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:

Silent synapses dictate cocaine memory destabilization and reconsolidation

Abstract

Cocaine-associated memories are persistent, but, on retrieval, become temporarily destabilized and vulnerable to disruptions, followed by reconsolidation. To explore the synaptic underpinnings for these memory dynamics, we studied AMPA receptor (AMPAR)-silent excitatory synapses, which are generated in the nucleus accumbens by cocaine self-administration, and subsequently mature after prolonged withdrawal by recruiting AMPARs, echoing acquisition and consolidation of cocaine memories. We show that, on memory retrieval after prolonged withdrawal, the matured silent synapses become AMPAR-silent again, followed by re-maturation ~6 h later, defining the onset and termination of a destabilization window of cocaine memories. These synaptic dynamics are timed by Rac1, with decreased and increased Rac1 activities opening and closing, respectively, the silent synapse-mediated destabilization window. Preventing silent synapse re-maturation within the destabilization window decreases cue-induced cocaine seeking. Thus, cocaine-generated silent synapses constitute a discrete synaptic ensemble dictating the dynamics of cocaine-associated memories and can be targeted for memory disruption.

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: Memory retrieval re-silences cocaine-generated synapses.
Fig. 2: Spine morphology correlate with memory destabilization and reconsolidation.
Fig. 3: Synapse re-silencing destabilizes cocaine memories.
Fig. 4: Decreased Rac1 activity primes cue-induced synaptic re-silencing.
Fig. 5: Increasing Rac1 activity prevents cue-induced synaptic re-silencing.
Fig. 6: Active Rac1 stabilizes synaptic states to regulate cocaine memory.

Similar content being viewed by others

Data availability

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

References

  1. Hyman, S. E., Malenka, R. C. & Nestler, E. J. Neural mechanisms of addiction: the role of reward-related learning and memory. Annu. Rev. Neurosci. 29, 565–598 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Wolf, M. E. Synaptic mechanisms underlying persistent cocaine craving. Nat. Rev. Neurosci. 17, 351–365 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nader, K. & Hardt, O. A single standard for memory: the case for reconsolidation. Nat. Rev. Neurosci. 10, 224–234 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Tronson, N. C. & Taylor, J. R. Molecular mechanisms of memory reconsolidation. Nat. Rev. Neurosci. 8, 262–275 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Lee, J. L. C., Di Ciano, P., Thomas, K. L. & Everitt, B. J. Disrupting reconsolidation of drug memories reduces cocaine-seeking behavior. Neuron 47, 795–801 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Miller, C. A. & Marshall, J. F. Molecular substrates for retrieval and reconsolidation of cocaine-associated contextual memory. Neuron 47, 873–884 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Torregrossa, M. M. & Taylor, J. R. Neuroscience of learning and memory for addiction medicine: from habit formation to memory reconsolidation. Prog. Brain Res. 223, 91–113 (2016).

    Article  PubMed  Google Scholar 

  8. Jobes, M. L. et al. Effects of prereactivation propranolol on cocaine craving elicited by imagery Script/Cue sets in opioid-dependent polydrug users: a randomized study. J. Addict. Med. 9, 491–498 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lonergan, M. et al. Reactivating addiction-related memories under propranolol to reduce craving: a pilot randomized controlled trial. J. Behav. Ther. Exp. Psychiatry 50, 245–249 (2016).

    Article  PubMed  Google Scholar 

  10. Dunbar, A. B. & Taylor, J. R. Reconsolidation and psychopathology: moving towards reconsolidation-based treatments. Neurobiol. Learn. Mem. 142, 162–171 (2017).

    Article  PubMed  Google Scholar 

  11. Dong, Y. & Nestler, E. J. The neural rejuvenation hypothesis of cocaine addiction. Trends Pharmacol. Sci. 35, 374–383 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Huang, Y. H. et al. In vivo cocaine experience generates silent synapses. Neuron 63, 40–47 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Brown, T. E. et al. A silent synapse-based mechanism for cocaine-induced locomotor sensitization. J. Neurosci. 31, 8163–8174 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Isaac, J. T., Nicoll, R. A. & Malenka, R. C. Evidence for silent synapses: implications for the expression of LTP. Neuron 15, 427–434 (1995).

    Article  CAS  PubMed  Google Scholar 

  15. Liao, D., Hessler, N. A. & Malinow, R. Activation of postsynaptically silent synapses during pairing-induced LTP in CA1 region of hippocampal slice. Nature 375, 400–404 (1995).

    Article  CAS  PubMed  Google Scholar 

  16. Lee, B. R. et al. Maturation of silent synapses in amygdala-accumbens projection contributes to incubation of cocaine craving. Nat. Neurosci. 16, 1644–1651 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ma, Y.-Y. et al. Bidirectional modulation of incubation of cocaine craving by silent synapse-based remodeling of prefrontal cortex to accumbens projections. Neuron 83, 1453–1467 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ma, Y.-Y. et al. Re-silencing of silent synapses unmasks anti-relapse effects of environmental enrichment. Proc. Natl Acad. Sci. USA 113, 5089–5094 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Grimm, J. W., Hope, B. T., Wise, R. A. & Shaham, Y. Neuroadaptation. Incubation of cocaine craving after withdrawal. Nature 412, 141–142 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Graziane, N. M. et al. Opposing mechanisms mediate morphine- and cocaine-induced generation of silent synapses. Nat. Neurosci. 19, 915–925 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hanse, E., Seth, H. & Riebe, I. AMPA-silent synapses in brain development and pathology. Nat. Rev. Neurosci. 14, 839–850 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Cull-Candy, S., Kelly, L. & Farrant, M. Regulation of Ca2+-permeable AMPA receptors: synaptic plasticity and beyond. Curr. Opin. Neurobiol. 16, 288–297 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Conrad, K. L. et al. Formation of accumbens GluR2-lacking AMPA receptors mediates incubation of cocaine craving. Nature 454, 118–121 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nader, K., Schafe, G. E. & LeDoux, J. E. Fear memories require protein synthesis in the amygdala for reconsolidation after retrieval. Nature 406, 722–726 (2000).

    Article  CAS  PubMed  Google Scholar 

  25. Holtmaat, A. & Svoboda, K. Experience-dependent structural synaptic plasticity in the mammalian brain. Nat. Rev. Neurosci. 10, 647–658 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Matsuzaki, M. et al. Dendritic spine geometry is critical for AMPA receptor expression in hippocampal CA1 pyramidal neurons. Nat. Neurosci. 4, 1086–1092 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lin, M. T. et al. Coupled activity-dependent trafficking of synaptic SK2 channels and AMPA receptors. J. Neurosci. 30, 11726–11734 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hayashi, Y. et al. Driving AMPA receptors into synapses by LTP and CaMKII: requirement for GluR1 and PDZ domain interaction. Science 287, 2262–2267 (2000).

    Article  CAS  PubMed  Google Scholar 

  29. Zhou, Z. et al. The C-terminal tails of endogenous GluA1 and GluA2 differentially contribute to hippocampal synaptic plasticity and learning. Nat. Neurosci. 21, 50–62 (2018).

  30. Cingolani, L. A. & Goda, Y. Actin in action: the interplay between the actin cytoskeleton and synaptic efficacy. Nat. Rev. Neurosci. 9, 344–356 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Dietz, D. M. et al. Rac1 is essential in cocaine-induced structural plasticity of nucleus accumbens neurons. Nat. Neurosci. 15, 891–896 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wu, Y. I. et al. A genetically encoded photoactivatable Rac controls the motility of living cells. Nature 461, 104–108 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wang, X., He, L., Wu, Y. I., Hahn, K. M. & Montell, D. J. Light-mediated activation reveals a key role for Rac in collective guidance of cell movement in vivo. Nat. Neurosci. 12, 591–597 (2010).

