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.

  • Letter
  • Published:

Basal forebrain projections to the lateral habenula modulate aggression reward

Abstract

Maladaptive aggressive behaviour is associated with a number of neuropsychiatric disorders1 and is thought to result partly from the inappropriate activation of brain reward systems in response to aggressive or violent social stimuli2. Nuclei within the ventromedial hypothalamus3,4,5, extended amygdala6 and limbic7 circuits are known to encode initiation of aggression; however, little is known about the neural mechanisms that directly modulate the motivational component of aggressive behaviour8. Here we established a mouse model to measure the valence of aggressive inter-male social interaction with a smaller subordinate intruder as reinforcement for the development of conditioned place preference (CPP). Aggressors develop a CPP, whereas non-aggressors develop a conditioned place aversion to the intruder-paired context. Furthermore, we identify a functional GABAergic projection from the basal forebrain (BF) to the lateral habenula (lHb) that bi-directionally controls the valence of aggressive interactions. Circuit-specific silencing of GABAergic BF–lHb terminals of aggressors with halorhodopsin (NpHR3.0) increases lHb neuronal firing and abolishes CPP to the intruder-paired context. Activation of GABAergic BF–lHb terminals of non-aggressors with channelrhodopsin (ChR2) decreases lHb neuronal firing and promotes CPP to the intruder-paired context. Finally, we show that altering inhibitory transmission at BF–lHb terminals does not control the initiation of aggressive behaviour. These results demonstrate that the BF–lHb circuit has a critical role in regulating the valence of inter-male aggressive behaviour and provide novel mechanistic insight into the neural circuits modulating aggression reward processing.

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

Figure 1: Individual differences in aggression-related reward behaviour.
Figure 2: GABAergic BF–lHb circuit is differentially activated by intruder interactions.
Figure 3: BF–lHb activity bi-directionally modulates aggression reward.
Figure 4: BF–lHb does not initiate attack but modulates aggression severity.

Similar content being viewed by others

References

  1. Anderson, D. J. Optogenetics, sex, and violence in the brain: implications for psychiatry. Biol. Psychiatry 71, 1081–1089 (2012)

    PubMed  Google Scholar 

  2. Decety, J., Michalska, K. J., Akitsuki, Y. & Lahey, B. B. Atypical empathic responses in adolescents with aggressive conduct disorder: a functional MRI investigation. Biol. Psychol. 80, 203–211 (2009)

    PubMed  Google Scholar 

  3. 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)

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Wasman, M. & Flynn, J. P. Directed attack elicited from hypothalamus. Arch. Neurol. 6, 220–227 (1962)

    CAS  PubMed  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  7. Yu, Q. et al. Optogenetic stimulation of DAergic VTA neurons increases aggression. Mol. Psychiatry 19, 635 (2014)

    CAS  PubMed  Google Scholar 

  8. Takahashi, A. & Miczek, K. A. Neurogenetics of aggressive behavior: studies in rodents. Curr. Top. Behav. Neurosci. 17, 3–44 (2013)

    Google Scholar 

  9. Kudryavtseva, N. N., Bakshtanovskaya, I. V. & Koryakina, L. A. Social model of depression in mice of C57BL/6J strain. Pharmacol. Biochem. Behav. 38, 315–320 (1991)

    CAS  PubMed  Google Scholar 

  10. Golden, S. A., Covington, H. E. III, Berton, O. & Russo, S. J. A standardized protocol for repeated social defeat stress in mice. Nat. Protocols 6, 1183–1191 (2011)

    CAS  PubMed  Google Scholar 

  11. Miczek, K. A., DeBold, J. F. & Thompson, M. L. Pharmacological, hormonal, and behavioral manipulations in analysis of aggressive behavior. Prog. Clin. Biol. Res. 167, 1–26 (1984)

    CAS  PubMed  Google Scholar 

  12. Glenn, A. L. & Yang, Y. The potential role of the striatum in antisocial behavior and psychopathy. Biol. Psychiatry 72, 817–822 (2012)

    PubMed  Google Scholar 

  13. Zahm, D. S., Parsley, K. P., Schwartz, Z. M. & Cheng, A. Y. On lateral septum-like characteristics of outputs from the accumbal hedonic “hotspot” of Peciña and Berridge with commentary on the transitional nature of basal forebrain “boundaries”. J. Comp. Neurol. 521, 50–68 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Callaway, E. M. & Luo, L. Monosynaptic circuit tracing with glycoprotein-deleted rabies viruses. J. Neurosci. 35, 8979–8985 (2015)

