Endocannabinoid System Changes throughout Life: Implications and Therapeutic Potential for Autism, ADHD, and Alzheimer’s Disease
Abstract
:1. Introduction
2. The Endocannabinoid System Changes throughout Life
3. Autism Spectrum Disorder
4. Attention-Deficit/Hyperactivity Disorder
5. Alzheimer’s Disease
6. Final Considerations
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Dos Santos Sampaio, M.F.; de Paiva, Y.B.; Sampaio, T.B.; Pereira, M.G.; Coimbra, N.C. Therapeutic applicability of cannabidiol and other phytocannabinoids in epilepsy, multiple sclerosis and Parkinson’s disease and in comorbidity with psychiatric disorders. Basic Clin. Pharmacol. Toxicol. 2024, 134, 574–601. [Google Scholar] [CrossRef] [PubMed]
- Pain, S. A potted history. Nature 2015, 525, S10–S11. [Google Scholar] [CrossRef]
- Khan, R.; Naveed, S.; Mian, N.; Fida, A.; Raafey, M.A.; Aedma, K.K. The therapeutic role of Cannabidiol in mental health: A systematic review. J. Cannabis. Res. 2020, 2, 2. [Google Scholar] [CrossRef] [PubMed]
- Citti, C.; Palazzoli, F.; Licata, M.; Vilella, A.; Leo, G.; Zoli, M.; Vandelli, M.A.; Forni, F.; Pacchetti, B.; Cannazza, G. Untargeted rat brain metabolomics after oral administration of a single high dose of cannabidiol. J. Pharm. Biomed. Anal. 2018, 161, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Muller-Vahl, K.R. Cannabinoids in the Treatment of Selected Mental Illnesses: Practical Approach and Overview of the Literature. Pharmacopsychiatry 2024, 57, 104–114. [Google Scholar] [CrossRef] [PubMed]
- Abate, G.; Uberti, D.; Tambaro, S. Potential and Limits of Cannabinoids in Alzheimer’s Disease Therapy. Biology 2021, 10, 542. [Google Scholar] [CrossRef]
- Zou, S.; Kumar, U. Cannabinoid Receptors and the Endocannabinoid System: Signaling and Function in the Central Nervous System. Int. J. Mol. Sci. 2018, 19, 833. [Google Scholar] [CrossRef]
- Fernandez-Moncada, I.; Rodrigues, R.S.; Fundazuri, U.B.; Bellocchio, L.; Marsicano, G. Type-1 cannabinoid receptors and their ever-expanding roles in brain energy processes. J. Neurochem. 2023, 168, 693–703. [Google Scholar] [CrossRef]
- Ye, L.; Cao, Z.; Wang, W.; Zhou, N. New Insights in Cannabinoid Receptor Structure and Signaling. Curr. Mol. Pharmacol. 2019, 12, 239–248. [Google Scholar] [CrossRef]
- Morales, P.; Lago-Fernandez, A.; Hurst, D.P.; Sotudeh, N.; Brailoiu, E.; Reggio, P.H.; Abood, M.E.; Jagerovic, N. Therapeutic Exploitation of GPR18: Beyond the Cannabinoids? J. Med. Chem. 2020, 63, 14216–14227. [Google Scholar] [CrossRef]
- Medina-Vera, D.; Zhao, H.; Bereczki, E.; Rosell-Valle, C.; Shimozawa, M.; Chen, G.; de Fonseca, F.R.; Nilsson, P.; Tambaro, S. The Expression of the Endocannabinoid Receptors CB2 and GPR55 Is Highly Increased during the Progression of Alzheimer’s Disease in App(NL-G-F) Knock-In Mice. Biology 2023, 12, 805. [Google Scholar] [CrossRef] [PubMed]
- Marichal-Cancino, B.A.; Fajardo-Valdez, A.; Ruiz-Contreras, A.E.; Mendez-Diaz, M.; Prospero-Garcia, O. Advances in the Physiology of GPR55 in the Central Nervous System. Curr. Neuropharmacol. 2017, 15, 771–778. [Google Scholar] [CrossRef] [PubMed]
- Armin, S.; Muenster, S.; Abood, M.; Benamar, K. GPR55 in the brain and chronic neuropathic pain. Behav. Brain Res. 2021, 406, 113248. [Google Scholar] [CrossRef] [PubMed]
- Hill, J.D.; Zuluaga-Ramirez, V.; Gajghate, S.; Winfield, M.; Sriram, U.; Rom, S.; Persidsky, Y. Activation of GPR55 induces neuroprotection of hippocampal neurogenesis and immune responses of neural stem cells following chronic, systemic inflammation. Brain Behav. Immun. 2019, 76, 165–181. [Google Scholar] [CrossRef] [PubMed]
- Pola, S.; Shah, S.R.; Pingali, H.; Zaware, P.; Thube, B.; Makadia, P.; Patel, H.; Bandyopadhyay, D.; Rath, A.; Giri, S.; et al. Discovery of a potent G-protein-coupled receptor 119 agonist for the treatment of type 2 diabetes. Bioorg. Med. Chem. 2021, 35, 116071. [Google Scholar] [CrossRef] [PubMed]
- Paredes, D.A.; Catlow, B.J.; Sanchez-Ramos, J. 402C14 The Endocannabinoid System in the Aging Brain and Neurodegenerative Diseases. In Integrative Neurology; McBurney, J.W., Ruhoy, I.S., Weil, A.T., Eds.; Oxford University Press: Oxford, UK, 2020. [Google Scholar] [CrossRef]
- Glass, M.; Dragunow, M.; Faull, R.L. Cannabinoid receptors in the human brain: A detailed anatomical and quantitative autoradiographic study in the fetal, neonatal and adult human brain. Neuroscience 1997, 77, 299–318. [Google Scholar] [CrossRef] [PubMed]
- Berrendero, F.; Garcia-Gil, L.; Hernandez, M.L.; Romero, J.; Cebeira, M.; de Miguel, R.; Ramos, J.A.; Fernandez-Ruiz, J.J. Localization of mRNA expression and activation of signal transduction mechanisms for cannabinoid receptor in rat brain during fetal development. Development 1998, 125, 3179–3188. [Google Scholar] [CrossRef] [PubMed]
- Berrendero, F.; Sepe, N.; Ramos, J.A.; Di Marzo, V.; Fernandez-Ruiz, J.J. Analysis of cannabinoid receptor binding and mRNA expression and endogenous cannabinoid contents in the developing rat brain during late gestation and early postnatal period. Synapse 1999, 33, 181–191. [Google Scholar] [CrossRef]
- Long, L.E.; Lind, J.; Webster, M.; Weickert, C.S. Developmental trajectory of the endocannabinoid system in human dorsolateral prefrontal cortex. BMC Neurosci. 2012, 13, 87. [Google Scholar] [CrossRef]
- Bisogno, T.; Howell, F.; Williams, G.; Minassi, A.; Cascio, M.G.; Ligresti, A.; Matias, I.; Schiano-Moriello, A.; Paul, P.; Williams, E.J.; et al. Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J. Cell Biol. 2003, 163, 463–468. [Google Scholar] [CrossRef]
- Ellgren, M.; Artmann, A.; Tkalych, O.; Gupta, A.; Hansen, H.S.; Hansen, S.H.; Devi, L.A.; Hurd, Y.L. Dynamic changes of the endogenous cannabinoid and opioid mesocorticolimbic systems during adolescence: THC effects. Eur. Neuropsychopharmacol. 2008, 18, 826–834. [Google Scholar] [CrossRef] [PubMed]
- Morishita, J.; Okamoto, Y.; Tsuboi, K.; Ueno, M.; Sakamoto, H.; Maekawa, N.; Ueda, N. Regional distribution and age-dependent expression of N-acylphosphatidylethanolamine-hydrolyzing phospholipase D in rat brain. J. Neurochem. 2005, 94, 753–762. [Google Scholar] [CrossRef] [PubMed]
- Lee, T.T.; Hill, M.N.; Hillard, C.J.; Gorzalka, B.B. Temporal changes in N-acylethanolamine content and metabolism throughout the peri-adolescent period. Synapse 2013, 67, 4–10. [Google Scholar] [CrossRef] [PubMed]
