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-to-hypothalamus circuit for social reward

Abstract

Social interactions and relationships are often rewarding, but the neural mechanisms through which social interaction drives positive experience remain poorly understood. In this study, we developed an automated operant conditioning system to measure social reward in mice and found that adult mice of both sexes display robust reinforcement of social interaction. Through cell-type-specific manipulations, we identified a crucial role for GABAergic neurons in the medial amygdala (MeA) in promoting the positive reinforcement of social interaction. Moreover, MeA GABAergic neurons mediate social reinforcement behavior through their projections to the medial preoptic area (MPOA) and promote dopamine release in the nucleus accumbens. Finally, activation of this MeA-to-MPOA circuit can robustly overcome avoidance behavior. Together, these findings establish the MeA as a key node for regulating social reward in both sexes, providing new insights into the regulation of social reward beyond the classic mesolimbic reward system.

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: Adult mice exhibit robust reinforcement for social reward in an automated operant task.
Fig. 2: MeApd Vgat+ neurons are required for social reward.
Fig. 3: Activation of MeApd Vgat+ neurons is sufficient to drive reinforcement.
Fig. 4: Dynamics of dopamine signals in the NAc during social operant task.
Fig. 5: Activating MeApd Vgat+ neurons triggers dopamine release in the NAc.
Fig. 6: The MPOA-projecting MeApd circuit mediates social reward.
Fig. 7: Activation of the MeApd-to-MPOA circuit overcomes avoidance behavior.

Similar content being viewed by others

Data availability

All data generated and analyzed during this study are either included in this published article or available from the corresponding author upon reasonable request.

Code availability

Behavior was analyzed using MATLAB code, available at https://pdollar.github.io/toolbox/. The code that supports these findings is available upon reasonable request from the corresponding author.

References

  1. Stanley, D. A. & Adolphs, R. Toward a neural basis for social behavior. Neuron 80, 816–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Umberson, D., Crosnoe, R. & Reczek, C. Social relationships and health behavior across the life course. Annu. Rev. Socio. 36, 139–157 (2010).

    Article  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Tamir, D. I. & Hughes, B. L. Social rewards: from basic social building blocks to complex social behavior. Perspect. Psychol. Sci. 13, 700–717 (2018).

    Article  PubMed  Google Scholar 

  5. Hu, H. Reward and aversion. Annu. Rev. Neurosci. 39, 297–324 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Chevallier, C., Kohls, G., Troiani, V., Brodkin, E. S. & Schultz, R. T. The social motivation theory of autism. Trends Cogn. Sci. 16, 231–239 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Zeeland, A. A. S.-V., Dapretto, M., Ghahremani, D. G., Poldrack, R. A. & Bookheimer, S. Y. Reward processing in autism. Autism Res. 3, 53–67 (2010).

    Google Scholar 

  8. Nieh, E. H. et al. Inhibitory input from the lateral hypothalamus to the ventral tegmental area disinhibits dopamine neurons and promotes behavioral activation. Neuron 90, 1286–1298 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hung, L. W. et al. Gating of social reward by oxytocin in the ventral tegmental area. Science 357, 1406–1411 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dölen, G., Darvishzadeh, A., Huang, K. W. & Malenka, R. C. Social reward requires coordinated activity of nucleus accumbens oxytocin and serotonin. Nature 501, 179 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  11. McHenry, J. A. et al. Hormonal gain control of a medial preoptic area social reward circuit. Nat. Neurosci. 20, 449–458 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Chen, P. B. et al. Sexually dimorphic control of parenting behavior by the medial amygdala. Cell 176, 1206–1221 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lin, D. et al. Functional identification of an aggression locus in the mouse hypothalamus. Nature 470, 221–226 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wu, Z., Autry, A. E., Bergan, J. F., Watabe-Uchida, M. & Dulac, C. G. Galanin neurons in the medial preoptic area govern parental behaviour. Nature 509, 325 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Falkner, A. L., Grosenick, L., Davidson, T. J., Deisseroth, K. & Lin, D. Hypothalamic control of male aggression-seeking behavior. Nat. Neurosci. 19, 596–604 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Miller, S. M., Marcotulli, D., Shen, A. & Zweifel, L. S. Divergent medial amygdala projections regulate approach-avoidance conflict behavior. Nat. Neurosci. 22, 565–575 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gutzeit, V. A. et al. Optogenetic reactivation of prefrontal social neural ensembles mimics social buffering of fear. Neuropsychopharmacology 45, 1068–1077 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Swanson, L. W. Cerebral hemisphere regulation of motivated behavior. Brain Res. 886, 113–164 (2000).

