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.

A cannabinoid link between mitochondria and memory

Abstract

Cellular activity in the brain depends on the high energetic support provided by mitochondria, the cell organelles which use energy sources to generate ATP1,2,3,4. Acute cannabinoid intoxication induces amnesia in humans and animals5,6, and the activation of type-1 cannabinoid receptors present at brain mitochondria membranes (mtCB1) can directly alter mitochondrial energetic activity7,8,9. Although the pathological impact of chronic mitochondrial dysfunctions in the brain is well established1,2, the involvement of acute modulation of mitochondrial activity in high brain functions, including learning and memory, is unknown. Here, we show that acute cannabinoid-induced memory impairment in mice requires activation of hippocampal mtCB1 receptors. Genetic exclusion of CB1 receptors from hippocampal mitochondria prevents cannabinoid-induced reduction of mitochondrial mobility, synaptic transmission and memory formation. mtCB1 receptors signal through intra-mitochondrial Gαi protein activation and consequent inhibition of soluble-adenylyl cyclase (sAC). The resulting inhibition of protein kinase A (PKA)-dependent phosphorylation of specific subunits of the mitochondrial electron transport system eventually leads to decreased cellular respiration. Hippocampal inhibition of sAC activity or manipulation of intra-mitochondrial PKA signalling or phosphorylation of the Complex I subunit NDUFS2 inhibit bioenergetic and amnesic effects of cannabinoids. Thus, the G protein-coupled mtCB1 receptors regulate memory processes via modulation of mitochondrial energy metabolism. By directly linking mitochondrial activity to memory formation, these data reveal that bioenergetic processes are primary acute regulators of cognitive functions.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Requirement of mtCB1 receptors for cannabinoid effects on mitochondrial respiration and mobility, synaptic transmission and memory.
Figure 2: Activation of mtCB1 receptors affects mitochondrial activity via Gαi/o proteins and sAC.
Figure 3: Blockade of sAC activity prevents the effects of cannabinoids on mitochondrial respiration and mobility, synaptic transmission and memory.
Figure 4: Activation of mtCB1 decreases PKA-dependent phosphorylation of mitochondrial proteins.

Similar content being viewed by others

References

  1. Mattson, M. P., Gleichmann, M. & Cheng, A. Mitochondria in neuroplasticity and neurological disorders. Neuron 60, 748–766 (2008)

    Article  CAS  Google Scholar 

  2. Sheng, Z. H. & Cai, Q. Mitochondrial transport in neurons: impact on synaptic homeostasis and neurodegeneration. Nat. Rev. Neurosci. 13, 77–93 (2012)

    Article  CAS  Google Scholar 

  3. Rangaraju, V., Calloway, N. & Ryan, T. A. Activity-driven local ATP synthesis is required for synaptic function. Cell 156, 825–835 (2014)

    Article  CAS  Google Scholar 

  4. Attwell, D. & Laughlin, S. B. An energy budget for signaling in the grey matter of the brain. J. Cereb. Blood Flow Metab. 21, 1133–1145 (2001)

    Article  CAS  Google Scholar 

  5. Broyd, S. J., van Hell, H. H., Beale, C., Yücel, M. & Solowij, N. Acute and chronic effects of cannabinoids on human cognition—a systematic review. Biol. Psychiatry 79, 557–567 (2016)

    Article  CAS  Google Scholar 

  6. Marsicano, G. & Lafenêtre, P. Roles of the endocannabinoid system in learning and memory. Curr. Top. Behav. Neurosci. 1, 201–230 (2009)

    Article  CAS  Google Scholar 

  7. Bénard, G. et al. Mitochondrial CB1 receptors regulate neuronal energy metabolism. Nat. Neurosci. 15, 558–564 (2012)

    Article  Google Scholar 

  8. Hebert-Chatelain, E. et al. Cannabinoid control of brain bioenergetics: Exploring the subcellular localization of the CB1 receptor. Mol. Metab. 3, 495–504 (2014)

    Article  CAS  Google Scholar 

  9. Koch, M. et al. Hypothalamic POMC neurons promote cannabinoid-induced feeding. Nature 519, 45–50 (2015)

    Article  CAS  ADS  Google Scholar 

  10. Claros, M. G. MitoProt, a Macintosh application for studying mitochondrial proteins. Comput. Appl. Biosci. 11, 441–447 (1995)

