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:

Preservation of a remote fear memory requires new myelin formation

Abstract

Experience-dependent myelination is hypothesized to shape neural circuit function and subsequent behavioral output. Using a contextual fear memory task in mice, we demonstrate that fear learning induces oligodendrocyte precursor cells to proliferate and differentiate into myelinating oligodendrocytes in the medial prefrontal cortex. Transgenic animals that cannot form new myelin exhibit deficient remote, but not recent, fear memory recall. Recording population calcium dynamics by fiber photometry, we observe that the neuronal response to conditioned context cues evolves over time in the medial prefrontal cortex, but not in animals that cannot form new myelin. Finally, we demonstrate that pharmacological induction of new myelin formation with clemastine fumarate improves remote memory recall and promotes fear generalization. Thus, bidirectional manipulation of myelin plasticity functionally affects behavior and neurophysiology, which suggests that neural activity during fear learning instructs the formation of new myelin, which in turn supports the consolidation and/or retrieval of remote fear memories.

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

Fig. 1: Fear learning induces OPC proliferation in the mPFC.
Fig. 2: Fear learning experience induces OPC maturation into myelinating OLs in the mPFC.
Fig. 3: OPC maturation into myelinating OLs occurs over several weeks in adult gray matter.
Fig. 4: Inhibition of new myelin formation impairs remote fear memory recall.
Fig. 5: Immediate early gene expression following remote recall is impaired in the absence of new myelin formation.
Fig. 6: Prefrontal population calcium dynamics in the mPFC are altered in the absence of new myelin formation.
Fig. 7: Induction of new myelin formation preserves remote fear memory recall.
Fig. 8: Induction of new myelin formation increases immediate early gene expression following remote fear memory recall.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

Code availability

Custom Matlab software written for extraction, analyses and visualization of photometry signals is available as Supplementary Software 13 and online at https://github.com/sp808/Fiber-photometry.

References

  1. Hill, R., Patel, K., Goncalves, C., Grutzendler, J. & Nishiyama, A. Modulation of oligodendrocyte generation during a critical temporal window after NG2 cell division. Nat. Neurosci. 17, 1518–1527 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  5. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Young, K. et al. Oligodendrocyte dynamics in the healthy adult CNS: evidence for myelin remodeling. Neuron 77, 873–885 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  9. Gibson, E. et al. Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 344, 1252304 (2014).

    PubMed  PubMed Central  Google Scholar 

  10. Mitew, S. et al. Pharmacogenetic stimulation of neuronal activity increases myelination in an axon-specific manner. Nat. Commun. 9, 306 (2018).

    PubMed  PubMed Central  Google Scholar 

  11. Geraghty, A. et al. Loss of adaptive myelination contributes to methotrexate chemotherapy-related cognitive impairment. Neuron 103, 250–265 (2019).

    CAS  PubMed  Google Scholar 

  12. Etxeberria, A. et al. Dynamic modulation of myelination in response to visual stimuli alters optic nerve conduction velocity. J. Neurosci. 36, 6937–6948 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Makinodan, M., Rosen, K. M., Ito, S. & Corfas, G. A critical period for social experience-dependent oligodendrocyte maturation and myelination. Science 337, 1357–1360 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Mangin, J. M., Li, P., Scafidi, J. & Gallo, V. Experience-dependent regulation of NG2 progenitors in the developing barrel cortex. Nat. Neurosci. 15, 1192–1194 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Liu, J. et al. Impaired adult myelination in the prefrontal cortex of socially isolated mice. Nat. Neurosci. 36, 957–962 (2012).

    Google Scholar 

  17. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Tripathi, R. B. et al. Remarkable stability of myelinating oligodendrocytes in mice. Cell Rep. 21, 316–323 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Pan, S. & Chan, J. R. Regulation and dysregulation of axon infrastructure by myelinating glia. J. Cell Biol. 216, 3903–3916 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. de Hoz, L. & Simons, M. The emerging functions of oligodendrocytes in regulating neuronal network behaviour. BioEssays 37, 60–69 (2015).

    PubMed  Google Scholar 

  21. Shalev, A., Liberzon, I. & Marmar, C. Post-traumatic stress disorder. N. Engl. J. Med. 376, 2459–2469 (2017).

    PubMed  Google Scholar 

  22. Tovote, P., Fadok, J. P. & Lüthi, A. Neuronal circuits for fear and anxiety. Nat. Rev. Neurosci. 16, 317–331 (2015).

    CAS  PubMed  Google Scholar 

  23. Cowansage, K. K. et al. Direct reactivation of a coherent neocortical memory of context. Neuron 84, 432–441 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. DeNardo, L. A. et al. Temporal evolution of cortical ensembles promoting remote memory retrieval. Nat. Neurosci. 22, 460–469 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kitamura, T. et al. Engrams and circuits crucial for systems consolidation of a memory. Science 356, 73–78 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Tanaka, K. Z. Cortical representations are reinstated by the hippocampus during memory retrieval. Neuron 84, 347–354 (2014).

