustrations of the research design. The expression of mGCaMP6s in myelinating oligodendrocytes was induced between P60–80. Oligodendrocyte Ca2+ activity in the visual cortex of head-fixed, awake mice was observed and recorded using the same condition as the recording of OPC Ca2+ activity (see Fig. 1). e, Representative images showing the Ca2+ activity detected using 2P microscopy (Sum of Ca2+ activity from a 6-minute recording) corresponds to local myelin sheath detected using SCoRe. Cyan arrowhead indicates auto-fluorescent vascular structures. f, Ca2+ events detected in e. g, Distribution of the Ca2+ event numbers detected in myelin sheaths within 6 minutes (n = total 215 sheaths from 3 mice). Note that about 85% of the myelin sheath did not generate any Ca2+ event during the recording. h, Example ΔF/F traces of oligodendrocyte membrane Ca2+ events in f. i, Quantification of average Ca2+ event frequency, size, duration, and amplitude (n = 3 mice). Black filled circles and error bars represent mean ± s.e.m.

Extended Data Fig. 10 PE promotes OPC proliferation in vitro.

a, Gene ontology (GO) terms that were significantly up-regulated (adjusted p value < 0.05) in primary OPCs after 1 hour of PE treatment (20 µM, n = 3 independent biological repeats, differential gene analysis by EdgeR 3.15 with k = 2, adjusted for multiple comparisons). Numbers in the bars indicate the number of genes that were significantly up-regulated after PE treatment within each GO term. If more than 5 GO terms were significantly enriched within the subontology (BP: Biological process; MF: Molecular function. CC: Cellular component), only the top 5 GO terms were shown. b, GO terms that were significantly down-regulated (adjusted p value < 0.05) in a. c, Volcano plot showing differential gene expression in OPCs treated with PE for 1 hour compared to control (no treatment). FDR: false discovery rate. FC: fold change. Total variables: 19,820. d, Representative live cell tracking of primary cultured OPCs for 24 hours after PE treatment (+ PE) and without treatment (Ctrl). OPCs that did not proliferate within 24 hours were labeled in blue. OPCs that proliferated at least once within 24 hours were labeled in red. e, Quantification of d. n = 3 independent biological repeats. Black filled circles and error bars represent mean ± s.e.m. Student’s t-test, two-sided. f, The experimental design of OPC differentiation assay, and representative confocal images of the OPC/oligodendrocytes mixed cultures 2 days after PDGF-AA withdrawal (Day 4) in the absence (Ctrl) or with the presence of PE (+PE). Green arrow indicates an example of fully differentiated oligodendrocytes with strong MBP expression (MBPS). White arrow indicates an example of differentiating OPCs that have weak expression of both NG2 and MBP (NG2WMBPW). Magenta arrow indicates an example of OPCs that remain undifferentiated with strong NG2 expression. g, Quantification of f. n = 3 independent biological repeats. Black filled circles and error bars represent mean ± s.e.m. Student’s t-test, two-sided.

Supplementary information

Supplementary Information

Supplementary Fig. 1, Supplementary Video Legend 1, Supplementary Video Legend 2, Supplementary Video Legend 3, Supplementary Video Legend 4 and Supplementary Video Legend 5

Reporting Summary

Supplementary Video 1

Supplementary Video 1. Cytosolic OPC Ca2+ activity in the visual cortex in vivo. Left, OPC Ca2+ activity detected by 2P microscopy; right, output video from AQuA with randomly pseudo-colored Ca2+ events (5× speed).

Supplementary Video 2

Supplementary Video 2. Membrane OPC Ca2+ activity in the visual cortex in vivo. Left, OPC Ca2+ activity detected by 2P microscopy; right, output video from AQuA with randomly pseudo-colored Ca2+ events (5× speed).

Supplementary Video 3

Supplementary Video 3. Enforced locomotion stimulates OPC Ca2+ activity in the mouse visual cortex. The red dot indicates when the platter began to rotate (movie played at 5× speed).

Supplementary Video 4

Supplementary Video 4. PE evokes Ca2+ influx in OPCs in acute cortical slices. PE was superfused ~3 min after the recording begins (movie shown at 50× speed).

Supplementary Video 5

Supplementary Video 5. Myelinating oligodendrocytes exhibit infrequent Ca2+ activity in the visual cortex in vivo. Left, oligodendrocyte near membrane Ca2+ activity detected by 2P microscopy; right, output video from AQuA with randomly pseudo-colored Ca2+ events (5× speed).

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, TY., Hanumaihgari, P., Hsu, E.T. et al. Norepinephrine modulates calcium dynamics in cortical oligodendrocyte precursor cells promoting proliferation during arousal in mice. Nat Neurosci 26, 1739–1750 (2023). https://doi.org/10.1038/s41593-023-01426-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-023-01426-0

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