Skip to main content

Advertisement

Log in

Cannabinoids modulate proliferation, differentiation, and migration signaling pathways in oligodendrocytes

  • Original Paper
  • Published:
European Archives of Psychiatry and Clinical Neuroscience Aims and scope Submit manuscript

Abstract

Cannabinoid signaling, mainly via CB1 and CB2 receptors, plays an essential role in oligodendrocyte health and functions. However, the specific molecular signals associated with the activation or blockade of CB1 and CB2 receptors in this glial cell have yet to be elucidated. Mass spectrometry-based shotgun proteomics and in silico biology tools were used to determine which signaling pathways and molecular mechanisms are triggered in a human oligodendrocytic cell line (MO3.13) by several pharmacological stimuli: the phytocannabinoid cannabidiol (CBD); CB1 and CB2 agonists ACEA, HU308, and WIN55, 212–2; CB1 and CB2 antagonists AM251 and AM630; and endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG). The modulation of cannabinoid signaling in MO3.13 was found to affect pathways linked to cell proliferation, migration, and differentiation of oligodendrocyte progenitor cells. Additionally, we found that carbohydrate and lipid metabolism, as well as mitochondrial function, were modulated by these compounds. Comparing the proteome changes and upstream regulators among treatments, the highest overlap was between the CB1 and CB2 antagonists, followed by overlaps between AEA and 2-AG. Our study opens new windows of opportunities, suggesting that cannabinoid signaling in oligodendrocytes might be relevant in the context of demyelinating and neurodegenerative diseases. Proteomics data are available at ProteomeXchange (PXD031923).

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Data availability

The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE (PubMed ID: 34,723,319) partner repository with the dataset identifier PXD031923 [90].

Abbreviations

2-AG:

2-Arachidonoylglycerol

ACN:

Acetonitrile

AEA:

Anandamide

BSA:

Bovine serum albumin

CB1:

Cannabinoid receptor, type 1

CB2:

Cannabinoid receptor, type 2

CBD:

Cannabidiol

CNS:

Central nervous system

D2:

Dopamine receptor, type 2

DAGLA:

Diacylglycerol lipase alpha

DAGLB:

Diacylglycerol lipase beta

DEPs:

Differentially expressed proteins

DMEM:

Dulbecco’s modified Eagle medium

ECBs:

Endocannabinoids

EIF2:

Eukaryotic initiation factor 2

ESI:

Electrospray ionization

FAAH:

Fatty acid amide hydrolase

FDR:

False discovery rate

HBSS:

Hank’s balanced salt solution

ICC:

Immunocytochemistry

IPA:

Ingenuity pathway analysis

LC:

Liquid chromatography

MAGL:

Monoacylglycerol lipase

MAPK:

Mitogen-activated protein kinase

MBP:

Myelin binding protein

MS:

Mass spectrometry

MS-TOF:

Time-of-flight mass spectrometry

NAPE-PLD:

N-acyl phosphatidylethanolamine phospholipase D

OPC:

Oligodendrocyte precursor cell

PBS:

Phosphate-buffered saline

RT:

Room temperature

TBS:

Tris-buffered saline

THC:

Tetrahydrocannabinol

TRPV1:

Transient receptor potential cation channel, subfamily V, member 1

UPLC:

Ultra-performance liquid chromatography

VWM:

Vanishing white matter

References

  1. Howlett AC (1985) Cannabinoid inhibition of adenylate cyclase. Biochemistry of the response in neuroblastoma cell membranes. Mol Pharmacol 27:429–436

    CAS  PubMed  Google Scholar 

  2. Matsuda LA, Lolait SJ, Brownstein MJ et al (1990) Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 346:561–564

    CAS  PubMed  Google Scholar 

  3. Munro S, Thomas KL, Abu-Shaar M (1993) Molecular characterization of a peripheral receptor for cannabinoids. Nature 365:61–65