    CAS  PubMed  Google Scholar 

  11. Nakai, K. & Horton, P. PSORT: a program for detecting sorting signals in proteins and predicting their subcellular localization. Trends Biochem. Sci. 24, 34–36 (1999)

    Article  CAS  Google Scholar 

  12. Boesmans, W., Ameloot, K., van den Abbeel, V., Tack, J. & Vanden Berghe, P. Cannabinoid receptor 1 signalling dampens activity and mitochondrial transport in networks of enteric neurones. Neurogastroenterol. and Motil. 21, 958–968 (2009)

    Article  CAS  Google Scholar 

  13. Kano, M., Ohno-Shosaku, T., Hashimotodani, Y., Uchigashima, M. & Watanabe, M. Endocannabinoid-mediated control of synaptic transmission. Physiol. Rev. 89, 309–380 (2009)

    Article  CAS  Google Scholar 

  14. Puighermanal, E. et al. Cannabinoid modulation of hippocampal long-term memory is mediated by mTOR signaling. Nat. Neurosci. 12, 1152–1158 (2009)

    Article  CAS  Google Scholar 

  15. Benincá, C. et al. A new non-canonical pathway of Gαq protein regulating mitochondrial dynamics and bioenergetics. Cell. Signal. 26, 1135–1146 (2014)

    Article  Google Scholar 

  16. Lyssand, J. S. & Bajjalieh, S. M. The heterotrimeric G protein subunit Gαi is present on mitochondria. FEBS Lett. 581, 5765–5768 (2007)

    Article  CAS  Google Scholar 

  17. Acin-Perez, R. et al. Modulation of mitochondrial protein phosphorylation by soluble adenylyl cyclase ameliorates cytochrome oxidase defects. EMBO Mol. Med. 1, 392–406 (2009)

    Article  CAS  Google Scholar 

  18. Chen, Y. et al. Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor. Science 289, 625–628 (2000)

    Article  CAS  ADS  Google Scholar 

  19. Pierre, S., Eschenhagen, T., Geisslinger, G. & Scholich, K. Capturing adenylyl cyclases as potential drug targets. Nat. Rev. Drug Discov. 8, 321–335 (2009)

    Article  CAS  Google Scholar 

  20. Hess, K. C. et al. The ‘soluble’ adenylyl cyclase in sperm mediates multiple signaling events required for fertilization. Dev. Cell 9, 249–259 (2005)

    Article  CAS  Google Scholar 

  21. Acin-Perez, R. et al. Cyclic AMP produced inside mitochondria regulates oxidative phosphorylation. Cell Metab. 9, 265–276 (2009)

    Article  CAS  Google Scholar 

  22. Livigni, A. et al. Mitochondrial AKAP121 links cAMP and src signaling to oxidative metabolism. Mol. Biol. Cell 17, 263–271 (2006)

    Article  CAS  Google Scholar 

  23. Chen, Q., Lin, R. Y. & Rubin, C. S. Organelle-specific targeting of protein kinase AII (PKAII). Molecular and in situ characterization of murine A kinase anchor proteins that recruit regulatory subunits of PKAII to the cytoplasmic surface of mitochondria. J. Biol. Chem. 272, 15247–15257 (1997)

    Article  CAS  Google Scholar 

  24. Niswender, C. M. et al. Cre recombinase-dependent expression of a constitutively active mutant allele of the catalytic subunit of protein kinase A. Genesis 43, 109–119 (2005)

    Article  CAS  Google Scholar 

  25. Amanchy, R. et al. A curated compendium of phosphorylation motifs. Nat. Biotechnol. 25, 285–286 (2007)

    Article  CAS  Google Scholar 

  26. Neuberger, G., Schneider, G. & Eisenhaber, F. pkaPS: prediction of protein kinase A phosphorylation sites with the simplified kinase-substrate binding model. Biol. Direct 2, 1–23 (2007)

    Article  Google Scholar 

  27. Lin, A. L. et al. Decreased in vitro mitochondrial function is associated with enhanced brain metabolism, blood flow, and memory in Surf1-deficient mice. J. Cereb. Blood Flow Metab. 33, 1605–1611 (2013)

    Article  CAS  Google Scholar 

  28. Roubertoux, P. L. et al. Mitochondrial DNA modifies cognition in interaction with the nuclear genome and age in mice. Nat. Genet. 35, 65–69 (2003)

