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:

5-HT release in nucleus accumbens rescues social deficits in mouse autism model

Abstract

Dysfunction in prosocial interactions is a core symptom of autism spectrum disorder. However, the neural mechanisms that underlie sociability are poorly understood, limiting the rational development of therapies to treat social deficits. Here we show in mice that bidirectional modulation of the release of serotonin (5-HT) from dorsal raphe neurons in the nucleus accumbens bidirectionally modifies sociability. In a mouse model of a common genetic cause of autism spectrum disorder—a copy number variation on chromosome 16p11.2—genetic deletion of the syntenic region from 5-HT neurons induces deficits in social behaviour and decreases dorsal raphe 5-HT neuronal activity. These sociability deficits can be rescued by optogenetic activation of dorsal raphe 5-HT neurons, an effect requiring and mimicked by activation of 5-HT1b receptors in the nucleus accumbens. These results demonstrate an unexpected role for 5-HT action in the nucleus accumbens in social behaviours, and suggest that targeting this mechanism may prove therapeutically beneficial.

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: Activation of DR neurons or their NAc projections increases sociability.
Fig. 2: Bidirectional modulation of DR 5-HT neuron activity modifies sociability.
Fig. 3: 16p11.2 deletion in DR and 5-HT neurons decreases sociability.
Fig. 4: 16p11.2 deletion in DR 5-HT neurons decreases their activity.
Fig. 5: Rescue of social deficits in 16p11.2 deletion mice by 5-HT activity in the NAc.

Similar content being viewed by others

References

  1. Christensen, D. L. et al. Prevalence and characteristics of autism spectrum disorder among children aged 8 years–autism and developmental disabilities monitoring network, 11 Sites, United States, 2012. MMWR Surveill. Summ. 65, 1–23 (2016).

    Article  PubMed  Google Scholar 

  2. 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 

  3. Dölen, G., Darvishzadeh, A., Huang, K. W. & Malenka, R. C. Social reward requires coordinated activity of nucleus accumbens oxytocin and serotonin. Nature 501, 179–184 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  4. Brown, S.-L. & Praag, H. M. v. The Role of Serotonin in Psychiatric Disorders (Brunner/Mazel, New York, 1991).

    Google Scholar 

  5. Charney, D. S., Sklar, P. B., Buxbaum, J. D. & Nestler, E. J. Charney & Nestler’s Neurobiology of Mental Illness 5th edn (Oxford Univ. Press, 2018).

  6. Furay, A. R., McDevitt, R. A., Miczek, K. A. & Neumaier, J. F. 5-HT1B mRNA expression after chronic social stress. Behav. Brain Res. 224, 350–357 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Kane, M. J. et al. Mice genetically depleted of brain serotonin display social impairments, communication deficits and repetitive behaviors: possible relevance to autism. PLoS One 7, e48975 (2012).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  8. Challis, C. et al. Raphe GABAergic neurons mediate the acquisition of avoidance after social defeat. J. Neurosci. 33, 13978–13988, 13988a (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Li, Y. et al. Serotonin neurons in the dorsal raphe nucleus encode reward signals. Nat. Commun. 7, 10503 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  10. Muller, C. L., Anacker, A. M. J. & Veenstra-VanderWeele, J. The serotonin system in autism spectrum disorder: From biomarker to animal models. Neuroscience 321, 24–41 (2016).

    Article  PubMed  CAS  Google Scholar 

  11. Schain, R. J. & Freedman, D. X. Studies on 5-hydroxyindole metabolism in autistic and other mentally retarded children. J. Pediatr. 58, 315–320 (1961).

    Article  PubMed  CAS  Google Scholar 

  12. Weiss, L. A. et al. Association between microdeletion and microduplication at 16p11.2 and autism. N. Engl. J. Med. 358, 667–675 (2008).

