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:

An amygdala circuit that suppresses social engagement

Abstract

Innate social behaviours, such as mating and fighting, are fundamental to animal reproduction and survival1. However, social engagements can also put an individual at risk2. Little is known about the neural mechanisms that enable appropriate risk assessment and the suppression of hazardous social interactions. Here we identify the posteromedial nucleus of the cortical amygdala (COApm) as a locus required for the suppression of male mating when a female mouse is unhealthy. Using anatomical tracing, functional imaging and circuit-level epistatic analyses, we show that suppression of mating with an unhealthy female is mediated by the COApm projections onto the glutamatergic population of the medial amygdalar nucleus (MEA). We further show that the role of the COApm-to-MEA connection in regulating male mating behaviour relies on the neuromodulator thyrotropin-releasing hormone (TRH). TRH is expressed in the COApm, whereas the TRH receptor (TRHR) is found in the postsynaptic MEA glutamatergic neurons. Manipulating neural activity of TRH-expressing neurons in the COApm modulated male mating behaviour. In the MEA, activation of the TRHR pathway by ligand infusion inhibited mating even towards healthy female mice, whereas genetic ablation of TRHR facilitated mating with unhealthy individuals. In summary, we reveal a neural pathway that relies on the neuromodulator TRH to modulate social interactions according to the health status of the reciprocating individual. Individuals must balance the cost of social interactions relative to the benefit, as deficits in the ability to select healthy mates may lead to the spread of disease.

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: COApm is activated by female mice treated with LPS.
Fig. 2: COApm mediates suppression of mating behaviours towards unhealthy females.
Fig. 3: COApm projections to MEA-Vglut2+ neurons mediate suppression of mating towards LPS females.
Fig. 4: Suppression of social behaviours engages COApm-TRH+ neurons.

Similar content being viewed by others

Data availability

Source data are provided in the Supplementary Information. Sequencing data sets are publicly available in NCBI Gene Expression Omnibus (GEO) under accession GSE167176. All data are available from the corresponding author upon request.

References

  1. Tinbergen, N. The Study of Instinct (Oxford Univ. Press, 1951).

  2. Altizer, S. et al. Social organization and parasite risk in mammals: integrating theory and empirical studies. Annu. Rev. Ecol. Evol. Syst. 34, 517–547 (2003).

    Article  Google Scholar 

  3. Hart, B. L. Behavioral adaptations to pathogens and parasites: five strategies. Neurosci. Biobehav. Rev. 14, 273–294 (1990).

    Article  CAS  PubMed  Google Scholar 

  4. Ehman, K. D. & Scott, M. E. Female mice mate preferentially with non-parasitized males. Parasitology 125, 461–466 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chamero, P. et al. Identification of protein pheromones that promote aggressive behaviour. Nature 450, 899–902 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Hashikawa, K., Hashikawa, Y., Falkner, A. & Lin, D. The neural circuits of mating and fighting in male mice. Curr. Opin. Neurobiol. 38, 27–37 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Stowers, L., Holy, T. E., Meister, M., Dulac, C. & Koentges, G. Loss of sex discrimination and male–male aggression in mice deficient for TRP2. Science 295, 1493–1500 (2002).

    Article  ADS  CAS  PubMed  Google Scholar 

  9. Leypold, B. G. et al. Altered sexual and social behaviors in trp2 mutant mice. Proc. Natl Acad. Sci. USA 99, 6376–6381 (2002).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Medzhitov, R. Toll-like receptors and innate immunity. Nat. Rev. Immunol. 1, 135–145 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Boillat, M. et al. The vomeronasal system mediates sick conspecific avoidance. Curr. Biol. 25, 251–255 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Rivière, S., Challet, L., Fluegge, D., Spehr, M. & Rodriguez, I. Formyl peptide receptor-like proteins are a novel family of vomeronasal chemosensors. Nature 459, 574–577 (2009).

    Article  ADS  PubMed  CAS  Google Scholar 

  13. Liberles, S. D. et al. Formyl peptide receptors are candidate chemosensory receptors in the vomeronasal organ. Proc. Natl Acad. Sci. USA 106, 9842–9847 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kavaliers, M., Choleris, E., Agmo, A. & Pfaff, D. W. Olfactory-mediated parasite recognition and avoidance: linking genes to behavior. Horm. Behav. 46, 272–283 (2004).