    Article  CAS  Google Scholar 

  29. Bockaert, J. & Marin, P. mTOR in brain physiology and pathologies. Physiol. Rev. 95, 1157–1187 (2015)

    Article  CAS  Google Scholar 

  30. Pacher, P., Bátkai, S. & Kunos, G. The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol. Rev. 58, 389–462 (2006)

    Article  CAS  Google Scholar 

  31. Marsicano, G. et al. The endogenous cannabinoid system controls extinction of aversive memories. Nature 418, 530–534 (2002)

    Article  CAS  ADS  Google Scholar 

  32. Martín-Couce, L. et al. Chemical probes for the recognition of cannabinoid receptors in native systems. Angew. Chem. Int. Ed. Engl. 51, 6896–6899 (2012)

    Article  Google Scholar 

  33. Narayana, N., Cox, S., Shaltiel, S., Taylor, S. S. & Xuong, N. Crystal structure of a polyhistidine-tagged recombinant catalytic subunit of cAMP-dependent protein kinase complexed with the peptide inhibitor PKI(5-24) and adenosine. Biochemistry 36, 4438–4448 (1997)

    Article  CAS  Google Scholar 

  34. McClure, C., Cole, K. L., Wulff, P., Klugmann, M. & Murray, A. J. Production and titering of recombinant adeno-associated viral vectors. J. Vis. Exp. 27, e3348 (2011)

    Google Scholar 

  35. Soria-Gómez, E. et al. The endocannabinoid system controls food intake via olfactory processes. Nat. Neurosci. 17, 407–415 (2014)

    Article  Google Scholar 

  36. Chiarlone, A. et al. A restricted population of CB1 cannabinoid receptors with neuroprotective activity. Proc. Natl Acad. Sci. USA 111, 8257–8262 (2014)

    Article  CAS  ADS  Google Scholar 

  37. López-Doménech, G. et al. The Eutherian Armcx genes regulate mitochondrial trafficking in neurons and interact with Miro and Trak2. Nat. Commun. 3, 814 (2012)

    Article  ADS  Google Scholar 

  38. Jose, C. et al. AICAR inhibits cancer cell growth and triggers cell-type distinct effects on OXPHOS biogenesis, oxidative stress and Akt activation. Biochim. Biophys. Acta 1807, 707–718 (2011)

    Article  CAS  Google Scholar 

  39. Hébert Chatelain, E., Dupuy, J. W., Letellier, T. & Dachary-Prigent, J. Functional impact of PTP1B-mediated Src regulation on oxidative phosphorylation in rat brain mitochondria. Cell. Mol. Life Sci. 68, 2603–2613 (2011)

    Article  Google Scholar 

  40. Pocaly, M. et al. Proteomic analysis of an imatinib-resistant K562 cell line highlights opposing roles of heat shock cognate 70 and heat shock 70 proteins in resistance. Proteomics 8, 2394–2406 (2008)

    Article  CAS  Google Scholar 

  41. Käll, L., Canterbury, J. D., Weston, J., Noble, W. S. & MacCoss, M. J. Semi-supervised learning for peptide identification from shotgun proteomics datasets. Nat. Methods 4, 923–925 (2007)

    Article  Google Scholar 

  42. Zala, D. et al. Vesicular glycolysis provides on-board energy for fast axonal transport. Cell 152, 479–491 (2013)

    Article  CAS  Google Scholar 

  43. De Vos, K. J. & Sheetz, M. P. Visualization and quantification of mitochondrial dynamics in living animal cells. Methods Cell Biol. 80, 627–682 (2007)

    Article  CAS  Google Scholar 

  44. Courchet, J. et al. Terminal axon branching is regulated by the LKB1-NUAK1 kinase pathway via presynaptic mitochondrial capture. Cell 153, 1510–1525 (2013)

    Article  CAS  Google Scholar 

  45. Rodríguez-Puertas, R., Barreda-Gómez, G., Giralt, M. & Fernández-Pastor, B. Method of quantifying the G protein-coupled receptor (GPCR)/G protein coupling using a cell membrane array. Patent US 8017346 B2. Google Patents (2013)

  46. Rodriguez-Puertas, R. et al. Method for the surface treatment of solid substrates. Patent EP 2048534 A1 Google Patents (2009)

  47. Busquets-Garcia, A. et al. Differential role of anandamide and 2-arachidonoylglycerol in memory and anxiety-like responses. Biol. Psychiatry 70, 479–486 (2011)

