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:

Early growth response 2 in the mPFC regulates mouse social and cooperative behaviors

Abstract

Adolescent social neglect impairs social performance, but the underlying molecular mechanisms remain unclear. Here we report that isolation rearing of juvenile mice caused cooperation defects that were rescued by immediate social reintroduction. We also identified the transcription factor early growth response 2 (Egr2) in the medial prefrontal cortex (mPFC) as a major target of social isolation and resocialization. Isolation rearing increased corticosteroid production, which reduced the expression of Egr2 in the mPFC, including in oligodendrocytes. Overexpressing Egr2 ubiquitously in the mPFC, but not specifically in neurons nor in oligodendroglia, protected mice from the isolation rearing-induced cooperation defect. In addition to synapse integrity, Egr2 also regulated the development of oligodendroglia, specifically the transition from undifferentiated oligodendrocyte precursor cells to premyelinating oligodendrocytes. In conclusion, this study reveals the importance of mPFC Egr2 in the cooperative behavior that is modulated by social experience, and its unexpected role in oligodendrocyte development.

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: The effect of isolation rearing on social interaction and cooperation ability of mice.
Fig. 2: Identification of potential genes regulating cooperative behavior.
Fig. 3: Knocking down Egr2 impairs mice’s sociability, cooperation behaviors and myelination.
Fig. 4: Overexpressing Egr2 in the mPFC of SI mice mitigates sociability and cooperation defects and restores synaptic and myelination impairments.
Fig. 5: Specifically overexpressing Egr2 in oligodendrocytes in the mPFC of SI mice restored the social interaction but not cooperation performance.
Fig. 6: Elevated corticosterone level in SI mice potentially downregulates Egr2 expression.
Fig. 7: Egr2 is expressed in early- and late-stage oligodendrocytes and Egr2 knockdown dampens oligodendrocyte differentiation.

Similar content being viewed by others

Data availability

The authors declare that all data generated or analyzed during this study are included in this published article and Supplementary Information, or available from the authors upon reasonable request. Source data are provided with this paper.

References

  1. Ioannou, C. C., Guttal, V. & Couzin, I. D. Predatory fish select for coordinated collective motion in virtual prey. Science 337, 1212–1215 (2012).

    Article  CAS  Google Scholar 

  2. Kanter, J. W., Manos, R. C., Busch, A. M. & Rusch, L. C. Making behavioral activation more behavioral. Behav. Modif. 32, 780–803 (2008).

    Article  Google Scholar 

  3. Segrin, C. Social skills deficits associated with depression. Clin. Psychol. Rev. 20, 379–403 (2000).

    Article  CAS  Google Scholar 

  4. Lai, M. C., Lombardo, M. V. & Baron-Cohen, S. Autism. Lancet 383, 896–910 (2014).

    Article  Google Scholar 

  5. Lahera, G. et al. Hostile attributions in bipolar disorder and schizophrenia contribute to poor social functioning. Acta Psychiatr. Scand. 131, 472–482 (2015).

    Article  CAS  Google Scholar 

  6. Thaler, N. S., Allen, D. N., Sutton, G. P., Vertinski, M. & Ringdahl, E. N. Differential impairment of social cognition factors in bipolar disorder with and without psychotic features and schizophrenia. J. Psychiatr. Res. 47, 2004–2010 (2013).

    Article  Google Scholar 

  7. Strikwerda-Brown, C. et al. The interplay of emotional and social conceptual processes during moral reasoning in frontotemporal dementia. Brain 144, 938–952 (2021).

    Article  Google Scholar 

  8. Ko, J. Neuroanatomical substrates of rodent social behavior: the medial prefrontal cortex and its projection patterns. Front. Neural Circuits https://doi.org/10.3389/fncir.2017.00041 (2017).

  9. Chugani, H. T. et al. Local brain functional activity following early deprivation: a study of postinstitutionalized romanian orphans. NeuroImage 14, 1290–1301 (2001).

    Article  CAS  Google Scholar 

  10. Eluvathingal, T. J. et al. Abnormal brain connectivity in children after early severe socioemotional deprivation: a diffusion tensor imaging study. Pediatrics 117, 2093–2100 (2006).