    Article  CAS  PubMed  Google Scholar 

  20. 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 

  21. 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 

  22. Bergan, J. F., Ben-Shaul, Y. & Dulac, C. Sex-specific processing of social cues in the medial amygdala. eLife 3, e02743 (2014).

  23. 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 

  24. Unger, E. K. et al. Medial amygdalar aromatase neurons regulate aggression in both sexes. Cell Rep. 10, 453–462 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Yao, S., Bergan, J., Lanjuin, A. & Dulac, C. Oxytocin signaling in the medial amygdala is required for sex discrimination of social cues. eLife 6, e31373 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Demir, E. et al. The pheromone darcin drives a circuit for innate and reinforced behaviours. Nature 578, 137–141 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Golden, S. A. et al. Compulsive addiction-like aggressive behavior in mice. Biol. Psychiatry 82, 239–248 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Venniro, M. et al. Volitional social interaction prevents drug addiction in rat models. Nat. Neurosci. 21, 1520–1529 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Martin, L. & Iceberg, E. Quantifying social motivation in mice using operant conditioning. J. Vis. Exp. e53009 (2015).

  30. Jacobs, S., Huang, F., Tsien, J. & Wei, W. Social recognition memory test in rodents. Bio-protocol 6, e1804 (2016).

    Google Scholar 

  31. Hlinák, Z. & Krejcí, I. Social recognition in male rats: age differences and modulation by MIF-I and Alaptide. Physiol. Res. 40, 59–67 (1991).

    PubMed  Google Scholar 

  32. Berke, J. D. What does dopamine mean? Nat. Neurosci. 21, 787–793 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Volkow, N. D., Wise, R. A. & Baler, R. The dopamine motive system: implications for drug and food addiction. Nat. Rev. Neurosci. 18, 741–752 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Fang, Y.-Y., Yamaguchi, T., Song, S. C., Tritsch, N. X. & Lin, D. A hypothalamic midbrain pathway essential for driving maternal behaviors. Neuron 98, 192–207.e10 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kohl, J. et al. Functional circuit architecture underlying parental behaviour. Nature 556, 326–331 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hennessy, M. B., Kaiser, S. & Sachser, N. Social buffering of the stress response: diversity, mechanisms, and functions. Front. Neuroendocrinol. 30, 470–482 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Nardou, R. et al. Oxytocin-dependent reopening of a social reward learning critical period with MDMA. Nature 569, 116–120 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Schultz, W. Dopamine reward prediction-error signalling: a two-component response. Nat. Rev. Neurosci. 17, 183–195 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Collins, A. L. et al. Dynamic mesolimbic dopamine signaling during action sequence learning and expectation violation. Sci. Rep. 6, 20231 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Prévost-Solié, C., Girard, B., Righetti, B., Tapparel, M. & Bellone, C. Dopamine neurons of the VTA encode active conspecific interaction and promote social learning through social reward prediction error. Prerint at bioRxiv https://doi.org/10.1101/2020.05.27.118851 (2020).