- Egertova, M.; Cravatt, B.F.; Elphick, M.R. Comparative analysis of fatty acid amide hydrolase and cb(1) cannabinoid receptor expression in the mouse brain: Evidence of a widespread role for fatty acid amide hydrolase in regulation of endocannabinoid signaling. Neuroscience 2003, 119, 481–496. [Google Scholar] [CrossRef] [PubMed]
- Heng, L.; Beverley, J.A.; Steiner, H.; Tseng, K.Y. Differential developmental trajectories for CB1 cannabinoid receptor expression in limbic/associative and sensorimotor cortical areas. Synapse 2011, 65, 278–286. [Google Scholar] [CrossRef] [PubMed]
- Yamagishi, S.; Iga, Y.; Nakamura, M.; Takizawa, C.; Fukumoto, D.; Kakiuchi, T.; Nishiyama, S.; Ohba, H.; Tsukada, H.; Sato, K.; et al. Upregulation of cannabinoid receptor type 2, but not TSPO, in senescence-accelerated neuroinflammation in mice: A positron emission tomography study. J. Neuroinflamm. 2019, 16, 208. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez de Fonseca, F.; Ramos, J.A.; Bonnin, A.; Fernandez-Ruiz, J.J. Presence of cannabinoid binding sites in the brain from early postnatal ages. Neuroreport 1993, 4, 135–138. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez de Fonseca, F.; Cebeira, M.; Ramos, J.A.; Martin, M.; Fernandez-Ruiz, J.J. Cannabinoid receptors in rat brain areas: Sexual differences, fluctuations during estrous cycle and changes after gonadectomy and sex steroid replacement. Life Sci. 1994, 54, 159–170. [Google Scholar] [CrossRef] [PubMed]
- El-Talatini, M.R.; Taylor, A.H.; Konje, J.C. The relationship between plasma levels of the endocannabinoid, anandamide, sex steroids, and gonadotrophins during the menstrual cycle. Fertil. Steril. 2010, 93, 1989–1996. [Google Scholar] [CrossRef]
- Meyer, H.C.; Lee, F.S.; Gee, D.G. The Role of the Endocannabinoid System and Genetic Variation in Adolescent Brain Development. Neuropsychopharmacology 2018, 43, 21–33. [Google Scholar] [CrossRef]
- Di Marzo, V.; Stella, N.; Zimmer, A. Endocannabinoid signalling and the deteriorating brain. Nat. Rev. Neurosci. 2015, 16, 30–42. [Google Scholar] [CrossRef] [PubMed]
- Marquez-Caraveo, M.E.; Rodriguez-Valentin, R.; Perez-Barron, V.; Vazquez-Salas, R.A.; Sanchez-Ferrer, J.C.; De Castro, F.; Allen-Leigh, B.; Lazcano-Ponce, E. Children and adolescents with neurodevelopmental disorders show cognitive heterogeneity and require a person-centered approach. Sci. Rep. 2021, 11, 18463. [Google Scholar] [CrossRef] [PubMed]
- Fusar-Poli, L.; Cavone, V.; Tinacci, S.; Concas, I.; Petralia, A.; Signorelli, M.S.; Diaz-Caneja, C.M.; Aguglia, E. Cannabinoids for People with ASD: A Systematic Review of Published and Ongoing Studies. Brain Sci. 2020, 10, 572. [Google Scholar] [CrossRef] [PubMed]
- Aran, A.; Eylon, M.; Harel, M.; Polianski, L.; Nemirovski, A.; Tepper, S.; Schnapp, A.; Cassuto, H.; Wattad, N.; Tam, J. Lower circulating endocannabinoid levels in children with autism spectrum disorder. Mol. Autism. 2019, 10, 2. [Google Scholar] [CrossRef] [PubMed]
- Karhson, D.S.; Krasinska, K.M.; Dallaire, J.A.; Libove, R.A.; Phillips, J.M.; Chien, A.S.; Garner, J.P.; Hardan, A.Y.; Parker, K.J. Plasma anandamide concentrations are lower in children with autism spectrum disorder. Mol. Autism. 2018, 9, 18. [Google Scholar] [CrossRef]
- Kerr, D.M.; Downey, L.; Conboy, M.; Finn, D.P.; Roche, M. Alterations in the endocannabinoid system in the rat valproic acid model of autism. Behav. Brain Res. 2013, 249, 124–132. [Google Scholar] [CrossRef]
- Wei, D.; Dinh, D.; Lee, D.; Li, D.; Anguren, A.; Moreno-Sanz, G.; Gall, C.M.; Piomelli, D. Enhancement of Anandamide-Mediated Endocannabinoid Signaling Corrects Autism-Related Social Impairment. Cannabis. Cannabinoid. Res. 2016, 1, 81–89. [Google Scholar] [CrossRef]
- Ferreira, L.F.; Pathapati, N.; Schultz, S.T.; Nunn, M.C.; Pierce, B.L.; Sanchez, Y.R.; Murrell, M.D.; Ginsburg, B.C.; Onaivi, E.S.; Gould, G.G. Acute cannabidiol treatment enhances social interaction in adult male mice. Adv. Drug Alcohol. Res. 2023, 3, 11163. [Google Scholar] [CrossRef]
- Manduca, A.; Buzzelli, V.; Rava, A.; Feo, A.; Carbone, E.; Schiavi, S.; Peruzzi, B.; D’Oria, V.; Pezzullo, M.; Pasquadibisceglie, A.; et al. Cannabidiol and positive effects on object recognition memory in an in vivo model of Fragile X Syndrome: Obligatory role of hippocampal GPR55 receptors. Pharmacol. Res. 2024, 203, 107176. [Google Scholar] [CrossRef]
- Staben, J.; Koch, M.; Reid, K.; Muckerheide, J.; Gilman, L.; McGuinness, F.; Kiesser, S.; Oswald, I.W.H.; Koby, K.A.; Martin, T.J.; et al. Cannabidiol and cannabis-inspired terpene blends have acute prosocial effects in the BTBR mouse model of autism spectrum disorder. Front. Neurosci. 2023, 17, 1185737. [Google Scholar] [CrossRef]
- Poleg, S.; Kourieh, E.; Ruban, A.; Shapira, G.; Shomron, N.; Barak, B.; Offen, D. Behavioral aspects and neurobiological properties underlying medical cannabis treatment in Shank3 mouse model of autism spectrum disorder. Transl. Psychiatry 2021, 11, 524. [Google Scholar] [CrossRef] [PubMed]
- Kurz, R.; Blaas, K. Use of dronabinol (delta-9-THC) in autism: A prospective single-case-study with an early infantile autistic child. Cannabinoids 2010, 5, 4–6. [Google Scholar]
- Kuester, G.; Vergara, K.; Ahumada, A.; Gazmuri, A.M. Oral cannabis extracts as a promising treatment for the core symptoms of autism spectrum disorder: Preliminary experience in Chilean patients. J. Neurol. Sci. 2017, 381, 932–933. [Google Scholar] [CrossRef]
- Barchel, D.; Stolar, O.; De-Haan, T.; Ziv-Baran, T.; Saban, N.; Fuchs, D.O.; Koren, G.; Berkovitch, M. Oral Cannabidiol Use in Children With Autism Spectrum Disorder to Treat Related Symptoms and Co-morbidities. Front. Pharmacol. 2018, 9, 1521. [Google Scholar] [CrossRef] [PubMed]
- Aran, A.; Cassuto, H.; Lubotzky, A.; Wattad, N.; Hazan, E. Brief Report: Cannabidiol-Rich Cannabis in Children with Autism Spectrum Disorder and Severe Behavioral Problems-A Retrospective Feasibility Study. J. Autism. Dev. Disord. 2019, 49, 1284–1288. [Google Scholar] [CrossRef] [PubMed]
- Bar-Lev Schleider, L.; Mechoulam, R.; Saban, N.; Meiri, G.; Novack, V. Real life Experience of Medical Cannabis Treatment in Autism: Analysis of Safety and Efficacy. Sci. Rep. 2019, 9, 200. [Google Scholar] [CrossRef] [PubMed]
- Fleury-Teixeira, P.; Caixeta, F.V.; Ramires da Silva, L.C.; Brasil-Neto, J.P.; Malcher-Lopes, R. Effects of CBD-Enriched Cannabis sativa Extract on Autism Spectrum Disorder Symptoms: An Observational Study of 18 Participants Undergoing Compassionate Use. Front. Neurol. 2019, 10, 1145. [Google Scholar] [CrossRef] [PubMed]