    Article  CAS  Google Scholar 

  34. Monfils, M.-H., Cowansage, K. K., Klann, E. & LeDoux, J. E. Extinction-reconsolidation boundaries: key to persistent attenuation of fear memories. Science 324, 951–955 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lee, J. L. C. Memory reconsolidation mediates the strengthening of memories by additional learning. Nat. Neurosci. 11, 1264–1266 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Sesack, S. R. & Grace, A. A. Cortico-basal ganglia reward network: microcircuitry. Neuropsychopharmacology 35, 27–47 (2010).

    Article  PubMed  Google Scholar 

  37. Cruz, F. C. et al. New technologies for examining the role of neuronal ensembles in drug addiction and fear. Nat. Rev. Neurosci. 14, 743–754 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Neumann, P. A. et al. Cocaine-induced synaptic alterations in thalamus to nucleus accumbens projection. Neuropsychopharmacology 41, 2399–2410 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Yang, G., Pan, F. & Gan, W.-B. Stably maintained dendritic spines are associated with lifelong memories. Nature 462, 920–924 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liao, Z. et al. Fear conditioning downregulates Rac1 activity in the basolateral amygdala astrocytes to facilitate the formation of fear memory. Front. Mol. Neurosci. 10, 396 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Das, A., Dines, M., Alapin, J. M. & Lamprecht, R. Affecting long-term fear memory formation through optical control of Rac1 GTPase and PAK activity in lateral amygdala. Sci. Rep. 7, 13930 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Nishiyama, J. & Yasuda, R. Biochemical computation for spine structural plasticity. Neuron 87, 63–75 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ghosh, M. et al. Cofilin promotes actin polymerization and defines the direction of cell motility. Science 304, 743–746 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Yasuda, R. Biophysics of biochemical signaling in dendritic spines: implications in synaptic plasticity. Biophys. J. 113, 2152–2159 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Obashi, K., Matsuda, A., Inoue, Y. & Okabe, S. Precise temporal regulation of molecular diffusion within dendritic spines by actin polymers during structural plasticity. Cell Rep. 27, 1503–1515.e1508 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Wells, A. M. et al. Extracellular signal-regulated kinase in the basolateral amygdala, but not the nucleus accumbens core, is critical for context-response-cocaine memory reconsolidation in rats. Neuropsychopharmacology 38, 753–762 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Theberge, F. R. M., Milton, A. L., Belin, D., Lee, J. L. C. & Everitt, B. J. The basolateral amygdala and nucleus accumbens core mediate dissociable aspects of drug memory reconsolidation. Learn. Mem. 17, 444–453 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Everitt, B. J. & Robbins, T. W. Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nat. Neurosci. 8, 1481–1489 (2005).

    Article  CAS  PubMed  Google Scholar 

  49. Taylor, J. R., Olausson, P., Quinn, J. J. & Torregrossa, M. M. Targeting extinction and reconsolidation mechanisms to combat the impact of drug cues on addiction. Neuropharmacology 56, 186–195 (2009).

    Article  CAS  PubMed  Google Scholar 

  50. Wang, S.-H., de Oliveira Alvares, L. & Nader, K. Cellular and systems mechanisms of memory strength as a constraint on auditory fear reconsolidation. Nat. Neurosci. 12, 905–912 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Mu, P. et al. Exposure to cocaine dynamically regulates the intrinsic membrane excitability of nucleus accumbens neurons. J. Neurosci. 30, 3689–3699 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Brebner, K. et al. Nucleus accumbens long-term depression and the expression of behavioral sensitization. Science 310, 1340–1343 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Mardilovich, K. et al. LIM kinase inhibitors disrupt mitotic microtubule organization and impair tumor cell proliferation. Oncotarget 6, 38469–38486 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  54. LaPlant, Q. et al. Dnmt3a regulates emotional behavior and spine plasticity in the nucleus accumbens. Nat. Neurosci. 13, 1137–1143 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Maze, I. et al. Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science 327, 213–216 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Barrot, M. et al. CREB activity in the nucleus accumbens shell controls gating of behavioral responses to emotional stimuli. Proc. Natl Acad. Sci. USA 99, 11435–11440 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yu, J. et al. Nucleus accumbens feedforward inhibition circuit promotes cocaine self-administration. Proc. Natl Acad. Sci. USA 56, 201707822 (2017).

    Google Scholar 

  58. Huang, Y. H., Schluter, O. M. & Dong, Y. Silent synapses speak up: updates of the neural rejuvenation hypothesis of drug addiction. Neuroscientist 21, 451–459 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Winters, B. D. et al. Cannabinoid receptor 1-expressing neurons in the nucleus accumbens. Proc. Natl Acad. Sci. USA 109, E2717–E2725 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dumitriu, D., Rodriguez, A. & Morrison, J. H. High-throughput, detailed, cell-specific neuroanatomy of dendritic spines using microinjection and confocal microscopy. Nat. Protoc. 6, 1391–1411 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Dumitriu, D. et al. Subregional, dendritic compartment, and spine subtype specificity in cocaine regulation of dendritic spines in the nucleus accumbens. J. Neurosci. 32, 6957–6966 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Robinson, T. E. & Kolb, B. Alterations in the morphology of dendrites and dendritic spines in the nucleus accumbens and prefrontal cortex following repeated treatment with amphetamine or cocaine. Eur. J. Neurosci. 11, 1598–1604 (1999).

Download references

Acknowledgements

We thank K. Tang and K. Churn for excellent technical support, as well as M. Varkey, M. Mulloth, S. Beriwal, S. Maddukkuri, Y. Jung, O. Ikwuegbu, R. Moazzam and A. Kang for assistance with behavioral training. This work was supported by NIH grant nos. NS007433 (W.J.W.), DA043940 (W.J.W.), DA023206 (Y.D.), DA044538 (Y.D.), DA040620 (Y.D., E.J.N.), DA047861 (Y.D.), DA035805 (Y.H.H.), MH101147 (Y.H.H.), DA008227 (E.J.N.) and DA014133 (E.J.N.); the NIDA Intramural Research Program (Y.S.); and a Mellon Fellowship (W.J.W.).

Author information

Authors and Affiliations

Authors

Contributions

W.J.W., N.M.G., Y.H.H., Y.S., O.M.S., E.J.N. and Y.D. designed the experiments and analyses and wrote the manuscript. W.J.W., N.M.G. and P.A.N. performed electrophysiology experiments. W.J.W., N.M.G., L.F., A.S., N.M.-B. and K.I. performed behavioral training and testing. N.M.G. performed ELISAs. W.J.W. performed spine analysis, immunohistochemistry and confocal microscopy. P.J.H. and H.M.C. made the HSVs.

Corresponding author

Correspondence to Yan Dong.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Rita A. Fuchs, Raphael Lamprecht and Manuel Mameli 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.

Integrated supplementary information

Supplementary Fig. 1 Training results and example traces for silent synapse experiments in Fig. 1.