    Article  PubMed  CAS  Google Scholar 

  13. Kumar, R. A. et al. Recurrent 16p11.2 microdeletions in autism. Hum. Mol. Genet. 17, 628–638 (2008).

    Article  PubMed  CAS  Google Scholar 

  14. Sanders, S. J. et al. Multiple recurrent de novo CNVs, including duplications of the 7q11.23 Williams syndrome region, are strongly associated with autism. Neuron 70, 863–885 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Christoffel, D. J. et al. IκB kinase regulates social defeat stress-induced synaptic and behavioral plasticity. J. Neurosci. 31, 314–321 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Walsh, J. J. et al. Stress and CRF gate neural activation of BDNF in the mesolimbic reward pathway. Nat. Neurosci. 17, 27–29 (2014).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Francis, T. C. et al. Nucleus accumbens medium spiny neuron subtypes mediate depression-related outcomes to social defeat stress. Biol. Psychiatry 77, 212–222 (2015).

    Article  PubMed  Google Scholar 

  19. Wallace, D. L. et al. CREB regulation of nucleus accumbens excitability mediates social isolation-induced behavioral deficits. Nat. Neurosci. 12, 200–209 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Luo, M., Zhou, J. & Liu, Z. Reward processing by the dorsal raphe nucleus: 5-HT and beyond. Learn. Mem. 22, 452–460 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Gong, S. et al. Targeting Cre recombinase to specific neuron populations with bacterial artificial chromosome constructs. J. Neurosci. 27, 9817–9823 (2007).

    Article  PubMed  CAS  Google Scholar 

  22. Veenstra-VanderWeele, J. et al. Autism gene variant causes hyperserotonemia, serotonin receptor hypersensitivity, social impairment and repetitive behavior. Proc. Natl Acad. Sci. USA 109, 5469–5474 (2012).

    Article  ADS  PubMed  Google Scholar 

  23. Portmann, T. et al. Behavioral abnormalities and circuit defects in the basal ganglia of a mouse model of 16p11.2 deletion syndrome. Cell Reports 7, 1077–1092 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Burns, K. A. et al. Nestin-CreER mice reveal DNA synthesis by nonapoptotic neurons following cerebral ischemia hypoxia. Cereb. Cortex 17, 2585–2592 (2007).

    Article  PubMed  Google Scholar 

  25. Horev, G. et al. Dosage-dependent phenotypes in models of 16p11.2 lesions found in autism. Proc. Natl Acad. Sci. USA 108, 17076–17081 (2011).

    Article  ADS  PubMed  Google Scholar 

  26. Grimm, D. et al. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 82, 5887–5911 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Tsai, H. C. et al. Phasic firing in dopaminergic neurons is sufficient for behavioral conditioning. Science 324, 1080–1084 (2009).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  28. Stuber, G. D., Britt, J. P. & Bonci, A. Optogenetic modulation of neural circuits that underlie reward seeking. Biol. Psychiatry 71, 1061–1067 (2012).

    Article  PubMed  Google Scholar 

  29. Steinberg, E. E. & Janak, P. H. Establishing causality for dopamine in neural function and behavior with optogenetics. Brain Res. 1511, 46–64 (2013).

    Article  PubMed  CAS  Google Scholar 

  30. Lammel, S., Lim, B. K. & Malenka, R. C. Reward and aversion in a heterogeneous midbrain dopamine system. Neuropharmacology 76 Pt B, 351–359 (2014).

    Article  PubMed  CAS  Google Scholar 

  31. Steinbusch, H. W., van der Kooy, D., Verhofstad, A. A. & Pellegrino, A. Serotonergic and non-serotonergic projections from the nucleus raphe dorsalis to the caudate-putamen complex in the rat, studied by a combined immunofluorescence and fluorescent retrograde axonal labeling technique. Neurosci. Lett. 19, 137–142 (1980).

    Article  PubMed  CAS  Google Scholar 

  32. Steinbusch, H. W. Distribution of serotonin-immunoreactivity in the central nervous system of the rat-cell bodies and terminals. Neuroscience 6, 557–618 (1981).

    Article  PubMed  CAS  Google Scholar 

  33. Hornung, J. P. The human raphe nuclei and the serotonergic system. J. Chem. Neuroanat. 26, 331–343 (2003).

    Article  PubMed  CAS  Google Scholar 

  34. Michelsen, K. A., Prickaerts, J. & Steinbusch, H. W. The dorsal raphe nucleus and serotonin: implications for neuroplasticity linked to major depression and Alzheimer’s disease. Prog. Brain Res. 172, 233–264 (2008).