    Article  Google Scholar 

  11. Xin, W. & Chan, J. R. Myelin plasticity: sculpting circuits in learning and memory. Nat. Rev. Neurosci. 21, 682–694 (2020).

    Article  CAS  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).

    Article  CAS  Google Scholar 

  13. Mount, C. W. & Monje, M. Wrapped to adapt: experience-dependent myelination. Neuron 95, 743–756 (2017).

    Article  CAS  Google Scholar 

  14. 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).

    Article  CAS  Google Scholar 

  15. Liu, J. et al. Impaired adult myelination in the prefrontal cortex of socially isolated mice. Nat. Neurosci. 15, 1621–1623 (2012).

    Article  CAS  Google Scholar 

  16. 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).

    Article  CAS  Google Scholar 

  17. Noë, R. Cooperation experiments: coordination through communication versus acting apart together. Anim. Behav. 71, 1–18 (2006).

    Article  Google Scholar 

  18. Sokolowski, M. B. Social interactions in ‘simple’ model systems. Neuron 65, 780–794 (2010).

    Article  CAS  Google Scholar 

  19. Martinez-Moreno, M. et al. Regulation of peripheral myelination through transcriptional buffering of Egr2 by an antisense long non-coding RNA. Cell Rep. 20, 1950–1963 (2017).

    Article  CAS  Google Scholar 

  20. Feng, W., et al. A water-reward task assay for evaluating mouse mutualistic cooperative behavior. Preprint at bioRxiv https://doi.org/10.1101/2021.02.06.430037 (2021).

  21. Skelly, M. J., Chappell, A. E., Carter, E. & Weiner, J. L. Adolescent social isolation increases anxiety-like behavior and ethanol intake and impairs fear extinction in adulthood: possible role of disrupted noradrenergic signaling. Neuropharmacology 97, 149–159 (2015).

    Article  CAS  Google Scholar 

  22. Quan, M. N., Tian, Y. T., Xu, K. H., Zhang, T. & Yang, Z. Post weaning social isolation influences spatial cognition, prefrontal cortical synaptic plasticity and hippocampal potassium ion channels in Wistar rats. Neuroscience 169, 214–222 (2010).

    Article  CAS  Google Scholar 

  23. Cao, M. et al. Early enriched physical environment reverses impairments of the hippocampus, but not medial prefrontal cortex, of socially-isolated mice. Brain Behav. Immun. 64, 232–243 (2017).

    Article  Google Scholar 

  24. Karasek, M., Swiltoslawski, J. & Zieliniska, A. Ultrastructure of the central nervous system: the basics. Folia Neuropathol. 42, 1–9 (2004).

    Google Scholar 

  25. De, S. & Turman, J. E. Jr. Krox-20 gene expression: influencing hindbrain–craniofacial developmental interactions. Arch. Histol. Cytol. 68, 227–234 (2005).

    Article  CAS  Google Scholar 

  26. Svaren, J. & Meijer, D. The molecular machinery of myelin gene transcription in Schwann cells. Glia 56, 1541–1551 (2008).

    Article  Google Scholar 

  27. Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014.e1022 (2018).

    Article  CAS  Google Scholar 

  28. Floriddia, E. M. et al. Distinct oligodendrocyte populations have spatial preference and different responses to spinal cord injury. Nat. Commun. 11, 5860 (2020).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Mittelstadt, P. R. & Ashwell, J. D. Inhibition of AP-1 by the glucocorticoid-inducible protein GILZ *. J. Biol. Chem. 276, 29603–29610 (2001).

    Article  CAS  Google Scholar 

  31. Snyder, J. S., Soumier, A., Brewer, M., Pickel, J. & Cameron, H. A. Adult hippocampal neurogenesis buffers stress responses and depressive behaviour. Nature 476, 458–461 (2011).

    Article  CAS  Google Scholar 

  32. Choi, G. E. et al. BNIP3L/NIX-mediated mitophagy protects against glucocorticoid-induced synapse defects. Nat. Commun. 12, 487 (2021).

    Article  CAS  Google Scholar 

  33. Wang, Z., Frederick, J. & Garabedian, M. J. Deciphering the phosphorylation ‘code’ of the glucocorticoid receptor in vivo. J. Biol. Chem. 277, 26573–26580 (2002).