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Herkenham, M. & Nauta, W. J. Afferent connections of the habenular nuclei in the rat. A horseradish peroxidase study, with a note on the fiber-of-passage problem. J. Comp. Neurol. 173, 123–145 (1977)

    CAS  PubMed  Google Scholar 

  17. Sutherland, R. J. The dorsal diencephalic conduction system: a review of the anatomy and functions of the habenular complex. Neurosci. Biobehav. Rev. 6, 1–13 (1982)

    CAS  PubMed  Google Scholar 

  18. Lecca, S., Meye, F. J. & Mameli, M. The lateral habenula in addiction and depression: an anatomical, synaptic and behavioral overview. Eur. J. Neurosci. 39, 1170–1178 (2014)

    PubMed  Google Scholar 

  19. Shelton, L. et al. Mapping pain activation and connectivity of the human habenula. J. Neurophysiol. 107, 2633–2648 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hikosaka, O. The habenula: from stress evasion to value-based decision-making. Nature Rev. Neurosci. 11, 503–513 (2010)

    CAS  Google Scholar 

  21. Lee, E. H. & Huang, S. L. Role of lateral habenula in the regulation of exploratory behavior and its relationship to stress in rats. Behav. Brain Res. 30, 265–271 (1988)

    CAS  PubMed  Google Scholar 

  22. Maroteaux, M. & Mameli, M. Cocaine evokes projection-specific synaptic plasticity of lateral habenula neurons. J. Neurosci. 32, 12641–12646 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, B. et al. Synaptic potentiation onto habenula neurons in the learned helplessness model of depression. Nature 470, 535–539 (2011)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yetnikoff, L., Cheng, A. Y., Lavezzi, H. N., Parsley, K. P. & Zahm, D. S. Sources of input to the rostromedial tegmental nucleus, ventral tegmental area, and lateral habenula compared: a study in rat. J. Comp. Neurol. 523, 2426–2456 (2015)

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Quina, L. A. et al. Efferent pathways of the mouse lateral habenula. J. Comp. Neurol. 523, 32–60 (2015)

    PubMed  Google Scholar 

  26. Felton, T. M., Linton, L., Rosenblatt, J. S. & Morell, J. I. First and second order maternal behavior related afferents of the lateral habenula. Neuroreport 10, 883–887 (1999)

    CAS  PubMed  Google Scholar 

  27. Beck, A., Heinz, A. J. & Heinz, A. Translational clinical neuroscience perspectives on the cognitive and neurobiological mechanisms underlying alcohol-related aggression. Curr. Top. Behav. Neurosci. 17, 443–474 (2014)

    Google Scholar 

  28. Martin, L. A., Neighbors, H. W. & Griffith, D. M. The experience of symptoms of depression in men vs women: analysis of the National Comorbidity Survey Replication. JAMA Psychiatry 70, 1100–1106 (2013)

    Google Scholar 

  29. Bewernick, B. H., Kayser, S., Sturm, V. & Schlaepfer, T. E. Long-term effects of nucleus accumbens deep brain stimulation in treatment-resistant depression: evidence for sustained efficacy. Neuropsychopharmacology 37, 1975–1985 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Sartorius, A. & Henn, F. A. Deep brain stimulation of the lateral habenula in treatment resistant major depression. Med. Hypotheses 69, 1305–1308 (2007)

    PubMed  Google Scholar 

  31. Kudryavtseva, N. N., Bondar, N. P. & Avgustinovich, D. F. Association between experience of aggression and anxiety in male mice. Behav. Brain Res. 133, 83–93 (2002)

    PubMed  Google Scholar 

  32. Russo, S. J. et al. Nuclear factor kappa B signaling regulates neuronal morphology and cocaine reward. J. Neurosci. 29, 3529–3537 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Golden, S. A. et al. Epigenetic regulation of RAC1 induces synaptic remodeling in stress disorders and depression. Nature Med. 19, 337–344 (2013)