    Article  PubMed  Google Scholar 

  15. Scalia, F. & Winans, S. S. The differential projections of the olfactory bulb and accessory olfactory bulb in mammals. J. Comp. Neurol. 161, 31–55 (1975).

    Article  CAS  PubMed  Google Scholar 

  16. Boehm, U. The vomeronasal system in mice: from the nose to the hypothalamus- and back! Semin. Cell Dev. Biol. 17, 471–479 (2006).

    Article  PubMed  Google Scholar 

  17. Choi, G. B. et al. Lhx6 delineates a pathway mediating innate reproductive behaviors from the amygdala to the hypothalamus. Neuron 46, 647–660 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ishii, K. K. et al. A labeled-line neural circuit for pheromone-mediated sexual behaviors in mice. Neuron 95, 123–137 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Hong, W., Kim, D. W. & Anderson, D. J. Antagonistic control of social versus repetitive self-grooming behaviors by separable amygdala neuronal subsets. Cell 158, 1348–1361 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wickersham, I. R., Finke, S., Conzelmann, K. K. & Callaway, E. M. Retrograde neuronal tracing with a deletion-mutant rabies virus. Nat. Methods 4, 47–49 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Heiman, M. et al. A translational profiling approach for the molecular characterization of CNS cell types. Cell 135, 738–748 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Suzuki, M., Sugano, H., Matsumoto, K., Yamamura, M. & Ishida, R. Synthesis and central nervous system actions of thyrotropin-releasing hormone analogues containing a dihydroorotic acid moiety. J. Med. Chem. 33, 2130–2137 (1990).

    Article  CAS  PubMed  Google Scholar 

  24. Hart, B. L. Biological basis of the behavior of sick animals. Neurosci. Biobehav. Rev. 12, 123–137 (1988).

    Article  CAS  PubMed  Google Scholar 

  25. Lopes, P. C., Block, P. & König, B. Infection-induced behavioural changes reduce connectivity and the potential for disease spread in wild mice contact networks. Sci. Rep. 6, 31790 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Van Kerckhove, K., Hens, N., Edmunds, W. J. & Eames, K. T. The impact of illness on social networks: implications for transmission and control of influenza. Am. J. Epidemiol. 178, 1655–1662 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Stockmaier, S., Bolnick, D. I., Page, R. A. & Carter, G. G. Sickness effects on social interactions depend on the type of behaviour and relationship. J. Anim. Ecol. 89, 1387–1394 (2020).

    Article  PubMed  Google Scholar 

  28. Fischer, S. & Ehlert, U. Hypothalamic-pituitary-thyroid (HPT) axis functioning in anxiety disorders. A systematic review. Depress. Anxiety 35, 98–110 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Baumgartner, A. Thyroxine and the treatment of affective disorders: an overview of the results of basic and clinical research. Int. J. Neuropsychopharmacol. 3, 149–165 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Krashes, M. J. et al. An excitatory paraventricular nucleus to AgRP neuron circuit that drives hunger. Nature 507, 238–242 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, F. et al. Optogenetic interrogation of neural circuits: technology for probing mammalian brain structures. Nat. Protoc. 5, 439–456 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ung, K. & Arenkiel, B. R. Fiber-optic implantation for chronic optogenetic stimulation of brain tissue. J. Vis. Exp. 68, e50004 (2012).

    Google Scholar 

  34. Armbruster, B. N., Li, X., Pausch, M. H., Herlitze, S. & Roth, B. L. Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc. Natl Acad. Sci. USA 104, 5163–5168 (2007).

    Article  ADS  PubMed  CAS  PubMed Central  Google Scholar 

  35. Pankevich, D. E., Baum, M. J. & Cherry, J. A. Olfactory sex discrimination persists, whereas the preference for urinary odorants from estrous females disappears in male mice after vomeronasal organ removal. J. Neurosci. 24, 9451–9457 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. McClure, C., Cole, K. L., Wulff, P., Klugmann, M. & Murray, A. J. Production and titering of recombinant adeno-associated viral vectors. J. Vis. Exp. 57, e3348 (2011).