    Article  CAS  Google Scholar 

  34. Marin-Husstege, M. et al. Multiple roles of Id4 in developmental myelination: predicted outcomes and unexpected findings. Glia 54, 285–296 (2006).

    Article  Google Scholar 

  35. Qi, Y. C. et al. Control of oligodendrocyte differentiation by the Nkx2.2 homeodomain transcription factor. Development 128, 2723–2733 (2001).

    Article  CAS  Google Scholar 

  36. Bujalka, H., et al. MYRF is a membrane-associated transcription factor that autoproteolytically cleaves to directly activate myelin genes. PLoS Biol. https://doi.org/10.1371/journal.pbio.1001625 (2013).

  37. Shin, J. & Ko, J. Protocol for quantitative assessment of social cooperation in mice. STAR Protoc. 2, 100305 (2021).

    Article  Google Scholar 

  38. Conde-Moro, A. R., Rocha-Almeida, F., Sánchez-Campusano, R., Delgado-García, J. M. & Gruart, A. The activity of the prelimbic cortex in rats is enhanced during the cooperative acquisition of an instrumental learning task. Prog. Neurobiol. 183, 101692 (2019).

    Article  Google Scholar 

  39. Zhang, Y. et al. miR-124 regulates early isolation-induced social abnormalities via inhibiting myelinogenesis in the medial prefrontal cortex. Cell. Mol. Life Sci. 79, 507 (2022).

    Article  CAS  Google Scholar 

  40. Wang, Z. et al. Miconazole promotes cooperative ability of a mouse model of Alzheimer’s disease. Int. J. Neuropsychopharmacol. https://doi.org/10.1093/ijnp/pyac061 (2022).

  41. Sock, E. & Wegner, M. Transcriptional control of myelination and remyelination. Glia 67, 2153–2165 (2019).

    Article  Google Scholar 

  42. Nagarajan, R. et al. EGR2 mutations in inherited neuropathies dominant-negatively inhibit myelin gene expression. Neuron 30, 355–368 (2001).

    Article  CAS  Google Scholar 

  43. Parkinson, D. B. et al. Krox-20 inhibits Jun-NH2-terminal kinase/c-Jun to control Schwann cell proliferation and death. J. Cell Biol. 164, 385–394 (2004).

    Article  CAS  Google Scholar 

  44. Zawadzka, M. et al. CNS-resident glial progenitor/stem cells produce Schwann cells as well as oligodendrocytes during repair of CNS demyelination. Cell Stem Cell 6, 578–590 (2010).

    Article  CAS  Google Scholar 

  45. Xie, X.-M. et al. Co-transplantation of MRF-overexpressing oligodendrocyte precursor cells and Schwann cells promotes recovery in rat after spinal cord injury. Neurobiol. Dis. 94, 196–204 (2016).

    Article  CAS  Google Scholar 

  46. Dubois, J. et al. The early development of brain white matter: a review of imaging studies in fetuses, newborns and infants. Neuroscience 276, 48–71 (2014).

    Article  CAS  Google Scholar 

  47. Lee, S. L., Tourtellotte, L. C., Wesselschmidt, R. L. & Milbrandt, J. Growth and differentiation proceeds normally in cells deficient in the immediate early gene NGFI-A. J. Biol. Chem. 270, 9971–9977 (1995).

    Article  CAS  Google Scholar 

  48. Williams, J. et al. Krox20 may play a key role in the stabilization of long-term potentiation. Brain Res. Mol. Brain Res. 28, 87–93 (1995).

    Article  CAS  Google Scholar 

  49. Raffety, B. D., Smith, R. E. & Ptacek, J. T. Facilitating and debilitating trait anxiety, situational anxiety, and coping with an anticipated stressor: a process analysis. J. Pers. Soc. Psychol. 72, 892–906 (1997).

    Article  CAS  Google Scholar 

  50. Schneier, F. R. Social anxiety disorder. Brit. Med. J. 327, 515–516 (2003).

    Article  Google Scholar 

  51. Stein, M. B. & Kean, Y. M. Disability and quality of life in social phobia: epidemiologic findings. Am. J. Psychiatry 157, 1606–1613 (2000).