    Article  PubMed  CAS  Google Scholar 

  35. Politte, L. C., Henry, C. A. & McDougle, C. J. Psychopharmacological interventions in autism spectrum disorder. Harv. Rev. Psychiatry 22, 76–92 (2014).

    Article  PubMed  Google Scholar 

  36. Heifets, B. D. & Malenka, R. C. MDMA as a probe and treatment for social behaviors. Cell 166, 269–272 (2016).

    Article  PubMed  CAS  Google Scholar 

  37. Volkow, N. D., Fowler, J. S., Wang, G. J. & Swanson, J. M. Dopamine in drug abuse and addiction: results from imaging studies and treatment implications. Mol. Psychiatry 9, 557–569 (2004).

    Article  PubMed  CAS  Google Scholar 

  38. Qi, J. et al. A glutamatergic reward input from the dorsal raphe to ventral tegmental area dopamine neurons. Nat. Commun. 5, 5390 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Liu, Z. et al. Dorsal raphe neurons signal reward through 5-HT and glutamate. Neuron 81, 1360–1374 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. McDevitt, R. A. et al. Serotonergic versus nonserotonergic dorsal raphe projection neurons: differential participation in reward circuitry. Cell Reports 8, 1857–1869 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Warden, M. R. et al. A prefrontal cortex-brainstem neuronal projection that controls response to behavioural challenge. Nature 492, 428–432 (2012).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  43. Fonseca, M. S., Murakami, M. & Mainen, Z. F. Activation of dorsal raphe serotonergic neurons promotes waiting but is not reinforcing. Curr. Biol. 25, 306–315 (2015).

    Article  PubMed  CAS  Google Scholar 

  44. Correia, P. A. et al. Transient inhibition and long-term facilitation of locomotion by phasic optogenetic activation of serotonin neurons. eLife 6, e20975 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Marcinkiewcz, C. A. et al. Serotonin engages an anxiety and fear-promoting circuit in the extended amygdala. Nature 537, 97–101 (2016).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  46. Franklin, K. B. J. & Paxinos, G. The Mouse Brain in Stereotaxic Coordinates 4th edn (Academic, 2012).

  47. Sharp, T., Bramwell, S. R., Clark, D. & Grahame-Smith, D. G. In vivo measurement of extracellular 5-hydroxytryptamine in hippocampus of the anaesthetized rat using microdialysis: changes in relation to 5-hydroxytryptaminergic neuronal activity. J. Neurochem. 53, 234–240 (1989).

    Article  PubMed  CAS  Google Scholar 

  48. Hayashi, K., Nakao, K. & Nakamura, K. Appetitive and aversive information coding in the primate dorsal raphé nucleus. J. Neurosci. 35, 6195–6208 (2015).

    Article  PubMed  CAS  Google Scholar 

  49. Kaidanovich-Beilin, O., Lipina, T., Vukobradovic, I., Roder, J. & Woodgett, J. R. Assessment of social interaction behaviors. J. Vis. Exp. 48, 2473 (2011).

    Google Scholar 

  50. Lammel, S. et al. Input-specific control of reward and aversion in the ventral tegmental area. Nature 491, 212–217 (2012).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This study was supported by the Simons Foundation Autism Research Initiative (award 305112 to R.C.M.) and NIMH (F32 MH103949 to J.J.W.).

Reviewer information

Nature thanks G. Feng, M. Lobo and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

J.J.W. performed the majority of experiments. D.J.C. performed the electrophysiology experiments. B.D.H. performed the fibre photometry experiments. G.A.B.-D., A.S. and L.W.H. assisted in surgeries and behavioural assays. J.J.W. and R.C.M. designed the experiments, interpreted results and wrote the paper, which was edited by all authors.

Corresponding author

Correspondence to Robert C. Malenka.

Ethics declarations

Competing interests

R.C.M. and K.D. are cofounders and on the scientific advisory board of Circuit Therapeutics, Inc.