    CAS  PubMed  Google Scholar 

  27. Vetere, G. et al. Chemogenetic interrogation of a brain-wide fear memory network in mice. Neuron 94, 363–374 (2017).

    CAS  PubMed  Google Scholar 

  28. Frankland, P. W. & Bontempi, B. The organization of recent and remote memories. Nat. Rev. Neurosci. 6, 119–130 (2005).

    CAS  PubMed  Google Scholar 

  29. Tonegawa, S., Morrissey, M. & Kitamura, T. The role of engram cells in the systems consolidation of memory. Nat. Rev. Neurosci. 19, 485–498 (2018).

    CAS  PubMed  Google Scholar 

  30. Emery, B. et al. Myelin gene regulatory factor is a critical transcriptional regulator required for CNS myelination. Cell 138, 172–185 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Giustino, T. F., Fitzgerald, P. J. & Maren, S. Fear expression suppresses medial prefrontal cortical firing in rats. PLoS One 11, e0165256 (2016).

    PubMed  PubMed Central  Google Scholar 

  32. Halladay, L. R. & Blair, H. T. Distinct ensembles of medial prefrontal cortex neurons are activated by threatening stimuli that elicit excitation vs. inhibition of movement. J. Neurophysiol. 114, 793–807 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Mei, F. et al. Micropillar arrays as a high-throughput screening platform for therapeutics in multiple sclerosis. Nat. Med. 20, 954–960 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Mei, F. et al. Accelerated remyelination during inflammatory demyelination prevents axonal loss and improves functional recovery. eLife 5, e18246 (2016).

    PubMed  PubMed Central  Google Scholar 

  35. Green, A. J., Gelfand, J. M., Cree, B. A. & Lancet, B.-C. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): a randomised, controlled, double-blind, crossover trial. Lancet 390, 2481–2489 (2017).

    CAS  PubMed  Google Scholar 

  36. Liu, J. et al. Clemastine enhances myelination in the prefrontal cortex and rescues behavioral changes in socially isolated mice. J. Neurosci. 36, 957–962 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Wang, F. et al. Enhancing oligodendrocyte myelination rescues synaptic loss and improves functional recovery after chronic hypoxia. Neuron 99, 689–701 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Guo, N. et al. Dentate granule cell recruitment of feedforward inhibition governs engram maintenance and remote memory generalization. Nat. Med. 24, 438–449 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Freeman, S. A. et al. Acceleration of conduction velocity linked to clustering of nodal components precedes myelination. Proc. Natl Acad. Sci. USA 112, 321–328 (2015).

    Google Scholar 

  40. Fünfschilling, U. et al. Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 485, 517–522 (2012).

    PubMed  PubMed Central  Google Scholar 

  41. Lee, Y. et al. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature 487, 443–448 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Xin, W. et al. Oligodendrocytes support neuronal glutamatergic transmission via expression of glutamine synthetase. Cell Rep. 27, 2262–2271 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Frankland, P. W., Bontempi, B., Talton, L. E., Kaczmarek, L. & Silva, A. J. The involvement of the anterior cingulate cortex in remote contextual fear memory. Science 304, 881–883 (2004).

    CAS  PubMed  Google Scholar 

  44. Maviel, T., Durkin, T., Menzaghi, F. & Bontempi, B. Sites of neocortical reorganization critical for remote spatial memory. Science 305, 96–99 (2004).

    CAS  PubMed  Google Scholar 

  45. Benchenane, K. et al. Coherent theta oscillations and reorganization of spike timing in the hippocampal–prefrontal network upon learning. Neuron 66, 921–936 (2010).

    CAS  PubMed  Google Scholar 

  46. Karalis, N. et al. 4-Hz oscillations synchronize prefrontal–amygdala circuits during fear behavior. Nat. Neurosci. 19, 605–612 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Xia, F. et al. Parvalbumin-positive interneurons mediate neocortical–hippocampal interactions that are necessary for memory consolidation. eLife 6, e27868 (2017).

