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:

Cocaine- and stress-primed reinstatement of drug-associated memories elicit differential behavioral and frontostriatal circuit activity patterns via recruitment of L-type Ca2+ channels

A Correction to this article was published on 06 December 2019

This article has been updated

Abstract

Cocaine-associated memories are critical drivers of relapse in cocaine-dependent individuals that can be evoked by exposure to cocaine or stress. Whether these environmental stimuli recruit similar molecular and circuit-level mechanisms to promote relapse remains largely unknown. Here, using cocaine- and stress-primed reinstatement of cocaine conditioned place preference to model drug-associated memories, we find that cocaine drives reinstatement by increasing the duration that mice spend in the previously cocaine-paired context whereas stress increases the number of entries into this context. Importantly, both forms of reinstatement require Cav1.2 L-type Ca2+ channels (LTCCs) in cells of the prelimbic cortex that project to the nucleus accumbens core (PrL→NAcC). Utilizing fiber photometry to measure circuit activity in vivo in conjunction with the LTCC blocker, isradipine, we find that LTCCs drive differential recruitment of the PrL→ NAcC pathway during cocaine- and stress-primed reinstatement. While cocaine selectively activates PrL→NAcC cells prior to entry into the cocaine-paired chamber, a measure that is predictive of duration in that chamber, stress increases persistent activity of this projection, which correlates with entries into the cocaine-paired chamber. Using projection-specific chemogenetic manipulations, we show that PrL→NAcC activity is required for both cocaine- and stress-primed reinstatement, and that activation of this projection in Cav1.2-deficient mice restores reinstatement. These data indicate that LTCCs are a common mediator of cocaine- and stress-primed reinstatement. However, they engage different patterns of behavior and PrL→NAcC projection activity depending on the environmental stimuli. These findings establish a framework to further study how different environmental experiences can drive relapse, and supports further exploration of isradipine, an FDA-approved LTCC blocker, as a potential therapeutic for the prevention of relapse in cocaine-dependent individuals.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Change history

References

  1. Stewart J. Review. Psychological and neural mechanisms of relapse. Philos Trans R Soc Lond B Biol Sci. 2008;363:3147–58.

    PubMed  PubMed Central  Google Scholar 

  2. White JA, McKinney BC, John MC, Powers PA, Kamp TJ, Murphy GG. Conditional forebrain deletion of the L-type calcium channel Ca V 1.2 disrupts remote spatial memories in mice. Learn Mem. 2008;15:1–5.

    CAS  PubMed  Google Scholar 

  3. Moosmang S, Haider N, Klugbauer N, Adelsberger H, Langwieser N, Muller J, et al. Role of hippocampal Cav1.2 Ca2+ channels in NMDA receptor-independent synaptic plasticity and spatial memory. J Neurosci. 2005;25:9883–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The physiology, pathology, and pharmacology of voltage-gated calcium channels and their future therapeutic potential. Pharmacol Rev. 2015;67:821–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Heyes S, Pratt WS, Rees E, Dahimene S, Ferron L, Owen MJ, et al. Genetic disruption of voltage-gated calcium channels in psychiatric and neurological disorders. Prog Neurobiol. 2015;134:36–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kabir ZD, Martinez-Rivera A, Rajadhyaksha AM. From gene to behavior: L-type calcium channel mechanisms underlying neuropsychiatric symptoms. Neurotherapeutics. 2017;14:588–613.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Cassidy F, Ahearn EP, Carroll BJ. Substance abuse in bipolar disorder. Bipolar Disord. 2001;3:181–8.

    CAS  PubMed  Google Scholar 

  8. Green AI, Brown ES. Comorbid schizophrenia and substance abuse. J Clin Psychiatry. 2006;67:e08.

    PubMed  Google Scholar 

  9. Post RM, Kalivas P. Bipolar disorder and substance misuse: pathological and therapeutic implications of their comorbidity and cross-sensitisation. Br J Psychiatry. 2013;202:172–6.

    PubMed  PubMed Central  Google Scholar 

  10. Lancaster TM, Heerey EA, Mantripragada K, Linden DE. CACNA1C risk variant affects reward responsiveness in healthy individuals. Transl Psychiatry. 2014;4:e461.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Wessa M, Linke J, Witt SH, Nieratschker V, Esslinger C, Kirsch P, et al. The CACNA1C risk variant for bipolar disorder influences limbic activity. Mol Psychiatry. 2010;15:1126–7.

