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:

Dynamics of social representation in the mouse prefrontal cortex

An Author Correction to this article was published on 03 March 2020

This article has been updated

Abstract

The prefrontal cortex (PFC) plays an important role in regulating social functions in mammals, and its dysfunction has been linked to social deficits in neurodevelopmental disorders. Yet little is known of how the PFC encodes social information and how social representations may be altered in such disorders. Here, we show that neurons in the medial PFC of freely behaving male mice preferentially respond to socially relevant olfactory cues. Population activity patterns in this region differed between social and nonsocial stimuli and underwent experience-dependent refinement. In mice lacking the autism-associated gene Cntnap2, both the categorization of sensory stimuli and the refinement of social representations were impaired. Noise levels in spontaneous population activity were higher in Cntnap2 knockouts and correlated with the degree to which social representations were disrupted. Our findings elucidate the encoding of social sensory cues in the medial PFC and provide a link between altered prefrontal dynamics and autism-associated social dysfunction.

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: Experimental setup for presentation of social and nonsocial olfactory cues to freely behaving mice.
Fig. 2: Social tuning in mPFC unit response.
Fig. 3: Distinct representation of social cues in the mPFC neuronal population code.
Fig. 4: Cntnap2–/– mice display impaired social behavior, but intact olfaction.
Fig. 5: Altered dynamics of social representation in the Cntnap2–/– mouse model of autism.
Fig. 6: Decoding of stimulus identity and social category from mPFC population code.
Fig. 7: Elevated neural noise correlates with deficits in social processing in Cntnap2–/ mice.

Similar content being viewed by others

Data availability

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

Code availability

The custom written analysis codes are available from the corresponding author upon reasonable request.

Change history

  • 03 March 2020

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Anderson, D. J. Circuit modules linking internal states and social behaviour in flies and mice. Nat. Rev. Neurosci. 17, 692–704 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Chen, P. & Hong, W. Neural circuit mechanisms of social behavior. Neuron 98, 16–30 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Euston, D. R., Gruber, A. J. & McNaughton, B. L. The role of medial prefrontal cortex in memory and decision making. Neuron 76, 1057–1070 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Adolphs, R. The social brain: neural basis of social knowledge. Annu. Rev. Psychol. 60, 693–716 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Bicks, L. K., Koike, H., Akbarian, S. & Morishita, H. Prefrontal cortex and social cognition in mouse and man. Front. Psychol. 6, 1–15 (2015).

    Article  Google Scholar 

  6. Watson, K. K. & Platt, M. L. Social signals in primate orbitofrontal cortex. Curr. Biol. 22, 2268–2273 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhou, T. et al. History of winning remodels thalamo–PFC circuit to reinforce social dominance. Science 357, 162–168 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Murugan, M. et al. Combined social and spatial coding in a descending projection from the prefrontal cortex. Cell 171, 1663–1677.e16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lee, E. et al. Enhanced neuronal activity in the medial prefrontal cortex during social approach behavior. J. Neurosci. 36, 6926–6936 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rigotti, M. et al. The importance of mixed selectivity in complex cognitive tasks. Nature 497, 585–590 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Parthasarathy, A. et al. Mixed selectivity morphs population codes in prefrontal cortex. Nat. Neurosci. 20, 1770–1779 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Zikopoulos, B. & Barbas, H. Changes in prefrontal axons may disrupt the network in autism. J. Neurosci. 30, 14595–14609 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Oblak, A., Gibbs, T. T. T. & Blatt, G. J. J. Decreased GABAA receptors and benzodiazepine binding sites in the anterior cingulate cortex in autism. Autism Res. 2, 205–219 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Carper, R. A. & Courchesne, E. Localized enlargement of the frontal cortex in early autism. Biol. Psychiatry 57, 126–133 (2005).