    PubMed  PubMed Central  Google Scholar 

  48. Pajevic, S., Basser, P. J. & Fields, R. D. Role of myelin plasticity in oscillations and synchrony of neuronal activity. Neuroscience 276, 135–147 (2014).

    CAS  PubMed  Google Scholar 

  49. Steadman, P. E. et al. Disruption of oligodendrogenesis impairs memory consolidation in adult mice. Neuron https://doi.org/10.1016/j.neuron.2019.10.013 (2019).

  50. Chao, L., Tosun, D., Woodward, S., Kaufer, D. & Neylan, T. Preliminary evidence of increased hippocampal myelin content in veterans with posttraumatic stress disorder. Front. Behav. Neurosci. 9, 333 (2015).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to K. Hoi for providing reagents, insightful discussion, technical assistance and comments on the manuscript. We also thank members of the Chan and Kheirbek laboratories for technical assistance and scientific discussion, and V. Sohal, S. Pleasure, K. Poskanzer and A. Molofsky for insightful discussions. This work was supported by the National Institutes of Health/National Institute of Neurological Disorders and Stroke (R01NS062796, R01NS097428 and R01NS095889), the Adelson Medical Research Foundation (ANDP grant A130141) and the Rachleff Family Endowment to J.R.C., and the NIMH (R01 MH108623, R01 MH111754 and R01 MH117961), a Weill Scholar Award, the Pew Charitable Trusts, the Esther A. and Joseph Klingenstein Fund, and an IMHRO/One Mind Rising Star Award to M.A.K.

Author information

Authors and Affiliations

Authors

Contributions

S.P. conceptualized the project. S.P. designed and performed all the experiments. M.A.K. and J.R.C. provided technical expertise, funding and advice. S.P., M.A.K. and J.R.C. drafted and edited the manuscript. S.P. wrote all of the code for photometry signal processing and analyses, performed the quantification and data analyses and prepared the figures. S.R.M. prepared the electron microscopy samples and performed image quantification. H.S.C. assisted with histological preparations and quantification.

Corresponding authors

Correspondence to Jonah R. Chan or Mazen A. Kheirbek.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Ekaterina Likhtik, Stephen Maren, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Proliferation, differentiation, and maturation of OPCs following fear learning.

