1
|
Mori K, Togo A, Yamashita K, Sakuragi S, Bannai H, Umezawa T, Ohta K, Asahi T, Nozaki C, Kataoka K. Mitochondrial damage and ER stress in CB1 receptor antagonist-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neuropharmacology 2025; 273:110440. [PMID: 40185361 DOI: 10.1016/j.neuropharm.2025.110440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/13/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Cannabinoid receptor type 1 (CB1R) is the key modulator of neuronal viability. CB1R antagonists provide neuroprotective effects on neurotoxicity caused by e.g. neuronal injury. However, the underlying mechanisms and potential limitations of CB1R antagonism remain unclear. Here we investigated the impact of environmental conditions on CB1R antagonist effects. We have found that cell-permeable CB1R antagonists, rimonabant and AM251, induced cell death in human neuroblastoma SH-SY5Y cells under serum-free conditions. Mitochondrial morphological analysis revealed mitochondrial swelling characterized by their network fragmentation and cristae reduction. Phosphoproteomics analysis showed the ER stress signaling pathway PERK/eIF2α/ATF4/CHOP, leading to caspase-dependent apoptosis. These results suggest that CB1R antagonists promote apoptosis via mitochondrial damage and ER stress under serum-free conditions in SH-SY5Y cells. Our findings indicate that while CB1R antagonists may be neuroprotective in certain conditions, they may also pose a neurotoxic risk in environments characterized by cellular stress or nutrient deprivation.
Collapse
Affiliation(s)
- Kazuaki Mori
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Kota Yamashita
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Shigeo Sakuragi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Hiroko Bannai
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Taishi Umezawa
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Keisuke Ohta
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Toru Asahi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan
| | - Chihiro Nozaki
- Global Center for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Kosuke Kataoka
- Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan; Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan.
| |
Collapse
|
2
|
Tseng KY, Molla HM. Cannabinoid CB1 receptor-sensitive neurodevelopmental processes and trajectories. Mol Psychiatry 2025:10.1038/s41380-025-03057-2. [PMID: 40389627 DOI: 10.1038/s41380-025-03057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 05/12/2025] [Indexed: 05/21/2025]
Abstract
As high-potency cannabis (with high Δ9-Tetrahydrocannabinol content) becomes easily accessible and widespread, it is of extreme importance for public health that a scientific platform is used to implement practical guidelines, particularly for at-risk populations. Many reviews have been written in the past decade summarizing the impact of cannabis in the developing brain. One critical concept frequently mentioned but not discussed in detail is whether there are sensitive neurodevelopmental events driving the age-specific sensitivity to cannabis, particularly those mediated by cannabinoid type 1 receptor signaling. By integrating available data from humans and animal models, the goal of the present expert review article is to provide a mechanistic overview on how cannabis exposure during sensitive periods of neural circuit plasticity and development can result in lasting consequences. Here we used the frontal cortex as a proxy to align the trajectory of the brain cannabinoid system between humans and rodents. Both the strengths and limitations of available mechanistic studies on the effects of cannabis and cannabinoids were discussed using a developmental framework from which neural circuit adaptations during sensitive periods are considered. Such an approach is needed to align key neurodevelopmental variables through the lifespan, which in turn will provide valuable insights applicable to the human brain by defining the underpinning mechanisms of sensitive periods and how the impact of cannabis changes from childhood to adolescence, and thereafter through young adulthood.
Collapse
Affiliation(s)
- Kuei Y Tseng
- Department of Anatomy & Cell Biology, University of Illinois at Chicago - College of Medicine, Chicago, IL, USA.
| | - Hanna M Molla
- Department of Psychiatry & Behavioral Neuroscience, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Tyler J, Park Y, Lu H, Roeder N, Richardson B, Gold MS, Blum K, Pinhasov A, Baron D, Thanos PK. High-intensity interval training exercise decreases brain CB1 receptor levels in male and female adult rats. Neuroscience 2025; 573:254-263. [PMID: 40122443 DOI: 10.1016/j.neuroscience.2025.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
The numerous health benefits of exercise are well-documented, including its efficacy in treating substance use disorders (SUDs). Several exercise regimens have been proposed; however, the most effective regimen for patients with addiction has yet to be elucidated. High-intensity interval training (HIIT) exhibits considerable potential compared to aerobic and resistance exercise. Dopamine signaling is recognized as a key neurobiological mechanism contributing to HIIT's therapeutic potential for SUDs; however, the role of the endocannabinoid system in this context is not well understood. The present study investigated the effects of HIIT exercise on endocannabinoid signaling by measuring cannabinoid receptor 1 (CB1R) binding in the brains of male and female rats using [3H]SR141716A autoradiography. Male and female rats were separated into sedentary and HIIT exercise groups. For six weeks, exercise was completed daily on a treadmill for 30 min (10 3-min intervals) progressively increasing speed to 0.8 mph (21.5 m/min). The HIIT program significantly reduced CB1R binding in both sexes across multiple brain regions, including the striatum, thalamus, and distinct areas of the cortex. Sex differences were observed wherein males exhibited greater CB1R binding than females across brain regions, including the cerebellum, striatum, and parts of the cortex. Males experienced an increase in mean cerebellum CB1R binding due to HIIT, whereas females showed no effect in this region. The results suggest HIIT exercise can modulate endocannabinoid signalling by way of decreased CB1R binding. These findings further support the intensity dependence of endocannabinoid modulation and highlight potential pathways for exercise-induced neurobiological and behavioural change.