- Pretzsch, C.M.; Freyberg, J.; Voinescu, B.; Lythgoe, D.; Horder, J.; Mendez, M.A.; Wichers, R.; Ajram, L.; Ivin, G.; Heasman, M.; et al. Effects of cannabidiol on brain excitation and inhibition systems; a randomised placebo-controlled single dose trial during magnetic resonance spectroscopy in adults with and without autism spectrum disorder. Neuropsychopharmacology 2019, 44, 1398–1405. [Google Scholar] [CrossRef]
- Pretzsch, C.M.; Voinescu, B.; Lythgoe, D.; Horder, J.; Mendez, M.A.; Wichers, R.; Ajram, L.; Ivin, G.; Heasman, M.; Edden, R.A.E.; et al. Effects of cannabidivarin (CBDV) on brain excitation and inhibition systems in adults with and without Autism Spectrum Disorder (ASD): A single dose trial during magnetic resonance spectroscopy. Transl. Psychiatry 2019, 9, 313. [Google Scholar] [CrossRef]
- Pretzsch, C.M.; Voinescu, B.; Mendez, M.A.; Wichers, R.; Ajram, L.; Ivin, G.; Heasman, M.; Williams, S.; Murphy, D.G.; Daly, E.; et al. The effect of cannabidiol (CBD) on low-frequency activity and functional connectivity in the brain of adults with and without autism spectrum disorder (ASD). J. Psychopharmacol 2019, 33, 1141–1148. [Google Scholar] [CrossRef]
- Ponton, J.A.; Smyth, K.; Soumbasis, E.; Llanos, S.A.; Lewis, M.; Meerholz, W.A.; Tanguay, R.L. A pediatric patient with autism spectrum disorder and epilepsy using cannabinoid extracts as complementary therapy: A case report. J. Med. Case Rep. 2020, 14, 162. [Google Scholar] [CrossRef] [PubMed]
- Aran, A.; Harel, M.; Cassuto, H.; Polyansky, L.; Schnapp, A.; Wattad, N.; Shmueli, D.; Golan, D.; Castellanos, F.X. Cannabinoid treatment for autism: A proof-of-concept randomized trial. Mol. Autism. 2021, 12, 6. [Google Scholar] [CrossRef] [PubMed]
- Efron, D.; Freeman, J.L.; Cranswick, N.; Payne, J.M.; Mulraney, M.; Prakash, C.; Lee, K.J.; Taylor, K.; Williams, K. A pilot randomised placebo-controlled trial of cannabidiol to reduce severe behavioural problems in children and adolescents with intellectual disability. Br. J. Clin. Pharmacol. 2021, 87, 436–446. [Google Scholar] [CrossRef] [PubMed]
- Ma, L.; Platnick, S.; Platnick, H. Cannabidiol in Treatment of Autism Spectrum Disorder: A Case Study. Cureus 2022, 14, e28442. [Google Scholar] [CrossRef] [PubMed]
- Raz, N.; Heller, I.; Lombardi, T.; Marino, G.; Davidson, E.M.; Eyal, A.M. Terpene-Enriched CBD oil for treating autism-derived symptoms unresponsive to pure CBD: Case report. Front. Pharmacol. 2022, 13, 979403. [Google Scholar] [CrossRef] [PubMed]
- Hacohen, M.; Stolar, O.E.; Berkovitch, M.; Elkana, O.; Kohn, E.; Hazan, A.; Heyman, E.; Sobol, Y.; Waissengreen, D.; Gal, E.; et al. Children and adolescents with ASD treated with CBD-rich cannabis exhibit significant improvements particularly in social symptoms: An open label study. Transl. Psychiatry 2022, 12, 375. [Google Scholar] [CrossRef] [PubMed]
- Stolar, O.; Hazan, A.; Vissoker, R.E.; Kishk, I.A.; Barchel, D.; Lezinger, M.; Dagan, A.; Treves, N.; Meiri, D.; Berkovitch, M.; et al. Medical cannabis for the treatment of comorbid symptoms in children with autism spectrum disorder: An interim analysis of biochemical safety. Front. Pharmacol. 2022, 13, 977484. [Google Scholar] [CrossRef] [PubMed]
- Schnapp, A.; Harel, M.; Cayam-Rand, D.; Cassuto, H.; Polyansky, L.; Aran, A. A Placebo-Controlled Trial of Cannabinoid Treatment for Disruptive Behavior in Children and Adolescents with Autism Spectrum Disorder: Effects on Sleep Parameters as Measured by the CSHQ. Biomedicines 2022, 10, 1685. [Google Scholar] [CrossRef] [PubMed]
- Montagner, P.S.S.; Medeiros, W.; da Silva, L.C.R.; Borges, C.N.; Brasil-Neto, J.; de Deus Silva Barbosa, V.; Caixeta, F.V.; Malcher-Lopes, R. Individually tailored dosage regimen of full-spectrum Cannabis extracts for autistic core and comorbid symptoms: A real-life report of multi-symptomatic benefits. Front. Psychiatry 2023, 14, 1210155. [Google Scholar] [CrossRef]
- Siani-Rose, M.; McKee, R.; Cox, S.; Goldstein, B.; Abrams, D.; Taylor, M.; Kurek, I. The Potential of Salivary Lipid-Based Cannabis-Responsive Biomarkers to Evaluate Medical Cannabis Treatment in Children with Autism Spectrum Disorder. Cannabis. Cannabinoid. Res. 2023, 8, 642–656. [Google Scholar] [CrossRef]
- Silva, E.A.D.J.; Medeiros, W.M.B.; Santos, J.; Sousa, J.M.M.; Costa, F.B.D.; Pontes, K.M.; Borges, T.C.; Espinola, C.N.S.; Andrade, E.S.A.H.; Nunes, E.L.G.; et al. Evaluation of the efficacy and safety of cannabidiol-rich cannabis extract in children with autism spectrum disorder: Randomized, double-blind, and placebo-controlled clinical trial. Trends. Psychiatry Psychother. 2024, 46, e20210396. [Google Scholar] [CrossRef] [PubMed]
- Fayyad, J.; Sampson, N.A.; Hwang, I.; Adamowski, T.; Aguilar-Gaxiola, S.; Al-Hamzawi, A.; Andrade, L.H.; Borges, G.; de Girolamo, G.; Florescu, S.; et al. The descriptive epidemiology of DSM-IV Adult ADHD in the World Health Organization World Mental Health Surveys. Atten. Defic. Hyperact. Disord. 2017, 9, 47–65. [Google Scholar] [CrossRef] [PubMed]
- Posner, J.; Polanczyk, G.V.; Sonuga-Barke, E. Attention-deficit hyperactivity disorder. Lancet 2020, 395, 450–462. [Google Scholar] [CrossRef] [PubMed]
- Gujska, J.H.; Silczuk, A.; Madejek, R.; Szulc, A. Exploring the Link Between Attention-Deficit Hyperactivity Disorder and Cannabis Use Disorders: A Review. Med. Sci. Monit. 2023, 29, e939749. [Google Scholar] [CrossRef] [PubMed]
- Adriani, W.; Caprioli, A.; Granstrem, O.; Carli, M.; Laviola, G. The spontaneously hypertensive-rat as an animal model of ADHD: Evidence for impulsive and non-impulsive subpopulations. Neurosci. Biobehav. Rev. 2003, 27, 639–651. [Google Scholar] [CrossRef] [PubMed]
- Pandolfo, P.; Pamplona, F.A.; Prediger, R.D.; Takahashi, R.N. Increased sensitivity of adolescent spontaneously hypertensive rats, an animal model of attention deficit hyperactivity disorder, to the locomotor stimulation induced by the cannabinoid receptor agonist WIN 55,212-2. Eur. J. Pharmacol. 2007, 563, 141–148. [Google Scholar] [CrossRef] [PubMed]
- Centonze, D.; Bari, M.; Di Michele, B.; Rossi, S.; Gasperi, V.; Pasini, A.; Battista, N.; Bernardi, G.; Curatolo, P.; Maccarrone, M. Altered anandamide degradation in attention-deficit/hyperactivity disorder. Neurology 2009, 72, 1526–1527. [Google Scholar] [CrossRef] [PubMed]