    Article  PubMed  Google Scholar 

  15. Rubenstein, J. L. R. & Merzenich, M. M. Model of autism: increased ratio of excitation/inhibition in key neural systems. Genes Brain Behav. 2, 255–267 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lazaro, M. T. et al. Reduced prefrontal synaptic connectivity and disturbed oscillatory population dynamics in the CNTNAP2 model of autism. Cell Rep. 27, 2567–2578.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Antoine, M. W., Langberg, T., Schnepel, P. & Feldman, D. E. Increased excitation–inhibition ratio stabilizes synapse and circuit excitability in four autism mouse models. Neuron 101, 648–661.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rubin, R., Abbott, L. F. & Sompolinsky, H. Balanced excitation and inhibition are required for high-capacity, noise-robust neuronal selectivity. Proc. Natl Acad. Sci. USA 114, E9366–E9375 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Scott-Van Zeeland, A. A. et al. Altered functional connectivity in frontal lobe circuits is associated with variation in the autism risk gene CNTNAP2. Sci. Transl Med. 2, 56ra80 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Baum, S. H., Stevenson, R. A. & Wallace, M. T. Behavioral, perceptual, and neural alterations in sensory and multisensory function in autism spectrum disorder. Prog. Neurobiol. 134, 140–160 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Dinstein, I. et al. Unreliable evoked responses in autism. Neuron 75, 981–991 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Peñagarikano, O. et al. Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities, and core autism-related deficits. Cell 147, 235–246 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Stowers, L. & Kuo, T.-H. Mammalian pheromones: emerging properties and mechanisms of detection. Curr. Opin. Neurobiol. 34, 103–109 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Root, C. M., Denny, C. A., Hen, R. & Axel, R. The participation of cortical amygdala in innate, odour-driven behaviour. Nature 515, 269–273 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kobayakawa, K. et al. Innate versus learned odour processing in the mouse olfactory bulb. Nature 450, 503–508 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Schneidman, E., Berry, M. J., Segev, R. & Bialek, W. Weak pairwise correlations imply strongly correlated network states in a neural population. Nature 440, 1007–1012 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Granot-Atedgi, E., Tkačik, G., Segev, R. & Schneidman, E. Stimulus-dependent maximum entropy models of neural population codes. PLoS Comput. Biol. 9, e1002922 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tkacik, G., Granot-Atedgi, E., Segev, R. & Schneidman, E. Retinal metric: a stimulus distance measure derived from population neural responses. Phys. Rev. Lett. 110, 058104 (2013).

    Article  PubMed  CAS  Google Scholar 

  29. Alarcon, M. et al. Linkage, association, and gene-expression analyses identify CNTNAP2 as an autism-susceptibility gene. Am. J. Hum. Genet. 82, 150–159 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bourgeron, T. From the genetic architecture to synaptic plasticity in autism spectrum disorder. Nat. Rev. Neurosci. 16, 551–563 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Guiraud, J. A. et al. Differential habituation to repeated sounds in infants at high risk for autism. Neuroreport 22, 845–849 (2011).

    PubMed  Google Scholar 

  32. Auerbach, B. D., Osterweil, E. K. & Bear, M. F. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature 480, 63–68 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liska, A. et al. Homozygous loss of autism-risk gene CNTNAP2 results in reduced local and long-range prefrontal functional connectivity. Cereb. Cortex 28, 1141–1153 (2018).

    Article  PubMed  Google Scholar 

  34. Dickinson, A., Jones, M. & Milne, E. Measuring neural excitation and inhibition in autism: different approaches, different findings and different interpretations. Brain Res. 1648, 277–289 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Nelson, S. B. & Valakh, V. Excitatory/inhibitory balance and circuit homeostasis in autism spectrum disorders. Neuron 87, 684–698 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Litwin-Kumar, A. & Doiron, B. Slow dynamics and high variability in balanced cortical networks with clustered connections. Nat. Neurosci. 15, 1498–1505 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Shadlen, M. N. & Newsome, W. T. The variable discharge of cortical neurons: implications for connectivity, computation, and information coding. J. Neurosci. 18, 3870–3896 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gerstein, G. L. & Mandelbrot, B. Random walk models for the spike activity of a single neuron. Biophys. J. 4, 41–68 (1964).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Stella, F., Baracskay, P., O’Neill, J. & Csicsvari, J. Hippocampal reactivation of random trajectories resembling Brownian diffusion. Neuron 102, 450–461.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Otis, J. M. et al. Prefrontal cortex output circuits guide reward seeking through divergent cue encoding. Nature 543, 103–107 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Milad, M. R. & Quirk, G. J. Neurons in medial prefrontal cortex signal memory for fear extinction. Nature 420, 70–74 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. Iurilli, G. & Datta, S. R. Population coding in an innately relevant olfactory area. Neuron 93, 1180–1197.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bolding, K. A. & Franks, K. M. Recurrent cortical circuits implement concentration-invariant odor coding. Science 361, eaat6904 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Vertes, R. P. Differential projections of the infralimbic and prelimbic cortex in the rat. Synapse 51, 32–58 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Price, J. L. et al. in Olfaction: A Model System for Computational Neuroscience (eds Davis, J. L. & Eichenbaum, H.) 101–120 (MIT Press, 1991).