Collapse
Affiliation(s)
- John Tyler
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
| | - Youmin Park
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Huy Lu
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Nicole Roeder
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA
| | - Brittany Richardson
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Mark S Gold
- Department of Psychiatry, Washington University in St. Louis Euclid Ave, St. Louis, MO 63110, USA
| | - Kenneth Blum
- Division of Addiction Research and Education, Center for Sports, Exercise, and Mental Health, Western University Health Sciences, Pomona, CA 91766, USA; Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - David Baron
- Division of Addiction Research and Education, Center for Sports, Exercise, and Mental Health, Western University Health Sciences, Pomona, CA 91766, USA; Department of Psychiatry, Stanford University School of Medicine, Palo Alto, CA. 94305-5101 USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; Department of Psychology, University at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
4
|
Pereira CF, Boileau I, Kloiber S. Effects of pharmacological inhibition of fatty acid amide hydrolase on corticosterone release: a systematic review of preclinical studies. DISCOVER MENTAL HEALTH 2025; 5:51. [PMID: 40195219 PMCID: PMC11977098 DOI: 10.1007/s44192-025-00155-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/02/2025] [Indexed: 04/09/2025]
Abstract
Psychiatric conditions are often linked to dysfunction of the Hypothalamic-Pituitary-Adrenal (HPA) axis. The Endocannabinoid System (ECS) plays a significant role in stress and anxiety and interacts with the HPA axis. The ECS metabolizing enzyme, Fatty Acid Amide Hydrolase (FAAH), may be integral for HPA axis response to stress by reducing levels of the endocannabinoid anandamide (AEA). However, there is conflicting evidence regarding the effects of FAAH inhibition on stress-related hormone changes, and no comprehensive evaluation of this literature exists. This review aims to synthesize the literature on the impact of pharmacological FAAH inhibition on corticosterone levels in rodents. A systematic search of PubMed/MEDLINE, APA PsychInfo, and Embase up to July 2024 was conducted. Articles reporting the effects of FAAH inhibition on corticosterone levels in rodents were included. Risk of Bias was assessed using SYRCLE's Risk of Bias tool. This review included 21 articles. FAAH inhibition showed limited effects depending on type of FAAH inhibitor, stress exposure, and rodent age. Selective FAAH inhibition did not significantly affect corticosterone levels in the absence of stress and showed minimal effects following acute stress. After chronic stress, these compounds showed more pronounced effects, reducing corticosterone in 40% of studies. Limited studies employing flavonoid-based and dual FAAH/TRPV1 inhibitors suggested blunted corticosterone after acute, but not chronic stress. This review found that FAAH inhibition has inconsistent effects on corticosterone regulation, highlighting the complex and context-dependent role of FAAH inhibition in modulating stress hormone responses, warranting further investigation to clarify its therapeutic potential in stress-related disorders.
Collapse
Affiliation(s)
- Christina F Pereira
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Stefan Kloiber
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Kansagara D, Hill KP, Yost J, Humphrey LL, Shaw B, Obley AJ, Haeme R, Akl EA, Qaseem A, Dunn AS, Jackson CD, Jokela JA, Lee RA, Mackey K, Saini SD, Tschanz MP, Wilt TJ, Etxeandia-Ikobaltzeta I, Shamliyan T, Vigna C. Cannabis or Cannabinoids for the Management of Chronic Noncancer Pain: Best Practice Advice From the American College of Physicians. Ann Intern Med 2025. [PMID: 40183677 DOI: 10.7326/annals-24-03319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
DESCRIPTION The American College of Physicians' Population Health and Medical Science Committee (PHMSC) developed this best practice advice to inform clinicians about what is currently known about the benefits and harms of cannabis or cannabinoids in the management of chronic noncancer pain and to provide advice for clinicians counseling patients seeking this therapy. METHODS The PHMSC considers areas where evidence is uncertain or emerging or practice does not follow the evidence to provide clinical advice based on a review and assessment of scientific work, including systematic reviews and individual studies. Sources of evidence included a living systematic review on cannabis and cannabinoid treatments for chronic noncancer pain and a series of living systematic reviews and primary studies. BEST PRACTICE ADVICE 1A Clinicians should counsel patients about the benefits and harms of cannabis or cannabinoids when patients are considering whether to start or continue to use cannabis or cannabinoids to manage their chronic noncancer pain. BEST PRACTICE ADVICE 1B Clinicians should counsel the following subgroups of patients that the harms of cannabis or cannabinoid use for chronic noncancer pain are likely to outweigh the benefits: young adult and adolescent patients, patients with current or past substance use disorder, patients with serious mental illness, and frail patients and those at risk for falling. BEST PRACTICE ADVICE 2 Clinicians should advise against starting or continuing to use cannabis or cannabinoids to manage chronic noncancer pain in patients who are pregnant or breastfeeding or actively trying to conceive. BEST PRACTICE ADVICE 3 Clinicians should advise patients against the use of inhaled cannabis to manage chronic noncancer pain.