- Mansell, H.; Quinn, D.; Kelly, L.E.; Alcorn, J. Cannabis for the Treatment of Attention Deficit Hyperactivity Disorder: A Report of 3 Cases. Med. Cannabis. Cannabinoids. 2022, 5, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Hupli, A.M.M. Medical Cannabis for Adult Attention Deficit Hyperactivity Disorder: Sociological Patient Case Report of Cannabinoid Therapeutics in Finland. Med. Cannabis. Cannabinoids. 2019, 1, 112–118. [Google Scholar] [CrossRef]
- Ittiphakorn, P.; Erridge, S.; Holvey, C.; Coomber, R.; Rucker, J.J.; Sodergren, M.H. UK Medical Cannabis Registry: An analysis of clinical outcomes of medicinal cannabis therapy for attention-deficit/hyperactivity disorder. Neuropsychopharmacol. Rep. 2023, 43, 596–606. [Google Scholar] [CrossRef]
- Stueber, A.; Cuttler, C. Self-Reported Effects of Cannabis on ADHD Symptoms, ADHD Medication Side Effects, and ADHD-Related Executive Dysfunction. J. Atten. Disord. 2022, 26, 942–955. [Google Scholar] [CrossRef] [PubMed]
- Cooper, R.E.; Williams, E.; Seegobin, S.; Tye, C.; Kuntsi, J.; Asherson, P. Cannabinoids in attention-deficit/hyperactivity disorder: A randomised-controlled trial. Eur. Neuropsychopharmacol. 2017, 27, 795–808. [Google Scholar] [CrossRef] [PubMed]
- Hergenrather, J.Y.; Aviram, J.; Vysotski, Y.; Campisi-Pinto, S.; Lewitus, G.M.; Meiri, D. Cannabinoid and Terpenoid Doses are Associated with Adult ADHD Status of Medical Cannabis Patients. Rambam. Maimonides. Med. J. 2020, 11, e0001. [Google Scholar] [CrossRef] [PubMed]
- Sampaio, T.B.; Savall, A.S.; Gutierrez, M.E.Z.; Pinton, S. Neurotrophic factors in Alzheimer’s and Parkinson’s diseases: Implications for pathogenesis and therapy. Neural Regen. Res. 2017, 12, 549–557. [Google Scholar] [CrossRef] [PubMed]
- Esposito, G.; De Filippis, D.; Carnuccio, R.; Izzo, A.A.; Iuvone, T. The marijuana component cannabidiol inhibits beta-amyloid-induced tau protein hyperphosphorylation through Wnt/beta-catenin pathway rescue in PC12 cells. J. Mol. Med. 2006, 84, 253–258. [Google Scholar] [CrossRef] [PubMed]
- Esposito, G.; De Filippis, D.; Maiuri, M.C.; De Stefano, D.; Carnuccio, R.; Iuvone, T. Cannabidiol inhibits inducible nitric oxide synthase protein expression and nitric oxide production in beta-amyloid stimulated PC12 neurons through p38 MAP kinase and NF-kappaB involvement. Neurosci. Lett. 2006, 399, 91–95. [Google Scholar] [CrossRef] [PubMed]
- Iuvone, T.; Esposito, G.; Esposito, R.; Santamaria, R.; Di Rosa, M.; Izzo, A.A. Neuroprotective effect of cannabidiol, a non-psychoactive component from Cannabis sativa, on beta-amyloid-induced toxicity in PC12 cells. J. Neurochem. 2004, 89, 134–141. [Google Scholar] [CrossRef] [PubMed]
- Eubanks, L.M.; Rogers, C.J.; Beuscher, A.E.t.; Koob, G.F.; Olson, A.J.; Dickerson, T.J.; Janda, K.D. A molecular link between the active component of marijuana and Alzheimer’s disease pathology. Mol. Pharm. 2006, 3, 773–777. [Google Scholar] [CrossRef] [PubMed]
- Esposito, G.; Scuderi, C.; Savani, C.; Steardo, L., Jr.; De Filippis, D.; Cottone, P.; Iuvone, T.; Cuomo, V.; Steardo, L. Cannabidiol in vivo blunts beta-amyloid induced neuroinflammation by suppressing IL-1beta and iNOS expression. Br. J. Pharmacol. 2007, 151, 1272–1279. [Google Scholar] [CrossRef]
- Esposito, G.; Scuderi, C.; Valenza, M.; Togna, G.I.; Latina, V.; De Filippis, D.; Cipriano, M.; Carratu, M.R.; Iuvone, T.; Steardo, L. Cannabidiol reduces Abeta-induced neuroinflammation and promotes hippocampal neurogenesis through PPARgamma involvement. PLoS ONE 2011, 6, e28668. [Google Scholar] [CrossRef]
- Martin-Moreno, A.M.; Reigada, D.; Ramirez, B.G.; Mechoulam, R.; Innamorato, N.; Cuadrado, A.; de Ceballos, M.L. Cannabidiol and other cannabinoids reduce microglial activation in vitro and in vivo: Relevance to Alzheimer’s disease. Mol. Pharmacol. 2011, 79, 964–973. [Google Scholar] [CrossRef]
- Janefjord, E.; Maag, J.L.; Harvey, B.S.; Smid, S.D. Cannabinoid effects on beta amyloid fibril and aggregate formation, neuronal and microglial-activated neurotoxicity in vitro. Cell Mol. Neurobiol. 2014, 34, 31–42. [Google Scholar] [CrossRef]
- Scuderi, C.; Steardo, L.; Esposito, G. Cannabidiol promotes amyloid precursor protein ubiquitination and reduction of beta amyloid expression in SHSY5YAPP+ cells through PPARgamma involvement. Phytother. Res. 2014, 28, 1007–1013. [Google Scholar] [CrossRef]
- Cheng, D.; Low, J.K.; Logge, W.; Garner, B.; Karl, T. Chronic cannabidiol treatment improves social and object recognition in double transgenic APPswe/PS1E9 mice. Psychopharmacology 2014, 231, 3009–3017. [Google Scholar] [CrossRef] [PubMed]
- Cheng, D.; Spiro, A.S.; Jenner, A.M.; Garner, B.; Karl, T. Long-term cannabidiol treatment prevents the development of social recognition memory deficits in Alzheimer’s disease transgenic mice. J. Alzheimers Dis. 2014, 42, 1383–1396. [Google Scholar] [CrossRef]
- Aso, E.; Sanchez-Pla, A.; Vegas-Lozano, E.; Maldonado, R.; Ferrer, I. Cannabis-based medicine reduces multiple pathological processes in AbetaPP/PS1 mice. J. Alzheimers Dis. 2015, 43, 977–991. [Google Scholar] [CrossRef] [PubMed]
- Aso, E.; Andres-Benito, P.; Carmona, M.; Maldonado, R.; Ferrer, I. Cannabinoid Receptor 2 Participates in Amyloid-beta Processing in a Mouse Model of Alzheimer’s Disease but Plays a Minor Role in the Therapeutic Properties of a Cannabis-Based Medicine. J. Alzheimers Dis. 2016, 51, 489–500. [Google Scholar] [CrossRef] [PubMed]
- Aso, E.; Andres-Benito, P.; Ferrer, I. Delineating the Efficacy of a Cannabis-Based Medicine at Advanced Stages of Dementia in a Murine Model. J. Alzheimers Dis. 2016, 54, 903–912. [Google Scholar] [CrossRef]
- Hughes, B.; Herron, C.E. Cannabidiol Reverses Deficits in Hippocampal LTP in a Model of Alzheimer’s Disease. Neurochem. Res. 2019, 44, 703–713. [Google Scholar] [CrossRef]
- Schubert, D.; Kepchia, D.; Liang, Z.; Dargusch, R.; Goldberg, J.; Maher, P. Efficacy of Cannabinoids in a Pre-Clinical Drug-Screening Platform for Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 7719–7730. [Google Scholar] [CrossRef]
- Watt, G.; Chesworth, R.; Przybyla, M.; Ittner, A.; Garner, B.; Ittner, L.M.; Karl, T. Chronic cannabidiol (CBD) treatment did not exhibit beneficial effects in 4-month-old male TAU58/2 transgenic mice. Pharmacol. Biochem. Behav. 2020, 196, 172970. [Google Scholar] [CrossRef]