a, EdU+ cell density of home cage (HC), fear conditioned (CFC), no shock (NS), and immediate shock (IS) animals 24 hours post-conditioning; unpaired two-tailed t-tests comparing HC vs. CFC for PL (difference: 2.060 ± 0.8565, 95% CI: 0.1942 to 3.927, t12 = 2.406, p = 0.0332), IL (difference: 2.239 ± 0.8566, 95% CI: 0.3731 to 4.106, t12 = 2.614, p = 0.0226), ACC (difference: 1.176 ± 0.5811, 95% CI: -0.09067 to 2.442, t12 = 2.406, p = 0.0760), BLA (difference: 3.865 ± 1.395, 95% CI: 0.8506 to 6.880, t12 = 2.77, p = 0.0159), dHPC (difference: 2.060 ± 0.8565, 95% CI: 0.1942 to 3.927, t12 = 2.406, p = 0.6694), vHPC (difference: 0.2300 ± 0.5265, 95% CI: -0.9075 to 1.368, t12 = 0.4369, p = 0.3128), paired two-tailed t-test comparing PL vs. IL for CFC animals (difference: -0.1025 ± 0.7635, 95% CI: -1.971 to 1.766, t6 = 0.1343, p = 0.8975). b, EdU+/Olig2+ cell density at 24 hours post-conditioning; unpaired two-tailed t-tests comparing HC vs. CFC for PL (difference: 1.801 ± 0.7408, 95% CI: 0.1867 to 3.415, t12 = 2.431, p = 0.0317), IL (difference: 1.547 ± 0.6297, 95% CI: 0.1750 to 2.919, t12 = 2.457, p = 0.0302), ACC (difference: 0.2161 ± 0.5518, 95% CI: -0.9863 to 1.418, t12 = 0.3916, p = 0.7022), BLA (difference: 3.619 ± 1.338, 95% CI: 0.7295 to 6.509, t12 = 2.706, p = 0.018), dHPC (difference: 0.2882 ± 0.4868, 95% CI: -0.7634 to 1.340, t12 = 0.592, p = 0.564), vHPC (difference: -0.008014 ± 0.5297, 95% CI: -1.162 to 1.146, t12 = 0.01513, p = 0.9882), paired two-tailed t-test comparing PL vs. IL for CFC animals (difference: -0.2322 ± 0.4552, 95% CI: -1.346 to 0.8817, t12 = 0.5102, p = 0.6289). For (a-b), n = 7 mice (HC), 8 mice (CFC), 7 mice (NS), 8 mice (IS). c, EdU+/ASPA+ density post-conditioning; unpaired two-tailed t-tests HC vs. CFC for PL (difference: 3.576 ± 1.006, 95% CI: 1.404 to 5.749, t13 = 3.556, p = 0.035), IL (difference: 2.472 ± 0.7097, 95% CI: 0.9503 to 3.995, t13 = 0.5102, p = 0.037), ACC (difference 0.9596 ± 0.5516, 95% CI: -0.2234 to 2.143, t13 = 0.5102, p = 0.1038), BLA (difference: 1.520 ± 2.071, 95% CI: -3.165 to 6.204, t13 = 0.7339, p = 0.4817), dHPC (difference: 0.3704 ± 0.5950, 95% CI: -0.9150 to 1.656, t13 = 0.6225, p = 0.5444), vHPC (difference: 0.0536 ± 0.6227, 95% CI: -1.303 to 1.410, t13 = 0.086, p = 0.9328), paired two-tailed t-test comparing PL vs. IL for CFC animals (difference: -0.9171 ± 0.7719, 95% CI: -2.742 to 0.9081, t13 = 1.188, p = 0.5466). d, EdU+/Olig2+ density 30 days post-conditioning; unpaired two-tailed t-tests HC vs. CFC for PL (difference: 4.512 ± 5.558, 95% CI: -7.599 to 16.62, t13 = 0.8117, p = 0.4328), IL (difference: 4.514 ± 6.705, 95% CI: -10.10 to 19.12, t13 = 0.6731, p = 0.5136), ACC (difference: 3.086 ± 6.324, 95% CI: -10.69 to 16.86, t13 = 0.488, p = 0.6343), BLA (difference: 19.00 ± 11.32, 95% CI: -5.452 to 43.45, t13 = 1.679, p = 0.1171), dHPC (difference: 4.325 ± 2.933, 95% CI: -2.011 to 10.66, t13 = 1.475, p = 0.1641), vHPC (difference: -0.8542 ± 3.854, 95% CI: -9.251 to 7.543, t13 = 0.2216, p = 0.8283), paired two-tailed t-test comparing PL vs. IL for CFC animals (difference: -4.630 ± 6.858, 95% CI: -21.41 to 12.15, t13 = 0.6751, p = 0.5248). For (c-d), n = 7 mice (HC) and 8 mice (CFC). Quantification of GFP+/MBP+ (e), GFP+/MBP (f), and EdU+/ASPA+ (g) cell density in the dHPC and BLA; n = 6 mice (7 days), 9 mice (14 days), and 7 mice (30 days). e-g One-way ANOVA with Sidak’s post hoc tests comparing cell densities across days; (e) BLA: F2,19 = 4.369, p = 0.0275, 7 vs. 14 day, (difference: -3.760 ± 1.672, 95% CI: -7.818 to 0.2981, p = 0.0718), 7 vs. 30 day (difference: -5.041 ± 1.765, 95% CI: -9.324 to -0.7572, p = 0.0201), dHPC: F2,22 = 5.397, p = 0.0124, 7 vs. 14 day, (difference: -3.108 ± 1.241, 95% CI: -6.086 to -0.1300, p = 0.0399), 7 vs. 30 day (difference: -4.124 ± 1.303, 95% CI: -7.251 to -0.9968, p = 0.009); (f) BLA: F2,19 = 0.07572, p = 0.9274, 7 vs. 14 day, (difference: 2.354 ± 6.908, 95% CI: -14.41 to 19.12, p = 0.9308), 7 vs. 30 day (difference: 2.540 ± 7.292, 95% CI: -15.16 to 20.24, p = 0.9279), dHPC: F2,20 = 0.4852, p = 0.6226, 7 vs. 14 day, (difference: 1.629 ± 3.759, 95% CI: -7.456 to 10.71, p = 0.8907), 7 vs. 30 day (difference: -1.900 ± 4.049, 95% CI: -11.69 to 7.888, p = 0.8733); (g) BLA: F2,18 = 47.49, p < 0.0001, 7 vs. 14 day, (difference: -0.6155 ± 0.6763, 95% CI: -2.265 to 1.034, p = 0.6092), 7 vs. 30 day (difference: -5.632 ± 0.6565, 95% CI: -7.233 to -4.031, p < 0.0001), dHPC: F2,16 = 46.74, p < 0.0001, 7 vs. 14 day, (difference: -0.03272 ± 0.7020, 95% CI: -1.764 to 1.699, p = 0.9987), 7 vs. 30 day (difference: -6.038 ± 0.7285, 95% CI: -7.835 to -4.242, p < 0.0001). h, Proportion of GFP+/MBP+ cells that are ASPA+ (left bars) and the proportion of ASPA+/GFP+ cells that are MBP+ (right bars) at 30 days post-conditioning; n = 7 mice. Representative DAPI (gray) images the sub-regional dHPC (i) and amygdalar (j) cytoarchitecture, scale bars: 250 μm (k) Schematic of the sampled area within the mPFC (green rectangle); dashed lines demarcate the approximate region cut for electron microscopy. For box-and-whisker plots, the center, boxes, and whiskers represent the median, interquartile range, and the 10th and 90th percentiles, with asterisks indicating the following p-value ranges: * ≤ 0.05, ** ≤ 0.01, *** ≤ 0.001, **** ≤ 0.0001.