Additional information

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 Activation of DR neurons or their NAc projections increases sociability, and bidirectional modulation of DR 5-HT neuron activity bidirectionally modifies sociability.

a, b, Quantification of chamber time (a: eYFP, F6,45 = 0.6823, P = 0.6647, n = 6; ChR2, F6,72 = 17.21, P < 0.01; n = 9) and proximity time (b: eYFP, F3,30 = 0.7517, P = 0.5300, n = 6; ChR2, F3,48 = 32.96, P < 0.01, n = 9) in the three-chamber assay. c, Quantification of centre time in the locomotion assay (c: F5,65 = 1.263, P = 0.2908, n = 6–9). d, e, Quantification of chamber time (d: eYFP, F6,54 = 0.2602, P = 0.9529, n = 7; ChR2, F6,72 = 19.36, P < 0.01, n = 9) and proximity time (e: eYFP, F3,36 = 0.3456, P = 0.7925, n = 7; ChR2, F3,48 = 26.44, P < 0.01, n = 9) in the three-chamber assay. f, Quantification of centre time in the locomotion assay (F5,40 = 0.7786, P = 0.5710, n = 5). g, h, Quantification of chamber time (g: eYFP, F6,54 = 0.7293, P = 0.6280, n = 7; ChR2, F6,72 = 3.812, P < 0.05, n = 9) and proximity time (h: eYFP, F3,36 = 0.9256, P = 0.4383, n = 7; ChR2, F3,48 = 4.844, P < 0.05, n = 9) in the three-chamber assay. i, j, Quantification of chamber time (i: eYFP, F6,54 = 1.058, P = 0.3989, n = 7; NpHR, F6,81 = 13.04, P < 0.01, n = 10) and proximity time (j: eYFP, F3,36 = 1.661, P = 0.1926, n = 7; NpHR, F3,54 = 16.29, P < 0.01, n = 10) in the three-chamber assay. k, l, Quantification of novel object interaction assay (k: F1,52 = 0.01018, P = 0.9200, n = 13–15), locomotion assay (l: F1,52 = 0.7626, P = 0.3865, n = 13–15), and centre time (m: F5,130 = 0.766, P = 0.5759, n = 13–15) in Sert-cre mice expressing DIO-eYFP or DIO-ChR2 in DR receiving soma stimulation. np, Quantification of novel object interaction assay (n: F1,30 = 0.04112, P = 0.8407, n = 7–10), locomotion assay (o: F1,30 = 0.3837, P = 0.5403, n = 7–10), and centre time (p: F5,80 = 1.195, P = 0.3190, n = 8–10) in Sert-cre mice expressing DIO-eYFP or DIO-NpHR in DR receiving soma stimulation. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant.

Source Data

Extended Data Fig. 2 Bidirectional modulation of DR-to-NAc 5-HT terminals modifies sociability, but not control behaviours.

a, b, Quantification of chamber time (a: eYFP, F6,63 = 1.383, P = 0.2352, n = 8; ChR2, F6,72 = 4.891, P < 0.05, n = 9) and proximity time (b: eYFP, F3,42 = 0.9652, P = 0.4181, n = 8; ChR2, F3,48 = 7.565, P < 0.05, n = 9) in the three-chamber assay. c, d, Quantification of chamber time (c: eYFP, F6,81 = 1.626, P = 0.1506, n = 10; NpHR, F6,81 = 6.253, P < 0.05, n = 10) and proximity time (d: eYFP, F3,54 = 2.304, P = 0.0872, n = 10; NpHR, F3,54 = 7.821, P < 0.05, n = 10) in the three-chamber assay. eg, Quantification of novel object interaction assay (e: F1,30 = 0.00206, P = 0.9641, n = 8–9), locomotion assay (f: F1,30 = 0.03023, P = 0.8631, n = 8–9), and centre time (g: F5,75 = 1.205, P = 0.3151, n = 8–9) in Sert-cre mice expressing DIO-eYFP or DIO-ChR2 in DR receiving DR-to-NAc terminal stimulation. hj, Quantification of novel object interaction assay (h: F1,38 = 0.213, P = 0.6471, n = 10–11), locomotion assay (i: F1,34 = 1.077, P = 0.3066, n = 9–10), and centre time (j: F5,90 = 0.1646, P = 0.9749, n = 9–10) in Sert-cre mice expressing DIO-eYFP or DIO-NpHR in DR receiving DR-to-NAc terminal stimulation. Data are mean ± s.e.m. *P < 0.05, **P < 0.01; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant.