  41. Padilla, S. L. et al. Agouti-related peptide neural circuits mediate adaptive behaviors in the starved state. Nat. Neurosci. 19, 734–741 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Berke, J. D. & Hyman, S. E. Addiction, dopamine, and the molecular mechanisms of memory. Neuron 25, 515–532 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Golden, S. A. et al. Basal forebrain projections to the lateral habenula modulate aggression reward. Nature 534, 688 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Beny-Shefer, Y. et al. Nucleus accumbens dopamine signaling regulates sexual preference for females in male mice. Cell Rep. 21, 3079–3088 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Kingsbury, L. et al. Cortical representations of conspecific sex shape social behavior. Neuron 107, 941–953 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wu, Y. E., Pan, L., Zuo, Y., Li, X. & Hong, W. Detecting activated cell populations using single-cell RNA-seq. Neuron 96, 313–329 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Kupferberg, A., Bicks, L. & Hasler, G. Social functioning in major depressive disorder. Neurosci. Biobehav. Rev. 69, 313–332 (2016).

    Article  PubMed  Google Scholar 

  48. Matthews, G. A. et al. Dorsal raphe dopamine neurons represent the experience of social isolation. Cell 164, 617–631 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Vong, L. et al. Leptin action on GABAergic neurons prevents obesity and reduces inhibitory tone to POMC neurons. Neuron 71, 142–154 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yang, C. F. et al. Sexually dimorphic neurons in the ventromedial hypothalamus govern mating in both sexes and aggression in males. Cell 153, 896–909 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Mahn, M. et al. High-efficiency optogenetic silencing with soma-targeted anion-conducting channelrhodopsins. Nat. Commun. 9, 4125 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Dana, H. et al. High-performance calcium sensors for imaging activity in neuronal populations and microcompartments. Nat. Methods 16, 649–657 (2019).

    Article  CAS  PubMed  Google Scholar 

  53. Broussard, G. J. et al. In vivo measurement of afferent activity with axon-specific calcium imaging. Nat. Neurosci. 21, 1272–1280 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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 

  55. Patriarchi, T. et al. Ultrafast neuronal imaging of dopamine dynamics with designed genetically encoded sensors. Science 360, eaat4422 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Dimidschstein, J. et al. A viral strategy for targeting and manipulating interneurons across vertebrate species. Nat. Neurosci. 19, 1743–1749 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Wassum, P. Golshani, L. Kingsbury, T. Raam, D. Wei, L. Gu, F. Sun and M. Zhang for critical comments on this manuscript and P. Chen, S. Hu, H. Li, Y. Wu and P. Zhao for technical assistance. This work was supported, in part, by NIH R01 NS113124, a Searle Scholars Award, a Klingenstein-Simons fellowship, a Brain Research Foundation grant, a Packard Foundation fellowship, a McKnight Scholar Award, a Keck Foundation Award, a Vallee Scholars Award and a Mallinckrodt Scholar Award to W.H. and a Marion Bowen postdoctoral grant to R.K.H.

Author information

Authors and Affiliations

Authors

Contributions

R.K.H. and W.H. conceptualized and designed the study. R.K.H., Y.Z., T.L. and J.W. performed optogenetics and behavioral experiments. R.K.H. and T.L. performed fiber photometry and histology. R.K.H. analyzed behavioral and neural data. R.K.H. performed ex vivo electrophysiology experiments. P.M. provided suggestions on electrophysiology. R.K.H., Y.E.W. and W.H. wrote the manuscript. W.H. supervised the entire study, provided resources and acquired funding.

Corresponding author

Correspondence to Weizhe Hong.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Peer review information Nature Neuroscience thanks Sam Golden, Olga Penagarikano and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Characterizations of behaviors in free social interaction and social operant task.

a-c, Fraction of time spent on seven distinct social behaviors displayed by adult animals (a, all animals; b, males; c, females) towards juvenile animals during free social interaction. Both male and female adult animals predominantly display close social investigation (sniffing) towards juvenile animals. The behavioral characterizations of these male and female animals in the social operant task are included in Extended Data Fig. 2. d, e, Cumulative distribution of the numbers of nose pokes in social and null ports in social operant task in one representative animal on each day. The assay was performed in the presence (d) or absence (e) of the target animal. f, The majority of social port pokes are followed by close social interaction between subject and target animals (across the barrier). P = 0.2149, Paired t test (two-sided). g, Percentage of animals showing consistent preference for the social port (defined as showing >20% preference on both days 6 and 7) in the presence or absence of the target animal. h, Full distribution of nose poke preferences in individual animals in Fig. 1i. Grey lines and dots indicate preferences of individual mice and orange line indicates the average across all animals. In (a-c), n = 12 males and 8 females; in (f), n = 17 mice; (g), n = 17 mice (with target animals) and 25 mice (without target animals); in (h), n = 17 mice. (a-c, f), boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 2 Both male and female adult mice exhibit social reward behavior.