    CAS  PubMed  Google Scholar 

  12. Addy NA, Nunes EJ, Hughley SM, Small KM, Baracz SJ, Haight JL, et al. The L-type calcium channel blocker, isradipine, attenuates cue-induced cocaine-seeking by enhancing dopaminergic activity in the ventral tegmental area to nucleus accumbens pathway. Neuropsychopharmacology. 2018;43:2361–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Du C, Volkow ND, You J, Park K, Allen CP, Koob GF et al. Cocaine-induced ischemia in prefrontal cortex is associated with escalation of cocaine intake in rodents. Mol Psychiatry. 2018. https://doi.org/10.1038/s41380-018-0261-8. [Epub ahead of print].

  14. Degoulet M, Stelly CE, Ahn KC, Morikawa H. L-type Ca(2)(+) channel blockade with antihypertensive medication disrupts VTA synaptic plasticity and drug-associated contextual memory. Mol Psychiatry. 2016;21:394–402.

    CAS  PubMed  Google Scholar 

  15. Burgdorf CE, Schierberl KC, Lee AS, Fischer DK, Van Kempen TA, Mudragel V, et al. Extinction of contextual cocaine memories requires Cav1.2 within D1R-expressing cells and recruits hippocampal Cav1.2-dependent signaling mechanisms. J Neurosci. 2017;37:11894–911.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Schierberl K, Hao J, Tropea TF, Ra S, Giordano TP, Xu Q, et al. Cav1.2 L-type Ca(2)(+) channels mediate cocaine-induced GluA1 trafficking in the nucleus accumbens, a long-term adaptation dependent on ventral tegmental area Ca(v)1.3 channels. J Neurosci. 2011;31:13562–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Giordano TP, Tropea TF, Satpute SS, Sinnegger-Brauns MJ, Striessnig J, Kosofsky BE, et al. Molecular switch from L-type Ca v 1.3 to Ca v 1.2 Ca2+ channel signaling underlies long-term psychostimulant-induced behavioral and molecular plasticity. J Neurosci. 2010;30:17051–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Terrillion CE, Dao DT, Cachope R, Lobo MK, Puche AC, Cheer JF, et al. Reduced levels of Cacna1c attenuate mesolimbic dopamine system function. Genes Brain Behav. 2017;16:495–505.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Dedic N, Pohlmann ML, Richter JS, Mehta D, Czamara D, Metzger MW, et al. Cross-disorder risk gene CACNA1C differentially modulates susceptibility to psychiatric disorders during development and adulthood. Mol Psychiatry. 2018;23:533–43.

    CAS  PubMed  Google Scholar 

  20. Bavley CC, Fischer DK, Rizzo BK, Rajadhyaksha AM. Cav1.2 channels mediate persistent chronic stress-induced behavioral deficits that are associated with prefrontal cortex activation of the p25/Cdk5-glucocorticoid receptor pathway. Neurobiol Stress. 2017;7:27–37.

    PubMed  PubMed Central  Google Scholar 

  21. Terrillion CE, Francis TC, Puche AC, Lobo MK, Gould TD. Decreased nucleus accumbens expression of psychiatric disorder risk gene Cacna1c promotes susceptibility to social stress. Int J Neuropsychopharmacol. 2017;20:428–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. McFarland K, Lapish CC, Kalivas PW. Prefrontal glutamate release into the core of the nucleus accumbens mediates cocaine-induced reinstatement of drug-seeking behavior. J Neurosci. 2003;23:3531–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Park WK, Bari AA, Jey AR, Anderson SM, Spealman RD, Rowlett JK, et al. Cocaine administered into the medial prefrontal cortex reinstates cocaine-seeking behavior by increasing AMPA receptor-mediated glutamate transmission in the nucleus accumbens. J Neurosci. 2002;22:2916–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. McFarland K, Kalivas PW. The circuitry mediating cocaine-induced reinstatement of drug-seeking behavior. J Neurosci. 2001;21:8655–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. McFarland K, Davidge SB, Lapish CC, Kalivas PW. Limbic and motor circuitry underlying footshock-induced reinstatement of cocaine-seeking behavior. J Neurosci. 2004;24:1551–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Soeiro-de-Souza MG, Lafer B, Moreno RA, Nery FG, Chile T, Chaim K, et al. The CACNA1C risk allele rs1006737 is associated with age-related prefrontal cortical thinning in bipolar I disorder. Transl Psychiatry. 2017;7:e1086.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Kabir ZD, Lee AS, Rajadhyaksha AM. L-type Ca(2+) channels in mood, cognition and addiction: integrating human and rodent studies with a focus on behavioural endophenotypes. J Physiol. 2016;594:5823–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Lee AS, Ra S, Rajadhyaksha AM, Britt JK, De Jesus-Cortes H, Gonzales KL, et al. Forebrain elimination of cacna1c mediates anxiety-like behavior in mice. Mol Psychiatry. 2012;17:1054–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Bavley CC, Rice RC, Fischer DK, Fakira AK, Byrne M, Kosovsky M, et al. Rescue of learning and memory deficits in the human nonsyndromic intellectual disability cereblon knock-out mouse model by targeting the AMP-activated protein kinase-mTORC1 translational pathway. J Neurosci. 2018;38:2780–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Milner TA, Waters EM, Robinson DC, Pierce JP. Degenerating processes identified by electron microscopic immunocytochemical methods. Methods Mol Biol. 2011;793:23–59.