    Article  CAS  Google Scholar 

  48. Puighermanal, E. et al. Dissociation of the pharmacological effects of THC by mTOR blockade. Neuropsychopharmacology 38, 1334–1343 (2013)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Gonzales,N. Aubailly and all the personnel of the Animal Facility of the NeuroCentre Magendie, M. Biguerie of the technical service of the NeuroCentre Magendie, the personnel from the Bordeaux Imaging Center and V. Morales for continuous help. We thank G. Manfredi (Cornell University) for the sAC–HA, A. Feliciello (University of Napoli) for anti-AKAP121 antiserum and for AKAP121 shRNA, and M. Montcouquiol, N. Piguel (INSERM U1215, Bordeaux) and R. Rossignol (University of Bordeaux) for help with experiments. We thank F. Francia, P. V. Piazza, A. Bacci, G. Ferreira, F. Chaouloff and M. Guzman for critical reading and the members of Marsicano’s laboratory for discussions. This work was supported by INSERM (G.M., D.C.), EU–Fp7 (PAINCAGE, HEALTH-603191, G.M. and FP7-PEOPLE-2013-IEF-623638, A.B.-G.), European Research Council (Endofood, ERC–2010–StG–260515 and CannaPreg, ERC-2014-PoC-640923, G.M.), Fondation pour la Recherche Medicale (DRM20101220445, G.M., SPF20121226369, R.S. and ARF20140129235, L.B.), Fondation pour la Recherche en Psychiatrie et en Santé Mentale (FRPSM, G.M.), Human Frontiers Science Program (RGP0036/2014, G.M.), Region Aquitaine (G.M.), AFM Telethon Trampoline Grant (16474, G.B.), Agence Nationale de la Recherche (ANR Blanc NeuroNutriSens ANR-13-BSV4-0006, G.M., D.C., BRAIN ANR-10-LABX-0043, G.M., D.C., F.M. and ANR-10-IDEX-03-02, A.B.-G.), Dulbecco Telethon Institute post-doc fellowship (E.H.-C.), NSERC (RGPIN-2015-05880, E.H.-C.), Fyssen Foundation (E.S.-G.), EMBO post-doc fellowship (L.B.), CONACyT (E.S.-G.), Zabalduz pre-doc fellowship (M.D.G.-F.), the Basque Government (IT764-13, P.G.), MINECO/FEDER (SAF2015-65034-R, P.G.), University of the Basque Country (UPV/EHU UFI11/41, P.G.), Red de Trastornos Adictivos—Instituto de Salud Carlos III (RD12/0028/0004, RD16/0017/0012, P.G.).

Author information

Authors and Affiliations

Authors

Contributions

E.H.-C., T.D., R.S. and L.B. performed biochemical, molecular biology, behavioural and cellular experiments; E.S.-G., A.B.-G. and L.M.R. helped with behavioural experiments and analyses; E.S.-G., A.C.P.Z., A.C., A.D., P.V. and M.V. helped with biochemistry and molecular biology; G.T., M.C., F.D., W.M., D.C. and F.M. performed electrophysiological studies; M.D.G.-F. and G.B.-G. performed G protein signalling and binding experiments; N.P., L.R., I.E. and P.G. performed electron microscopy experiments; J.-W.D. provided proteomics experiments; M.-L.L.-R. provided reagents; G.B. and G.M. supervised the work; E.H.-C., T.D., G.B. and G.M. wrote the manuscript; all authors discussed results and edited the manuscript.

Corresponding author

Correspondence to Giovanni Marsicano.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information

Nature thanks M. Mattson 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 The mutant DN22-CB1 receptor is functional, but it does not mediate cannabinoid-induced alterations of mitochondrial activity and mobility.

a, Acute treatment with HU210 (100 nM) induces changes in cellular oxygen consumption in intact primary fibroblasts (MFs) from CB1−/− mice electroporated with a CB1-expressing plasmid, whereas no alterations were observed in DN22-CB1-expressing or mock-electroporated (mCherry) CB1−/− mouse fibroblasts (n = 6–10). b, HU210 (200 nM) treatment decreases HEK293 cellular respiration in cells expressing CB1, but not mCherry or DN22-CB1 receptors (n = 4). c, Representative immunoblotting showing the effect of HU210 (100 nM) on pERK and ERK levels in HEK293 cells transfected with plasmids expressing mCherry, CB1 or DN22-CB1 (n = 7–13). Quantitative data are shown in Fig. 1b of main text. d, Representative confocal images (green, GFP; red, pDsred2–mito) and kymographs corresponding to the results shown in Fig. 1d; showing how HU210 (1 μM) reduces mitochondrial mobility in the primary neurons from CB1−/− mice transfected with vectors expressing CB1, but not GFP or DN22-CB1. Scale bar, 10 μM. Data, mean ± s.e.m.; *P < 0.05. For statistics, see Supplementary Tables 1–3.