    Article  CAS  Google Scholar 

  52. Doremus-Fitzwater, T. L., Varlinskaya, E. I. & Spear, L. P. Social and non-social anxiety in adolescent and adult rats after repeated restraint. Physiol. Behav. 97, 484–494 (2009).

    Article  CAS  Google Scholar 

  53. Leclerc, N. et al. Opposing effects of glucocorticoids and Wnt signaling on Krox20 and mineral deposition in osteoblast cultures. J. Cell. Biochem. 103, 1938–1951 (2008).

    Article  CAS  Google Scholar 

  54. Zalewska, K., et al. Corticosterone administration alters white matter tract structure and reduces gliosis in the sub-acute phase of experimental stroke. Int. J. Mol. Sci. https://doi.org/10.3390/ijms22136693 (2021).

  55. Alonso, G. Prolonged corticosterone treatment of adult rats inhibits the proliferation of oligodendrocyte progenitors present throughout white and gray matter regions of the brain. Glia 31, 219–231 (2000).

    Article  CAS  Google Scholar 

  56. Bhat, R. V., Worley, P. F., Cole, A. J. & Baraban, J. M. Activation of the zinc finger encoding gene krox-20 in adult rat brain: comparison with zif268. Brain Res. Mol. Brain Res. 13, 263–266 (1992).

    Article  CAS  Google Scholar 

  57. Beckmann, A. M., Davidson, M. S., Goodenough, S. & Wilce, P. A. Differential expression of Egr-1-like DNA-binding activities in the naive rat brain and after excitatory stimulation. J. Neurochem. 69, 2227–2237 (1997).

    Article  CAS  Google Scholar 

  58. Zelikowsky, M. et al. The neuropeptide Tac2 controls a distributed brain state induced by chronic social isolation stress. Cell 173, 1265–1279 e1219 (2018).

    Article  CAS  Google Scholar 

  59. Felix-Ortiz, A. C. & Tye, K. M. Amygdala inputs to the ventral hippocampus bidirectionally modulate social behavior. J. Neurosci. 34, 586–595 (2014).

    Article  CAS  Google Scholar 

  60. Leung, C., Kim, J. C. & Jia, Z. Three-chamber social approach task with optogenetic stimulation (mice). Bio Protoc. 8, e3120 (2018).

    Article  CAS  Google Scholar 

  61. Wang, L. et al. Deep cervical lymph node ligation aggravates AD-like pathology of APP/PS1 mice. Brain Pathol. 29, 176–192 (2019).

    Article  CAS  Google Scholar 

  62. Gao, J. et al. A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature 466, 1105–1109 (2010).

    Article  CAS  Google Scholar 

  63. Lei, Y. et al. SIRT1 in forebrain excitatory neurons produces sexually dimorphic effects on depression-related behaviors and modulates neuronal excitability and synaptic transmission in the medial prefrontal cortex. Mol. Psychiatry 25, 1094–1111 (2020).

    Article  CAS  Google Scholar 

  64. Flores-Obando, R. E., Freidin, M. M. & Abrams, C. K. Rapid and specific immunomagnetic isolation of mouse primary oligodendrocytes. J. Vis. Exp. https://doi.org/10.3791/57543 (2018).

  65. Kinn Rod, A. M., Harkestad, N., Jellestad, F. K. & Murison, R. Comparison of commercial ELISA assays for quantification of corticosterone in serum. Sci Rep. 7, 6748 (2017).

    Article  Google Scholar 

  66. Xu, Z. et al. Deletion of aquaporin-4 in APP/PS1 mice exacerbates brain Aβ accumulation and memory deficits. Mol. Neurodegener. 10, 58 (2015).

    Article  CAS  Google Scholar 

  67. Castejon, O.J. Low resolution scanning electron microscopy of cerebellar neurons and neuroglial cells of the granular layer. Scan Electron Microsc. 1391–1400 (1984).

  68. Castejon, O. J., Castejon, H. V. & Castellano, A. Oligodendroglial cell damage and demyelination in infant hydrocephalus. An electron microscopic study. J. Submicrosc. Cytol. Pathol. 33, 33–40 (2001).

    CAS  Google Scholar 

  69. Garcia-Cabezas, M.A., John, Y.J., Barbas, H. & Zikopoulos, B. Distinction of neurons, glia and endothelial cells in the cerebral cortex: an algorithm based on cytological features. Front. Neuroanat. https://doi.org/10.3389/fnana.2016.00107 (2016).