    CAS  PubMed  Google Scholar 

  4. Devane WA, Dysarz FA 3rd, Johnson MR et al (1988) Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol 34:605–613

    CAS  PubMed  Google Scholar 

  5. Di Marzo V, Piscitelli F (2015) The endocannabinoid system and its modulation by phytocannabinoids. Neurotherapeutics 12:692–698

    PubMed  PubMed Central  Google Scholar 

  6. Howlett AC (1998) The CB1 cannabinoid receptor in the brain. Neurobiol Dis 5:405–416

    CAS  PubMed  Google Scholar 

  7. Howlett AC, Abood ME (2017) CB and CB Receptor Pharmacology. Adv Pharmacol 80:169–206

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Van Sickle MD, Duncan M, Kingsley PJ et al (2005) Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science 310:329–332

    PubMed  Google Scholar 

  9. Lisboa SF, Gomes FV, Guimaraes FS, Campos AC (2016) Microglial Cells as a Link between Cannabinoids and the Immune Hypothesis of Psychiatric Disorders. Front Neurol 7:5

    PubMed  PubMed Central  Google Scholar 

  10. Rodrigues LCM, Gobira PH, de Oliveira AC et al (2014) Neuroinflammation as a possible link between cannabinoids and addiction. Acta Neuropsychiatr 26:334–346

    PubMed  Google Scholar 

  11. Huerga-Gómez A, Aguado T, Sánchez-de la Torre A et al (2021) Δ -Tetrahydrocannabinol promotes oligodendrocyte development and CNS myelination in vivo. Glia 69:532–545

    PubMed  Google Scholar 

  12. de Almeida V, Martins-de-Souza D (2018) Cannabinoids and glial cells: possible mechanism to understand schizophrenia. Eur Arch Psychiatry Clin Neurosci 268:727–737

    PubMed  Google Scholar 

  13. Katona I, Sperlágh B, Sı́k A, et al (1999) Presynaptically located CB1 cannabinoid receptors regulate GABA release from axon terminals of specific hippocampal interneurons. J Neurosci 19:4544–4558

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Stella N (2010) Cannabinoid and cannabinoid-like receptors in microglia, astrocytes, and astrocytomas. Glia 58:1017–1030

    PubMed  PubMed Central  Google Scholar 

  15. Fünfschilling U, Supplie LM, Mahad D et al (2012) Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 485:517–521

    PubMed  PubMed Central  Google Scholar 

  16. Lee Y, Morrison BM, Li Y et al (2012) Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature 487:443–448

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Molina-Holgado E, Vela JM, Arévalo-Martín A et al (2002) Cannabinoids promote oligodendrocyte progenitor survival: involvement of cannabinoid receptors and phosphatidylinositol-3 kinase/Akt signaling. J Neurosci 22:9742–9753

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Solbrig MV, Fan Y, Hermanowicz N et al (2010) A synthetic cannabinoid agonist promotes oligodendrogliogenesis during viral encephalitis in rats. Exp Neurol 226:231–241

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Gomez O, Sanchez-Rodriguez A, Le M et al (2011) Cannabinoid receptor agonists modulate oligodendrocyte differentiation by activating PI3K/Akt and the mammalian target of rapamycin (mTOR) pathways. Br J Pharmacol 163:1520–1532

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Tomas-Roig J, Wirths O, Salinas-Riester G, Havemann-Reinecke U (2016) The cannabinoid CB1/CB2 agonist WIN55212.2 promotes oligodendrocyte differentiation in vitro and neuroprotection during the cuprizone-induced central nervous system demyelination. CNS Neurosci Ther 22:387–395

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Moreno-Luna R, Esteban PF, Paniagua-Torija B et al (2021) Heterogeneity of the endocannabinoid system between cerebral cortex and spinal cord oligodendrocytes. Mol Neurobiol 58:689–702

    CAS  PubMed  Google Scholar 

  22. Ilyasov AA, Milligan CE, Pharr EP, Howlett AC (2018) The Endocannabinoid system and oligodendrocytes in health and disease. Front Neurosci 12:733