    CAS  PubMed  Google Scholar 

  31. Rajadhyaksha A, Husson I, Satpute SS, Kuppenbender KD, Ren JQ, Guerriero RM, et al. L-type Ca2+ channels mediate adaptation of extracellular signal-regulated kinase 1/2 phosphorylation in the ventral tegmental area after chronic amphetamine treatment. J Neurosci. 2004;24:7464–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Gunaydin LA, Grosenick L, Finkelstein JC, Kauvar IV, Fenno LE, Adhikari A, et al. Natural neural projection dynamics underlying social behavior. Cell. 2014;157:1535–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Lerner TN, Shilyansky C, Davidson TJ, Evans KE, Beier KT, Zalocusky KA, et al. Intact-brain analyses reveal distinct information carried by SNc dopamine subcircuits. Cell. 2015;162:635–47.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, Cusher DS, et al. Rapid, reversible activation of AgRP neurons drives feeding behavior in mice. J Clin Investig. 2011;121:1424–8.

    CAS  PubMed  Google Scholar 

  35. Hell JW, Westenbroek RE, Warner C, Ahlijanian MK, Prystay W, Gilbert MM, et al. Identification and differential subcellular localization of the neuronal class C and class D L-type calcium channel alpha 1 subunits. J Cell Biol. 1993;123:949–62.

    CAS  PubMed  Google Scholar 

  36. Sinnegger-Brauns MJ, Huber IG, Koschak A, Wild C, Obermair GJ, Einzinger U, et al. Expression and 1,4-dihydropyridine-binding properties of brain L-type calcium channel isoforms. Mol Pharmacol. 2009;75:407–14.

    CAS  PubMed  Google Scholar 

  37. Seisenberger C, Specht V, Welling A, Platzer J, Pfeifer A, Kuhbandner S, et al. Functional embryonic cardiomyocytes after disruption of the L-type alpha1C (Cav1.2) calcium channel gene in the mouse. J Biol Chem. 2000;275:39193–9.

    CAS  PubMed  Google Scholar 

  38. Capriles N, Rodaros D, Sorge RE, Stewart J. A role for the prefrontal cortex in stress- and cocaine-induced reinstatement of cocaine seeking in rats. Psychopharmacology. 2003;168:66–74.

    CAS  PubMed  Google Scholar 

  39. Zavala AR, Weber SM, Rice HJ, Alleweireldt AT, Neisewander JL. Role of the prelimbic subregion of the medial prefrontal cortex in acquisition, extinction, and reinstatement of cocaine-conditioned place preference. Brain Res. 2003;990:157–64.

    CAS  PubMed  Google Scholar 

  40. Augur IF, Wyckoff AR, Aston-Jones G, Kalivas PW, Peters J. Chemogenetic activation of an extinction neural circuit reduces cue-induced reinstatement of cocaine seeking. J Neurosci. 2016;36:10174–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Peters J, LaLumiere RT, Kalivas PW. Infralimbic prefrontal cortex is responsible for inhibiting cocaine seeking in extinguished rats. J Neurosci. 2008;28:6046–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Gutman AL, Nett KE, Cosme CV, Worth WR, Gupta SC, Wemmie JA, et al. Extinction of cocaine seeking requires a window of infralimbic pyramidal neuron activity after unreinforced lever presses. J Neurosci. 2017;37:6075–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Wang F, McIntosh AM, He Y, Gelernter J, Blumberg HP. The association of genetic variation in CACNA1C with structure and function of a frontotemporal system. Bipolar Disord. 2011;13:696–700.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Paulus FM, Bedenbender J, Krach S, Pyka M, Krug A, Sommer J, et al. Association of rs1006737 in CACNA1C with alterations in prefrontal activation and fronto-hippocampal connectivity. Hum Brain Mapp. 2014;35:1190–1200.

    PubMed  Google Scholar 

  45. Berendse HW, Galis-de Graaf Y, Groenewegen HJ. Topographical organization and relationship with ventral striatal compartments of prefrontal corticostriatal projections in the rat. J Comp Neurol. 1992;316:314–47.