    Google Scholar 

  37. Paxinos, G. & Franklin, K. B. J. The Mouse Brain in Stereotaxic Coordinates 2nd edn (Elsevier/Academic Press, 2004).

  38. Gunaydin, L. A. et al. Natural neural projection dynamics underlying social behavior. Cell 157, 1535–1551 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Byers, S. L., Wiles, M. V., Dunn, S. L. & Taft, R. A. Mouse estrous cycle identification tool and images. PLoS ONE 7, e35538 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bankhead, P. et al. QuPath: Open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  41. Cantu, D. A. et al. EZcalcium: open-source toolbox for analysis of calcium imaging data. Front. Neural Circuits 14, 25 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Heiman, M., Kulicke, R., Fenster, R. J., Greengard, P. & Heintz, N. Cell type-specific mRNA purification by translating ribosome affinity purification (TRAP). Nat. Protoc. 9, 1282–1291 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014). 8

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Hornung, V. et al. 5′-Triphosphate RNA is the ligand for RIG-I. Science 314, 994–997 (2006).

    Article  ADS  PubMed  Google Scholar 

  46. Huang, K. W. & Sabatini, B. L. Single-cell analysis of neuroinflammatory responses following intracranial injection of G-deleted rabies viruses. Front. Cell. Neurosci. 14, 65 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44 (W1), W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Choe, H. K. et al. Oxytocin mediates entrainment of sensory stimuli to social cues of opposing valence. Neuron 87, 152–163 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank N. Soares, M. Andina, K. Ronayne and I. D. Mejia for assistance with experiments; B. Noro and M. Trombly for critical reading of the manuscript; J. Huh for his contribution to the conceptual development of the project; B. Lowell for generously sharing the Trh-Cre mouse line; and H. Choi for the fibre photometry setup. This work was supported by the National Institute of Mental Health grants R01 MH122270 and R01 MH106497 (G.B.C.), the JPB Foundation (M.H. and G.B.C), Simons Center for the Social Brain Postdoctoral Fellowship (J.-T.K. and H.K.C.) and the Picower Fellows (J.-T.K. and H.L.).

Author information

Authors and Affiliations

Authors

Contributions

J.-T.K. and G.B.C. conceptualized the study. J.-T.K., H.L., C.R., M.H. and G.B.C. designed the experiments and/or provided advice and technical expertise. J.-T.K., H.L., C.R., A.S., J.F., D.H.C., S.B., H.A.S. and H.K.C. performed the experiments. I.R.W. provided reagents. J.-T.K. and G.B.C. wrote the manuscript with inputs from the co-authors.

Corresponding author

Correspondence to Gloria B. Choi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Dayu Lin, Ruslan Medzhitov 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 figures and tables

Extended Data Fig. 1 Male mice avoid mounting sick females.

a, b, Male mice were presented with a pair of oestrus female mice each injected intraperitoneally with either PBS (PBS female) or LPS (LPS female) (a). Mounting time during a 10-min test (b) (n = 9; from 2 independent experiments). c, Mounting time for male mice presented with two untreated, healthy females (n = 8; from 2 independent experiments). d, Investigation time of PBS and LPS females during a 10-min three-chamber assay (n = 5; from 2 independent experiments). ei, These data are associated with Fig. 1a–e. Duration of other typical male behaviours while engaged in direct interactions with a PBS or LPS female (e, f). Percentage of individual female behaviours during males’ mounting attempts (g) and the number of cage crossings (h). Representative traces of male and female behaviours during direct interactions (i). **P < 0.01, ***P < 0.001 and ****P < 0.0001 calculated by paired two-tailed t-test (b), two-way ANOVA with Sidak’s post hoc test (f, g) and unpaired two-tailed t-test (h). Data are mean ± s.e.m. Pvalues are described in the Supplementary Statistical Information.