  46. Beny, Y. & Kimchi, T. Innate and learned aspects of pheromone-mediated social behaviours. Anim. Behav. 97, 301–311 (2014).

    Article  Google Scholar 

  47. Remedios, R. et al. Social behaviour shapes hypothalamic neural ensemble representations of conspecific sex. Nature 550, 388–392 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Li, Y. et al. Neuronal representation of social information in the medial amygdala of awake behaving mice. Cell 171, 1176–1190.e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Selimbeyoglu, A. et al. Modulation of prefrontal cortex excitation/inhibition balance rescues social behavior in CNTNAP2-deficient mice. Sci. Transl Med. 9, eaah6733 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Varea, O. et al. Synaptic abnormalities and cytoplasmic glutamate receptor aggregates in contactin associated protein-like 2 /Caspr2 knockout neurons. Proc. Natl Acad. Sci. USA 112, 6176–6181 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Franklin, K. B. J. & Paxinos, G. The Mouse Brain in Stereotaxic Coordinates 3rd ed. (Academic Press, 2008).

  52. Ellegood, J. & Crawley, J. N. Behavioral and neuroanatomical phenotypes in mouse models of autism. Neurotherapeutics 12, 521–533 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yang, M. & Crawley, J. N. Simple behavioral assessment of mouse olfaction. Curr. Protoc. Neurosci. Chapter 8, Unit 8.24 (2009).

    PubMed  Google Scholar 

  54. Parabucki, A. et al. Odor concentration change coding in the olfactory bulb. eNeuro 6, ENEURO.0396–18.2019 (2019).

    Article  Google Scholar 

  55. Schneidman, E. Towards the design principles of neural population codes. Curr. Opin. Neurobiol. 37, 133–140 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Maoz, O. & Schneidman, E. maxent_toolbox: Maximum Entropy Toolbox for MATLAB v1.02. https://doi.org/10.5281/zenodo.191625 (2017).

  57. Segev, R., Ganmor, E. & Schneidman, E. A thesaurus for a neural population code. eLife 4, e06134 (2015).

    Article  PubMed Central  Google Scholar 

  58. Lin, J. Divergence measures based on the Shannon entropy. IEEE Trans. Inf. Theory 37, 145–151 (1991).

    Article  Google Scholar 

  59. Yu, B. M. et al. Gaussian-process factor analysis for low-dimensional single-trial analysis of neural population activity. J. Neurophysiol. 102, 614–635 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the Simons Foundation, the European Research Council (ERC 819496 PrefrontalMap and ERC 311238 NEURO-POPCODE), the Israel Science Foundation, the Israel–US Binational Science Foundation, the Adelis Foundation, the Lord Sieff of Brimpton Memorial Fund, and the Candice Appleton Family Trust. O.Y. is supported by a NARSAD Young Investigator Grant from the Brain & Behavior Research Foundation and by the Gertrude and Philip Nollman Career Development Chair. E.S. receives support as the Joseph and Bessie Feinberg Professorial Chair.

Author information

Authors and Affiliations

Authors

Contributions

D.R.L. and O.Y. designed the study, and D.R.L. and A.W. built the experimental setup. D.R.L. performed all experiments and analyzed the unit activity and behavioral data. A.P. and D.R.L. performed the sniffing experiments and analyses. D.R.L., T.T., O.Y. and E.S. performed the population coding analyses. M.K. contributed to the behavioral data analyses. D.R.L., O.Y., T.T. and E.S. wrote the manuscript.

Corresponding author

Correspondence to Ofer Yizhar.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Olfactometer calibrations and microarray electrode location.