    CAS  PubMed  Google Scholar 

  46. Vranjkovic O, Hang S, Baker DA, Mantsch JR. beta-adrenergic receptor mediation of stress-induced reinstatement of extinguished cocaine-induced conditioned place preference in mice: roles for beta1 and beta2 adrenergic receptors. J Pharmacol Exp Ther. 2012;342:541–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Ellwood IT, Patel T, Wadia V, Lee AT, Liptak AT, Bender KJ, et al. Tonic or phasic stimulation of dopaminergic projections to prefrontal cortex causes mice to maintain or deviate from previously learned behavioral strategies. J Neurosci. 2017;37:8315–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Finlay JM, Zigmond MJ, Abercrombie ED. Increased dopamine and norepinephrine release in medial prefrontal cortex induced by acute and chronic stress: effects of diazepam. Neuroscience. 1995;64:619–28.

    CAS  PubMed  Google Scholar 

  49. Zhang Z, Cordeiro Matos S, Jego S, Adamantidis A, Seguela P. Norepinephrine drives persistent activity in prefrontal cortex via synergistic alpha1 and alpha2 adrenoceptors. PLoS One. 2013;8:e66122.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Schwabe L, Hoffken O, Tegenthoff M, Wolf OT. Preventing the stress-induced shift from goal-directed to habit action with a beta-adrenergic antagonist. J Neurosci. 2011;31:17317–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Patriarchi T, Buonarati OR, Hell JW. Postsynaptic localization and regulation of AMPA receptors and Cav1.2 by beta2 adrenergic receptor/PKA and Ca(2+)/CaMKII signaling. EMBO J. 2018;37:e99771. https://doi.org/10.15252/embj.201899771.

  52. McReynolds JR, Doncheck EM, Li Y, Vranjkovic O, Graf EN, Ogasawara D, et al. Stress promotes drug seeking through glucocorticoid-dependent endocannabinoid mobilization in the prelimbic cortex. Biol Psychiatry. 2017;84:85–94.

Download references

Acknowledgements

We would like to thank Nii Addy, Kristen Pleil, and Francis Lee for their input and edits on the paper. This work was supported by grants to AMR from the National Institute of Drug Abuse (5R01DA029122), The New York Weill Cornell Alumni Council Award, and the Paul Fund along with grants to CL from the National Institute of Mental Health, the One Mind Institute, The Hartwell Foundation, the Rita Allen foundation, the Klingenstein–Simons Foundation Fund, and the Brain and Behavior Research Foundation. Grants to NVD from Kellen Jr Faculty (Anna-Maria and Stephen Kellen Foundation) and Affinito-Stewart (President’s Council of Cornell Women), grants to TLK from NIAAA (U24AA025475 and U01AA020911), and grants to TAM from NIH (DA08259 and HL098351). CCB was supported by a T32 grant from NIDA (T32DA039080-01), a TL1 grant from the National Center for Advancing Translation Sciences (TL1TR002386), and the Frank & Blanche Mowrer Memorial Fellowship. RNF was supported by a T32 grant from NIGMS (T32GM007739) and an NRSA from NIMH (F30MH115622). CEB was supported by a T32 grant from NIDA (T32DA039080-01) and the Frank & Blanche Mowrer Memorial Fellowship. BSH was supported by an NSF GRFP fellowship (ID# 2015174265, Award# 1257284) and the Jacques Cohenca Predoctoral Fellowship. RB was supported by a T32 grant from NIGMS (T32GM007739) and an NRSA from NIMH (F30MH117939).

Author information

Authors and Affiliations

Authors

Contributions

CCB, AMR, and CL designed experiments and wrote the paper. CCB, CEB, and DKF ran behavioral experiments. CCB and BH ran fiber photometry experiments. CCB performed stereotaxic surgeries. CCB and NS analyzed data for behavioral experiments. RNF analyzed data for fiber photometry experiments. APW, JH, and SA ran immunohistological experiments. TLK provided reagents. APW, NHC, and TAM performed RNAscope experiments. RB and NVD took confocal images for RNAscope experiments. NVD provided guidance for viral experiments.

Corresponding author

Correspondence to Anjali M. Rajadhyaksha.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bavley, C.C., Fetcho, R.N., Burgdorf, C.E. et al. Cocaine- and stress-primed reinstatement of drug-associated memories elicit differential behavioral and frontostriatal circuit activity patterns via recruitment of L-type Ca2+ channels. Mol Psychiatry 25, 2373–2391 (2020). https://doi.org/10.1038/s41380-019-0513-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-019-0513-2

This article is cited by

Search

Quick links