Extended Data Fig. 2 Overall myelination status and microglial density are unchanged in the absence of new myelin formation.

a, Representative images of ASPA (green) staining in the PFC of cre-negative and Myrf icKO mice 30 days post-conditioning, quantified in (b); unpaired two-tailed t-test, difference: -26.50 ± 26.20, 95% CI: -81.15 to 28.15, t20 = 1.012, p = 0.3238, n = 11 mice per genotype. c, Representative images of MBP (gray) staining in the PFC of cre-negative and Myrf icKO mice 30 days post-conditioning, quantified in (d); unpaired two-tailed t-test, difference: 0.1927 ± 12.13, 95% CI: -25.12 to 25.50, t20 = 2.406, p = 0.9875, n = 11 mice per genotype. e, Representative images of Iba1 (magenta) staining in the PFC of cre-negative and Myrf icKO mice 30 days post-conditioning, quantified in (f); unpaired two-tailed t-test, difference: -3.698 ± 29.55, 95% CI: -69.53 to 62.14, t10 = 0.1251, p = 0.9029, n = 6 mice per genotype. Scale bars: 100 μm (a,c,e). Data are presented as mean ± SEM.

Extended Data Fig. 3 Inhibition of new myelin formation impairs remote fear memory recall.

a, Extended experimental timeline for cohort represented in Fig. 4a,e, quantified in (e); two-way ANOVA (F4,80 = 3.437, p = 0.0121) with Sidak’s post hoc tests comparing cre-negative vs. Myrf icKO during conditioning (difference: -2.416 ± 5.790, 95% CI: -17.58 to 12.74, p = 0.9965), 24 hour (difference: -5.169 ± 5.790, 95% CI: -20.33 to 9.991, p = 0.9040), 7 day (difference: -9.901 ± 5.790, 95% CI: -25.06 to 5.260, p = 0.9040), 21 day (difference: -12.35 ± 5.790, 95% CI: -27.51 to 2.811, p = 0.3772), and 30 day (difference: -24.37 ± 5.790, 95% CI: -39.53 to -9.209, p = 0.0003), and comparing 7 day (difference: 5.685 ± 5.685, 95% CI: -6.004 to 17.37, p = 0.6277), 21 day (difference: 17.05 ± 17.05, 95% CI: 5.361 to 28.74, p = 0.0015), and 30 day (difference: 26.18 ± 26.18, 95% CI: 14.49 to 37.87, p < 0.0001) to 24 hour freezing within Myrf icKO animals, n = 11 animals per genotype. b, Extended experimental timeline for cohort represented in Fig. 4b,f,g, quantified in (f); two-way ANOVA (F4,80 = 1.435, p = 0.2291) with Sidak’s post hoc tests comparing cre-negative vs. Myrf icKO during conditioning (difference 1.467 ± 7.398, 95% CI: -13.19 to 16.13, p = 0.8431), 24 hour (difference: 0.9199 ± 7.398, 95% CI: -13.74 to 15.58, p = 0.9013), 7 day (difference: -1.946 ± 7.398, 95% CI: -16.61 to 12.71, p = 0.7930), 21 day (difference: 6.165 ± 7.398, 95% CI: -8.496 to 20.83, p = 0.4064), and 30 day (difference: 18.43 ± 7.398, 95% CI: 3.767 to 33.09, p = 0.0142), n = 14 cre-negative and 10 Myrf icKO mice. c, Experimental timeline for cohort in which tamoxifen was administered after 24 hour recall, quantified in (h); two-way ANOVA (F4,72 = 1.724, p = 0.1541), n = 10 animals per genotype. d, Experimental timeline for cohort in which tamoxifen was administered seven weeks prior to conditioning, quantified in (h); two-way ANOVA (genotype, F1,26 = 0.03642, p = 0.8501) with Sidak’s post hoc tests comparing cre-negative vs. Myrf icKO during conditioning (difference: 5.241 ± 6.245, 95% CI: -9.579 to 20.06, p = 0.6507) and 24 hour (difference: 3.556 ± 6.245, 95% CI: -11.26 to 18.37, p = 0.8185), n = 8 cre-negative and 7 Myrf icKO mice. i, Expanded quantification of Fos+ cell density across brain regions following 30-day retrieval sessions; unpaired two-tailed t-tests, PL (difference: 62.14 ± 24.56, 95% CI: 10.54 to 113.7, t21 = 72.53, p = 0.0209), IL (difference: 65.70 ± 28.64, 95% CI: 5.532 to 125.9, t21 = 2.294, p = 0.034), ACC (difference: 99.84 ± 21.22, 95% CI: 55.27 to 144.4, t21 = 4.706, p = 0.0002), BA (difference: 80.55 ± 24.87, 95% CI: 28.66 to 132.4, t21 = 3.238, p = 0.0041), LA (difference: 6.494 ± 9.138, 95% CI: -12.70 to 25.69, t21 = 0.7107, p = 0.4864), DG (difference: 163.5 ± 55.29, 95% CI: 48.50 to 278.5, t21 = 2.957, p = 0.0075), CA3 (difference: 175.4 ± 63.83, 95% CI: 42.70 to 308.2, t21 = 2.749, p = 0.012), NR (difference: 84.08 ± 22.50, 95% CI: 35.83 to 132.3, t21 = 3.737, p = 0.0022), vDG (difference: 88.20 ± 32.23, 95% CI: 18.58 to 157.8, t21 = 2.737, p = 0.017), vCA3 (difference: 55.39 ± 24.33, 95% CI: 2.831 to 108.0, t21 = 7.361, p = 0.0404), vPAG (difference: 105.9 ± 18.71, 95% CI: 66.04 to 145.8, t21 = 5.662, p < 0.0001), SCC (difference: 21.86 ± 53.48, 95% CI: -89.35 to 133.1, t21 = 0.4088, p = 0.6868), n = 12 cre-negative and 11 Myrf icKO mice. j, Within-genotype comparisons of Fos+ cell density between the PL and IL; paired two-tailed t-tests, cre-negative (difference: 35.68 ± 33.66, 95% CI: -40.47 to 111.8, t9 = 0.4088, p = 0.3168), Myrf icKO (difference: 3.320 ± 21.65, 95% CI: -45.65 to 52.29, t9 = 0.4088, p = 0.8815), n = 10 mice per genotype. Within-genotype comparisons of freezing responses between males and females for animals represented in a-e for cre-negative (k) and Myrf icKO animals (l); two-way ANOVA for cre-negative (F4,44 = 0.6815, p = 0.6085) and Myrf icKO animals (F4,44 = 1.649, p = 0.1833), n = 8 males and 5 females for cre-negative mice, n = 5 males and 6 females for Myrf icKO mice. Data are presented as mean ± SEM, with asterisks indicating the following p-value ranges: * ≤ 0.05, ** ≤ 0.01, *** ≤ 0.001, **** ≤ 0.0001.

Extended Data Fig. 4 Innate anxiety-like and locomotor behaviors are unchanged in the absence of new myelin formation.

Quantification of time spent in the closed arms of the elevated plus maze (EPM) (a) and the periphery of the open field test (OFT) (b); unpaired two-tailed t-test, EPM (difference: -16.38 ± 12.29, 95% CI: -42.59 to 9.818, t15 = 1.333, p = 0.2025), OFT (difference: 20.94 ± 22.43, 95% CI: -27.17 to 69.05, t15 = 0.9334, p = 0.3664). (c) Quantification of total distance traveled in the open field test; unpaired two-tailed t-test (difference: 788.3 ± 780.0, 95% CI: -884.6 to 2461, t15 = 1.011, p = 0.3293). Heatmaps representing relative frequency (red - frequent, blue - infrequent) of cre-negative and Myrf icKO animals in the elevated plus maze (d) (dashed lines demarcate the closed arms), and open field test (e). Data are presented as mean ± SEM. For a-c, n = 5 cre-negative and 11 Myrf icKO mice.