(a) Diagram showing the timeline of self-administration training with two withdrawal time points at which the behavioral and electrophysiological experiments were performed. (b-e) Self-administration training results from rats whose electrophysiology results are presented in Fig. 1f–h (saline 1w/d: infusions - d1 = 18.63 ± 3.73, d2 = 10.88 ± 2.82, d3 = 11.14 ± 2.31, d4 = 9.86 ± 5.62, d5 = 12.21 ± 3.01, inactive – d1 = 12.88 ± 2.93, d2 = 9.63 ± 3.29, d3 = 12.00 ± 3.98, d4 = 9.00 ± 5.46, d5 = 8.89 ± 2.37, n = 5 animals; cocaine 1w/d: infusions – d1 = 34.60 ± 3.71, d2 = 29.40 ± 5.84, d3 = 31.20 ± 3.85, d4 = 33.00 ± 2.88, d5 = 35.00 ± 4.00, inactive – d1 = 25.80 ± 10.52, d2 = 16.67 ± 9.27, d3 = 14.60 ± 5.73, d4 = 10.00 ± 2.59, d5 = 12.60 ± 3.44, n = 6 animals; saline 45w/d: infusion – d1=13.70 ± 2.60, d2 = 11.90 ± 2.60, d3= 8.40 ± 2.30, d4 = 8.90 ± 1.90, d5 = 9.30 ± 2.30, inactive – d1= 12.20 ± 3.0, d2 = 9.30 ± 2.00, d3 = 6.30 ± 1.60, d4 = 7.00 ± 1.20, d5 = 5.80 ± 1.10, n = 5 animals; cocaine 45w/d: infusions – d1 = 19.70 ± 3.03, d2 = 21.45 ± 3.26, d3 = 20.64 ± 3.37, d4 = 22.36 ± 4.00, d5 = 21.36 ± 1.38, inactive – d1 = 7.10 ± 3.49, d2 = 7.45 ± 2.53, d3 = 4.64 ± 1.43, d4 = 6.00 ± 1.22, d5 = 11.36 ± 2.62, n = 12 animals). (f-h) Example EPSCs over 100 trials from the minimal stimulation assay for the summarized results presented in Fig. 1h. (i) Diagram showing different time points after cue re-exposure at which silent synapses were assessed. (j-o) Self-administration training results of rats whose electrophysiology results are presented in Fig. 1j–m, s–v (saline re-exp: infusions – d1 = 13.80 ± 1.72, d2 = 12.40 ± 3.44, d3 = 7.20 ± 1.32, d4 = 8.60 ± 2.14, d5 = 10.40 ± 3.66, inactive – d1 = 10.20 ± 0.735, d2 = 11.80 ± 3.07, d3 = 7.60 ± 0.678, d4 = 6.60 ± 1.29, d5 = 6.60 ± 2.21, n = 5 animals; cocaine 45w/d: infusions – d1 = 27.73 ± 3.34, d2 = 23.60 ± 2.53, d3 = 24.73 ± 2.54, d4 = 22.00 ± 1.77, d5 = 23.53 = 2.03, inactive – d1= 5.71 ± 1.48, d2 = 8.13 ± 3.07, d3 = 7.14 ± 2.38, d4 = 5.86 ± 2.29, d5 = 4.33 ± 1.41, n = 16 animals; cocaine re-exp: infusions – d1 = 32.91 ± 5.17, d2 = 30.05 ± 3.78, d3 = 26.09 ± 2.06, d4 = 26.46 ± 2.33, d5 = 32.67 ± 4.34, inactive – d1 = 12.95 ± 4.10, d2 = 16.18 ± 5.16, d3 = 8.14 ± 2.79, d4 = 8.14 ± 2.80, d5 = 9.38 ± 3.15, n = 22 animals; cocaine 2hr: infusions – d1 = 40.20 ± 3.56, d2 = 36.40 ± 2.62, d3 = 30.20 ± 2.80, d4 = 34.80 ± 2.44, d5 = 36.20 ± 4.21, inactive – d1 = 7.00 ± 3.56, d2 = 8.00 ± 3.27, d3 = 8.00 ± 2.85, d4 = 8.00 ± 3.35, d5 = 6.60 ± 2.60, n = 5 animals; cocaine 4hr: infusions – d1 = 28.86 ± 1.49, d2 = 30.14 ± 4.26, d3 = 22.43 ± 1.07, d4 = 22.57 ± 2.26, d5 = 23.57 ± 2.99, inactive – d1= 13.71 ± 4.85, d2 = 11.86 ± 4.04, d3 = 6.57 ± 1.82, d4 = 9.34 ± 2.51, d5 = 8.14 ± 2.12, n = 7 animals; cocaine 6hr: infusions – d1 = 27.15 ± 3.52, d2 = 26.92 ± 2.18, d3 = 31.23 ± 6.20, d4 = 30.46 ± 4.26, d5 = 25.69 ± 2.36, inactive – d1 = 17.62 ± 11.29, d2 = 7.54 ± 4.49, d3 = 2.75 ± 1.18, d4 = 3.39 ± 2.03, d5 = 1.60 ± 0.743, n = 13 animals). (p) Summary showing no difference in nose poke responding during the 10-min cue re-exposure session in rats in which NAcSh silent synapses were assessed 10 min, 2 hr, 4 hr, and 6 hr after cue re-exposure (Fig. 1v) (10min = 29.45 ± 3.44, n = 22 animals; 2hr = 37.40 ± 3.98, n = 5 animals; 4hr = 21.86 ± 3.12, n = 7 animals; 6hr = 31.92 ± 2.68, n = 13 animals, F3,43=1.55, p=0.22, one-way ANOVA, n.s. > 0.05). (q-s) Example EPSCs over 100 trials from the minimal stimulation assay for the summarized results presented in Fig. 1m, v. Data presented as mean±SEM.

Supplementary Fig. 2 Training results for NMDAR and I-V experiments in Fig. 1.

(a-d) Self-administration training results of rats whose electrophysiology results are presented in Fig. 1n, o (saline 1w/d: infusions – d1 = 17.00 ± 3.62, d2 = 9.40 ± 2.54, d3 = 2.40 ± 3.12, d4 = 11.00 ± 3.08, d5 = 15.50 ± 4.29, inactive – d1 = 12.40 ± 2.50, d2 = 7.80 ± 1.32, d3 = 9.20 ± 2.04, d4 = 8.40 ± 3.44, d5 = 10.75 ± 3.95, n = 5 animals; cocaine 1w/d: infusions – d1 = 42.20 ± 11.43, d2= 32.00 ± 8.06, d3= 39.40 ± 9.88, d4= 40.40 ± 7.94, d5= 41.60 ± 7.71, inactive – d1= 20.40 ± 7.81, d2= 30.60 ± 9.94, d3= 13.00 ± 4.37, d4= 10.60 ± 2.93, d5 = 12.00 ± 3.65, n = 5 animals; saline 45w/d: infusions – d1= 17.00 ± 3.46, d2= 9.33 ± 6.33, d3= 5.33 ± 2.85, d4= 9.67 ± 5.67, d5= 10.67 ± 1.33, inactive – d1= 19.00 ± 8.02, d2= 8.33 ± 4.49, d3= 4.00 ± 3.06, d4= 7.00 ± 1.53, d5= 7.67 ± 2.33, n = 3 animals; cocaine 45w/d: infusions – d1= 33.00 ± 6.78, d2= 26.00 ± 1.92, d3= 26.00 ± 2.30, d4 = 28.40 ± 4.20, d5= 23.40 ± 1.83, inactive – d1 = 14.80 ± 3.76, d2= 15.00 ± 5.06, d3= 11.00 ± 1.30, d4= 11.20 ± 2.29, d5= 11.00 ± 4.55, n = 5 animals; cocaine re-exp: infusions – d1= 30.60 ± 4.76, d2= 28.80 ± 3.06, d3= 26.00 ± 2.67, d4= 30.20 ± 2.76, d5 = 31.20 ± 3.29, inactive – d1= 21.60 ± 14.94, d2= 7.20 ± 3.84, d3= 6.40 ± 3.36, d4 = 9.20 ± 2.65, d5= 8.00 ± 1.41, n = 5 animals). (d, right). Nose poke responding during the cue re-exposure session of rats whose electrophysiology results are presented in Fig. 1o (37.80 ± 7.70, n = 5 animals). (e,f) Self-administration training results of rats whose electrophysiology results are presented in Fig. 1p, q (saline: infusions – d1= 15.50 ± 2.39, d2= 10.67 ± 2.97, d3= 7.83 ± 2.97, d4= 9.50 ± 2.66, d5=8.00 ± 1.34, inactive – d1 = 15.00 ± 4.26, d2= 9.67 ± 2.28, d3= 7.00 ± 2.56, d4= 8.17 ± 1.17, d5= 6.17 ± 1.62, n = 6 animals; cocaine 45w/d: infusions – d1= 35.00 ± 11.24, d2= 26.00 ± 2.76, d3= 26.80 ± 2.81, d4= 28.20 ± 4.66, d5= 28.20 ± 2.46, inactive – d1= 15.80 ± 4.58, d2= 10.40 ± 2.29, d3= 9.20 ± 1.56, d4= 8.20 ± 2.56, d5= 12.00 ± 4.99, n = 5 animals; cocaine re-exp: infusions – d1= 45.75 ± 18.54, d2= 27.50 ± 2.40, d3= 23.25 ± 3.59, d4= 24.40 ± 5.39, d5= 25.25 ± 5.14, inactive – d1= 21.00 ± 11.23, d2= 13.75 ± 7.42, d3= 5.75 ± 2.59, d4= 5.25 ± 3.25, d5= 3.25 ± 2.29, n = 4 animals). (f, right). Nose poke responding during the cue re-exposure session of rats whose electrophysiology results are presented in Fig. 1p, q (20.50 ± 3.57, n = 4 animals). Data presented as mean±SEM.