- Alali, S.; Riazi, G.; Ashrafi-Kooshk, M.R.; Meknatkhah, S.; Ahmadian, S.; Hooshyari Ardakani, M.; Hosseinkhani, B. Cannabidiol Inhibits Tau Aggregation In Vitro. Cells 2021, 10, 3521. [Google Scholar] [CrossRef] [PubMed]
- Mooko, T.; Bala, A.; Tripathy, S.; Kumar, C.S.; Mahadevappa, C.P.; Chaudhary, S.K.; Matsabisa, M.G. Cannabis Sativa L. Flower and Bud Extracts Inhibited In vitro Cholinesterases and beta-Secretase Enzymes Activities: Possible Mechanisms of Cannabis Use in Alzheimer Disease. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 297–309. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Choi, P.; Park, Y.T.; Kim, T.; Ham, J.; Kim, J.C. The Cannabinoids, CBDA and THCA, Rescue Memory Deficits and Reduce Amyloid-Beta and Tau Pathology in an Alzheimer’s Disease-like Mouse Model. Int. J. Mol. Sci. 2023, 24, 6827. [Google Scholar] [CrossRef]
- Volicer, L.; Stelly, M.; Morris, J.; McLaughlin, J.; Volicer, B.J. Effects of dronabinol on anorexia and disturbed behavior in patients with Alzheimer’s disease. Int. J. Geriatr. Psychiatry 1997, 12, 913–919. [Google Scholar] [CrossRef]
- Walther, S.; Mahlberg, R.; Eichmann, U.; Kunz, D. Delta-9-tetrahydrocannabinol for nighttime agitation in severe dementia. Psychopharmacology 2006, 185, 524–528. [Google Scholar] [CrossRef]
- Walther, S.; Schupbach, B.; Seifritz, E.; Homan, P.; Strik, W. Randomized, controlled crossover trial of dronabinol, 2.5 mg, for agitation in 2 patients with dementia. J. Clin. Psychopharmacol. 2011, 31, 256–258. [Google Scholar] [CrossRef] [PubMed]
- Woodward, M.R.; Harper, D.G.; Stolyar, A.; Forester, B.P.; Ellison, J.M. Dronabinol for the treatment of agitation and aggressive behavior in acutely hospitalized severely demented patients with noncognitive behavioral symptoms. Am. J. Geriatr. Psychiatry 2014, 22, 415–419. [Google Scholar] [CrossRef]
- van den Elsen, G.A.; Ahmed, A.I.; Verkes, R.J.; Kramers, C.; Feuth, T.; Rosenberg, P.B.; van der Marck, M.A.; Olde Rikkert, M.G. Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: A randomized controlled trial. Neurology 2015, 84, 2338–2346. [Google Scholar] [CrossRef]
- van den Elsen, G.A.H.; Ahmed, A.I.A.; Verkes, R.J.; Feuth, T.; van der Marck, M.A.; Olde Rikkert, M.G.M. Tetrahydrocannabinol in Behavioral Disturbances in Dementia: A Crossover Randomized Controlled Trial. Am. J. Geriatr. Psychiatry 2015, 23, 1214–1224. [Google Scholar] [CrossRef]
- Shelef, A.; Barak, Y.; Berger, U.; Paleacu, D.; Tadger, S.; Plopsky, I.; Baruch, Y. Safety and Efficacy of Medical Cannabis Oil for Behavioral and Psychological Symptoms of Dementia: An-Open Label, Add-On, Pilot Study. J. Alzheimers Dis. 2016, 51, 15–19. [Google Scholar] [CrossRef] [PubMed]
- van den Elsen, G.A.; Tobben, L.; Ahmed, A.I.; Verkes, R.J.; Kramers, C.; Marijnissen, R.M.; Olde Rikkert, M.G.; van der Marck, M.A. Effects of tetrahydrocannabinol on balance and gait in patients with dementia: A randomised controlled crossover trial. J. Psychopharmacol. 2017, 31, 184–191. [Google Scholar] [CrossRef] [PubMed]
- Ruver-Martins, A.C.; Bicca, M.A.; de Araujo, F.S.; de Noronha Sales Maia, B.H.L.; Pamplona, F.A.; da Silva, E.G.; Nascimento, F.P. Cannabinoid extract in microdoses ameliorates mnemonic and nonmnemonic Alzheimer’s disease symptoms: A case report. J. Med. Case Rep. 2022, 16, 277. [Google Scholar] [CrossRef] [PubMed]
- Palmieri, B.; Vadala, M. Oral THC: CBD cannabis extract in main symptoms of Alzheimer disease: Agitation and weight loss. Clin. Ter. 2023, 174, 53–60. [Google Scholar] [CrossRef] [PubMed]
- Libro, R.; Diomede, F.; Scionti, D.; Piattelli, A.; Grassi, G.; Pollastro, F.; Bramanti, P.; Mazzon, E.; Trubiani, O. Cannabidiol Modulates the Expression of Alzheimer’s Disease-Related Genes in Mesenchymal Stem Cells. Int. J. Mol. Sci. 2016, 18, 26. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, A.I.; van den Elsen, G.A.; Colbers, A.; Kramers, C.; Burger, D.M.; van der Marck, M.A.; Olde Rikkert, M.G. Safety, pharmacodynamics, and pharmacokinetics of multiple oral doses of delta-9-tetrahydrocannabinol in older persons with dementia. Psychopharmacology 2015, 232, 2587–2595. [Google Scholar] [CrossRef]
- Ruthirakuhan, M.; Lanctot, K.L.; Vieira, D.; Herrmann, N. Natural and Synthetic Cannabinoids for Agitation and Aggression in Alzheimer’s Disease: A Meta-Analysis. J. Clin. Psychiatry 2019, 80, 2246. [Google Scholar] [CrossRef]
Preclinical Studies | |||
Model | Treatment | Main Findings | References |
InsG3680 Shank3 mutant mice | CBD/THC ratio 25:1 oil (25 mg/kg CBD, 1 mg/kg THC), CBD oil (25 mg/kg CBD), or THC oil (1 mg/kg THC) or olive oil (5 mL/kg) intragastric, twice a week, for 3 consecutive weeks. | THC oil is preferable for treating autistic-like phenotypes. THC oil improved social deficits, repetitive grooming, and anxiety-like behaviors. CBD/THC ratio 25:1 oil decreased the glutamate concentration in the CSF. These effects were linked to CBR1 activation. | [42] |
BTBR T+ Itpr3tf/J (BTBR) mice | Inhaled CBD isolate oil. Specifically, 83.3 µL vapored for 6 s, every 5 min, totaling 30 min. | Inhaled CBD improves prosocial and anxiety-like behaviors. Only the terpene blend, also enhanced prosocial behavior, independently of CBD. | [41] |
Adult male BTBR, C57BL/6J and SERT KO mice | CBD 0.1, 1, or 10 mg/kg, i.p., 30 min before beginning the tests. | CBD (10 mg/kg) enhanced social interaction preference and attenuated social novelty preference in BTBR mice. No behavioral difference was observed in C57BL/6J or SERT KO mice treated with CBD. | [39] |
WT and Fmr1-Δexon 8 male rats | Acute treatment: CBD 10 mg/kg or vehicle, i.p., 2 h before testing, at PND40. Chronic treatment: CBD 10 mg/kg or vehicle, i.p., once daily for three weeks (PND 19 to PND39). | CBD reduced short-term object recognition deficits displayed by Fmr1-Δexon 8 rats, without inducing tolerance after repeated administration, through hippocampal GPR55 receptor stimulation. | [40] |
Clinical Studies | |||
Study Features | Treatment | Main Findings | References |
Case report of a child (6 years old) | THC (initial dosage of 0.62 mg, gradually increased to 3.62 mg). | THC enhanced hyperactivity, irritability, lethargy, stereotype, and inappropriate speech. | [43] |
Retrospective case series (n = 21; mean age = 9 years old) | Sublingual cannabis extracts (71.5% received balanced CBD:THC extract, 19% high CBD, and 9.5% high THC extracts) for at least three months. | Cannabis extracts improved at least one of ASD’s core symptoms and comorbidities, such as sensory difficulties, sleep disorders, and/or seizures. | [44] |