    PubMed  PubMed Central  Google Scholar 

  23. Mecha M, Carrillo-Salinas FJ, Feliú A et al (2020) Perspectives on cannabis-based therapy of multiple sclerosis: a mini-review. Front Cell Neurosci 14:34

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Jinsmaa Y, Isonaka R, Sharabi Y, Goldstein DS (2020) 3,4-Dihydroxyphenylacetaldehyde is more efficient than dopamine in oligomerizing and quinonizing -synuclein. J Pharmacol Exp Ther 372:157–165

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Iwata K, Café-Mendes CC, Schmitt A et al (2013) The human oligodendrocyte proteome. Proteomics 13:3548–3553

    CAS  PubMed  Google Scholar 

  26. Buntinx M, Vanderlocht J, Hellings N et al (2003) Characterization of three human oligodendroglial cell lines as a model to study oligodendrocyte injury: morphology and oligodendrocyte-specific gene expression. J Neurocytol 32:25–38

    CAS  PubMed  Google Scholar 

  27. Brandão-Teles C, de Almeida V, Cassoli JS, Martins-de-Souza D (2019) Biochemical pathways triggered by antipsychotics in human [corrected] oligodendrocytes: potential of discovering new treatment targets. Front Pharmacol 10:186

    PubMed  PubMed Central  Google Scholar 

  28. Seabra G, de Almeida V, Reis-de-Oliveira G et al (2020) Ubiquitin-proteasome system, lipid metabolism and DNA damage repair are triggered by antipsychotic medication in human oligodendrocytes: implications in schizophrenia. Sci Rep 10:12655

    PubMed  PubMed Central  Google Scholar 

  29. Cassoli JS, Brandão-Teles C, Santana AG et al (2018) Ion mobility-enhanced data-independent acquisitions enable a deep proteomic landscape of oligodendrocytes. Proteomics. https://doi.org/10.1002/pmic.2018700015

    Article  PubMed  Google Scholar 

  30. Silva JC, Denny R, Dorschel CA et al (2005) Quantitative proteomic analysis by accurate mass retention time pairs. Anal Chem 77:2187–2200

    CAS  PubMed  Google Scholar 

  31. Snel B, Lehmann G, Bork P, Huynen MA (2000) STRING: a web-server to retrieve and display the repeatedly occurring neighbourhood of a gene. Nucleic Acids Res 28:3442–3444

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fabregat A, Sidiropoulos K, Garapati P et al (2016) The reactome pathway knowledgebase. Nucleic Acids Res 44:D481–D487

    CAS  PubMed  Google Scholar 

  33. Uhlen M, Ponten F, Lindskog C (2015) Charting the human proteome: Understanding disease using a tissue-based atlas. Science 347:1274–1274

    Google Scholar 

  34. Yu G, Wang L-G, Han Y, He Q-Y (2012) clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16:284–287

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Computing RFS (2013) R: A language and environment for statistical computing. R Core Team, Vienna

    Google Scholar 

  36. Tatomir A, Rao G, Boodhoo D et al (2020) Histone deacetylase SIRT1 mediates C5b-9-induced cell cycle in oligodendrocytes. Front Immunol 11:619

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Prozorovski T, Ingwersen J, Lukas D et al (2019) Regulation of sirtuin expression in autoimmune neuroinflammation: Induction of SIRT1 in oligodendrocyte progenitor cells. Neurosci Lett 704:116–125

    CAS  PubMed  Google Scholar 

  38. Hiratsuka D, Kurganov E, Furube E et al (2019) VEGF- and PDGF-dependent proliferation of oligodendrocyte progenitor cells in the medulla oblongata after LPC-induced focal demyelination. J Neuroimmunol 332:176–186