Extended Data Fig. 5 Supplementary data for fiber photometry experiments.

a, Schematic of fiber photometry recording setup. b, Representative trace of a recorded retrieval session depicting the raw GCaMP (blue) and isobestic (black) signal, with no detectable motion artifact. Quantification of the z-scored mean ΔF/F during the two seconds pre- and post-bout transition at 24 hours and 30 days for control (c) and Myrf icKO (d) animals; n = 10 bouts across 7 mice (controls) and 7 mice (Myrf icKO); paired two-tailed t-tests comparing z-scored signal intensity pre- and post-bout for control 24 hour (difference: -0.2016 ± 0.1564, 95% CI: -0.5095 to 0.1064, t138 = 1.289, p = 0.0202), control 30 day (difference 0.1051 ± 0.1118, 95% CI: -0.1150 to 0.3251, t138 = 0.9396, p = 0.055), Myrf icKO 24 hour (difference: -0.2216 ± 0.1650, 95% CI: -0.5494 to 0.1063, t138 = 1.343, p < 0.0001), and Myrf icKO 30 day (difference: -0.1837 ± 0.1502, 95% CI: -0.4810 to 0.1136, t138 = 1.223, p = 0.0032). e, Quantification of the mean ΔF/F over a five-minute home cage recording taken just prior to the 24-hour and 30-day retrieval sessions; n = 7 (controls) and 7 mice (Myrf icKO); unpaired two-tailed t-tests, 24 hour (difference: -0.0001284 ± 0.006472, 95% CI: -0.01423 to 0.01397, t13 = 0.01984, p = 0.6534), 30 day (difference: 0.004054 ± 0.01353, 95% CI: -0.03350 to 0.04161, t13 = 0.9334, p = 0.8892). f, Representative image of Iba1 staining under the implant site, quantified in (g); paired two-tailed t-test (difference: -32.40 ± 26.10, 95% CI: -92.58 to 27.78, t16 = 0.908, p = 0.2496), n = 9 mice. Scale bar: 100 μm. For box-and-whisker plots, the center, boxes, and whiskers represent the median, interquartile range, and the 10th and 90th percentiles, with asterisks indicating the following p-value ranges: * ≤ 0.05, ** ≤ 0.01, **** ≤ 0.0001.

Extended Data Fig. 6 Induction of new myelin formation preserves remote fear memory recall.