Observational study (n = 53; mean age = 11 years old) | CBD-rich oil at a daily dose of CBD 16 mg/kg (maximal daily dose 600 mg), and THC 0.8 mg/kg (maximal daily dose of 40 mg). | 20:1 CBD/THC was associated with improvements in various comorbidity symptoms of ASD, such as hyperactivity, self-injury and rage attacks, sleep problems, and anxiety. These results were not statistically different from conventional treatments. | [45] |
Retrospective study (n = 60; mean age = 11 years old) | 20:1 CBD/THC extract, two or three times a day, starting with 1 mg/kg/day of CBD and a maximal dose of 10 mg/kg/day of CBD. | CBD-based treatment improved the behavioral outbreaks in 61% of ASD children. Side effects included sleep disorders (14%), irritability (9%), and loss of appetite (9%). One serious psychotic event was observed after a higher dose of THC. | [46] |
Observational study (n = 188; mean age = 12 years old) | CBD-rich oil in a ratio of 20:1 (30% CBD and 1.5% THC). | CBD-rich oil is well tolerated, safe, and effective in relieving symptoms of ASD children, including seizures, tics, depression, restlessness, and rage attacks. Significant improvements were reported in the quality of life, mood, ability to perform daily activities, sleep, and concentration, after 6 months of treatment. Specifically, 34.3% of patients reduced the use of concomitant medications. Improvement in a child’s global assessment was reported by more than 80% of parents. | [47] |
Observational study (n = 18; mean age = 10 years old) | Approximately 75:1 CBD/THC administered in capsules containing, respectively, 25 or 50 mg of CBD and 0.34 or 0.68 mg of THC. | Among the 15 patients who adhered to the treatment, only one patient failed to improve in regard to their ASD symptoms after 6–9 months of treatment. The strongest ameliorations were seizures, ADHD, sleep disorders, communication, and social interaction deficits. | [48] |
Placebo-controlled, randomized, double-blind, repeated-measures, crossover case-control study (n = 34; mean age = 30 years old) | 600 mg of CBD, or 600 mg of CBDV, or placebo. | CBD modulates glutamate/GABA systems, but the prefrontal GABA system is decreased in ASD. CBD significantly increased the fractional amplitude of low-frequency fluctuations in the cerebellar vermis and the right fusiform gyrus in the ASD group, with no significant change in the controls, and altered vermal functional connectivity with several of its subcortical and cortical targets in the ASD group. CBDV also significantly increased glutamate in the basal ganglia of ASD patients. | [49,50,51] |
Case report of a teenager (15 years old) | 0.1 mL of 20:1 CBD/THC (2 mg CBD and 0.1 mg THC), twice a day, with an increase of 0.1 mL per dose if no effects were observed, up to a maximum of 0.5 mL (10 mg CBD and 0.5 mg THC). | Low doses of CBD/THC (4 mg CBD and 0.2 mg THC) improved anxiety, sleep, and social deficits. | [52] |
Randomized, double-blind, placebo-controlled trial (n = 150; mean age = 11 years old) | Oral placebo, CBD/THC at a 20:1 ratio (167 mg/mL CBD and 8.35 mg/mL THC), pure CBD and pure THC at the same concentration. | No significant differences in the total scores of the HSQ-ASD and the APSI among the treatment groups. Disruptive behavior, assessed by CGI-I, and median SRS total score showed a significant improvement in participants who received CBD/THC extract compared to the placebo. | [53] |
Phase I/II double-blind, parallel-group, randomized, placebo-controlled pilot study (n = 8; mean age = 13 years old) | Starting dose of CBD at 5 mg/kg/day (two doses). The dose was increased in increments of 5 mg/kg every 3 days for 9 days up to the maintenance dose of 20 mg/kg/day. A maximal dose of 1000 mg/day was administered to all participants weighing 50 kg or greater. | CBD was effective in reducing severe behavioral problems in children and adolescents with intellectual disability. | [54] |
Case report of a child (9-year-old) | High-CBD and low-THC oil (20 mg of CBD and <1 mg of THC/mL). The starting dose was 0.1ml, twice daily, increased every three to four days until 0.5 mL, twice daily. | High-CBD and low-THC oil treatment reduced the behavioral deficits, including violent outbursts, self-injurious behaviors, and sleep disruptions, and improved social interactions, concentration, and emotional stability, without side effects. | [55] |
Case report of a teenager (17 years old) | Terpene-enriched CBD oil, three times/day, corresponding to 11.2 mg CBD/day and 0.19 mg/kg. | Terpene-enriched CBD oil was more effective than CBD oil alone in treating aggression associated with ASD. | [56] |
Open-label study (n = 110; mean age = 9 years old) | CBD/THC oil 20:1, starting with one daily drop containing 5.7 mg CBD and 0.3 mg THC, with a gradual increase in dosage until parents perceived improvements. | Significant improvements were observed in ADOS-2, SRS, and Vineland scores after a 6-month treatment. Participants with more severe initial symptoms showed larger improvements following the intervention, regardless of age or final cannabis dosage. | [57] |
Prospective cohort (n = 59; mean age = 10 years old) | CBD/THC oil 20:1, starting with one daily drop containing 5.7 mg CBD and 0.3 mg THC, with a gradual increase in dosage until parents perceived improvements. | No significant changes were observed in total blood count, urea, creatinine, liver enzymes, thyroid hormones, or other hormones after 3 months of treatment. LDH and TSH levels were significantly lower after 3 months of treatment compared to the baseline, while the free T4 levels were significantly higher. There were no significant differences in the biochemical parameters between the group receiving concomitant medications and the group receiving only the cannabis oil, except for a higher potassium level in the group not receiving additional medications. All values remained within the normal range. | [58] |
Placebo-controlled trial (n = 150; mean age = 11 years old) | Whole-plant extract (CBD/THC ratio 20:1), pure cannabinoids (CBD/THC at a 20:1 ratio), and placebo. The starting dose was 1 and 0.05 mg/kg/day of CBD and THC, respectively, or placebo. The dose was increased every other day, up to 10 and 0.5 mg/kg/day of CBD and THC, respectively, for children weighing 20–40 kg; or 7.5 and 0.375 mg/kg/day of CBD and THC, respectively, for weight >40 kg until 420 mg CBD and 21 mg THC/day, divided into three daily doses. | CBD-rich cannabinoid treatment did not improve sleep disturbances more than the placebo, accessed by CSHQ. However, the total CSHQ scores were associated with improvements in the ASD core symptoms, as indicated by the SRS scores. | [59] |