    CAS  PubMed  Google Scholar 

  39. Mindos T, Dun X-P, North K et al (2017) Merlin controls the repair capacity of Schwann cells after injury by regulating Hippo/YAP activity. J Cell Biol 216:495–510

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Azevedo MM, Domingues HS, Cordelières FP et al (2018) Jmy regulates oligodendrocyte differentiation via modulation of actin cytoskeleton dynamics. Glia 66:1826–1844

    PubMed  Google Scholar 

  41. Thomason EJ, Escalante M, Osterhout DJ, Fuss B (2019) The oligodendrocyte growth cone and its actin cytoskeleton: A fundamental element for progenitor cell migration and CNS myelination. Glia. https://doi.org/10.1002/glia.23735

    Article  PubMed  PubMed Central  Google Scholar 

  42. Nicaise AM, Johnson KM, Willis CM et al (2019) TIMP-1 promotes oligodendrocyte differentiation through receptor-mediated signaling. Mol Neurobiol 56:3380–3392

    CAS  PubMed  Google Scholar 

  43. Harlow DE, Saul KE, Komuro H, Macklin WB (2015) Myelin proteolipid protein complexes with αv integrin and AMPA receptors In vivo and regulates ampa-dependent oligodendrocyte progenitor cell migration through the modulation of cell-surface GluR2 expression. J Neurosci 35:12018–12032

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Linneberg C, Harboe M, Laursen LS (2015) Axo-glia interaction preceding CNS myelination is regulated by bidirectional eph-ephrin signaling. ASN Neuro. https://doi.org/10.1177/1759091415602859

    Article  PubMed  PubMed Central  Google Scholar 

  45. Câmara J, Wang Z, Nunes-Fonseca C et al (2009) Integrin-mediated axoglial interactions initiate myelination in the central nervous system. J Cell Biol 185:699–712

    PubMed  PubMed Central  Google Scholar 

  46. Jagielska A, Lowe AL, Makhija E et al (2017) Mechanical strain promotes oligodendrocyte differentiation by global changes of gene expression. Front Cell Neurosci 11:93

    PubMed  PubMed Central  Google Scholar 

  47. Harboe M, Torvund-Jensen J, Kjaer-Sorensen K, Laursen LS (2018) Ephrin-A1-EphA4 signaling negatively regulates myelination in the central nervous system. Glia 66:934–950

    PubMed  Google Scholar 

  48. Liang X, Draghi NA, Resh MD (2004) Signaling from integrins to Fyn to Rho family GTPases regulates morphologic differentiation of oligodendrocytes. J Neurosci 24:7140–7149

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Ackerman SD, Garcia C, Piao X et al (2015) The adhesion GPCR Gpr56 regulates oligodendrocyte development via interactions with Gα12/13 and RhoA. Nat Commun 6:6122

    CAS  PubMed  Google Scholar 

  50. Thurnherr T, Benninger Y, Wu X et al (2006) Cdc42 and Rac1 signaling are both required for and act synergistically in the correct formation of myelin sheaths in the CNS. J Neurosci 26:10110–10119

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Seixas AI, Azevedo MM, Paes de Faria J et al (2019) Evolvability of the actin cytoskeleton in oligodendrocytes during central nervous system development and aging. Cell Mol Life Sci 76:1–11

    CAS  PubMed  Google Scholar 

  52. Maki T, Choi YK, Miyamoto N et al (2018) A-kinase anchor protein 12 is required for oligodendrocyte differentiation in adult white matter. Stem Cells 36:751–760

    CAS  PubMed  Google Scholar 

  53. Ghidinelli M, Poitelon Y, Shin YK et al (2017) Laminin 211 inhibits protein kinase A in Schwann cells to modulate neuregulin 1 type III-driven myelination. PLoS Biol 15:e2001408

    PubMed  PubMed Central  Google Scholar 

  54. Yang Y, Wang H, Zhang J et al (2013) Cyclin dependent kinase 5 is required for the normal development of oligodendrocytes and myelin formation. Dev Biol 378:94–106