Source Data

Extended Data Fig. 3 16p11.2 deletion decreases sociability, but not an anxiety-related behaviour.

a, b, Quantification of chamber time (a: control, F2,45 = 39.5, P < 0.001, n = 16; heterozygous, F2,21 = 23.39, P < 0.001, n = 8; homozygous, F2,21 = 31.54, P < 0.001, n = 8) and proximity time (b: control, F1,30 = 14.61, P < 0.001, n = 16; heterozygous, F1,14 = 10.14, P < 0.01, n = 8; homozygous, F1,14 = 11.88, P < 0.01, n = 8) in the three-chamber assay. c, Quantification of centre time (c: F10,135 = 1.03, P = 0.4215, n = 7–15) in control, heterozygous 16p11.2flx:Nes-creER and homozygous 16p11.2flx:Nes-creER mice. d, e, Quantification of chamber time in the three-chamber assay (d: 16p11.2flx:∆cre, F2,24 = 121.2, P < 0.001, n = 9; 16p11.2flx:cre, F2,24 = 27.86, P < 0.001, n = 9; Sert-cre:16p11.2flx, F2,21 = 84.95, P < 0.001, n = 8) and proximity time (e: 16p11.2flx:∆cre, F1,16 = 26.13, P < 0.001, n = 9; 16p11.2flx:cre, F1,16 = 6.885, P < 0.05, n = 9; Sert-cre:16p11.2flx, F1,14 = 44.00, P < 0.001, n = 8). f, Quantification of centre time in (c: F2,22 = 0.6019, P = 0.5565, n = 8–9) 16p11.2flx:∆cre, 16p11.2flx:cre and Sert-cre:16p11.2flx mice. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant.

Source Data

Extended Data Fig. 4 Optogenetic activation of DR neurons reverses social deficits induced by 16p11.2 deletion, but does not alter control behaviours.

a, Schematic of AAV-DJ-Cre and DIO-ChR2 injected into and optic fibre implanted above the DR in 16p11.2flx mice. b, Timeline of behavioural experiments. c, d, Quantification of sociability during juvenile interaction (cF1,36 = 62.43, P < 0.001, n = 10) and the three-chamber sociability assay (dF1,34 = 2.298, P < 0.05, n = 9–10) in 16p11.2flx mice expressing DIO-eYFP or DIO-ChR2 and AAV-DJ-Cre in DR receiving soma stimulation. e, f, Quantification of chamber and proximity time in the three-chamber assay (e: eYFP, F2,24 = 92.48, P < 0.001, n = 9 (left); F1,16 = 17.53, P < 0.05, n = 9 (right); f: ChR2, F6,81 = 3.085, P < 0.05, n = 10 (left); F3,54 = 3.493, P < 0.05, n = 10 (right)). gi, Quantification of novel object interaction assay (g: F1,36 = 0.956, P = 0.3424, n = 10), locomotion assay (h: F1,36 = 1.962, P = 0.1698, n = 10), and centre time (i: F5,90 = 0.7668, P = 0.5761, n = 10) in 16p11.2flx mice. Data are mean ± s.e.m. *P < 0.05, ***P < 0.001; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant. The schematic of the mouse brain in this figure has been adapted with permission from Franklin & Paxinos46.

Source Data

Extended Data Fig. 5 Optogenetic activation of DR 5-HT neurons or DR-to-NAc 5-HT terminals rescues social deficits induced by 16p11.2 deletion, but does not alter control behaviours.