a, b, Preference for social port and numbers of pokes in females (a) and males (b) in the presence of a target animal. Both male and female animals develop strong preference for the social port in the presence of target animals. Experiment was performed over a 10-day period, which consists of 1 day of baseline session, 7 days of training sessions with target animals, and 2 days of post-training sessions. c, d, Preference for social port and numbers of pokes in females (c) and males (d) in the absence of a target animal. Neither male nor female animals develop preference for the social port. Experiment was performed over an 8-day period, which consists of 1 day of baseline session and 7 days of training sessions without target animals. e, Percentage of male and female mice exhibiting consistent preference for social port (defined as showing >20% preference on both days 6 and 7). In (a, b, e), with target animals, n = 17 females and 20 males; these include 9 females and 8 males presented in Fig. 1 as well as an additional 8 females and 12 males presented in Extended Data Fig. 1. In (c, d, e), without target animals, n = 15 females and 10 males. In (a-d), left panels, one-way repeated measures ANOVA with Bonferroni post-hoc correction (*P < 0.05, **P < 0.01, ***P < 0.001); right panels, two-way repeated measures ANOVA with Bonferroni post-hoc correction (**P < 0.01, ***P < 0.001). (a-d), mean ± SEM. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 3 Additional characterizations of genetic ablation and optogenetic inhibition experiments.

a, b, Full distribution of nose poke preferences in individual animals in Fig. 2e (a) and Fig. 2g (b). Grey lines and dots indicate preferences of individual mice and colored line indicates the average across all animals. c, Representative trace showing robust silencing of Vgat+ neurons during light illumination. d, Firing rates before, during, and after light illumination. One-way repeated measures ANOVA with Bonferroni post-hoc correction (***P < 0.001). e, g, Fraction of time spent on seven distinct social behaviors displayed by subject animals (e, GtACR-expressing animals, or g, control animals) towards juvenile animals. Two-way repeated measures ANOVA with Bonferroni post-hoc (**P < 0.01, ***P < 0.001). f, h, Percentage of total time spent on all social behaviors displayed by subject animals (f, GtACR-expressing animals or h, control animals). **P = 0.0046 (f), P = 0.1766 (h), paired t test (two-sided). (a, b) n = 8 control mice and 10 Caspase mice; (c, d) n = 10 trials from 3 cells (2 mice); (e-h), n = 6 control mice and 5 GtACR-expressing mice. (d-h) boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 4 Fiber photometry of calcium signals in response to social and nonsocial rewarding stimuli.

MeApd Vgat+ neurons and their projections to the MPOA are active in response to social stimuli, but not to nonsocial rewarding stimuli, such as sucrose and chocolate. a, c, Dynamics of Ca2+ signals (ΔF/F) in MeApd Vgat+ neurons (a) or in the axonal projections of MeApd Vgat+ neurons to the MPOA (c) in response to social stimuli (juveniles) or during consumption of sucrose solution or chocolate. b, d, AUC per second under different conditions in a and c. Controls were done by measuring Ca2+ signals from EYFP-expressing MeApd Vgat+ neurons or EYFP-expressing MeApd Vgat+ neuron projections to the MPOA during exposure to social stimuli. One-way ANOVA with Bonferroni post-hoc correction (*P < 0.05, **P < 0.01, ***P < 0.001). (a, b), n = 15 trials (social), 20 trials (sucrose), and 11 trials (chocolate) from 6 GCaMP7f-expressing mice; n = 22 trials (social) from 4 EYFP-expressing control mice. (c, d), n = 31 trials (social), 48 trials (sucrose), and 37 trials (chocolate) from 7 GCaMP6s-expressing mice; n = 17 trials (social) from 4 EYFP-expressing control mice. (a, c), mean ± SEM; (b, d), boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 5 The MeApd-to-MPOA circuit does not mediate food reward.