Supplementary Fig. 3 Cocaine-induced changes in spine morphology and training results for Fig. 2.

(a) Example NAcSh dendrites from saline-trained rats (upper) and cocaine-trained rats (lower) after one day from the 5-day self-administration procedure. Scale bars, 2.5 µm. (b) Summary showing that cocaine-trained rats exhibited an increase in the density of total dendritic spines on withdrawal day 1 after cocaine self-administration compared to saline-trained rats (saline = 11.29 ± 0.40, n = 4 animals; cocaine = 16.46 ± 0.298, n = 4 animals, t6=10.45, p<0.0001, two-tail, unpaired t-test). (c) Summary showing no difference in the density of mushroom-like spines between cocaine- and saline-trained rats on withdrawal day 1, there was (saline = 3.89 ± 0.044, n = 4 animals; cocaine = 4.12 ± 0.150, n = 4 animals, t6=1.41, p=0.21, two-tail, unpaired t-test). (d) Summary showing an increase in the density of thin spines in cocaine-trained rats compared to saline-trained rats on withdrawal day 1 (saline = 5.97 ± 0.450, n = 4 animals; cocaine = 10.19 ± 0.292, n = 4 animals, t6=7.87, p=0.0002, two-tail, unpaired t-test). (e) Summary showing an increased in the density of stubby spines in cocaine-trained rats compared to saline-trained rats on withdrawal day 1 (saline = 1.43 ± 0.092, n = 4 animals; cocaine = 2.15 ± 0.092, n = 4 animals, t6=5.55, p=0.0014, two-tail, unpaired t-test). (f,g) Self-administration training results of rats used for spine analysis on withdrawal day 1 (Supplementary Fig. 3a–e) (saline 1w/d: infusions – d1= 22.25 ± 1.38, d2= 17.00 ± 2.65, d3= 18.25 ± 3.90, d4= 14.00± 3.58, d5= 15.00 ± 2.16, inactive – d1= 32.25 ± 5.59, d2= 23.50± 6.96, d3= 15.75 ± 3.75, d4= 13.50 ± 2.18, d5= 15.00 ± 4.97, n = 4 animals; cocaine 1w/d: infusions – d1= 42.75 ± 5.41, d2= 42.25±3.52, d3= 41.00 ± 4.06, d4= 42.00 ± 6.44, d5= 37.25 ± 1.84, inactive – d1=17.50 ± 5.98, d2= 14.50 ± 6.51, d3= 12.50 ± 5.35, d4= 13.75 ± 3.97, d5= 8.50 ± 3.78, n = 4 animals). (h-k) Self-administration training results of rats whose morphological results are presented in Fig. 2d–h (saline re-exp: infusions – d1= 9.50 ± 3.01, d2= 10.00 ± 2.35, d3= 14.25 ± 5.14, d4= 8.50 ± 2.60, d5= 13.00 ± 4.66, inactive – d1= 7.25 ± 1.11, d2= 5.75 ± 1.38, d3= 8.25 ± 0.854, d4= 6.75 ± 1.65, d5= 6.75 ± 1.93, n = 4 animals; cocaine 45w/d: infusions – d1= 48.43 ± 7.46, d2= 34.29 + 2.45, d3= 38.71 ± 1.89, d4= 38.57 ± 1.60, d5= 36.14 ± 2.25, inactive – d1= 8.71 ± 2.75, d2= 4.57 ± 0.812, d3= 4.86 ± 1.58, d4= 2.75 ± 1.07, d5= 1.86 ± 0.884, n = 7 animals; cocaine re-exp: infusions – d1= 45.57 ± 5.05, d2= 41.86 ± 3.67, d3= 43.14 ± 2.65, d4= 41.71 ± 4.01, d5= 43.29 ± 4.28, inactive – d1= 30.86 ± 9.12, d2= 15.57 ± 6.29, d3= 7.86 ± 1.64, d4= 3.71 ± 2.06, d5= 4.86± 3.21, n = 7 animals; cocaine 6hr: infusions- d1= 35.50 ± 4.03, d2= 32.75 ± 2.72, d3= 32.00 ± 2.55, d4= 28.25 ± 2.75, d5= 34.75 ± 4.01, inactive – d1= 18.50 ± 15.86, d2= 8.50 ± 4.48, d3= 3.25 ± 2.63, d4= 2.75 ± 2.14, d5= 3.50 ± 3.50, n = 4 animals). (l) Nose poke responding during the cue re-exposure session of rats whose morphological results are presented in Fig. 2d–h. Cocaine-trained rats exhibited higher levels of nose pokes compared to saline-trained rats. There was no difference in cocaine-trained rats examined 10 min or 6hr after cue re-exposure (saline = 12.25 ± 1.81, n = 4 animals; cue re-exp = 31.71 ± 2.07, n = 7 animals; 6hr post = 33.50 ± 2.99, n = 4 animals, F2,12=23.65, p<0.0001, one-way ANOVA; **p<0.01, Bonferroni posttest). See Supplementary Table 1 for exact p values for all comparisons made during posthoc tests. Data presented as mean±SEM.

Supplementary Fig. 4 Peptide validation and training results of rats presented in Fig. 3.