a, b, Quantification of chamber and proximity time in the three-chamber assay (a: eYFP, F2,21 = 84.95, P < 0.001, n = 8 (left); F1,14 = 44.00, P < 0.001, n = 8 (right); b: ChR2, F6,81 = 9.26, P < 0.001, n = 10 (left); F3,54 = 11.54, P < 0.001, n = 10 (right)). ce, Quantification of the novel object interaction assay (c: F1,32 = 0.03819, P = 0.8463, n = 9), locomotion assay (d: F1,32 = 0.141, P = 0.7097, n = 8–10), and centre time (e: F5,80 = 0.195, P = 0.9636, n = 8–10) in Sert-cre:16p11.2flx mice expressing DIO-eYFP or DIO-ChR2 in DR receiving soma stimulation. f, g, Quantification of chamber and proximity time in the three-chamber assay (f: eYFP, F2,27 = 73.89, P < 0.001, n = 10 (left); F1,18 = 63.38, P < 0.001, n = 10 (right); g: ChR2, F6,81 = 11.33, P < 0.001, n = 10 (left); F3,54 = 14.55, P < 0.001, n = 10 (right)). hj, Quantification of novel object interaction assay (h: F1,36 = 0.01038, P = 0.9194, n = 10), locomotion assay (i: F1,32 = 0.3655, P = 0.5497, n = 9), and centre time (j: F5,90 = 0.9092, P = 0.4788, n = 10) in Sert-cre:16p11.2flx mice expressing DIO-eYFP or DIO-ChR2 in DR receiving DR-to-NAc 5-HT terminal stimulation. Data are mean ± s.e.m. ***P < 0.001; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant.

Source Data

Extended Data Fig. 6 5-HT release in the NAc is not acutely reinforcing.

a, b, Quantification of chamber time in the real-time CPP assay for 0 Hz (a) and 20 Hz (b) stimulation for initial (left) and reversal (right) stimulations (a: 0 Hz initial F2,16 = 10.22, P < 0.05, n = 9; 0 Hz reversal F2,16 = 29.01, P < 0.001, n = 9; b: 20 Hz initial F2,16 = 19.37, P < 0.001, n = 9; 20 Hz reversal F2,16 = 7.53, P < 0.01, n = 9) in Sert-cre mice receiving DR-to-NAc terminal stimulation. c, d, Quantification of chamber time in the real-time CPP assay for 0 Hz (c) and 20 Hz (d) stimulation for initial (left) and reversal (right) stimulations (c: 0 Hz initial F2,16 = 12.44, P < 0.001, n = 9; 0 Hz reversal F2,16 = 19.92, P < 0.001, n = 9; d: 20 Hz initial F2,16 = 8.56, P < 0.01, n = 9; 20 Hz reversal F2,16 = 9.517, P < 0.001, n = 9) in Sert-cre:16p11.2flx mice receiving DR-to-NAc terminal stimulation. e, Quantification of nosepokes for active and inactive ports for Sert-cre and Sert-cre:16p11.2flx mice (Sert-cre F2,32 = 1.9, P = 0.1655, n = 9; Sert-cre:16p11.2flx F2,32 = 0.25, P = 0.7821, n = 9). f, g, Quantification of chamber time (left) and proximity time (right) in high-fat food three-chamber assay for Sert-cre mice (F6,72 = 0.6713, P = 0.6731, n = 9; F3,48 = 1.495, P = 0.2279, n = 10) and Sert-cre:16p11.2flx mice (F6,72 = 0.4006, P = 0.8763, n = 10; F3,48 = 0.4157, P = 0.7425, n = 10) (g). Post-hoc analysis showed no significant difference between stimulated and non-stimulated chambers. *P < 0.05, **P < 0.01, ***P < 0.001; repeated measures, one-way (ad) or two-way (f, g) ANOVA with Tukey’s multiple comparison post hoc test, or one-way ANOVA with Sidak’s multiple comparison post hoc test comparing active to inactive nosepokes (e). Comparisons with no asterisk had P > 0.05 and were considered not significant.

Source Data

Extended Data Fig. 7 Activation of DR 5-HT terminals in the dorsal striatum does not enhance sociability nor rescue social deficits induced by 16p11.2 deletion.