a-d, Ablation of MeApd Vgat+ neurons does not affect food reward in an operant task. Animals are trained to lever press to obtain food pellets (see Methods). Control: animals expressing mCherry in MeApd Vgat+ neurons, Caspase 3: animals expressing Caspase 3 in MeApd Vgat+ neurons. e-h, Ablation of MPOA-projecting MeApd neurons does not affect food reward in an operant task. Control: animals expressing mCherry in MPOA-projecting MeApd neurons, Caspase 3: animals expressing Caspase 3 in MPOA-projecting MeApd neurons. a, b, e, f, cumulative distribution of lever presses in the operant task for food reward. c, g, number of lever presses. Two-way repeated measures ANOVA with Bonferroni post-hoc correction (***P < 0.001). d, h, preferences for the lever associated with food reward. Mann-Whitney test (two-sided). (a-d), n = 7 control mice and 6 Caspase mice; (e-h), n = 5 control mice and 6 Caspase mice; (a-h), mean ± SEM. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 6 Activation of MeApd Vgat+ neurons promotes reinforcement behavior in both males and females.

a, Activation of MeApd Vgat+ neurons in the intracranial self-stimulation assay. ChR2-expressing animals spend greater time in the stimulation-coupled active port (total time spent in the port), whereas control animals do not. Two-way repeated measures ANOVA with Bonferroni post-hoc correction (***P < 0.001). b, e, In a real-time place preference assay, ChR2-expressing animals display a positive preference towards the stimulation-coupled chamber compared to the EYFP-expressing controls in both females (b) and males (e). *P = 0.0242 (b), *P = 0.0121 (e), Mann-Whitney test (two-sided). c, f, In an intracranial self-stimulation assay, ChR2-expressing female (c) and male (f) animals exhibit a greater number of pokes towards the active port compared to the inactive port, whereas control animals do not. Two-way repeated measures ANOVA with Bonferroni post-hoc correction (**p < 0.01, ***p < 0.001). d, g, ChR2-expressing female (d) and male (g) animals spend greater time in the active port (total time spent in the port), whereas control animals do not. Two-way repeated measures ANOVA with Bonferroni post-hoc correction (*P < 0.05, **P < 0.01, ***P < 0.001). (a), n = 6 control mice and 10 ChR2 mice; (b), n = 3 control mice and 8 ChR2 mice; (c-d), n = 3 control mice and 7 ChR2 mice; (e), n = 3 control mice and 8 ChR2 mice; (f-g), n = 3 control mice and 3 ChR2 mice. (a) boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile; (b-g) mean ± SEM. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 7 Optogenetic activation of MeApd Vgat+ neurons or MeApd-to-MPOA projections drives reinforcement.

a-d, Cumulative distribution of the numbers of nose pokes to the optogenetic ‘social’ port or the null port in a modified social operant task. Representative data from a control or ChR2 animal (a, b, MeApd cell bodies; c, d, MeApd-to-MPOA projections) on each day. e-h, Numbers of pokes to the optogenetic ‘social’ port or the null port on each day. For MeApd cell bodies, control (e) and ChR2-expressing (f) animals; for MeApd-to-MPOA projections, control (g) and ChR2-expressing (h) animals. Two-way repeated measures ANOVA with Bonferroni post-hoc correction (**P < 0.01, ***P < 0.001). i, Activation of MeApd-to-MPOA projections in ChR2-expressing animals spend greater time in the active port (total time spent in the port), whereas control animals do not. Two-way repeated measures ANOVA with Bonferroni post-hoc correction ((*P < 0.05, **P < 0.01). (e, f), n = 6 mice (control) and n = 8 mice (ChR2); (g, h), n = 8 mice (control) and n = 8 mice (ChR2); (i), n = 5 mice (control) and 7 mice (ChR2). (e-h), mean ± SEM; (i), boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 8 Optogenetic activation of MeApd Vglut2+ neurons promotes place aversion.