(a) (left and bottom) Diagram showing experimental setup for the induction of LTP at Schaffer collateral to CA1 synapses, with example field EPSPs before and after LTP induction. (right) Summarized results showing that this form of LTP, which was sensitive to the NMDAR-selective antagonist APV, was abolished in sliced pre-incubated with TGL, but not AGL (at time 60min: control = 1.48 ± 0.10, n = 8 cells; APV = 0.969 ± 0.05, n = 6 cells; AGL = 1.45 ± 0.092, n = 7 cells; TGL = 1.174 ± 0.113, n = 8 cells, F207, 1725=3.07, p<0.0001, two-way RM ANOVA; *p<0.05, Bonferroni posttest, cell-based statistics). (b,c) Self-administration training results of rats whose electrophysiology are presented in Fig. 3c–f (saline AGL: infusions – d1= 11.20 ± 3.60, d2= 7.20 ± 2.52, d3= 7.40 ± 1.91, d4= 11.00 ± 2.97, d5= 11.00 ± 2.35, inactive – d1= 12.40 ± 4.20, d2= 5.20 ± 1.69, d3= 3.80 ± 0.97, d4= 7.00 + 1.41, d5= 5.00 ± 1.79, n = 5 animals; saline TGL: infusions – d1= 10.00 ± 1.95, d2= 6.80± 1.07, d3= 7.40 ± 1.44, d4= 6.40 ± 1.72, d5= 5.60 ± 1.17, inactive – d1= 10.60 ± 3.47, d2= 5.80 ± 2.01, d3= 5.80 ± 1.69, d4= 4.40 ± 0.68, d5= 5.60 ± 1.89, n =5 animals; cocaine AGL: infusions – d1= 44.73 ± 3.96, d2= 40.27 ± 6.37, d3= 41.82 ± 5.47, d4= 35.36 ± 4.60, d5= 37.00 ± 7.60, inactive – d1= 34.45 ± 18.39, d2= 14.45 ± 5.17, d3= 18.45 ± 9.14, d4= 8.46 ± 2.40, d5= 9.18 ± 2.24, n = 11 animals; cocaine TGL: infusions – d1= 41.00 ± 7.18, d2= 35.29 ± 5.07, d3= 31.71 ± 3.78, d4= 28.71 ± 3.16, d5= 30.00 ± 4.71, inactive – d1= 12.43 ± 4.07, d2= 13.86 ± 4.38, d3= 15.83 ± 4.73, d4= 9.71 ± 1.49, d5= 11.57 ± 3.56, n = 7 animals). (d) Summary showing no difference in nose poking responding during the 10-min cue re-exposure session in rats presented in Fig. 3c–f (saline AGL = 7.00 ± 0.775, n =5 animals; saline TGL = 13.40 ± 3.30, n = 5 animals, t8=1.89, p=0.09, saline-AGL vs saline-TGL; cocaine AGL = 24.00 ± 2.34, n = 11 animals; cocaine TGL = 22.57 ± 2.94, t16=0.38, p=0.71, cocaine-AGL vs cocaine-TGL, two-tail, unpaired t-test, n.s. > 0.05). (e-g) Example EPSCs over 100 trials from the minimal stimulation assay for the summarized results presented in Fig. 3f. (h-i) Self-administration training results of rats whose morphology results are presented in Fig. 3g–l (saline AGL: infusions – d1= 9.67 ± 1.86, d2= 15.33 ± 2.91, d3= 11.67 ± 2.03, d4= 7.67 ± 0.882, d5= 8.33 ± 1.86, inactive – d1= 16.33 ± 4.63, d2= 7.00 ± 2.52, d3= 6.67 ± 1.76, d4= 4.67 ± 1.67, d5= 7.33 ± 1.86, n = 3 animals; saline TGL: infusions – d1= 7.00 ± 2.00, d2= 14.00 ± 1.53, d3= 20.00 ± 9.50, d4= 11.33 ± 2.03, d5= 12.67 ± 4.18, inactive – d1= 4.67 ± 2.33, d2= 11.33 ± 4.26, d3= 8.33 ± 4.84, d4= 7.67 ± 1.33, d5= 8.00 ± 1.53, n = 3 animals; cocaine AGL: infusions – d1= 43.00 ± 4.22, d2= 43.50 ± 7.26, d3= 41.00 ± 7.63, d4= 46.75 ± 5.76, d5= 43.75 ± 6.08, inactive – d1= 29.50 ± 24.24, d2= 4.00 ± 0.816, d3= 3.50 ± 1.66, d4= 4.25 ± 1.84, d5= 1.25 ± 0.946, n = 4 animals; cocaine TGL: infusions – d1= 44.00 ± 2.86, d2= 46.75 ± 4.11, d3= 54.50 ± 8.91, d4= 41.75 ± 4.91, d5= 38.75 ± 4.11, inactive – d1= 6.25 ± 3.97, d2= 3.00 ± 2.35, d3= 7.75 ± 2.50, d4= 5.50 ± 1.26, d5= 2.75 ± 1.32, n = 4 animals). (j) Summary showing no difference in nose poking responding during the 10-min cue re-exposure session in rats presented in Fig. 3g–l (saline AGL = 12.33 ± 5.04, n =3 animals; saline TGL = 13.00 ± 2.52, n = 3 animals, t4=0.118, p=0.9116, saline-AGL vs saline-TGL; cocaine AGL = 29.50 ± 1.56, n = 4 animals; cocaine TGL = 30.00 ± 5.12, t6=0.094, p=0.9285, cocaine-AGL vs cocaine-TGL, two-tail, unpaired t-test, n.s. > 0.05). See Supplementary Table 1 for exact p values for all comparisons made during posthoc tests. Data presented as mean±SEM.

Supplementary Fig. 5 Additional behavioral controls for experiments in Fig. 3.

(a,b) Summary showing that nose poke responding (seeking) in saline (a)- and cocaine (b)-trained rats was not altered when tested 6 hr after cue re-exposure (saline active = 24.18 ± 2.69, n = 11 animals; saline inactive = 10.82 ± 1.16, n =11 animals; cocaine active = 85.20 ± 4.38, n = 10 animals; cocaine inactive = 22.30 ± 3.86, n =11 animals). (c) Summary showing that cocaine-trained rats exhibited higher levels of nose poke responding during the 10-min cue re-exposure session compared to saline-trained rats on withdrawal day 45 (saline = 14.33 ± 2.10, n = 11 animals; coaine = 37.40 ± 2.66, n = 10 animals, t17=6.69, p<0.0001, two-tail, unpaired t-test). (d) Summary showing that intra-NAcSh infusions of AGL or TGL 2 hr after cue re-exposure did not affect cue-induced nose poke responding in saline-trained rats, measured 6 hr after re-exposure (AGL active = 17.87 ± 4.14, n = 8 animals; TGL active = 12.25 ± 2.07, n =8 animals; AGL inactive = 7.00 ± 1.65, n = 8 animals, TGL in active = 7.00 ± 1.71, n = 8 animals, F1,7=4.95, p=0.0614, RM two-way ANOVA, withdrawal day 45 peptide x hole interaction). Nose poke responding during the 10-min cue re-exposure session (right) was also not different between AGL and TGL rats (AGL = 13.00 ± 2.73, n = 8 animals; TGL = 13.00 ± 2.27, n =8 animals, t14=0.000, p=1.00, two-tail unpaired t-test). (e) Summary showing that intra-NAcSh infusions of AGL or TGL 6 hr after cue re-exposure did not affect cue-induced nose poke responding, tested 6.5 hr after cue re-exposure, in saline-trained rats (AGL active = 22.13 ± 4.88, n = 8 animals; TGL active = 20.88 ± 5.92, n = 8 animals; AGL inactive = 6.75 ± 1.37, n = 8 animals; TGL inactive = 7.13 ± 1.46, n = 8 animals, F1,7=0.0, p=0.7986, RM two-way ANOVA, withdrawal day 45 peptide x hole interaction). Nose poke responding during the 10-min cue re-exposure session (right) was also not different between AGL and TGL rats (AGL = 16.25 ± 2.72, n = 8 animals; TGL = 12.13 ± 2.91, n = 8 animals, t14=1.036, p=0.3179, two-tail, unpaired t-test). (f) Summary of nose poke responding during the 10-min cue re-exposure session in cocaine-trained rats whose behavioral results are presented in Fig. 3m, n. Nose poke responding was similar between rats with intra-NAcSh AGL vs rats with intra-NAcSh TGL when the peptides were injected either 2 hr (left) or 6 hr (right) after cue re-exposure (2 hr: AGL = 29.08 ± 3.96, n = 12 animals; TGL = 27.58 ± 3.58, n = 12 animals, t22=0.2811, p=0.7812; 6 hr: AGL = 32.10 ± 1.20, n = 10 animals; TGL = 29.30 ± 2.01, n = 10 animals, t18=1.196, p=0.2471, two-tail, unpaired t-test). (g-j) Diagrams showing the localization of the infusion needle tips for all rats included in the data analysis in Fig. 3m, n and Supplementary Fig. 5d, e. Data presented as mean±SEM.