a, Schematic of DIO-ChR2-eYFP injected into the DR and optic fibre implanted above the dorsal striatum (DS) in Sert-cre or Sert-cre:16p11.2flx mice. b, Timeline of behavioural experiments. c, d, Quantification of sociability during the juvenile interaction assay (cF1,14 = 0.5429, P = 0.4734, n = 4–5) and sociability in the three-chamber assay (dF1,14 = 0.055, P = 0.8177, n = 4–5) in Sert-cre or Sert-cre:16p11.2flx mice expressing DIO-eYFP or DIO-ChR2 with terminal stimulation of DR 5-HT neurons in the dorsal striatum. e, f, Quantification of the three-chamber assay showing chamber (left) and proximity (right) time (e: Sert-cre, F6,36 = 1.078, P = 0.3939, n = 5 (left); F3,24 = 2.009, P = 0.1395, n = 5 (right); f: Sert-cre:16p11.2flx, F2,9 = 16.44, P < 0.001, n = 4 (left); F1,6 = 11.2, P < 0.01, n = 4 (right)). gi, Quantification of novel object interaction assay (g: F1,14 = 0.058, P = 0.8125, n = 4–5), locomotion assay (h: F1,14 = 0.6195, P = 0.4444, n = 4–5), and centre time (i: F1,7 = 8.292, P < 0.05, n = 4–5) in Sert-cre or Sert-cre:16p11.2flx mice. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and considered were not significant. The schematic of the mouse brain in this figure has been adapted with permission from Franklin & Paxinos46.

Source Data

Extended Data Fig. 8 Activation of DR-to-NAc 5-HT terminals does not elicit long-lasting effects on sociability.

aj, Quantification of sociability during juvenile interaction assay on day 1 of stimulation (a: F1,16 = 10.65, P < 0.001, n = 9), day 2 (b: F1,16 = 16.71, P < 0.01, n = 9), day 3 session 1 (c: F1,16 = 17.7, P < 0.01, n = 9), session 2 (d: F1,16 = 27.05, P < 0.05, n = 9), session 3 (e: F1,16 = 13.35, P < 0.01, n = 9), session 4 (f: F1,16 = 25.03, P < 0.05, n = 9) and day 4 session 1 (g: F1,16 = 17.41, P < 0.001, n = 9), session 2 (h: F1,16 = 39.76, P < 0.01, n = 9), session 3 (i: F1,16 = 35.49, P < 0.001, n = 9), and session 4 (j: F1,16 = 8.295, P < 0.01, n = 9) in Sert-cre or Sert-cre:16p11.2flx mice expressing DIO-ChR2 with DR-to-NAc 5-HT terminal stimulation. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test.

Source Data

Extended Data Fig. 9 5-HT1b receptor antagonist infusion into NAc blocks enhanced sociability due to DR 5-HT neuron stimulation.

a, Quantification of chamber and proximity time in the three-chamber assay (a: ChR2 + vehicle, F6,54 = 15.15, P < 0.001, n = 7 (left); F3,36 = 18.00, P < 0.001, n = 7 (right); b: ChR2 + NAS-181, F6,45 = 1.479, P = 0.2069, n = 6 (left); F3,30 = 1.926, P = 0.1467, n = 6 (right)) in Sert-cre mice expressing DIO-ChR2 in DR with either vehicle (red) or NAS-181 (pink) infused into the NAc before analysis of behaviour. c, d, Quantification of sociability during the juvenile interaction assay (c: F1,24 = 0.004638, P = 0.9463, n = 7) and the three-chamber assay (d: F1,22 = 0.1686, P = 0.1686, n = 6–7) in Sert-cre mice expressing DIO-eYFP in DR with either vehicle (black) or NAS-181 (grey) infused into NAc before behaviour. e, f, Quantification of the three-chamber assay showing chamber and proximity time (e: eYFP + vehicle, F6,54 = 12.36, P < 0.001, n = 7 (left); F3,36 = 15.98, P < 0.01, n = 7 (right); f: eYFP + NAS-181, F6,45 = 0.4512, P = 0.8403, n = 6 (left); F3,30 = 0.7365, P = 0.7365, n = 6 (right)) in Sert-cre mice expressing DIO-eYFP in DR with either vehicle (grey) or NAS-181 (blue) infused into the NAc before analysis of behaviour. gi, Quantification of the novel object interaction assay (g: F1,24 = 0.0791, P = 0.7809, n = 7), locomotion assay (h: F1,24 = 0.8954, P = 0.3535, n = 7), and centre time (i: F5,60 = 0.6042, P = 0.6969, n = 7) in Sert-cre mice expressing DIO-eYFP in DR with either vehicle (grey) or NAS-181 (blue) infused into the NAc before behaviour. Data are mean ± s.e.m. ***P < 0.001; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant.