Extended Data Fig. 2 Role of the vomeronasal pathway in mounting behaviour.

a, Mounting time for males with a sham surgery (sham) or the VNO removed (VNOX) towards LPS females (sham, n = 5 and VNOX, n = 7; from 2 independent experiments). bd, Virus encoding the anterograde trans-synaptic tracer (AAV1-hSyn-Cre) was targeted to the AOB in Ai14 reporter mice that express tdTomato in a Cre-dependent manner (b). Representative images (c) and quantification (d) of trans-synaptically labelled tdTomato+ neurons in BST, MEA and COApm at the specified anterior-posterior axis (n = 4; from 3 independent experiments). Scale bars, 500 μm. eg, Virus encoding ChR2 (AAV2-hSyn-hChR2-eYFP) was targeted to the AOB (e, f). g, Representative image of FOS expression in the COApm upon photoactivation of the AOB, from n = 3 mice. Scale bars, 500 μm. h, These data are associated with Fig. 1g. Number of FOS-expressing neurons in the vomeronasal pathway after interaction with PBS or LPS females. **P < 0.01, ***P < 0.001 and ****P < 0.0001 calculated by unpaired two-tailed t-test (a), one-way ANOVA with Bonferroni’s post hoc test (d) and two-way ANOVA with Sidak’s post hoc test (h). Graph indicates mean ± s.e.m. P values are described in the Supplementary Statistical Information.

Extended Data Fig. 3 Investigation of LPS females induces neural activity in the COApm of males.

ac, These data are associated with Fig. 1h–o. a, Representative image of GCaMP6s expression in the COApm. Scale bar, 300 μm. b, Individual traces of COApm bulk fluorescence signal during interactions with a PBS or LPS female. c, Heat map of normalized COApm responses to PBS or LPS females. Each row represents a single investigation event. Investigation events were pooled from 6 mice from 3 independent experiments. Time = 0 indicates initiation of investigation. d, e, Virus encoding GCaMP6s (AAV1-Syn-GCaMP6s) was targeted to the COApm for fibre photometry recordings. Male mice were sequentially presented with an oestrus or dioestrus female in counterbalanced sessions. Representative traces of COApm bulk fluorescence signal (d) and the mean z-score of the fluorescence during direct investigation of the oestrus or dioestrus female (e) (oestrus, n = 5 and dioestrus, n = 5; from 3 independent experiments). fj, Male mounting behaviours towards an oestrus or dioestrus, healthy female. Percentage of male mounting (f), mounting time (g), number of mounts (h), latency to mount (i) and percentage of female partners with mating plugs (j) (oestrus, n = 6 and dioestrus, n = 6; from 2 independent experiments). j, *P < 0.05 calculated by chi-square test of independence. Data are mean ± s.e.m. P values are described in the Supplementary Statistical Information.

Extended Data Fig. 4 LPS odour suppresses male mating behaviours.

ac, Male mice were presented with PBS or LPS odour (a). Representative images (b) and quantification (c) of FOS expression in the COApm of males after exposure to PBS or LPS odour (PBS odour, n = 6 and LPS odour, n = 8; from 2 independent experiments). Scale bar, 500 μm. d, e, Virus encoding GCaMP6s (AAV1-Syn-GCaMP6s) was targeted to the COApm for fibre photometry recordings. Male mice were sequentially presented with a PBS or LPS odour in counterbalanced sessions. Traces of COApm bulk fluorescence signal (solid line shows the average and shaded area indicates the s.e.m.) (d) and the mean z-score of the fluorescence during the first 20 s of direct investigation of the odour (e) (n = 6; from 3 independent experiments). fm, Male mice were presented with a healthy female painted with PBS or LPS odour (f). Percentage of male mounting (g), mounting time (h), number of mounts (i), latency to mount (j) and duration of additional male behaviours while engaged in direct interactions with the female (k). Percentage of female behaviours in response to males’ mounting attempts (l) and the number of cage crossings (m) (PBS odour, n = 10 and LPS odour, n = 11; from 2 independent experiments). *P < 0.05, **P < 0.01 and ****P < 0.0001 calculated by unpaired two-tailed t-test (c, hj), paired two-tailed t-test (e), chi-square test of independence (g) and two-way ANOVA with Sidak’s post hoc test (k). Data are mean ± s.e.m. P values are described in the Supplementary Statistical Information.