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Luo F, Burke K, Kantor C et al (2014) Cyclin-dependent kinase 5 mediates adult OPC maturation and myelin repair through modulation of Akt and GsK-3β signaling. J Neurosci 34:10415–10429

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Miyamoto Y, Yamauchi J, Chan JR et al (2007) Cdk5 regulates differentiation of oligodendrocyte precursor cells through the direct phosphorylation of paxillin. J Cell Sci 120:4355–4366

    CAS  PubMed  Google Scholar 

  57. Colognato H, Tzvetanova ID (2011) Glia unglued: how signals from the extracellular matrix regulate the development of myelinating glia. Dev Neurobiol 71:924–955

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Hussain R, Macklin WB (2017) Integrin-linked kinase (ILK) deletion disrupts oligodendrocyte development by altering cell cycle. J Neurosci 37:397–412

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Pronk JC, van Kollenburg B, Scheper GC, van der Knaap MS (2006) Vanishing white matter disease: a review with focus on its genetics. Ment Retard Dev Disabil Res Rev 12:123–128

    PubMed  Google Scholar 

  60. Carter CJ (2007) eIF2B and oligodendrocyte survival: where nature and nurture meet in bipolar disorder and schizophrenia? Schizophr Bull 33:1343–1353

    PubMed  PubMed Central  Google Scholar 

  61. Arévalo-Martín A, García-Ovejero D, Rubio-Araiz A et al (2007) Cannabinoids modulate Olig2 and polysialylated neural cell adhesion molecule expression in the subventricular zone of post-natal rats through cannabinoid receptor 1 and cannabinoid receptor 2. Eur J Neurosci 26:1548–1559

    PubMed  Google Scholar 

  62. Kuhn S, Gritti L, Crooks D, Dombrowski Y (2019) Oligodendrocytes in development, myelin generation and beyond. Cells. https://doi.org/10.3390/cells8111424

    Article  PubMed  PubMed Central  Google Scholar 

  63. Gaesser JM, Fyffe-Maricich SL (2016) Intracellular signaling pathway regulation of myelination and remyelination in the CNS. Exp Neurol 283:501–511

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Bernal-Chico A, Canedo M, Manterola A et al (2015) Blockade of monoacylglycerol lipase inhibits oligodendrocyte excitotoxicity and prevents demyelination in vivo. Glia 63:163–176

    PubMed  Google Scholar 

  65. Sanchez-Rodriguez MA, Gomez O, Esteban PF et al (2018) The endocannabinoid 2-arachidonoylglycerol regulates oligodendrocyte progenitor cell migration. Biochem Pharmacol 157:180–188

    CAS  PubMed  Google Scholar 

  66. Nave K-A, Werner HB (2014) Myelination of the nervous system: mechanisms and functions. Annu Rev Cell Dev Biol 30:503–533

    CAS  PubMed  Google Scholar 

  67. Hussain G, Wang J, Rasul A et al (2019) Role of cholesterol and sphingolipids in brain development and neurological diseases. Lipids Health Dis 18:26

    PubMed  PubMed Central  Google Scholar 

  68. Voskuhl RR, Itoh N, Tassoni A et al (2019) Gene expression in oligodendrocytes during remyelination reveals cholesterol homeostasis as a therapeutic target in multiple sclerosis. Proc Natl Acad Sci U S A 116:10130–10139

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Lin J-P, Mironova YA, Shrager P, Giger RJ (2017) LRP1 regulates peroxisome biogenesis and cholesterol homeostasis in oligodendrocytes and is required for proper CNS myelin development and repair. Elife. https://doi.org/10.7554/eLife.30498

    Article  PubMed  PubMed Central  Google Scholar 

  70. Oddi S, Caporali P, Dragotto J et al (2019) The endocannabinoid system is affected by cholesterol dyshomeostasis: Insights from a murine model of Niemann Pick type C disease. Neurobiol Dis 130:104531