  70. Jones, D. G. & Devon, R. M. An ultrastructural study into the effects of pentobarbitone on synaptic organization. Brain Res. 147, 47–63 (1978).

    Article  CAS  Google Scholar 

  71. Flygt, J. et al. Neutralization of interleukin-1β following diffuse traumatic brain injury in the mouse attenuates the loss of mature oligodendrocytes. J. Neurotrauma 35, 2837–2849 (2018).

    Article  Google Scholar 

Download references

Acknowledgements

We thank L. Zhu (Tongji Medical College of HUST) and Q. Zhou (Nanjing Medical University) for their insightful comments. We also thank Z. Xia for technical assistance with the EM and Y. Zhang for technical assistance with the oligodendrocyte culture. This work was supported by the grants from the National Natural Science Foundation of China (81801378 and 81871117).

Author information

Authors and Affiliations

Authors

Contributions

M.X. and C.S. designed the experiments. Y. Zhang and W.F. performed most of the experiments and analysis. Y. Zhang, W.F., Z.W., Y.P., Y.J., S.C., S.D., T.W., Y. Zou, P.S., Y.C., H.H. and H.F. performed the behavioral experiments. The manuscript was written by M.X., C.S., Y. Zhang and W.F. All authors discussed the results, made the figures and edited the manuscript. M.X., C.S. and H.H. directed and supervised all aspects of the study.

Corresponding authors

Correspondence to Chengyu Sheng or Ming Xiao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Lab Animal thanks the anonymous reviewers 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 The effect of SI on mouse locomotion, exploration, spatial working memory, anxiety-like behaviors, and fear memory.

Postnatal day 21 mice were housed singly or in groups for 3 weeks before the behavioral tests. a, Locomotion, and exploration behavior of GH and SI mice at P56 in the open field. b, The spatial working memory performance of P57 GH and SI mice in the Y-maze testing. c, The anxiety-like behavior of P58 GH and SI mice examined by the elevated plus-maze test. d, Fear memory performance of GH and SI mice at P58-59 in the fear conditioning test. Data are represented as medians and IQR (n = 18 in each group). Data were analyzed by two-tailed Student’s t-tests. GH: group housing; SI: social isolation.

Source data

Extended Data Fig. 2 The sociability, cooperation, locomotion, exploration, spatial working memory, anxiety-like behaviors, and fear memory defects of SI mice are rescued by resocialization.

a, Diagram showing the timeline of the experiment, including weanling, social isolation, resocialization, behavioral tests (shown in b-i), and then qPCR (shown in Fig. 2j), Western blot (shown in Fig. 2k, l and Extended Data Fig. 3g,h), EM (shown in Extended Data Fig. 3a-d), immunohistochemistry (shown in Extended Data Fig. 3e,f), immunofluorescence (shown in Fig. 6i,j and Supplementary Fig. 3a, b), or ELISA analysis (shown in Fig. 6e). b, c, The social behavior performance and social memory of mice in social test. d, e, Graphs showing the time before the first drinking (drinking latency), the times of drinking (drinking number), and the total time spent on drinking (drinking time) each day in the training (d) and testing (e) periods. f, Locomotion, and exploration behavior in the open field. g, The anxiety-like behavior examined by the elevated plus-maze test. h, The spatial working memory performance in the Y-maze testing. i, Fear memory performance in the fear conditioning test. (GH: n = 15 mice; GH-SI: n = 15 mice; SI: n = 14 mice); Data are represented as medians and IQR in c, f-i; and mean + SEM in d and e (n = 7 pairs of mice per group). Data in c and f-i were analyzed by One-way ANOVA followed by Tukey’s post hoc test; and data in d and e were analyzed by Repeated Measures ANOVA with post hoc Student-Newman-Keuls test. GH: group housing; SI: social isolation; GH-SI: resocialization of SI mice with GH mice.