Extended Data Fig. 5 COApm mediates suppression of mating behaviours towards unhealthy females.

ad, These data are associated with Fig. 2a–e. a, Representative image of ChR2 expression in the COApm. b, Total direct investigation time of healthy females in the presence (ON) and absence (OFF) of COApm photoactivation. Duration of self grooming (c) and percentage of photoactivation trials with self grooming (d). Scale bar, 1 mm. e, f, These data are associated with Fig. 2i–m. Representative image of hM4Di expression in the COApm (e) and total direct investigation time of LPS females (f). Scale bar, 1 mm. gk, Additional control experiments for data in Fig. 2i–m. Male mice expressing inhibitory DREADD (AAV2-hSyn-hM4D(Gi)-mCherry) in COApm were injected with either saline or CNO and tested for mounting behaviour towards LPS females (g). Percentage of male mounting (h), mounting time (i), number of mounts (j) and latency to mount (k) (saline, n = 8 and CNO, n = 7; from 2 independent experiments). lp, Male mice expressing mCherry (AAV2-hSyn-mCherry) or inhibitory DREADD (hM4Di, AAV2-hSyn-hM4D(Gi)-mCherry) in COApm were injected with CNO and tested for mounting behaviour towards untreated, healthy oestrus females (l). Percentage of male mounting (m), mounting time (n), number of mounts (o) and latency to mount (p) (mCherry, n = 8 and hM4Di, n = 8; from 2 independent experiments). qu, Male mice expressing inhibitory DREADD (AAV2-hSyn-hM4D(Gi)-mCherry) in COApm were injected with saline or CNO and tested for mounting behaviour towards untreated, healthy dioestrus females (q). Percentage of male mounting (r), mounting time (s), number of mounts (t), and latency to mount (u) (saline, n = 6 and CNO, n = 6; from 2 independent experiments). *P < 0.05, **P < 0.01 calculated by unpaired two-tailed t-test (c, i, j). Data are mean ± s.e.m. P values are described in the Supplementary Statistical Informatio.

Extended Data Fig. 6 COApm neurons preferentially project to MEA-Vglut2+ neurons.

a, b, Virus encoding the anterograde tracer (AAV2-hSyn-tdTomato) was targeted to the COApm. Total fluorescence intensity was measured in sub-regions receiving COApm axonal projections: MEA, BST, ventral hippocampus (HCv), lateral septum (LS), AOB and piriform cortex (PIR) (n = 4 mice; from 2 independent experiments). Scale bars, 500 μm. c, Virus encoding the anterograde trans-synaptic tracer (AAV1-hSyn-Cre) was targeted to the COApm of Ai14 reporter mice that express tdTomato in a Cre-dependent manner. Representative image of MEA post-synaptic neurons at AP −1.7 mm, from 3 independent experiments. Scale bar, 300 μm. d, mRNA expression of Vglut2 and Vgat in MEA (Image credit: Allen Institute). eg, ChR2 (AAV2-hSyn-ChR2-eYFP) was targeted to the COApm (e). Representative images of ChR2 expression in COApm (f) and FOS expression in MEA upon photoactivation of COApm (g) (n = 3; from 2 independent experiments). Scale bars, 300 μm. h, i, Virus encoding GCaMP6s (AAV1-Syn-GCaMP6s) was targeted to the MEA of Vglut2-Cre mice for fibre photometry recordings. Male mice were sequentially presented with a PBS or LPS female in counterbalanced sessions. Representative traces of MEA-vGlut2+ bulk fluorescence signal (h) and the mean z-score of the fluorescence during direct investigation of the PBS or LPS female (i) (n = 3; from 2 independent experiments). i, *P < 0.05 calculated by paired two-tailed t-test. Data are mean ± s.e.m. P values are described in the Supplementary Statistical Information.

Extended Data Fig. 7 COApm suppresses male mating behaviours by engaging MEA-Vglut2+ neurons.

These data are associated with Fig. 3f–i. Percentage of male mounting (ad), number of mounts (eh), latency to mount (il). Representative images of ChR2 expression in COApm (m) and hM4Di expression in MEA (n) of Vglut2-Cre mice with concurrent photoactivation of COApm-MEA projections and hM4Di-inhibition of MEA-Vglut2+ neurons. Scale bars, 500 μm. *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001 calculated by chi-square test of independence (ac) and unpaired two-tailed t-test (ek). Data are mean ± s.e.m. P values are described in the Supplementary Statistical Information.