Supplementary Fig. 6 Training and re-exposure results and viral localization for Fig. 4.

(a-b) Self-administration training results of rats whose electrophysiology results are presented in Fig. 4e–h (saline: infusions – d1= 9.17 ± 1.42, d2= 10.50 ± 2.45, d3= 8.33 ± 1.15, d4= 5.17 ± 1.20, d5= 6.50 ± 1.46, inactive – d1= 9.50 ± 3.54, d2= 13.83 ± 3.04, d3= 7.00 ± 1.77, d4= 3.33 ± 1.50, d5= 5.33 ± 1.50, n= 6 animals; cocaine: infusions – d1= 39.53 ± 5.03, d2= 36.07 ± 4.18, d3= 34.60 ± 3.18, d4= 36.47 ± 3.33, d5= 37.00 ± 4.75, inactive – d1= 14.80 ± 4.37, d2= 12.73 ± 3.92, d3= 8.47 ± 2.36, d4= 4.60 ± 1.10, d5= 7.40 ± 2.49, n = 15 animals). (c,d) Example EPSCs (inset) over trials in the minimal stimulation assay from rats whose electrophysiology results are presented in Fig. 4h. (e,f) Self-administration training results of rats whose electrophysiology results are presented in Fig. 4i–o (saline: infusions – d1= 7.00 ± 1.58, d2= 7.40 ± 1.50, d3= 6.80 ± 1.88, d4= 6.20 ± 1.11, d5= 7.20 ± 1.83, inactive – d1= 4.80 ± 0.800, d2= 7.20 ± 1.74, d3= 6.40 ± 1.03, d4= 3.60 ± 1.21, d5= 5.40 ± 2.29, n = 5 animals; cocaine: infusions – d1= 34.11 ± 4.57, d2= 39.67 ± 6.49, d3= 43.44 ± 6.58, d4= 42.56 ± 5.65, d5= 40.11 ± 5.16, inactive – d1= 32.33 ± 19.25, d2= 8.44 ± 3.35, d3= 4.78 ± 2.76, d4= 4.67 ± 1.80, d5= 6.11 ± 2.69, n = 9 animals). (i,j) Nose poke responding during the cue re-exposure session of rats whose behavioral results are presented in Fig. 4p, q (2hr post = 28.00 ± 3.20, n = 7 animals; pa-dnRac1 re-exp = 26.00 ± 1.84, n = 7 animals). (k-m) Example image showing HSV-mediated expression of pa-dnRac1 in the NAcSh and location of the optical fiber that delivered laser stimulation (k), and a magnified portion of the image showing viral expression in individual neurons (l) from a cocaine-trained rats used in behavioral experiments presented in Fig. 4q. Viral expression and fiber tip localization for all rats that were included in data analysis are presented in (m). (n-p) Example image showing HSV-mediated expression of pa-dnRac1 in the NAcSh and location of the optical fiber that delivered laser stimulation (n), and a magnified portion of the image showing viral expression in individual neurons (o) from a cocaine-trained rat used in behavioral experiments presented in Fig. 4q. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (p). Data presented as mean±SEM.

Supplementary Fig. 7 HSV-mediated expression of C450M, an inactive control for caRac1, does not affect silent synapse levels.

(a,b) Self-administration training results of rats whose electrophysiology results are presented in Supplementary Fig 7d–h (saline: infusions – d1= 15.40 ± 3.01, d2= 8.60 ± 1.44, d3= 9.60 ± 1.99, d4= 13.00 ± 2.24, d5= 9.00 ± 0.894, inactive – d1= 10.20 ± 1.02, d2= 6.40 ± 1.12, d3= 7.20 ± 0.80, d4= 8.80 ± 2.38, d5= 6.20 ± 1.72, n = 5 animals; cocaine: infusions – d1= 39.50 ± 2.74, d2= 34.75 ± 2.16, d3= 31.58 ± 1.82, d4= 32.50 ± 2.16, d5 = 36.25 ± 2.85, inactive – d1= 36.92 ± 10.46, d2= 13.42 ± 2.92, d3= 10.58 ± 3.26, d4= 6.58 ± 1.75, d5= 8.25 ± 2.15, n = 12 animals). (c) Diagram showing the experimental timeline for self-administration, withdrawal, HSV-mediated expression of C450M, and electrophysiology. (d-f) Example EPSCs (inset) over trials in the minimal stimulation assay of C450M-expressing NAcSh MSNs in saline (d)- and cocaine-trained rats (e), and the effects of CP-AMPAR inhibition (f). (h) Summary showing that C450M expression did not affect the % silent synapses in saline- or cocaine-trained rats, and the low % silent synapses in cocaine-trained rats was restored to high levels by naspm inhibition of CP-AMPARs (saline = 6.64 ± 4.15, n = 5 animals; cocaine = 6.04 ± 2.27, n = 6 animals; cocaine naspm = 36.76 ± 3.48, n = 6 animals, F2,14=29.20, p<0.0001, one-way ANOVA; **p<0.01, Bonferroni posttest). See Supplementary Table 1 for exact p values for all comparisons made during posthoc tests. Data presented as mean±SEM.

Supplementary Fig. 8 Self-administration training results for rats used in Fig. 5.