Retrospective cohort (n = 20; mean age = 14 years old) | Treatment starts with low to intermediate doses of a CBD-rich extract (25–50 mg CBD/day), which were slowly increased over 4 to 12 weeks between 50–150 mg CBD/day. | Overall, 18 out of 20 patients showed improvements in symptoms of ASD, such as behavioral disorders, communication, social interaction deficits, and sleep disorders, and in the quality of life for patients and their families. Side effects were mild and infrequent. | [60] |
Observational study (n = 24; mean age = 9 years old) | Individual treatment with cannabis products for at least one year. THC doses ranged from 0.05 to 50 mg in 40% of children and CBD from 7.5 to 200 mg in 60% of children. | Among 65 potential cannabis-responsive biomarkers, 22 were categorized as anti-inflammatory, bioenergy associated, neurotransmitters, amino acids, and endocannabinoids. These biomarkers shifted toward typical physiological levels in children with ASD after cannabinoid treatment. | [61] |
Randomized, double-blind, and placebo-controlled clinical trial (n = 60; mean age = 7 years old) | CBD-rich extracts at a concentration of 0.5%, in the ratio of 9CBD/1THC. The starting dose (2.5 mg/mL) was six drops, twice a day, with an increase of two drops daily twice a week, if necessary, up to a maximum dose of 70 drops daily. | CBD-rich extract treatment resulted in an improvement in social interaction, psychomotor agitation, number of meals, and anxiety. Concentration enhancement was only observed in mild ASD carriers. A total of 9.7% of the children experienced adverse effects that were mild and transient. | [62] |
Preclinical Studies | |||
Model | Treatment | Main Findings | References |
PC12 neuronal cells exposed to Aβ1–42 peptide | Pretreatment with CBD (10−7–10−4 M) | CBD prevented nitroxidative stress, apoptosis induction, tau protein hyperphosphorylation, and neurotoxicity induced by the Aβ1–42 peptide. Also, CBD inhibited iNOS via p38 MAPK and NF-κB. | [76,77,78] |
AChE-induced Aβ1–40 peptide aggregation in vitro | THC (6.25–50 µM) | THC competitively inhibited AChE activity, as well as prevented AChE-induced Aβ peptide aggregation. | [79] |
Intrahippocampal Aβ1–42 injection in mice | CBD (2.5 or 10 mg/kg, i.p.) for 7 days | CBD dose-dependently inhibited reactive gliosis and proinflammatory markers (iNOS and IL-1b) caused by the Aβ1–42 peptide. | [80] |
Intrahippocampal Aβ1–42 injection in rats | CBD (10 mg/kg, i.p.) for 15 days | CBD interacted with PPARγ, reducing reactive gliosis and stimulating hippocampal neurogenesis. | [81] |
Intracerebroventricular Aβ1–40 injection in mice | CBD (20 mg/kg, i.p.) daily for 7 days, following for 3 days/week for 2 weeks | CBD prevented learning deficit in the water maze task and IL-6 increased gene expression induced by Aβ1–40, but did not affect TNF-α gene expression. | [82] |
SH-SY5Y neuroblastoma cells BV-2 microglia cells | THC and CBD (10 µM) | THC and CBD protected against microglial-activated neurotoxicity, but only CBD prevented Aβ neurotoxicity. | [83] |
SH-SY5YAPP+ neuroblastoma cells | CBD (10−7–10−4 M) | Through PPARγ activation, CBD induced APP ubiquitination, resulting in decreased Aβ production and increased cell survival. | [84] |
AβPP/PS1 mice aged 2.5 months | CBD (20 mg/kg, i.p.) daily for 3 weeks | CBD reversed social and novel object recognition memory impairments without affecting anxiety-related behaviors. | [85] |
AβPP/PS1 mice aged 2.5 months | CBD (20 mg/kg, i.p.) daily for 8 months | CBD prevented the development of social recognition memory deficits, attenuated neuroinflammation, cholesterol, and dietary phytosterol retention, and did not impact anxiety, associative learning, Aβ load, or oxidative stress. | [86] |
AβPP/PS1 mice aged 6 months (early symptomatic phase) | THC (0.75 mg/kg, i.p.); CBD (0.75 mg/kg, i.p.); THC + CBD (0.75 mg/kg, i.p., each) for 5 weeks | THC + CBD reduced learning impairment in object recognition and active avoidance tasks. Also, THC + CBD decreased soluble Aβ1–42 levels, changed plaque composition, and reduced astrogliosis, microgliosis, and proinflammatory molecules, more marked than with either THC or CBD. | [87] |
APP/PS1 with CB2R knockout mice aged 3 and 6 months | THC + CBD (0.75 mg/kg, i.p., each) for 5 weeks | The CB2R absence exacerbated cortical Aβ deposition and soluble Aβ1–40 levels, without affecting the mice’s viability, tau hyperphosphorylation, memory impairment, and the positive cognitive effect demonstrated by THC + CBD. | [88] |
AβPP/PS1 mice aged 12 months (advanced stages) | THC + CBD (0.75 mg/kg, i.p., each) for 5 weeks | THC + CBD was effective against memory impairment but did not modify Aβ processing or reduce glial reactivity. Mnemonic improvement was linked to reduced GluR2/3 and increased levels of GABA-A Rα1. | [89] |
Hippocampal slices from C57BL6 mice exposed to Aβ1–42 peptide | CBD (10 µM) | Pretreatment with CBD restored the Aβ1–42-mediated deficit in LTP in the hippocampal CA1 region. The PPARγ antagonist blocked this effect. | [90] |
MC65 neuronal cells exposed to Aβ HT22 hippocampal cells, primary neurons BV2 microglia cells | Cannabinoid compounds (250 nM–10 μM) | CBDA, THC, CBDV, Δ8-THC, CBG, CBC, CBN, and CBD removed intraneuronal Aβ, reduced oxidative damage, and offered protection from the loss of energy, trophic support withdrawal, and inflammation. THC and CBD lead to a synergistic neuroprotective effect. | [91] |
TAU58/2 mice aged 4 months | CBD (50 mg/kg, i.p.) for 3 weeks | CBD did not affect the behavioral changes observed (anxiolytic like, motor impairment, and increased freezing in the fear conditioning paradigm). | [92] |
Recombinant human tau protein 1N/4R isoform | Different CBD concentrations (0–40 μM) | CBD inhibits tau fibrils formation, by binding tau protein in Tyr residues. The tau–CBD complex formation is an exothermic process that occurs through hydrogen bonds and van der Waals forces in a spontaneous interaction. | [93] |