    CAS  PubMed  Google Scholar 

  71. Suo N, Guo Y-E, He B et al (2019) Inhibition of MAPK/ERK pathway promotes oligodendrocytes generation and recovery of demyelinating diseases. Glia 67:1320–1332

    PubMed  PubMed Central  Google Scholar 

  72. Wong YL, LeBon L, Basso AM et al (2019) eIF2B activator prevents neurological defects caused by a chronic integrated stress response. Elife. https://doi.org/10.7554/eLife.42940

    Article  PubMed  PubMed Central  Google Scholar 

  73. Melas PA, Qvist JS, Deidda M et al (2018) Cannabinoid modulation of eukaryotic initiation factors (eIF2α and eIF2B1) and behavioral cross-sensitization to cocaine in adolescent rats. Cell Rep 22:2909–2923

    CAS  PubMed  Google Scholar 

  74. Terumitsu-Tsujita M, Kitaura H, Miura I et al (2019) Glial pathology in a novel spontaneous mutant mouse of the Eif2b5 gene: a vanishing white matter disease model. J Neurochem. https://doi.org/10.1111/jnc.14887

    Article  PubMed  Google Scholar 

  75. Vrechi TA, Crunfli F, Costa AP, Torrão AS (2018) Cannabinoid receptor Type 1 Agonist ACEA protects neurons from death and attenuates endoplasmic reticulum stress-related apoptotic pathway signaling. Neurotox Res 33:846–855

    CAS  PubMed  Google Scholar 

  76. Harris JJ, Attwell D (2012) The energetics of CNS white matter. J Neurosci 32:356–371

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Rinholm JE, Vervaeke K, Tadross MR et al (2016) Movement and structure of mitochondria in oligodendrocytes and their myelin sheaths. Glia 64:810–825

    PubMed  Google Scholar 

  78. Rao VTS, Khan D, Cui Q-L et al (2017) Distinct age and differentiation-state dependent metabolic profiles of oligodendrocytes under optimal and stress conditions. PLoS ONE 12:e0182372

    PubMed  PubMed Central  Google Scholar 

  79. Scarante FF, Ribeiro MA, Almeida-Santos AF et al (2020) Glial cells and their contribution to the mechanisms of action of cannabidiol in neuropsychiatric disorders. Front Pharmacol 11:618065

    CAS  PubMed  Google Scholar 

  80. Barley K, Dracheva S, Byne W (2009) Subcortical oligodendrocyte- and astrocyte-associated gene expression in subjects with schizophrenia, major depression and bipolar disorder. Schizophr Res 112:54–64

    PubMed  Google Scholar 

  81. Morabito S, Miyoshi E, Michael N et al (2021) Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease. Nat Genet 53:1143–1155

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Jäkel S, Agirre E, Mendanha Falcão A et al (2019) Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566:543–547

    PubMed  PubMed Central  Google Scholar 

  83. Osorio-Querejeta I, Sáenz-Cuesta M, Muñoz-Culla M, Otaegui D (2017) Models for studying myelination, demyelination and remyelination. Neuromolecular Med 19:181–192

    CAS  PubMed  Google Scholar 

  84. Campos AC, Ortega Z, Palazuelos J et al (2013) The anxiolytic effect of cannabidiol on chronically stressed mice depends on hippocampal neurogenesis: involvement of the endocannabinoid system. Int J Neuropsychopharmacol 16:1407–1419

    CAS  PubMed  Google Scholar 

  85. Levin R, Peres FF, Almeida V et al (2014) Effects of cannabinoid drugs on the deficit of prepulse inhibition of startle in an animal model of schizophrenia: the SHR strain. Front Pharmacol 5:10

    PubMed  PubMed Central  Google Scholar 

  86. Almeida V, Peres FF, Levin R et al (2014) Effects of cannabinoid and vanilloid drugs on positive and negative-like symptoms on an animal model of schizophrenia: The SHR strain. Schizophr Res 153:150–159