Source data

Extended Data Figure 2 Characterization of CB1+/+(GFP), CB1−/−(GFP), CB1−/−(CB1) and CB1−/−(DN22-CB1) mice.

a, Representative immunofluorescence micrographs showing CB1-receptor expression in hippocampal brain sections from CB1+/+(GFP), CB1−/−(GFP), CB1−/−(CB1) and CB1−/−(DN22-CB1) mice, respectively. b, [3H]CP55,940 CB1-receptor binding experiments, showing similar levels of CB1 protein expression in hippocampal extracts from CB1−/−(CB1) and CB1−/−(DN22-CB1) mice, both significantly higher than control CB1−/−(GFP) (n = 7–24). c, Dose–response curves of [35S]GTPγS binding in membranes isolated from hippocampi of CB1−/−(GFP), CB1−/−(CB1) and CB1−/−(DN22-CB1) mice treated with WIN (n = 9–24). d, e, CB1-receptor-dependent activation of G proteins is similar in hippocampi from CB1−/−(CB1) and CB1−/−(DN22-CB1) mice, as shown by EC50 dose and maximal activation values calculated from [35S]GTPγS binding experiments after treatment with HU210 (d) or WIN (e). f, Total exploration time in NOR experiments as described in Fig. 1i. Data, mean ± s.e.m.; ***P < 0.001; NS, not significant. For statistics, see Supplementary Tables 1–3.

Source data

Extended Data Figure 3 Activation of mtCB1 receptor affects mitochondrial activity via Gαi/o signalling.

a, THC (1 μM) or WIN (1 μM) decrease cellular respiration of primary hippocampal cultures derived from CB1+/+ but not from CB1−/− mice (n = 5–15). b, THC (1 μM) or WIN (1 μM) reduce both total and mitochondrial ATP levels of primary hippocampal cultures derived from CB1+/+ but not from CB1−/− mice (n = 6–14). c, Quantification of data presented in Fig. 2b, showing the release of the G protein upon activation of mtCB1 receptors (n = 3). d, e, Effects of pertussis toxin (PTX, 1 μg ml−1) and THC (800 nM) on cAMP levels in purified brain mitochondria obtained from CB1+/+ mice (d) and CB1−/− littermates (e) (n = 4). f, g, Effects of PTX (1 μg ml−1) and THC (800 nM) on PKA activity in purified brain mitochondria obtained from CB1+/+ mice (f) and CB1−/− littermates (g) (n = 5–6). h, PTX (1 μg ml−1) blocks the effect of THC (800 nM) and WIN (100 nM) on complex-I activity of purified brain mitochondria obtained from CB1+/+ mice (n = 3–6). i, Representative trypsin-sensitivity assay (3 independent experiments) showing the intra-mitochondrial localization of Gα, sAC and PKA. NDUFA9, a subunit of complex I; TOM20, translocase of outer membrane subunit 20. Data, mean ± s.e.m.; *P < 0.05; **P < 0.01; ***P < 0.001; NS, not significant. For statistics, see Supplementary Tables 1–3.

Source data

Extended Data Figure 4 sAC mediates the mitochondrial effects of mtCB1-receptor activation.