(a,b) Self-administration training results of rats whose electrophysiology results are presented in Fig. 5b–e (saline: infusions – d1= 14.14 ± 2.70, d2= 12.14 ± 2.44, d3= 9.43 ± 2.33, d4= 11.14 ± 3.23, d5= 8.71 ± 2.12, inactive – d1= 12.29 ± 1.43, d2= 8.29 ± 1.41, d3= 6.71 ± 1.38, d4= 10.71 ± 3.26, d5= 10.14 ± 2.06, n = 7 animals; cocaine: infusions – d1= 37.72 ± 2.26, d2= 32.22 ± 1.14, d3= 34.56 ± 2.24, d4= 40.33 ± 3.64, d5= 40.33 ± 3.02, inactive – d1= 19.56 ± 3.74, d2= 8.67 ± 1.67, d3= 7.17 ± 1.64, d4= 10.00 ± 3.59, d5= 7.89 ± 2.07, n = 18 animals). (c) Summary of nose poke responding during the 10-min cue re-exposure session from rats whose electrophysiology results are presented in Fig. 5b–e. Cocaine-trained rats exhibited higher levels of cue-induced nose poke responding compared to saline-trained rats (saline = 12.29 ± 2.39, n = 7 animals; cocaine = 28.67 ± 1.69, n = 18 animals, t23=5.29, p<0.0001, two-tail, unpaired t-test). (d,e) Example EPSCs (inset) over trials in the minimal stimulation assay from rats whose electrophysiology results are presented in Fig. 4e. (f,g) Self-administration training results of rats whose electrophysiology results are presented in Fig. 5i–l (saline: infusions – d1= 9.40 ± 1.50, d2= 10.00 ± 3.52, d3= 6.60 ± 2.09, d4= 7.80 ± 2.59, d5= 6.60 ± 1.57, inactive – d1= 7.20 ± 1.88, d2= 8.00 + 2.57, d3= 10.20 ± 3.20, d4= 7.20 ± 1.49, d5= 8.00 ± 1.14, n = 5 animals; cocaine: infusions – d1= 44.42 ± 4.14, d2= 35.75 ± 3.39, d3= 37.83 ± 3.19, d4= 39.33 ± 2.59, d5= 36.00 ± 2.31, inactive – d1= 21.25 ± 5.05, d2= 11.17 ± 3.78, d3= 8.75 ± 2.32, d4= 6.33 ± 2.02, d5= 8.58 ± 2.74, n = 12 animals). (h) Summary of nose poke responding during the 10-min cue re-exposure session from rats whose electrophysiology results are presented in Fig. 5i–l. The nose poke responding was higher in cocaine-trained rats compared to saline-trained rats (saline = 4.60 ± 0.51, n= 5 animals; cocaine = 21.08 ± 1.62, n = 12 animals, t15=6.41, p<0.0001, two-tail, unpaired t-test). (i,j) Self-administration training results of rats whose electrophysiology results are presented in Fig. 5t–v (vehicle: infusions – d1= 33.25 ± 9.76, d2= 40.25 ± 6.65, d3= 30.25 ± 1.91, d4= 34.75 ± 3.20, d5= 38.75 ± 2.78, inactive – d1= 35.75 ± 28.13, d2= 21.00 ± 13.02, d3= 9.75 ± 6.22, d4= 6.75 ± 3.82, d5= 6.00 ± 4.38, n = 4 animals; LIMKi: infusions – d1= 44.75 ± 1.89, d2= 31.00 ± 2.12, d3= 37.75 ± 6.36, d4= 43.00 ± 5.82, d5= 41.25 ± 6.38, inactive – d1= 11.50 ± 2.78, d2= 14.00 ±6.38, d3= 5.75 ± 5.11, d4= 7.50 ± 3.79, d5= 5.50 ± 5.17, n = 4 animals) . (k) Summary of nose poke responding during the 10-min cue re-exposure session from rats whose electrophysiology results are presented in Fig. 5i–l. The nose poke responding was not different in animals treated with vehicle or LIMKi (vehicle = 23.50 ± 3.80, n = 4 animals; LIMKi = 19.25 ± 1.03, n = 4 animals, t6=1.08, p=0.3215, two-tail,unpaired t-test). (l,m) Self-administration training results of rats whose morphology results are presented in Fig. 5m–r (saline: infusions – d1= 9.50 ± 0.645, d2= 8.50 ± 1.56, d3= 5.50 ± 2.18, d4= 10.50 ± 1.94, d5= 6.00 ± 2.48, inactive – d1= 6.25 ± 0.629, d2= 4.75 ± 2.18, d3= 3.50 ± 0.866, d4= 8.25 ± 0.250, d5= 3.00 ± 0.707, n = 4 animals; cocaine: infusions – d1= 40.25 ± 1.93, d2= 30.00 ± 5.31, d3= 31.75 ± 4.13, d4= 40.50 ± 1.04, d5 = 40.00 ± 4.88, inactive – d1= 20.50 ± 10.91, d2= 10.25 ± 7.92, d3= 9.50 ± 7.86, d4= 1.25 ± 0.629, d5= 1.00 ± 0.707, n = 4 animals). (n) Summary of nose poke responding during the 10-min cue re-exposure session from rats whose morphology results are presented in Fig. 5m–r. The nose poke responding was higher in cocaine-trained rats compared to saline-trained rats (saline = 5.50 ± 1.19, n = 4 animals; cocaine = 22.00 ± 3.24, n = 4 animals, t6=4.78, p=0.0031, two-tail, unpaired t-test). Data presented as mean±SEM.

Supplementary Fig. 9 Self-administration training results for rats used in Fig. 6.

(a,b) Self-administration training results of rats whose electrophysiology results are presented in Fig. 6c–f (saline: infusions – d1= 14.00 ± 2.12, d2= 3.50 ± 2.96, d3= 11.75 ± 0.854, d4= 13.50 ± 2.90, d5= 15.25 ± 2.56, inactive – d1= 10.25 ± 3.19, d2= 10.50 ± 1.94, d3= 7.75 ± 2.14, d4= 12.75 ± 4.88, d5 = 5.25 ± 1.84, n = 4 animals; cocaine: infusions – d1= 40.00 ± 4.03, d2= 44.75 ± 4.07, d3= 41.58 ± 2.81, d4= 42.42 ± 2.90, d5= 43.67 ± 3.45, inactive – d1= 26.00 ± 5.81, d2= 12.92 ± 4.63, d3= 9.83 ± 3.79, d4= 4.75 ± 1.71, d5= 3.67 ± 1.12, n = 12 animals). (c) Summary of nose poke responding during the 10-min cue re-exposure session from rats whose electrophysiology results are presented in Fig. 6c–f. The nose poke responding was higher in cocaine-trained rats compared to saline-trained rats (saline = 8.50 ± 1.26, n = 4 animals; cocaine = 26.50 ± 2.13, n = 12 animals, t14=4.69, p=0.0003, two-tail, unpaired t-test). (d,e) Self-administration training results of rats whose electrophysiology results are presented in Fig. 6n–p (vehicle: infusions – d1= 32.00 ± 9.34, d2= 27.75 ± 5.19, d3= 33.50 ± 7.53, d4= 33.24 ± 7.98, d5= 30.50 ± 4.84, inactive – d1= 22.50 ± 10.66, d2= 5.25 ± 3.97, d3= 7.00 ± 2.67, d4= 7.00 ± 2.86, d5= 8.25 ± 4.39, n = 4 animals; LIMKi: infusions – d1= 50.75 ± 5.47, d2= 45.25 ± 3.90, d3= 41.50 ± 5.90, d4= 46.00 ± 11.57, d5= 39.50 ± 2.53, inactive – d1= 53.50 ± 8.97, d2= 20.00 ± 10.75, d3= 13.75 ± 4.73, d4= 6.25 ± 3.28, d5= 4.00 ± 2.12, n = 4 animals). (f) Summary showing nose poke responding during the 10-min cue re-exposure session from rats whose electrophysiology results are presented in Fig. 6n–p. The nose poke responding was not different in animals treated with vehicle or LIMKi (vehicle = 24.00 ± 4.18, n = 4 animals; LIMKi = 26.00 ± 1.73, n = 4 animals, t6=0.44, p=0.6742, two-tail, unpaired t-test). (g,h) Self-administration training results of rats whose morphology results are presented in Fig. 6g–l (saline: infusions – d1= 7.25 ± 0.629, d2= 7.00 ± 3.72, d3= 14.50 ± 3.12, d4= 3.50 ± 0.645, d5= 12.25 + 2.18, inactive – d1= 7.00 ± 2.61, d2= 5.50 ± 3.18, d3= 17.25, ± 5.49, d4= 11.00 ± 7.08, d5= 10.25 ± 2.18, n = 4 animals; cocaine: infusions – d1= 28.00 ± 5.86, d2= 35.00 ± 9.85, d3= 35.33 ± 8.95, d4= 35.33 ± 9.49, d5= 38.67 ± 6.17, inactive – d1= 8.67 ± 5.18, d2= 5.33 ± 2.60, d3= 7.67 ± 3.76, d4= 3.67 ± 1.86, d5= 4.00 ± 2.08, n = 3 animals). (i) Summary showing nose poke responding during the 10-min cue re-exposure session from rats whose morphology results are presented in Fig. 6g–l. The nose poke responding was higher in cocaine-trained rats compared to saline-trained rats (saline = 7.00 ± 2.27, n = 4 animals; cocaine = 27.00 ± 2.65, n = 3 animals, t5=5.74, p =0.0022, two-tail, unpaired t-test). Data presented as mean±SEM.