    ADS  CAS  PubMed  Google Scholar 

  34. Krishnan, V. et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell 131, 391–404 (2007)

    CAS  PubMed  Google Scholar 

  35. Yang, M., Silverman, J. L. & Crawley, J. N. Automated three-chambered social approach task for mice. Curr. Protoc. Neurosci. Chapter 8, Unit 8 26 (2011)

  36. Golde, W. T., Gollobin, P. & Rodriguez, L. L. A rapid, simple, and humane method for submandibular bleeding of mice using a lancet. Lab Anim. (NY) 34, 39–43 (2005)

    Google Scholar 

  37. Chaudhury, D. et al. Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature 493, 532–536 (2013)

    ADS  CAS  PubMed  Google Scholar 

  38. Friedman, A. K. et al. Enhancing depression mechanisms in midbrain dopamine neurons achieves homeostatic resilience. Science 344, 313–319 (2014)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lobo, M. K. et al. ΔFosB induction in striatal medium spiny neuron subtypes in response to chronic pharmacological, emotional, and optogenetic stimuli. J. Neurosci. 33, 18381–18395 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lobo, M. K. et al. Cell type-specific loss of BDNF signaling mimics optogenetic control of cocaine reward. Science 330, 385–390 (2010)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Stamatakis, A. M. & Stuber, G. D. Activation of lateral habenula inputs to the ventral midbrain promotes behavioral avoidance. Nature Neurosci. 15, 1105–1107 (2012)

    CAS  PubMed  Google Scholar 

  42. Chandra, R. et al. Optogenetic inhibition of D1R containing nucleus accumbens neurons alters cocaine-mediated regulation of Tiam1. Front. Mol. Neurosci. 6, 13 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Aquili, L., Liu, A. W., Shindou, M., Shindou, T. & Wickens, J. R. Behavioral flexibility is increased by optogenetic inhibition of neurons in the nucleus accumbens shell during specific time segments. Learn. Mem. 21, 223–231 (2014)

    PubMed  PubMed Central  Google Scholar 

  44. Kadir, S. N., Goodman, D. F. M. & Harris, K. D. High-dimensional cluster analysis with the masked EM algorithm. Neural Comput. 26, 2379–2394 (2014)

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by US National Institutes of Health grants R01 MH090264, P50 MH096890 and P50 AT008661-01 (S.J.R.), R01 MH092306 (M.H.H.), T32 MH087004 (M.L.P., M.H. and M.F.), T32 MH096678 (M.L.P.), F30 MH100835 (M.H.), F31 MH105217 (M.L.P.), National Institute of General Medical Sciences 1FI2GM117583-01 (S.A.G.) and the Natural National Science Foundation of China 81200862 (H.Z.). We would like to thank K. Miczek and Y. Shaham for their input.

Author information

Authors and Affiliations

Authors

Contributions

S.A.G. and S.J.R. designed and wrote the manuscript. S.A.G., D.J.C., M.H., C.M., J.J.W., M.L.P., N.R. H.A., G.E.H., M.F., D.B., L.K., J.T. and B.K. collected behavioural and immunohistochemistry data and aided in data analysis. H.Z., M.-H.H., D.C., K.G. and M.L.S. designed, carried out and analysed electrophysiological experiments.

Corresponding author

Correspondence to Scott J. Russo.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks O. Hikosaka and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Social behaviours exhibited by resident CD-1 and intruder C57 mice during aggression screening.

a, Experimental schematic of aggression screening procedure used in a subset (40 residents and 40 intruders) of mice to quantify social behaviours. be, Bouts of attacks (F2,156 = 13.10, two-way ANOVA ***P < 0.0001; post-hoc test ***P < 0.001; n = 40 per group) (b), pursuits (c), withdrawals (F2,156 = 5.745, two-way repeated measures ANOVA **P < 0.001; post-hoc test ***P < 0.001; n = 40 per group) (d) and non-aggressive social approaches (e). fh, Duration of attacks (F2,156 = 7.069, two-way repeated measures ANOVA **P < 0.001; post-hoc test ***P < 0.001; n = 40 per group) (f), pursuits (g), withdrawals (h) and non-aggressive social approaches (e). All data are presented as mean ± s.e.m.