a, Quantification of data presented in Fig. 2e, indicating higher interaction between Gα proteins and sAC after mtCB1 activation (n = 3). b, The transmembrane adenylyl cyclase activator forskolin (10 μM) does not alter basal levels and does not reverse the effect of THC (800 nM) on cAMP content of CB1+/+ brain mitochondria (n = 4). c, Forskolin (10 μM) has no effect on basal cAMP levels of CB1−/− brain mitochondria (n = 3–4). d, The sAC activator HCO3 (5 mM) reverses the effect of THC (800 nM) on cAMP content in purified brain mitochondria from CB1+/+ mice (n = 4). e, HCO3 (5 mM) and THC (800 nM) have no effect on cAMP levels (n = 3–4) of CB1−/− brain mitochondria. f, Representative trypsin sensitivity assay in mitochondrial fractions of 3T3 cells (3 independent experiments) showing intra-mitochondrial localization of CB1 receptors, Gα proteins, AKAP121, PKA and sAC in 3T3 cells. SDHA, succinate dehydrogenase A (subunit of complex II); TOM20, translocase of outer membrane subunit 20. g, Representative trypsin-sensitivity assay (3 independent experiments) showing that sAC–HA and mtsAC–HA are localized inside mitochondria in transfected 3T3 cells. h, Representative immunoblot (3 independent experiments) showing the expression of sAC–HA in transfected and non-transfected primary hippocampal cultures. i, Expression of sAC–HA in primary hippocampal cultures blocks the effect of THC (1 μM) on basal cellular respiration (n = 8–10). Data, mean ± s.e.m.; P < 0.05; **P < 0.01; ***P < 0.001; NS, not significant. For statistics, see Supplementary Tables 1–3.

Source data

Extended Data Figure 5 The sAC inhibitor KH7 prevents the effects of mtCB1 activation.

a, The effect of KH7 (5 μM) on mitochondrial respiration in purified brain mitochondria obtained from CB1+/+ mice (n = 5–6). b, KH7 (5 μM) blocks the effect of THC (800 nM) on PKA activity in purified brain mitochondria obtained from CB1+/+ mice (n = 5–11). c, KH7 (5 μM) blocks the effect of THC (800 nM) and WIN (100 nM) on complex-I activity in purified brain mitochondria obtained from CB1+/+ mice (n = 3–6). d, Representative confocal images (green, CB1-GFP; red, pDsred2–mito) of analysed axonal tracts and kymographs of the results shown in Fig. 3b, representing the effect of KH7 (5 μM) on the reduction in mitochondrial mobility in primary hippocampal neurons induced by HU210 (1 μM). Scale bar, 10 μM. e, mtsAC–HA blocks the decrease in mitochondrial mobility in CB1-expressing neurons induced by HU210 (1 μM) (n = 7–18). f, The effect of KH7 (10 μM) on the decrease in fEPSP at CA3–CA1 synapses from C57BL/6N mice induced by WIN (5 μM). Left, plots of normalized fEPSP slopes in vitro with representative fEPSP traces before (1) and after (2) WIN incubation. Vehicle (n = 6) or KH7 (n = 5) were pre-incubated 10 min before cannabinoid application. Right, histogram summarizing the average changes in the percentage of the fEPSP slope before (100% baseline, dotted line) and after WIN treatment in the presence or absence of KH7. g, KH7 (10 μM) alone does not affect the fEPSP slope at CA3–CA1 synapses from C57BL/6N mice. Plots of normalized fEPSP slopes in vitro before and after KH7 application (n = 5). h, Total exploration time in NOR experiments as described in Fig. 3d. Data, mean ± s.e.m.; *P < 0.05; ***P < 0.001; NS, not significant. For statistics, see Supplementary Tables 1–3.

Source data

Extended Data Figure 6 Mitochondrial PKA activity mediates the effects of mtCB1 receptors.

a, The PKA inhibitor H89 (800 nM) blocks the effect of THC (800 nM) and WIN (100 nM) on complex-I activity in CB1+/+ purified brain mitochondria (n = 3–6). b, H89 blocks the effect of THC on PKA activity in CB1+/+ purified brain mitochondria (n = 6). c, H89 and THC have no effect on PKA activity in CB1−/− brain mitochondria (n = 3–5). d, Quantification of data presented in Fig. 4c (n = 3), indicating a lower mitochondrial PKA content after silencing of AKAP121. e, f, Silencing of AKAP121 lowers mitochondrial PKA activity. e, Representative immunoblots of proteins phosphorylated by PKA (PKA-dependent phosphorylation) in 3T3 cells transfected with control vector or shAKAKP121. SHDA, succinate dehydrogenase A (a subunit of complex II). f, Quantification of data presented in e (n = 12). gi, Silencing of AKAP121 blocks the effect of THC on cellular respiration. g, Silencing of AKAP121 blocks the effect of THC (800 nM) on basal cellular respiration in 3T3 cells (n = 4). h, Representative immunoblotting of AKAP121 expression in extracts of hippocampal primary cultures in the presence or absence of shAKAP121. i, shAKAP121 inhibits the effect of THC on basal cellular respiration of primary hippocampal cultures (n = 5–10). Data, mean ± s.e.m.; *P < 0.05; **P < 0.01; ***P < 0.001; NS, not significant. For statistics, see Supplementary Tables 1–3.