(a) Latency to odor infusion from each of seven available odor ports. A pressure sensor was used to determine initiation of airflow into the chamber following TTL input indicating the opening of the appropriate solenoid. Each solenoid was tested 5 consecutive times (represented by individual circles). Mean ± s.e.m is presented. (b) Change in odor concentration at the center of the chamber. Measurements were taken using a volatile organic compound (VOC) meter following 5 s infusion of vapor from a 70% ethanol solution followed by infusion of clean air (5 repetitions). Shaded area marks stimulus presentation times. Mean ± s.e.m is presented. (c) Representative image depicting the location of an electrolytic lesion used to verify electrode position in the mPFC. Arrow indicates lesion location in the infralimbic cortex. (d) Schematic representation of electrode placement in recorded mice from all experimental groups. Location was determined using the most ventral end of localization lesion or electrode track. Average AP coordinates are marked.

Extended Data Fig. 2 Social tuning in the mPFC unit responses.

(a) Stimulus-evoked response across all recorded units per stimulus, sorted by response magnitude as calculated by the absolute change in firing rate from baseline. Color gradient represents the normalized change in firing rate calculated over 250 ms bins. Arrowheads mark the time of stimulus onset and offset. (b) Response Z-score distribution calculated for all recorded units in response to social (M/F) and nonsocial (B/P/H) cues. Circles represent maximum response of individual units to each stimulus category. Color code represents response specificity. Z score threshold (|Z| > 2) is represented by a dashed line. Units with Z > 15 were assigned with Z = 15 for presentation purposes. (c) Number of units significantly increasing (dark) and decreasing (bright) their firing rates in response to each presented stimulus. Color represents stimulus identity. (d) Stimulus specificity overlap within social (top) and nonsocial (bottom) units. Number of units in each category is indicated on the figure. Circle sizes are scaled to the relative number of units responding to each stimulus. (e) Relative ratio between social and nonsocial units, calculated using a continuous range of Z score thresholds. Arrow represents Z = 2. Note that the number of social units consistently exceeds that of nonsocial units starting at Z > 0.6 (f) Response magnitude for all presented stimuli for units significantly increasing their firing rate in response to stimulus presentations. Box plot depicts the interquartile range (IQR) and median, whiskers mark ± 1.5*IQR. One-way ANOVA, Fstimulus(4,131) = 1.603, P = 0.177. (nM = 45 units, nF = 46 units, nB = 9 units, nP = 12 units, nH = 24 units). For all panels: M, male; F, female; B, banana; P, peanut butter; H, hexanal; CA, clean air.

Extended Data Fig. 3 Social categorization generalizes over a range of stimulus concentrations.

Overall unit tuning, presented as normalized firing rate in response to male cues at three different concentrations (ML, MM, MH), female (F) banana (B) and peanut butter (P) stimuli. RM ANOVA with Bonferroni corrected post hoc comparisons. Fstimulus(5,440) = 7.458, P = 9.8 × 10−7 (n = 89 responsive units). Superscripts mark significant post hoc comparisons. Mean ± s.e.m (shaded area) is presented. (b) Overall unit tuning, presented as normalized firing rate in response to male (M) and female (F) stimuli, banana oil at three different concentrations (BL, BM, BH) and peanut butter oil (P). RM ANOVA with Bonferroni corrected post hoc comparisons. Fstimulus(5,285) = 9.856, P = 1.1 × 10−8 (n = 58 responsive units). Superscripts mark significant post hoc comparisons. Mean ± s.e.m (shaded area) is presented. For detailed statistics information see Supplementary Table 2. (c) Similarity matrices depicting the distance between population responses to the stimulus panel used in a (n = 4 C57BL/6 J mice). (d) Same as c but for the stimulus panel used in b (n = 3 C57BL/6 J mice). Norm. res.- normalized response.

Extended Data Fig. 4 Maximum entropy models accurately describe the encoding distributions of the stimuli.