Extended Data Figure 2 Detailed ethological analysis of AGG aggression-related behaviours.

a, Experimental schematic of aggression screening procedure used in a sample (448 mice total; 138 NON and 310 AGG) of mice. b, Histogram of attack latency frequency using 10-s bins. ce, Mean distribution across screening sessions (left) and individual screening sessions (right) for latency to aggression (F2,1338 = 49.37, two-way repeated measures ANOVA P < 0.001; post-hoc test, *P < 0.001; n = 138–310) (c), number of attack bouts (F2,1338 = 21.03, two-way repeated measures ANOVA P < 0.001; post-hoc test, *P < 0.001; n = 138–310) (d) and mean attack duration (F2,1338 = 11.96, two-way repeated measures ANOVA P < 0.001; post-hoc test, *P < 0.001; n = 138–310) (e). f, g, Correlation of mean latency to initial aggression with mean attack bouts (r = −0.78, P < 0.0001) (f) and mean duration of attack bouts (r = −0.40, P < 0.0001) (g). Distribution plots are presented as the median with interquartile range and normality determined by D’Agostino–Pearson, Shapiro–Wilk and Kolmogorov–Smirnov normality tests (P < 0.0001). Summary data are represented as mean ± s.e.m.

Extended Data Figure 3 Aggression CPP behaviour.

a, Experimental schematic of aggression CPP procedure. b, c, Individual duration spent in the intruder-paired context for AGG (t7 = 3.106, *P < 0.05; two-tailed paired t-test, n = 8 per group) (b) and NON (t7 = 2.918, *P < 0.05; two-tailed paired t-test, n = 8 per group) (c). d, Duration spent in the middle neutral chamber during pre-test and test sessions. e, Experimental schematic of sensory CPP procedure. f, g, Individual duration spent in the intruder-paired context for AGG (f) and NON (g). h, Duration spent in the middle neutral chamber during pre-test and test sessions. Summary data are represented as mean ± s.e.m.

Extended Data Figure 4 BF–lHb circuit tracing and GABAergic cell-type specificity.

a, Schematic of viral tracing strategy. b, Representative BF viral infection with AAV2-hSyn-eYFP. Scale bar: 500 μm. c, Histological analysis of viral infection with AAV2-hSyn-eYFP (F3,11 = 223.0, one-way ANOVA ***P < 0.0001, post-hoc test, ***P < 0.0001; n = 3 mice, 3 slices per mouse) across adjacent anatomical regions. d, e, Whole-cell electrophysiological recordings (d) and representative traces of lHb neurons photostimulated with AAV2-hSyn-ChR2.0 in the absence or presence of bath-applied GABAA receptor antagonist gabazine (2 μm; F2,7 = 220, one-way ANOVA P < 0.05; post-hoc test, ***P < 0.001, n = 4, 2, 2 cells from 2 mice) (e). f, Optically evoked IPSC response delay (n = 21 oIPSC events, 2 mice). g, Representative images of eYFPBF→lHb terminal colocalization between vesicular GABA transporter (top), and not vesicular glutamate transporter 1 (bottom). Scale bars: 10 μm; white arrows indicate colocalization within insets. MS, medial septum; pLS, posterior lateral septum. Summary data are represented as mean ± s.e.m.

Extended Data Figure 5 Multiunit anaesthetized optrode recordings.

a, Schematic of in vivo anaesthetized multi-unit optrode recording procedure (left) and representative optrode placement in lHb (right; scale bar: 200 μm). b, c, Heatmaps of normalized firing rates for lHb neurons in response to BF terminal stimulation with ChR2BF→lHb (b) or NpHR3BF→lHb (c) and averaged spike wave-form shown below for pre-stimulation, stimulation and post-stimulation epochs. d, Percentage of cells by firing response (top) and average normalized lHb firing rate (bottom) after BF–lHb terminal stimulation with ChR2BF→lHb for all identified cells (F2,134 = 8.249, one-way repeated-measure ANOVA P < 0.001; post-hoc test, *P < 0.05; n = 68 cells from 3 mice) and cells that significantly decreased firing during the stimulation epoch (F7,105 = 8.868, one-way repeated-measure ANOVA P < 0.0001; post-hoc test, *P < 0.05; n = 16/68 cells from 3 mice). e, Percentage of cells by firing response (top) and average normalized lHb firing rate (bottom) after BF–lHb terminal stimulation with NpHR3BF→lHb for all identified cells (F2,128 = 10.32, one-way repeated-measure ANOVA P < 0.0001; post-hoc test, *P < 0.05; n = 65/65 cells from 3 mice) and cells that significantly increased firing during the stimulation epoch (F7,203 = 17.58, one-way repeated-measure ANOVA P < 0.0001; post-hoc test, *P < 0.05; n = 30/65 cells from 3 mice). mHb, medial habenula. Summary data are represented as mean ± s.e.m.