Source data

Extended Data Fig. 3 The mPFC impairment of SI mice is rescued by resocialization.

a, Representative EM images showing myelin (the top panel), oligodendrocyte nuclear heterochromatin (the middle panel), and synaptic morphology (the bottom panel) in the mPFC, respectively. Scale bar, 500 nm. b, The scatter plots of g ratios with linear least squares fitting (g ratio vs. axon caliber) in GH (106 axons), GH-SI (107axons), and SI (110 axons) mice (n = 4 per group). c, Graph showing the area percentage of nuclear heterochromatin in GH (17 nuclei), GH-SI (19 nuclei), and SI (15) mice (n = 4 per group). d, Postsynaptic density thickness, synaptic cleft width, presynaptic active zones length, and synaptic curvature in GH (70 synapses), GH-SI (68 synapses) and SI (63 synapses) mice (n = 4 per group). e-h Representative immunostaining images (e) and Western blot bands (g) showing MBP and PSD-95 expression in the mPFC of GH, GH-SI, and SI mice; corresponding quantification analyses are shown in f for e (n = 4 per group) and h for g (n = 8); scale bar, 50 µm. Data are represented as mean ± SEM in c and d; and as medians and IQR in f and h. Data in b-h were analyzed by One-way ANOVA followed by Tukey’s post hoc test. GH: group housing (P87); SI: social isolation (P87); GH-SI: resocialization of SI mice with GH mice (P87).

Source data

Extended Data Fig. 4 Knocking down Egr2 in the mPFC of GH mice does not affect exploring activities, spatial working memory, anxiety-like behavior, and fear memory.

a, Graphs showing the time before the first drinking (drinking latency), the times of drinking (drinking number), and the total time spent on drinking (drinking time) each day in different groups at P46-57 in the 7 consecutive training days. b, Locomotion, and exploration behavior in different groups at P58 in the open field. c, The anxiety-like extent examined in different groups at P60 by the elevated plus-maze test. d, The spatial working memory performance of mice at P59 in the Y-maze testing. e, Fear memory performance of mice at P60-61 in fear conditioning test. Data are represented as mean + SEM in a; and as medians and IQR in b-e (n = 20 per group). Data in a were analyzed by repeated-measures ANOVA with post hoc Student-Newman-Keuls test; and in be by Two-way ANOVA followed by Tukey’s post hoc test. GH-GFP: GH mice with AAV-GFP virus injection; GH-Egr2-RNAi: GH mice with AAV-Egr2-RNAi virus injection; SI-GFP: SI mice with AAV-GFP virus injection; SI-Egr2-RNAi: SI mice with AAV-Egr2-RNAi virus injection.

Source data

Extended Data Fig. 5 Overexpression of Egr2 in the mPFC of SI mice mitigates abnormal anxiety-like behavior, but not exploring activities, spatial working memory and fear memory.

a, Graphs showing the time before the first drinking (drinking latency), the times of drinking (drinking number), and the total time spent on drinking (drinking time) each day in different groups at P46-57 in the 7 consecutive training days. b, Locomotion and exploration behavior of P58 mice were tested in the open field. c, The anxiety-like behavior of P60 mice was examined in the elevated plus-maze test. d, The spatial working memory performance of P59 mice was tested in the Y-maze test. e, Fear memory performance in different groups at P60-61 in fear conditioning test. Data are represented as mean + SEM in a; and as medians and IQR in b-e (data from 18 mice in GH-GFP group, 22 mice in GH-Egr2 group, 30 mice in SI-GFP group and 28 mice in SI-Egr2 group, respectively). Data in a were analyzed by repeated-measures ANOVA with post hoc Student-Newman-Keuls test; and data in b-e were analyzed by Two-way ANOVA followed by Tukey’s post hoc test. GH-GFP: GH mice with AAV-GFP virus injection; GH-Egr2: GH mice with AAV-Egr2 virus injection; SI-GFP: SI mice with AAV-GFP virus injection; SI-Egr2: SI mice with AAV-Egr2 virus injection.