(a) Normalized difference between the log-likelihood values of the pairwise maximum entropy model (ME2) and conditionally independent model, for each mouse. Models were trained over seven trials of a specific stimulus and tested on one held-out trial per stimulus. Each dot corresponds to one held-out trial for one specific stimulus (6 stimuli × 8 trials = 48 dots per mouse). Positive values indicate larger likelihood for the independent model over the ME2; the most likely model for each trial was then used for decoding analysis (see Fig. 6). (b) The empirical probabilities of population activity patterns of cells recorded in one mouse in response to one odor are plotted against the probabilities predicted by different models (gray dots, independent model; orange dots, ME2 model). Each dot corresponds to a single activity pattern observed during the experiment. The funnel marked by the dashed \ gray line indicates 99% confidence interval of the empirical measurement. Black dashed line shows equality. (c) The Jensen–Shannon divergences between the empirical joint probability distribution of activity patterns and the different models – ME2 (orange) and conditionally independent (gray). Black line indicates equality of the distance of the models from the test data, and the distance between the training and test data. Models were trained using randomly chosen 1750 samples, similar to the number of training data samples used for the decoding analysis (7 trials of 5 s each). Analysis was done using all recorded units from each mouse (up to 20 units) and the mean over ten randomly chosen training sets is plotted. While no model is consistently better than the other in capturing the distribution across all mice, both models clearly outperform the empirical model. Arrow indicates the example mouse shown in panel b.

Extended Data Fig. 5 Behavioral analysis of Cntnap2-/- mice.

(a) Open field test. Left: Mean distance travelled during test for WT (black, n = 8) and Cntnap2-/- mice (teal, n = 8). Two-sided Mann-Whitney U test, U = 28, P = 0.721. Right: Mean duration in center of arena. Two-sided Mann-Whitney U test, U = 28, P = 0.721. (b) Baseline sniffing quantifications for WT (black, n = 7 recording sessions) and Cntnap2-/- mice (teal, n = 6 sessions). Left: baseline sniffing frequency. Mann-Whitney U test, U = 26, P= 0.52. Right: baseline sniff amplitude. Mann-Whitney U test, U = 4, P= 0.014. Circles represents individual recording sessions. (c) Mean duration to find a buried food pellet for WT (black, n = 8) and Cntnap2-/- mice (teal, n = 8). Two-sided Mann-Whitney U test, U = 20, P = 0.234. (d) Mean duration of odor exploration, calculated as time sniffing odor port for WT (left, n = 7) and Cntnap2-/- mice (right, n = 7 mice). Duration is presented for first and second day of experiment (left to right) for each odor and each mouse. Two-way RM ANOVA with Bonferroni corrections. For WT: Fstimulus(5,30) = 15.444, P = 1.6 × 10−7; Fday(1,6) = 2.756, P = 0.148; Fstimulus*day(5,30) = 1.777, P = 0.148; for Cntnap2-/-: Fstimulus(5,30) = 11.862, P = 2.2 × 10−8; Fday(1,6) = 0.593, P = 0.470; Fstimulus*day(5,30) = 1.777, P = 0.203; (e) Left: Average latency to odor-evoked orientation responses for WT (black, n = 5) and Cntnap2-/- (teal, n = 6) mice in odor infusion chamber. Circles represent individual mice. Mixed-design RM ANOVA. Fgenotype(1,9) = 0.959, P = 0.352; Fstimulus(5,45) = 2.449, P = 0.048 with Dunnett’s test against clean air; Fgenotype*stimulus(5,45) = 0.163, P = 0.974. Right: mean probability of odor-evoked orientation responses. Mixed-design RM ANOVA. Fgenotype(1,9) = 0.040, P = 0.844; Fstimulus(5,45) = 3.304, P = 0.013 with Dunnett’s test against clean air; Fgenotype*stimulus(5,45) = 0.115, P = 0.988 (f) Locomotion levels of WT (black, n = 5) and Cntnap2-/- mice (teal, n = 6) in the odor infusion chamber during baseline and stimulus presentation (averaged across all presented odors). Two-sided Mann-Whitney U Test, For baseline: U = 7, P = 0.171; For stimulus: U = 10, P = 0.411. Circles represent individual mice unless otherwise indicated. For all panels: Mean ± s.e.m is presented. Note that some individual data points and error bars are covered by the mark of the mean. *P < 0.05. For detailed statistics information see Supplementary Table 2. M, male; F, female; B, banana; P, peanut butter; H, hexanal; CA, clean air. Base., baseline; Stim., Stimulus.