Extended Data Figure 6 BF–lHb AAV infection and CPP locomotor behaviour.

a, Schematic of BF coronal slice (left), alongside representative AAV-ChR2-eYFP (top) and AAV-NpHR3.0-eYFP (bottom) infections. Scale bar: 500 μm. b, Schematic of lHb coronal slice (left), alongside representative images of BF terminal infection by AAV-ChR2-eYFP (middle top) and AAV-NpHR3.0-eYFP (middle bottom) within the lHb. Scale bar: 200 μm. Representative close-ups of terminal regions shown in insets on right. Scale bar: 50 μm. All representative images counterstained with DAPI. c, d, Histological analysis of BF infection in NON (c) and AGG (d) mice. e, f, Histological analysis of habenular viral infection in NON (e) and AGG mice (f). gj, Total distance travelled (g, h) and mean velocity (i, j) between NON and AGG during the CPP pre-test and test phase. All data are presented as mean ± s.e.m., and are not significant as determined by two-way ANOVA, P < 0.05. dStr, dorsal striatum; mHb, medial habenula; MS, medial septum; pLS, posterior lateral septum.

Extended Data Figure 7 Direct lHb stimulation bi-directionally modulates aggression reward.

a, Schematic of viral infection strategy. b, c, Representative images of lHb cell body infection in NON (b) and AGG (c). Scale bar: 200 μm. d, Histological analysis of lHb viral infection. e, Representative CPP traces of NON. NON::NpHRlHb cell body infection mimics the physiological effect of NON::ChR2BF→lHb terminal stimulation. f, Normalized CPP score (t15 = 2.834, *P < 0.05; two-tailed unpaired t-test, n = 8–9 per group) and subtracted CPP score (t15 = 3.058, **P < 0.01; two-tailed unpaired t-test, n = 8–9 per group) in NON::eYFPlHb and NON::NpHRlHb. g, Individual duration spent in the intruder-paired context for NON::eYFPlHb (t9 = 0.9129, P > 0.05; two-tailed paired t-test, n = 10 per group) and NON::NpHRlHb (t9 = 2.344, *P < 0.05; two-tailed paired t-test, n = 10 per group). h, Representative CPP traces of AGG::eYFPlHb and AGG::ChR2lHb. i, Normalized CPP score (t18 = 2.692, *P < 0.05; two-tailed unpaired t-test, n = 9–11 per group) and subtracted CPP score (t18 = 4.203, ***P < 0.01; two-tailed unpaired t-test, n = 9–11 per group) for the intruder-paired context in AGG::eYFPlHb and AGG::ChR2lHb. j, Individual duration spent in the intruder-paired context for AGG::eYFPlHb mice (t10 = 3.212, **P < 0.01; two-tailed paired t-test, n = 9 per group) and AGG::ChR2lHb mice (t8 = 1.348, P < 0.05; two-tailed paired t-test, n = 11 per group). Summary data are represented as mean ± s.e.m. dStr, dorsal striatum; mHb, medial habenula.

Extended Data Figure 8 BF–lHb stimulation modulates cocaine CPP.

a, Experimental timeline of general anxiety and cocaine CPP testing. be, BF–lHb stimulation during open field testing (b, c) and elevated plus maze testing (d, e). f, Subthreshold cocaine (10 mg kg−1, intraperitoneal) CPP procedure with BF–lHb stimulation during CPP test (t9 = 2.403, P < 0.05; two-tailed unpaired t-test, n = 5–6 per group).

Extended Data Table 1 Stress and anxiety behaviours in AGG and NON
Extended Data Table 2 Social approach behaviours in AGG and NON

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Golden, S., Heshmati, M., Flanigan, M. et al. Basal forebrain projections to the lateral habenula modulate aggression reward. Nature 534, 688–692 (2016). https://doi.org/10.1038/nature18601

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18601

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