Extended Data Fig. 8 Summary of gene-expression profiling in COApm neurons projecting to MEA-Vglut2+ neurons.

a, A combination of AAV1-Syn-FLEX-TTA and AAV-TRE-B19G, and RV∆G-L10a-eGFP were sequentially injected into the MEA of Vglut2-Cre mice to express L10a-eGFP in COApm neurons projecting to MEA-Vglut2+ neurons (COApm-proj). COApm tissue was collected and immediately used for TRAP analyses. Control mice were injected with AAV.PHP.eB-Syn-L10a-eGFP via retro-orbital injection in order to label COApm neurons with L10a-eGFP independently of their efferent projections (Total COApm). b, Volcano plot showing log2-fold change plotted against −log10 FDR for the labelled COApm neurons projecting to the MEA-Vglut2+ population (COApm-Proj.) compared to the total COApm. Differentially expressed genes that pass the threshold for the FDR are highlighted in red. c, Heat map showing COApm-differentially expressed genes that belong to the KEGG neuroactive ligand–receptor interaction pathway. P values are described in the Supplementary Statistical Information.

Extended Data Fig. 9 COApm-TRH+ neurons mediate the suppression of male mating towards unhealthy females.

ag, Trh-Cre male mice expressing tdTomato (AAV1/2-Ef1α-DO-DIO-tdTomato(tdT)-eGFP, Control) or ChR2 (AAV1/2-Ef1α-DO-ChR2-mCherry, ChR2) in Cre neurons were tested for mating behaviours towards healthy females with COApm photoactivation (a). Representative images (b) and quantification (c) of FOS expression in COApm. Percentage of male mounting (d), mounting time (e), number of mounts (f), and latency to mount (g) with photoactivation of COApm-TRH cells (control, n = 5 and ChR2, n = 5; from 2 independent experiments). Scale bars, 500 μm. hl, Calcium imaging of MEA brain slices from Vglut2-Cre mice expressing GCaMP7f (AAV1-hSyn-FLEX-GCaMP7f) in MEA-Vglut2+ neurons upon taltirelin (10 μM) application (h). i, Representative images of MEA slices before (−15 s) and after (+35 s) taltirelin application. Example traces of fluorescence signal from individual neurons (j) and the average of the fluorescence signal (solid line shows average and shaded area represents the s.e.m.) (k) upon taltirelin application. l, Area under the curve (AUC) of the average fluorescence signal from individual MEA slices binned every 30 s (n = 6 slices, from 3 mice). Scale bars, 50 μm. m, These data are associated with Fig. 4k–o. Total duration of direct investigation following microinjection of the TRH analogue taltirelin into MEA. *P < 0.05, **P < 0.01 and ***P < 0.001 calculated by unpaired two-tailed t-test (c, m) and Friedman test with Dunn’s multiple comparisons test (l). Data are mean ± s.e.m. P values are described in the Supplementary Statistical Information.

Extended Data Fig. 10 Suppression of mating engages TRH–TRHR signalling in the COApm-MEA projection.

a, Schematic depicting the targeting construct used to generate the Trhr conditional-knockout mouse line. b, c, Representative images (AP −1.8 mm) (b) and quantification (c) of Trhr mRNA expression in the MEApv of Trhrfl/fl mice with or without Cre expression (AAV1-hSyn-Cre) (Trhrfl/fl, n = 3 and Trhrfl/fl with Cre, n = 3; from 2 independent experiments). Scale bars, 20 μm. dg, In vivo recordings of MEA responses to COApm inputs in Trhr conditional-knockout mice. Trhrfl/fl mice were injected with AAV2-hSyn-ChR2-eYFP in COApm and either AAV1-hSyn-GFP (GFP) or AAV1-hSyn-Cre (Cre) in MEApv. d, Local field potentials evoked by a 10-ms photoactivation were recorded from MEApv in anaesthetized mice using an optrode. e, Representative image of electrode localization. Representative waveforms (f) and amplitudes (baseline-to-negative peak) of MEApv responses evoked by photoactivation of COApm inputs (g) (GFP, n = 6 and Cre, n = 6; from 6 independent experiments). Scale bar, 300 μm. ***P < 0.001 and ****P < 0.0001 calculated by unpaired two-tailed t-test (c, g). Data are mean ± s.e.m. P values are described in the Supplementary Statistical Information.

Supplementary information

Supplementary Data

This file contains Supplementary Statistical Information.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kwon, JT., Ryu, C., Lee, H. et al. An amygdala circuit that suppresses social engagement. Nature 593, 114–118 (2021). https://doi.org/10.1038/s41586-021-03413-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03413-6

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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