Extended Data Fig. 6 Altered response patterns to social and nonsocial stimuli in the mPFC of Cntnap2-/- mice.

(a) Stimulus-evoked responses across all recorded units per stimulus, sorted by response magnitude as calculated by the change in firing rate from baseline for WT (top) and Cntnap2-/- (bottom) mice. Color gradient represents the change in firing rate from baseline, calculated over 250 ms bins. Arrows mark the time of stimulus onset and offset. (b) Response Z-score distribution calculated for all recorded units in WT (left) and Cntnap2-/- mice (right) in response to social (M/F) and nonsocial (B/P/H) cues. Circles represent maximum response of individual units to each stimulus category. Color code represents response specificity. Z score threshold (|Z|  > 2) is represented by a dashed line. Units with Z > 15 were assigned with Z = 15 for presentation purposes. (c) Number of units significantly increasing (dark) and decreasing (bright) their firing rates in response to each presented stimulus in wt (black) and Cntnap2-/- (teal) mice. (d) Relative ratio between social and nonsocial units, calculated using a continuous range of Z score thresholds for wt (black) and Cntnap2-/- (teal) mice. Arrows represent Z = 2. Linear regression analysis (0 ≤ Z ≤ 3, n = 32 measurements for each genotype), FWT(1,30) = 1088.42, P= 3.9 × 10−25, R2WT = 0.973. FCntnap2-/-(1,30) = 652.294, P = 6.5 × 10−22, R2Cntnap2-/- = 0.956; BWT = 1.106, BCntnap2-/- = 0.425, with non-overlapping 95% confidence intervals as a measure of statistical significant difference between regression lines. For all panels: *P < 0.05, M, male; F, female; B, banana; P, peanut butter; H, hexanal; CA, clean air.

Extended Data Fig. 7 Experience-dependent changes in stimulus-evoked unit responses.

(a) Stimulus-evoked PSTHs portraying mean response Z-score of cue-responsive units in the first (left) and second (right) recording sessions, for wt (top) and Cntnap2-/- (bottom) mice. Color code represents stimulus identity. Shaded areas mark stimulus presentation time. Mean ± s.e.m is presented. (b) Stimulus specificity among cue responsive units in the first (left) and second (right) recording sessions, in WT (top) and Cntnap2-/- (bottom) mice. Colors represent stimulus identity. For WT, nday1 = 82 units, nday2 = 77 units; for Cntnap2-/- mice, nday1 = 74 units, nday2 = 57 units. For all panels: M, male; F, female; B, banana; P, peanut butter; H, hexanal; CA, clean air.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Supplementary Video 1

Time-resolved trajectory of odor-evoked population activity in the mPFC of a wild-type C57BL/6J mouse. Representative 2D projections of the neuronal population trajectories before, during and after stimulus presentation (15 s time-window, 5 s for each phase, where each point was estimated in 150-ms bins, n = 15 units). Colors represent odor identity. Here, the first two principal components accounted for 75% of the variance. Time indication is relative to stimulus onset.

Supplementary Video 2

Time-resolved trajectory of odor-evoked population activity in the mPFC of a Cntnap2+/+ mouse. Representative 2D projections of the neuronal population trajectories before, during and after stimulus presentation (15 s time-window, 5 s for each phase, where each point was estimated in 150-ms bins, n = 22 unis). Colors represent odor identity. Here, the first two principal components accounted for 78% of the variance. Time indication is relative to stimulus onset.

Supplementary Video 3

Time-resolved trajectory of odor-evoked population activity in the mPFC of a Cntnap2–/– mouse. Representative 2D projections of the neuronal population trajectories before, during and after stimulus presentation (15 s time-window, 5 s for each phase, where each point was estimated in 150-ms bins, n = 17 unis). Colors represent odor identity. Here, the first two principal components accounted for 82% of the variance. Time indication is relative to stimulus onset.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Levy, D.R., Tamir, T., Kaufman, M. et al. Dynamics of social representation in the mouse prefrontal cortex. Nat Neurosci 22, 2013–2022 (2019). https://doi.org/10.1038/s41593-019-0531-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-019-0531-z

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