Source data

Extended Data Fig. 6 Specifically overexpressing Egr2 in oligodendrocytes in the mPFC of SI mice restores myelin damage, but has no effect on synaptic damage.

a, b, Representative immunostaining images and quantification analysis of Egr2 expression in the mPFC of GH-CNP-GFP, GH-CNP-Egr2, SI-CNP-GFP and SI-CNP-Egr2 mice (n = 6). Scale bar, 50 µm. c, d, Western blot and quantification analysis showing Egr2 protein levels in the mPFC of GH-CNP-GFP, GH-CNP-Egr2, SI-CNP-GFP and SI-CNP-Egr2 mice (n = 5-6). e, f, Immunostaining images and quantification analysis of MBP and PSD-95 expression in the mPFC (n = 6 mice per group). Scale bar, 50 µm. g-i, Western blot and quantification analysis of MBP and PSD-95 expression in the mPFC of GH-CNP-GFP, GH-CNP-Egr2, SI-CNP-GFP, and SI-CNP-Egr2 mice (n = 5-6). Data are represented as medians and IQR, and analyzed by Two-way ANOVA followed by Tukey’s post hoc test. GH-CNP-GFP: GH mice with AAV-CNP-GFP virus injection (P61); GH-CNP-Egr2: GH mice with AAV-CNP-Egr2 virus injection (P61); SI-CNP-GFP: SI mice with AAV-CNP-GFP virus injection (P61); SI-CNP-Egr2: SI mice with AAV-CNP-Egr2 virus injection (P61).

Source data

Extended Data Fig. 7 Overexpression of Egr2 in oligodendrocytes in the mPFC of SI mice mitigates abnormal anxiety-like behavior, but has no effect on fear memory, exploring activities and spatial working memory.

a, Graphs showing the time before the first drinking (drinking latency), the times of drinking (drinking number), and the total time spent on drinking (drinking time) each day in different groups at P46-57 in the 7 consecutive training days. b, Locomotion and exploration behavior of P58 mice were tested in the open field. c, The anxiety-like behavior was examined in P60 mice using the elevated plus-maze test. d, The spatial working memory performance in each group of P59 mice was tested in the Y-maze test. e, Fear memory performance of P60-61 mice was tested using the fear conditioning test. Data are represented as mean + SEM in a; and as medians and IQR in b-e (data from 20 mice in GH-CNP-GFP group, 16 mice in GH-CNP-Egr2 group, 16 mice in SI-CNP-GFP group and 16 mice in SI-CNP-Egr2 group, respectively). Data in a were analyzed by repeated-measures ANOVA with post hoc Student-Newman-Keuls test; and data in b-e were analyzed by Two-way ANOVA followed by Tukey’s post hoc test. GH-CNP-GFP: GH mice with AAV-CNP-GFP virus injection, GH-CNP-Egr2: GH mice with AAV-CNP-Egr2 virus injection, SI-CNP-GFP: SI mice with AAV-CNP-GFP virus injection, SI-CNP-Egr2: SI mice with AAV-CNP-Egr2 virus injection.

Source data

Extended Data Fig. 8 Specifically overexpressing Egr2 in neurons in the mPFC of SI mice has no effect on sociability or cooperation performance.

a, Diagram showing the timeline of the experiment, including weanling, AAV virus injection, group housing or social isolation, and then behavioral tests (shown in Extended Data Fig. 8b–f and Extended Data Fig. 9a-d), and immunofluorescence, EM, immunohistochemistry or Western blot analysis (shown in Extended Data Fig. 8a and Extended Data Fig. 10a–h). b, Representative immunofluorescence images showing GFP expression in some Tuj1- or Neun-labeled neurons, but not in NG2- or PDGFRα-labeled oligodendrocyte of P61 mice in the mPFC. Scale bar, 50 µm. c, d, Heat maps and graphs showing the sociability performance and social memory in the three-chamber test. e, Graphs showing the time before the first drinking (drinking latency), the times of drinking (drinking number), and the total time spent on drinking (drinking time) each day in different groups in the 7 consecutive training days. f, hSyn-Egr2 in the mPFC of SI mice did not reverse their cooperation performance, as assessed by drinking latency, drinking number, and drinking time. Data are represented as medians and IQR in c and d (16 mice per group); and mean + SEM in e and f (8 pairs of mice per group). Data in c and d were analyzed by two-way ANOVA followed by Tukey’s post hoc test; and data in e and f were analyzed by repeated-measures ANOVA with post hoc Student-Newman-Keuls test. GH-hSyn-GFP: GH mice with AAV-hSyn-GFP virus injection; GH-hSyn-Egr2: GH mice with AAV-hSyn-Egr2 virus injection; SI-hSyn-GFP: SI mice with AAV-hSyn-GFP virus injection; SI-hSyn-Egr2: SI mice with AAV-hSyn-Egr2 virus injection.