    PubMed  Google Scholar 

  87. Almeida V, Levin R, Peres FF et al (2013) Cannabidiol exhibits anxiolytic but not antipsychotic property evaluated in the social interaction test. Prog Neuropsychopharmacol Biol Psychiatry 41:30–35

    CAS  PubMed  Google Scholar 

  88. Gomez O, Sanchez-Rodriguez MA, Ortega-Gutierrez S et al (2015) A basal tone of 2-arachidonoylglycerol contributes to early oligodendrocyte progenitor proliferation by activating phosphatidylinositol 3-kinase (PI3K)/AKT and the mammalian target of rapamycin (MTOR) pathways. J Neuroimmune Pharmacol 10:309–317

    PubMed  Google Scholar 

  89. Almeida V, Levin R, Peres FF et al (2019) Role of the endocannabinoid and endovanilloid systems in an animal model of schizophrenia-related emotional processing/cognitive deficit. Neuropharmacology 155:44–53

    CAS  PubMed  Google Scholar 

  90. Perez-Riverol Y, Bai J, Bandla C et al (2022) The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res 50:D543–D552

    CAS  PubMed  Google Scholar 

Download references

Funding

The authors thank FAPESP (São Paulo Research Foundation—grants 2017/18242–1, 2017/25588–1, 2018/25818–0, 2018/03673–0, 2019/00098-7), and CAPES (Coordination for the Improvement of Higher Education Personnel, grants 1656470 and 88887.495565/2020–00) for financial support.

Author information

Authors and Affiliations

Authors

Contributions

VA: Conceptualization, Methodology, Formal analysis, Investigation, Writing—Original Draft, Writing—Review & Editing and Funding acquisition. GS, GSZ, PR, and MF: Investigation. GRO: Formal analysis and Visualization. BJS: Writing—Review & Editing. ACC, AWZ, JEH, and JAC: Resources and Writing—Review & Editing. DMS: Conceptualization, Resources, Writing—Review & Editing, Supervision, Funding acquisition. All authors contributed to and approved the final version of the manuscript.

Corresponding authors

Correspondence to Valéria de Almeida or Daniel Martins-de-Souza.

Ethics declarations

Conflict of interest

JAC is a member of the International Advisory Board of the Australian Centre for Cannabinoid Clinical and Research Excellence (ACRE) – National Health and Medical Research Council (NHMRC). JAC and JEH have received travel support to attend scientific meetings and personal consultation fees from BSPG-Pharm. JAC, JEH, and AWZ are co-inventors of the patent “Fluorinated CBD compounds, compositions and uses thereof. Pub. No.: WO/2014/108899. International Application No.: PCT/IL2014/050023,” Def. US number Reg. 62193296; July 29, 2015; INPI on August 19, 2015 (BR1120150164927; Mechoulam R, Zuardi AW, Kapczinski F, Hallak JEC, Guimarães FS, Crippa JAS, Breuer A). Universidade de São Paulo (USP) has licensed this patent to Phytecs Pharm (USP Resolution No. 15.1.130002.1.1) and has an agreement with Prati-Donaduzzi to “develop a pharmaceutical product containing synthetic CBD and prove its safety and therapeutic efficacy in the treatment of epilepsy, schizophrenia, Parkinson’s disease, and anxiety disorders.” JAC, JEH, AWZ are co-inventors of the patent “Cannabinoid-containing oral pharmaceutical composition, a method for preparing and using the same,” INPI on September 16, 2016 (BR 112018005423–2).

Supplementary Information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Almeida, V., Seabra, G., Reis-de-Oliveira, G. et al. Cannabinoids modulate proliferation, differentiation, and migration signaling pathways in oligodendrocytes. Eur Arch Psychiatry Clin Neurosci 272, 1311–1323 (2022). https://doi.org/10.1007/s00406-022-01425-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00406-022-01425-5

Keywords

Navigation