Supplementary Fig. 10 Effects of paRac1 stimulation on cue-induced nose poke responding in saline controls.

(a) Diagram showing the experimental timeline and setup. (b) Summary showing that saline-trained rats with photostimulation of intra-NAcSh paRac1 and photostimulation of intra-NAcSh C450M (control) during cue re-exposure exhibited similar cue-induced seeking, measured 6 hr after cue re-exposure (C450 active = 17.88 ± 2.95, n = 8 animals; paRac1 active = 21.83 ± 4.31, n = 6 animals; C450 inactive = 5.13 ± 0.915, n = 8 animals; paRac1 inactive = 9.17 ± 2.60, n = 6 animals, F1,12=0.0002, p=0.9878, two-way RM ANOVA, withdrawal day 45 hole x virus interaction, n.s. > 0.05). (c) Nose poke responding during the 10-min cue re-exposure session in saline-trained rats whose behavioral results presented in Supplementary Fig. 10b. Rats with intra-NAcSh stimulation of paRac1 and intra-NAcSh stimulation of C450M exhibited similar nose poke responding during the cue re-exposure session (C450M = 6.00 ± 0.707, n = 8 animals; paRac1 = 5.67 ± 1.48, n = 6 animals, t12=0.22, p=0.8286, two-tail, unpaired t-test, n.s. > 0.05). (d) Nose poke responding during the 10-min cue re-exposure session in cocaine-trained rats whose behavioral results are presented in Fig. 6q. Rats with intra-NAcSh stimulation of paRac1 and intra-NAcSh stimulation of C450M exhibited similar nose poke responding during the cue re-exposure session (C450M = 18.78 ± 2.52, n = 9 animals; paRac1 = 18.38 ± 1.97, n = 8 animals, t15=0.12, p=0.9033, two-tail, unpaired t-test, n.s. > 0.05). (e) Diagram showing the experimental timeline and setup. (f) Summary showing that rats with NAcSh paRac1 and rats with NAcSh C450M (control) receiving intra-NAcSh photostimulation 2 hr after cue re-exposure exhibited similar cue-induced nose poke responding, measured 6 hr after cue re-exposure (C450 active = 23.43 ± 7.57, n = 7 animals; paRac1 active = 17.43 ± 3.72, n = 7 animals; C450 inactive = 7.57 ± 2.31, n = 7 animals; paRac1 inactive = 5.71 ± 1.96, n = 7 animals, F1,12=0.23, p=0.6370, two-way RM ANOVA, withdrawal day 45 hole x virus interaction, n.s. > 0.05). (g) Nose poke responding during the 10-min cue re-exposure session in saline-trained rats used in experiments in Supplementary Fig. 10f. Rats with intra-NAcSh paRac1 and intra-NAcSh C450M exhibited similar nose poke responding over the cue re-exposure session (C450M = 11.29 ± 1.92, n = 7 animals; paRac1 = 10.57 ± 1.96, n =7 animals, t12=0.26, p=0.7993, two-tail, unpaired t-test, n.s. > 0.05). (h) Nose poke responding during the 10-min cue re-exposure session in cocaine-trained rats whose behavioral results are presented in Fig. 6r. Rats with intra-NAcSh paRac1 and intra-NAcSh C450M exhibited similar nose poke responding during the cue re-exposure session (C450M = 28.63 ± 2.16, n = 8 animals; paRac1 = 25.80 ± 1.79, n = 10 animals, t16=1.02, p=0.3246, two-tail, unpaired t-test, n.s. > 0.05). (i) Nose poke responding during the 10-min cue re-exposure session in cocaine-trained rats whose behavioral results are presented in Fig. 6s. Rats with intra-NAcSh paRac1 and intra-NAcSh C450M exhibited similar nose poke responding during the cue re-exposure session (C450M = 27.50 ± 2.31, n = 8 animals; paRac1 = 26.00 ± 2.00, n = 8 animals, t14=0.42, p=0.6308, two-tail, unpaired t-test, n.s. > 0.05). Data presented as mean±SEM.

Supplementary Fig. 11 Viral localization and expression for cue re-exposure photostimulation in behavioral experiments.

(a-c) Example image showing HSV-mediated expression of C450M in the NAcSh and location of the optical fiber that delivered laser stimulation (a), and a magnified portion of the image showing viral expression in individual neurons (b) from a saline-trained rat used in behavioral experiments presented in Supplementary Fig. 10b. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (c). (d-f) Example image showing HSV-mediated expression of C450M in the NAcSh and location of the optical fiber that delivered laser stimulation (d), and a magnified portion of image showing viral expression in individual neurons (e) from a cocaine-trained rat used in behavioral experiments presented in Fig. 6q. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (f). (g-i) Example image showing HSV-mediated expression of paRac1 in the NAcSh and location of the optical fiber that delivered laser stimulation (g), and a magnified portion of image showing viral expression in individual neurons (h) from a saline-trained rat used in the behavioral experiments presented in Supplementary Fig. 10b. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (i). (j-l) Example image showing HSV-mediated expression of paRac1 in the NAcSh and location of the optical fiber that delivered later stimulation (j), and a magnified portion of the image showing viral expression in individual neurons (k) from a cocaine-trained rat used in behavioral experiments presented in Fig. 6q. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (l).

Supplementary Fig. 12 Viral localization and expression for 2 hr post photostimulation in behavioral experiments.

(a-c) Example image showing HSV-mediated expression of C450M in the NAcSh and location of the optical fiber that delivered laser stimulation (a), and a magnified portion of the image showing viral expression in individual neurons (b) from a saline-trained rats used in behavioral experiments presented in Supplementary Fig. 10f. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (c). (d-f) Example image showing HSV-mediated expression of C450M in the NAcSh and location of the optical fiber that delivered laser stimulation (d), and a magnified portion of the image showing viral expression in individual neurons (e) from a cocaine-trained rats used in behavioral experiments presented in Fig. 6r. Viral expression and fiber tip localization for all rats that were included in data analysis are presented in (f). (g-i) Example image showing HSV-mediated expression of paRac1 in the NAcSh and location of the optical fiber that delivered laser stimulation (g), and a magnified portion of image showing viral expression in individual neurons (h) from a saline-trained rat used in behavioral experiments presented in Supplementary Fig. 10f. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (i). (j-l) Example image showing HSV-mediated expression of paRac1 in the NAcSh and location of the optical fiber that delivered laser stimulation (j), and a magnified portion of the image showing viral expression in individual neurons (k) from a cocaine-trained rat used in behavioral experiments presented in Fig. 6r. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (l).

Supplementary Fig. 13 Viral localization and expression for photostimulation in long-term memory experiments.

(a-c) Example image showing HSV-mediated expression of C450M in the NAcSh and location of the optical fiber that delivered laser stimulation (a), and a magnified portion of the image showing viral expression in individual neurons (b) from a cocaine-trained rats used in behavioral experiments presented in Fig. 6s. Viral expression and fiber tip localization for all rats that were included in data analysis are presented in (c). (d-f) Example image showing HSV-mediated expression of paRac1 in the NAcSh and location of the optical fiber that delivered laser stimulation (d), and a magnified portion of the image showing viral expression in individual neurons (e) from a cocaine-trained rat used in behavioral experiments presented in Fig. 6s. Viral expression and fiber tip localizations for all rats that were included in data analysis are presented in (f).

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wright, W.J., Graziane, N.M., Neumann, P.A. et al. Silent synapses dictate cocaine memory destabilization and reconsolidation. Nat Neurosci 23, 32–46 (2020). https://doi.org/10.1038/s41593-019-0537-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-019-0537-6

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