Source Data

Extended Data Fig. 10 5-HT1b receptor antagonist infusion into the NAc does not alter control behaviours and blocks rescue of 16p11.2 deletion social deficits by DR 5-HT neuron stimulation.

ac, Quantification of the novel object interaction assay (a: F1,24 = 0.0579, P = 0.8120, n = 7), locomotion assay (b: F1,24 = 0.4764, P = 0.4967, n = 7), and centre time (c: F5,60 = 0.3936, P = 0.8513, n = 7) in Sert-cre mice expressing DIO-ChR2 in DR with either vehicle (red) or NAS-181 (pink) infused into the NAc before behaviour. d, e, Quantification of chamber and proximity time in the three-chamber assay (d: ChR2 + vehicle, F6,90 = 9.03, P < 0.05, n = 11 (left); F3,60 = 13.04, P < 0.05, n = 11 (right); e: ChR2 + NAS-181, F6,90 = 1.226, P = 0.3004, n = 11 (left); F3,60 = 1.69, P = 0.1786, n = 11 (right)) in Sert-cre:16p11.2flx mice expressing DIO-ChR2 in DR with either vehicle (red) or NAS-181 (aqua) infused into the NAc before behaviour assays during optogenetic stimulation. fh, Quantification of novel object interaction assay (f: F1,18 = 01.263, P = 0.2758, n = 5–6), locomotion assay (g: F1,18 = 0.0344, P = 0.8549, n = 5–6), and centre time (h: F5,45 = 1.22, P = 0.3155, n = 5–6) in Sert-cre:16p11.2flx mice expressing DIO-ChR2 in DR with either vehicle (black) or NAS-181 (blue) infused into the NAc before behaviour assays. Data are mean ± s.e.m. *P < 0.05; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant.

Source Data

Extended Data Fig. 11 Infusion of 5-HT1b receptor agonist into the NAc increases sociability, but does not alter control behaviours.

a, Schematic of 5-HT1b receptor agonist (CP93129) infusion into the NAc in Sert-cre or Sert-cre:16p11.2flx mice. b, Timeline of behavioural experiments. c, d, Quantification of chamber and proximity time in the three-chamber assay (c: vehicle, F2,15 = 45.82, P < 0.001 (left); F3,30 = 4.453, P < 0.05 (right), n = 6; d: CP93129, F2,15 = 63.11, P < 0.001 (left); F1,10 = 17.23, P < 0.05, (right), n = 6) in Sert-cre mice with either vehicle (red) or CP93129 (aqua) infused into the NAc before analysis of behaviour. e, f, Quantification of chamber and proximity time in the three-chamber assay (e: vehicle, F2,12 = 67.49, P < 0.001(left); F1,8 = 41.03, P < 0.001 (right), n = 5; f: CP93129, F2,12 = 29.73, P < 0.001 (left); F1,8 = 17.17, P < 0.01 (right), n = 5) in Sert-cre:16p11.2flx mice with either vehicle (blue) or CP93129 (pink) infused into the NAc before analysis of behaviour. gi, Quantification of the novel object interaction assay (g: F1,18 = 1.612, P = 0.2204, n = 5–6), locomotion assay (h: F1,18 = 0.0149, P = 0.9041, n = 5–6), and centre time (i: F5,50 = 1.492, P = 0.2093; F5,40 = 0.4561, P = 0.8063, n = 5–6) in Sert-cre and Sert-cre:16p11.2flx mice with either vehicle or NAS-181 infused into the NAc before behaviour. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, ***P < 0.001; repeated measures, two-way ANOVA with Tukey’s multiple comparison post hoc test. Comparisons with no asterisk had P > 0.05 and were considered not significant. The schematic of the mouse brain in this figure has been adapted with permission from Franklin & Paxinos46.

Source Data

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Walsh, J.J., Christoffel, D.J., Heifets, B.D. et al. 5-HT release in nucleus accumbens rescues social deficits in mouse autism model. Nature 560, 589–594 (2018). https://doi.org/10.1038/s41586-018-0416-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0416-4

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