Vero (CCL-81) pre-adipocytes (3T3-L1) (CL-173) | Cannabis extracts in hexane, DCM, DCM:MeOH (1:1), MeOH and water (1–100 μg/mL), CBD and THC (1–50 μg/mL), physostigmine (1–10 μg/mL) | Physostigmine and CBD inhibited AChE and BChE, while THC showed paltry inhibitory activity against both cholinesterases. Hexane, DCM, DCM:MeOH, MeOH, and water cannabis extracts did not inhibit AChE, but hexane and DCM cannabis extract inhibit BChE activity. All cannabis extracts inhibit β-secretase activity. Low cytotoxicity was observed. | [94] |
Primary neurons, female ICR mice (8 weeks old) exposed to Aβ1–42 | Primary neurons treated with Aβ1–42 and/or CBDA or THCA for 24 h, CBDA (6 μM, 3 μL/mouse), THCA (12 μM, 3 μL/mouse) | CBDA and THCA significantly suppressed neuronal cell death, ameliorated AD-like features by modulating Ca2+ levels, and reduced the APP increase, polymeric Aβ, oligomeric Aβ, and p-tau levels induced by Aβ1–42. CBDA or THCA induced a better performance in the Morris water maze and novel object recognition tests, reduced the increase in APP, polymeric Aβ, oligomeric Aβ, and p-tau levels induced by Aβ1–42, and significantly increased BDNF, p-TrkB, and p-CREB levels. | [95] |
Clinical Studies | |||
Study Features | Treatment | Main Findings | Reference |
Randomized placebo-controlled double-blind crossover study (n = 12) | Placebo or THC 2.5 mg, p.o., for 6 weeks each | THC increased body weight and decreased agitation (CMAI), even during the placebo treatment period. Adverse reactions included euphoria, somnolence, and tiredness, without needed medication discontinuation. | [96] |
Open label, prospective descriptive trial, single arm, single center (n = 6) | THC 2.5 mg, p.o., every evening for 2 weeks | THC reduced nocturnal motor activity and NPI score in 2 weeks, without significant impact on anxiety, apathy, delusions, hallucinations, or augmentative sedative medication use. | [97] |
Single center randomized placebo-controlled double-blind crossover study (n = 2) | Placebo or THC 2.5 mg, p.o., every evening for 4 weeks each | THC initially decreased nocturnal motor activity, reduced NPI score after 4 weeks, and improved circadian rhythms (NPCRA) during treatment. | [98] |
Retrospective cohort, single center (n = 40) | THC 7.03 mg/day for 4–7 days | THC decreased all domains of PAS, and improved CGI, sleep duration, and percentage of meals consumed during the treatment. There was no difference in GAF or change in weight. Sedation, delirium, urinary tract infection, and confusion were the most frequently documented adverse events. None of them led to medication discontinuation. | [99] |
Multicenter randomized placebo-controlled double-blind crossover study (n = 50) | Placebo or THC 1.5 mg, p.o., 3×/day 3 weeks | No significant differences were found in the NPI score, CMAI, quality of life, activities of daily living according to the Barthel Index, vital signs, weight, episodic memory, and adverse effects between the placebo and THC. | [100] |
Multicenter randomized placebo-controlled double-blind crossover study (n = 20) | Placebo and THC 0.75 mg, p.o., 2×/day, in random order, for 3 days, separated by a four-day washout for 6 weeks. After, the THC dosage was increased to 1.5 mg in the same design for 6 weeks. | THC did not reduce the NPI score compared to the placebo but was well tolerated, as assessed by adverse event monitoring, vital signs, and mobility. | [101] |
Open label, placebo-controlled, prospective descriptive trial (n = 10) | Cannabis oil (THC 2.5 to a maximum of 7.5 mg) 2×/day for 4 weeks | Cannabis oil decreased the NPI score (decreased domains: delusions, agitation/aggression, irritability, apathy, sleep, and caregiver distress) and CGI in 4 weeks. | [102] |
Randomized placebo-controlled double-blind crossover study (n = 18) | Placebo and THC 1.5 mg, p.o., 2×/day, in random order, for 3 days, separated by a 4-day washout. | THC elevated sway during standing eyes closed but not during standing eyes open, increased the stride length and trunk sway during preferred speed walking. No differences in the number and type of adverse events were found, and no falls occurred. | [103] |
Case report (n = 1) | Cannabis extract (THC:CBD 8:1) (300 µg–1000 µg THC) | Microdoses of THC:CBD 8:1 improved MMSE and ADAS-Cog score. Cognitive enhancement was stable for more than one year. No significant side effects or toxicity were observed. The most effective dose for AD symptom suppression appeared to be 500 µg of THC. | [104] |
Anecdotal, spontaneous, and observational study (n = 30) | Cannabis oil extract sublingually, twice daily for 12 weeks (1st–2nd week 15 drops/day; 3rd–4th week 23 drops/day; 5th–8th week 30 drops/day; 9th–10th week 23 drops/day; 11th–12th week 15 drops/day) | Reduction in typical AD behavioral problems, such as agitation, apathy, irritability, sleep disturbance, and eating disturbances, evaluated by the NPI questionnaire. Reduction in aggressive behavior, measured by the CMAI questionnaire. Overall, 45% of patients previously classified as severe cognitive decline were stratified as mild/moderate by the MMSE questionnaire. Positive feedback by the caregiver. | [105] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dallabrida, K.G.; de Oliveira Bender, J.M.; Chade, E.S.; Rodrigues, N.; Sampaio, T.B. Endocannabinoid System Changes throughout Life: Implications and Therapeutic Potential for Autism, ADHD, and Alzheimer’s Disease. Brain Sci. 2024, 14, 592. https://doi.org/10.3390/brainsci14060592
Dallabrida KG, de Oliveira Bender JM, Chade ES, Rodrigues N, Sampaio TB. Endocannabinoid System Changes throughout Life: Implications and Therapeutic Potential for Autism, ADHD, and Alzheimer’s Disease. Brain Sciences. 2024; 14(6):592. https://doi.org/10.3390/brainsci14060592
Chicago/Turabian StyleDallabrida, Kamila Gabrieli, Joyce Maria de Oliveira Bender, Ellen Schavarski Chade, Nathalia Rodrigues, and Tuane Bazanella Sampaio. 2024. "Endocannabinoid System Changes throughout Life: Implications and Therapeutic Potential for Autism, ADHD, and Alzheimer’s Disease" Brain Sciences 14, no. 6: 592. https://doi.org/10.3390/brainsci14060592
APA StyleDallabrida, K. G., de Oliveira Bender, J. M., Chade, E. S., Rodrigues, N., & Sampaio, T. B. (2024). Endocannabinoid System Changes throughout Life: Implications and Therapeutic Potential for Autism, ADHD, and Alzheimer’s Disease. Brain Sciences, 14(6), 592. https://doi.org/10.3390/brainsci14060592