a, Schematic showing the real-time place preference assay (RTPP). Light blue area (top) indicates the chamber paired with light stimulation when the mouse enters. Representative heatmaps showing locomotion trajectories for controls (middle) and ChR2-experessing (bottom) Vglut2-Cre mice in the RTPP test. b, Example image of viral expression in the MeApd of Vglut2-Cre animals. Scale bar = 200 μm. c, ChR2-expressing Vglut2-Cre animals display a negative preference towards the stimulation-couple chamber compared to EYFP-expressing controls. n = 4 mice (control) and 6 mice (ChR2). **P = 0.0095, Mann-Whitney test (two-sided). Boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 9 Behavioral functions of MeA-to-MPOA and MeA-to-PMV pathways.

a, Schematic showing optogenetic stimulation of MeApd axon terminals in the MPOA and lidocaine infusion in the MeApd cell bodies. b, Local infusion of lidocaine in the MeApd does not affect the behavioral effect of stimulating the MeApd-to-MPOA projection. P = 0.7612, paired t test (two-sided). c-d, Activating the MeApd-to-PMV projection does not promote positive reinforcement. c, Example images showing expression of ChR2 in the MeApd Vgat+ neurons (left) and fiber placement above their axon terminals in the PMV (right). Scale bar = 200 µm. d, Optogenetic stimulation of MeApd Vgat+ neuron terminals in the PMV does not produce positive place preference in a real-time place preference assay. P = 0.5103, paired t test (two-sided). e-g. Suppressing MeApd Vgat+ neuron activity reduces social preference in a three-chamber assay. e, Schematic showing the three-chamber assay. f-g, Optogenetic inhibition of MPOA-projecting MeA neurons in GtACR-expressing mice (g), but not in control mice (f), reduces social preference in a three-chamber social preference assay. P = 0.7183 (f), *P = 0.0471 (g), paired t test (two-sided). (b), n = 5 ChR2-expressing mice; (c, d), n = 6 ChR2-expressing mice; (f-g), n = 6 control mice and 5 GtACR-expressing mice. Boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile. For detailed statistics information, see Supplementary Table 1.

Extended Data Fig. 10 Characterization of MeApd neurons projecting to the MPOA.

a, Schematic showing injections of CTB-Alexa 647 into the MPOA and AAV-Dlx-NLS-Ruby2 and AAV-DIO-EYFP into the MeApd of Vglut2-Cre animals. b, Representative images showing labeling of CTB-Alexa 647 in the MPOA. Scale bar = 100 µm. c, Representative images showing retrogradely labeled CTB-Alexa 647 with the expression of AAV-Dlx-NLS-Ruby2 and AAV-DIO-EYFP in the MeApd of Vglut2-Cre animals. Scale bar = 20 µm. d, The fraction of overlap between Dlx+ and Vglut2+ neurons among all Dlx+ neurons. We confirm that the expression of Dlx and Vglut2 promotors shows little overlap in the MeApd. e, Fraction of MPOA-projecting MeApd neurons that are GABAergic. The majority of the MeApd neurons that project to the MPOA are GABAergic. f, Representative images showing expression of ChR2 in the MeApd Vglut2+ neurons (left) and fiber placement above their axon terminals in the MPOA (right) of Vglut2-Cre animals. Scale bar = 100 µm. g, Activating the MeApd Vglut2+ projections to the MPOA does not drive place preference. P = 0.1958, Paired t test (two-sided). (b-e), n = 3 mice; (f, g), n = 6 ChR2 mice. (d, e), mean ± SEM; (g), Boxplots: center = median, box = quartiles, whisker = 10 − 90 percentile. For detailed statistics information, see Supplementary Table 1.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, R.K., Zuo, Y., Ly, T. et al. An amygdala-to-hypothalamus circuit for social reward. Nat Neurosci 24, 831–842 (2021). https://doi.org/10.1038/s41593-021-00828-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-00828-2

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