a, Extended experimental timeline for cohort represented in Fig. 7a,e. b, Freezing responses in the conditioning context; two-way ANOVA (F4,108 = 5.884, p = 0.0003) with Sidak’s post hoc tests for conditioning (difference: 0.8941 ± 4.116, 95% CI: -9.860 to 11.65, p = 0.9928), 24 hour (difference: -0.8004 ± 4.116, 95% CI: -11.55 to 9.954, p = 0.9948), 7 day (difference: -11.91 ± 4.116, 95% CI: -22.67 to -1.160, p = 0.0221), 21 day (difference: -16.77 ± 4.116, 95% CI: -27.52 to -6.013, p = 0.004), and 30 day (difference: -17.49 ± 4.116, 95% CI: -16.00 to -0.3832, p = 0.0002), n = 15 vehicle and 14 clemastine mice. c, Freezing responses in the similar context; two-way ANOVA (F1,27 = 8.277, p = 0.0077) with Sidak’s post hoc tests for 7 day (difference: 0.4158 ± 3.894, 95% CI: -7.391 to 8.222, p = 0.9154) and 21 day (difference: -8.190 ± 3.894, 95% CI: -28.24 to -6.733, p = 0.401), or across days within vehicle- (difference: -5.845 ± 2.078, 95% CI: -10.77 to -0.9248, p = 0.018) and clemastine-treated (difference: -14.45 ± 2.151, 95% CI: -19.54 to -9.357, p < 0.0001) animals, n = 15 vehicle and 14 clemastine mice. d, Experiment assessing the effects of continuous vehicle injections/handling, compared against home cage animals; two-way ANOVA (F4,36 = 9.926, p < 0.0001) with Sidak’s post hoc tests for conditioning (difference: -0.6013 ± 5.615, 95% CI: -15.66 to 14.46, p > 0.9999), 24 hour (difference: 0.2373 ± 5.615, 95% CI: -14.82 to 15.30, p > 0.9999), 7 day (difference: 22.20 ± 5.615, 95% CI: 7.137 to 37.25, p = 0.0014), 21 day (difference: 31.96 ± 5.615, 95% CI: 16.91 to 47.02, p < 0.0001), and 30 day (difference: 26.78 ± 5.615, 95% CI: 11.72 to 41.83, p < 0.0001), n = 5 mice per treatment group. e, Expanded quantification of Fos+ cell density following 30-day retrieval sessions; unpaired two-tailed t-tests, PL (difference: 279.0 ± 43.19, 95% CI: 188.6 to 369.4, t19 = 6.459, p < 0.0001), IL (difference: 198.6 ± 68.81, 95% CI: 49.90 to 347.2, t19 = 2.886, p = 0.274), ACC (difference: 127.4 ± 51.52, 95% CI: 18.66 to 236.1, t19 = 2.472, p = 0.0069), BA (difference: 86.21 ± 19.27, 95% CI: 45.87 to 126.5, t19 = 4.473, p = 0.0044), LA (difference: 128.6 ± 44.43, 95% CI: 35.89 to 221.2, t19 = 2.894, p = 0.009), DG (difference: 268.7 ± 56.69, 95% CI: 150.0 to 387.3, t19 = 4.739, (p < 0.0001), CA3 (difference: 270.5 ± 77.34, 95% CI: 109.2 to 431.8, t19 = 3.497, p =0.0023), NR (difference: 110.7 ± 34.65, 95% CI: 37.59 to 183.8, t19 = 3.195, p = 0.0026), vDG (difference: 153.9 ± 34.77, 95% CI: 80.51 to 227.2, t19 = 4.426, p = 0.0013), vCA3 (difference: 139.2 ± 36.88, 95% CI: 61.42 to 217.0, t19 = 3.775, p = 0.0019), vPAG (difference: 193.0 ± 48.67, 95% CI: 90.28 to 295.6, t19 = 3.965, p = 0.0017), SSC (difference: 79.25 ± 55.73, 95% CI: -38.33 to 196.8, t19 = 1.422, p = 0.1731), n = 13 vehicle and 8 clemastine mice. f, Within-treatment group comparisons of Fos+ cell density between the PL and IL; unpaired two-tailed t-tests, vehicle (difference: 80.43 ± 48.18, 95% CI: -20.78 to 181.6, t18 = 1.67, p = 0.3657), n = 13 PL- and 7 IL-sampled mice, clemastine (difference: -24.05 ± 65.62, 95% CI: -162.5 to 114.4, t17 = 0.3666, p = 0.7185), n = 8 PL- and 11 IL-sampled mice. Freezing responses for experimental timeline described in (a) for home cage (g), no shock (h), and immediate shock (i) animals; (g) two-way ANOVA (F3,24 = 2.217, p = 0.1122) (h) two-way ANOVA (F4,32 = 2.674, p = 0.0496) (i) two-way ANOVA (F4,32 = 0.7811, p = 0.5496); for g-I, n = 5 animals per condition. j, Extended experimental timeline for clemastine injections in Myrf icKO animals. k, Individual fear expression for vehicle- and clemastine-treated Myrf icKO animals represented in (j); two-way ANOVA (F4,48 =1.357, p = 0.2629), n = 6 vehicle and 8 clemastine mice. l, Quantification of the z-scored mean ΔF/F during pre- and post-bout transitions at 30 days for vehicle- (left) and clemastine-treated (right) animals; paired two-tailed t-tests comparing pre- and post-transition for vehicle (difference: 1.075 ± 0.1560, 95% CI: 0.7685 to 1.382, t78 = 6.894, p < 0.0001) and clemastine (difference: 0.8792 ± 0.1548, 95% CI: 0.5745 to 1.184, t78 = 5.678, p < 0.0001). m, Quantification of the z-scored mean ΔF/F of the pre- and post-bout transition for vehicle- and clemastine-treated animals; unpaired two-tailed t-test (difference: -0.1963 ± 0.2099, 95% CI: -0.6094 to 0.2168, t78 = 0.9352, p = 0.3504). For l-m, n = 10 bouts per animal, 4 animals per treatment group. For box-and-whisker plots, the center, boxes, and whiskers represent the median, interquartile range, and the 10th and 90th percentiles. For dot plots, data are presented as mean ± SEM, with asterisks indicating the following p-value ranges: * ≤ 0.05, ** ≤ 0.01, *** ≤ 0.001, **** ≤ 0.0001.

Supplementary information

Reporting Summary

Supplementary Software 1

Subject management, used in conjunction with photometry_f_cond.m and photometry_f_v2.m.

Supplementary Software 2

Master function for conditioning session analyses.

Supplementary Software 3

Master function for retrieval session analyses.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pan, S., Mayoral, S.R., Choi, H.S. et al. Preservation of a remote fear memory requires new myelin formation. Nat Neurosci 23, 487–499 (2020). https://doi.org/10.1038/s41593-019-0582-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-019-0582-1

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