Source data

Extended Data Fig. 9 Specifically overexpressing Egr2 in neurons in the mPFC of SI mice mitigates abnormal anxiety-like behavior, but has no effect on fear memory, exploring activities and spatial working memory.

a, Locomotion, and exploration behavior were tested at P58 for each group of mice in the open field. b, The anxiety-like behavior was examined in P60 mice by the elevated plus-maze test. c, The spatial working memory performance was tested at P59 for each group mice in the Y-maze testing. d, Fear memory performance of P60-61 mice in the fear conditioning test. Data are represented as medians and IQR from 16 mice in every group, and analyzed by two-way ANOVA followed by Tukey’s post hoc test. GH-hSyn-GFP: GH mice with AAV-hSyn-GFP virus injection; GH-hSyn-Egr2: GH mice with AAV-hSyn-Egr2 virus injection; SI-hSyn-GFP: SI mice with AAV-hSyn-GFP virus injection; SI-hSyn-Egr2: SI mice with AAV-hSyn-Egr2 virus injection.

Source data

Extended Data Fig. 10 Specifically overexpressing Egr2 in neurons ameliorates impairments of synapses but not myelination in the mPFC of SI mice.

a, Representative EM images showing myelin (in the top panel), oligodendrocyte nuclear heterochromatin (in the middle panel) and synaptic morphology (in the bottom panel) in the mPFC, respectively. Scale bar, 500 nm. b, Scatter plot of g ratios with linear least squares fitting in GH-hSyn-GFP (175 axons), GH-hSyn-Egr2 (171 axons), SI-hSyn-GFP (193 axons), and SI-hSyn-Egr2 (157 axons) mice (n = 5 per group). c, Graphs showing altered percentage of oligodendrocyte nuclear heterochromatin in GH-hSyn-GFP (27 nuclei), GH-hSyn-Egr2 (30 nuclei), SI-hSyn-GFP (25 nuclei), and SI-hSyn-Egr2 (29 nuclei) mice (n = 5 per group). d, Graphs showing quantified analysis of postsynaptic dense, synaptic cleft width, length of the active zones and synaptic curvature in GH-hSyn-GFP (102 synapses), GH-hSyn-Egr2 (106 synapses), SI-hSyn-GFP (138 synapses), and SI-hSyn-Egr2 (125 synapses) mice (n = 5 per group). e-h, Immunohistology (e, f) and Western blot (g, h) showing MBP and PSD-95 expression in the mPFC (n = 6 for each group). Scale bar, 50 µm. Data are represented as mean ± SEM in c and d; and medians and IQR in f-h. Data in b-d and f-h were analyzed by two-way ANOVA followed by Tukey’s post hoc test. GH-hSyn-GFP: GH mice with AAV-hSyn-GFP virus injection (P61); GH-hSyn-Egr2: GH mice with AAV-hSyn-Egr2 virus injection (P61); SI-hSyn-GFP: SI mice with AAV-hSyn-GFP virus injection (P61); SI-hSyn-Egr2: SI mice with AAV-hSyn-Egr2 virus injection mice (P61).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–19.

Reporting Summary

Supplementary Video 1

Social cooperation training.

Supplementary Video 2

Social cooperation behavior.

Supplementary Video 3

Social uncooperation behavior.

Supplementary Table 1

PCR primers.

Supplementary Data 1

Statistical data for Supplementary Fig. 1.

Supplementary Data 2

Statistical data for Supplementary Fig. 3.

Supplementary Data 3

Statistical data for Supplementary Fig. 4.

Supplementary Data 4

Statistical data for Supplementary Fig. 5.

Supplementary Data 5

Statistical data for Supplementary Fig. 6.

Supplementary Data 6

Statistical data for Supplementary Fig. 7.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

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

Zhang, Y., Feng, W., Wang, Z. et al. Early growth response 2 in the mPFC regulates mouse social and cooperative behaviors. Lab Anim 52, 37–50 (2023). https://doi.org/10.1038/s41684-022-01090-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41684-022-01090-0

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