Source data

Extended Data Figure 7 Activation of mtCB1 decreases PKA-dependent phosphorylation of the complex-I subunit NDUFS2, eventually mediating the effects on respiration.

a, THC specifically decreases complex-I- but not complex-II- or complex-IV-dependent respiration in purified brain mitochondria in a CB1-dependent manner (n = 3–4). b, Representative two-dimension (BN-PAGE/SDS–PAGE) immunoblotting (4 independent experiments) showing PKA-dependent phosphorylation of protein extracts of brain mitochondria from CB1+/+ and CB1−/− mice treated with vehicle or THC (800 nM). c, Representative immunofluorescence of HeLa cells transfected with empty vector (mock), the mitochondrial-targeted constitutively active PKA (MLS–PKA-CA) or the phosphomimetic mutant of NDUFS2 (NDUFS2-PM). Green, myc tag staining; red, mitochondrial staining (TOM20). d, Baseline cellular respiration of HEK293 cells transfected with plasmids expressing CB1, CB1 + MLS–PKA-CA or CB1 + NDUFS2-PM (n = 4–6). e, The effect of vehicle or WIN on cellular respiration in HEK293 cells as in d (n = 4–6). f, Representative images of hippocampi injected with AAV–GFP, AAV–MLS–PKA-CA or AAV–NDUFS2-PM. Green, GFP fluorescence (left panel) or myc tag staining (central and right panel). Arrows, subunit of complex I, NDUFS2. ***P < 0.001 between genotypes (a) or between vehicle and WIN (d); ##P < 0.01; ###P < 0.001 as compared to control. For statistics details, see Supplementary Tables 1–3.

Source data

Extended Data Table 1 Theoretical in silico predictions of mitochondrial and plasma membrane localization of CB1 and DN22-CB1 receptor proteins, based on the programs MitoProt and PSORT

Supplementary information

Supplementary Information

This file contains Supplementary Figure 1, which shows uncropped Western Immunoblotting images used for the relative panels of main figures. (PDF 1222 kb)

Cannabinoids reduce mitochondrial mobility in hippocampal neurons via CB1 receptors

Effect of the CB1 receptor agonist HU210 on mitochondrial mobility in CB1-/- hippocampal neurons re-expressing CB1. Representative time-lapse live imaging of axonal mitochondria before and after treatment of HU210 (1µM) for 15 min in a primary hippocampal neuron from CB1-/- mice, transfected with CB1-GFP. See Fig. 1e. (MOV 643 kb)

Cannabinoid-induced reduction of mitochondrial mobility depends on mtCB1 receptors

Re-expression of DN22-CB1 fails to rescue the HU210-dependent decrease of mitochondrial mobility in CB1-/- hippocampal neurons. Representative time-lapse live imaging of axonal mitochondria before and after treatment of HU210 (1µM) for 15 min in a primary hippocampal neuron from CB1-/- mice, transfected with DN22-CB1-GFP. See Fig. 1e. (MOV 700 kb)

Cannabinoid-induced reduction of mitochondrial mobility depends on sAC signaling.

Representative time-lapse live imaging of axonal mitochondria before and after treatment of HU210 (1µM) and vehicle for 15 min in a primary hippocampal neuron from CB1-/- mice, transfected with CB1-GFP. See Fig. 3b. (MOV 731 kb)

Cannabinoid-induced reduction of mitochondrial mobility depends on sAC signaling.

The sAC inhibitor KH7 blocks the reduction of mitochondrial mobility induced by HU210. Representative time-lapse live imaging of axonal mitochondria before and after treatment of HU210 (1µM) for 15min in the presence of KH7 (5µM) in a primary hippocampal neuron from CB1-/- mice, transfected with CB1-GFP. See Fig. 3b. (MOV 771 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hebert-Chatelain, E., Desprez, T., Serrat, R. et al. A cannabinoid link between mitochondria and memory. Nature 539, 555–559 (2016). https://doi.org/10.1038/nature20127

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature20127

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