Collapse
Affiliation(s)
- Devan Kansagara
- Oregon Health & Science University, Portland, Oregon (D.K., L.L.H., A.J.O.)
| | - Kevin P Hill
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts (K.P.H.)
| | - Jennifer Yost
- American College of Physicians, Philadelphia, and Villanova University, Villanova, Pennsylvania (J.Y.)
| | - Linda L Humphrey
- Oregon Health & Science University, Portland, Oregon (D.K., L.L.H., A.J.O.)
| | - Beth Shaw
- Center for Evidence-based Policy, Oregon Health & Science University, Portland, Oregon (B.S.)
| | - Adam J Obley
- Oregon Health & Science University, Portland, Oregon (D.K., L.L.H., A.J.O.)
| | - Ray Haeme
- Granite Falls, North Carolina (R.H.)
| | - Elie A Akl
- Department of Internal Medicine, American University of Beirut, Beirut, Lebanon, and Department of Health and Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada (E.A.A.)
| | - Amir Qaseem
- American College of Physicians, Philadelphia, Pennsylvania (A.Q.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Flores-López M, Herrera-Imbroda J, Requena-Ocaña N, García-Marchena N, Araos P, Verheul-Campos J, Ruiz JJ, Pastor A, de la Torre R, Bordallo A, Pavón-Morón FJ, Rodríguez de Fonseca F, Serrano A. Exploratory study on plasma Acylglycerol and Acylethanolamide dysregulation in substance use and attention-deficit/hyperactivity disorder: Implications for novel biomarkers in dual diagnosis. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111350. [PMID: 40188983 DOI: 10.1016/j.pnpbp.2025.111350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025]
Abstract
Substance use disorder (SUD) is a major global public health challenge, frequently co-occurring with psychiatric conditions such as attention-deficit/hyperactivity disorder (ADHD). Endocannabinoid system (ECS) dysregulation has been implicated in both SUD and ADHD, but the interplay between these conditions remains poorly understood. This study investigates plasma concentrations of endocannabinoid-congeners in individuals with SUD, with and without comorbid ADHD, to identify potential biomarkers. This exploratory study included 469 participants divided into three groups: (1) healthy controls (n = 136), (2) patients with SUD without ADHD (n = 267), and (3) patients with SUD and comorbid ADHD (n = 66). Plasma concentrations of 12 endocannabinoid-related molecules, including acylglycerols (2-AG, 2-LG, 2-OG) and acylethanolamides (AEA, DEA, DHEA, DGLEA, LEA, OEA, PEA, POEA, and SEA), were quantified using high-performance liquid chromatography-mass spectrometry (HPLC-MS/MS). A multinomial Elastic Net regression model was applied to assess their biomarker potential. Patients with SUD exhibited significantly lower plasma concentrations of 2-AG and 2-LG compared to controls, while most acylethanolamides were elevated, except for POEA and SEA. ADHD comorbidity was associated with lower concentrations of 2-AG, 2-LG, AEA, DGLEA, DHEA, and SEA, while PEA was elevated. Machine learning analysis identified AEA, OEA, PEA, and SEA as key biomarkers, achieving an accuracy of 72.1 % and an ROC-AUC of 0.77. This study suggests distinct ECS alterations in SUD and comorbid ADHD, highlighting endocannabinoid-congeners as potential biomarkers. Future research should validate these findings in larger cohorts and explore ECS-targeted therapeutic interventions for dual-diagnosis populations.
Collapse
Affiliation(s)
- María Flores-López
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, 29590 Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Jesús Herrera-Imbroda
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, 29590 Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Nerea Requena-Ocaña
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, 29590 Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Nuria García-Marchena
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28223 Pozuelo de Alarcón, Spain
| | - Pedro Araos
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain
| | - Julia Verheul-Campos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, 29590 Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Juan Jesús Ruiz
- Centro Provincial de Drogodependencias de Málaga, Diputación Provincial de Málaga, 29010 Málaga, Spain
| | - Antoni Pastor
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rafael de la Torre
- Hospital del Mar Research Institute, 08003 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Antonio Bordallo
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Francisco Javier Pavón-Morón
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, 29590 Málaga, Spain; Unidad Clínica Área del Corazón, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, 29590 Málaga, Spain; Servicio de Neurología, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), 29010 Malaga, Spain.
| | - Antonia Serrano
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina - IBIMA Plataforma BIONAND, 29590 Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| |
Collapse
|
7
|
Watkins J, Aradi P, Hahn R, Makriyannis A, Mackie K, Katona I, Hohmann AG. CB 1 cannabinoid receptor agonists induce acute respiratory depression in awake mice. Pharmacol Res 2025; 214:107682. [PMID: 40064359 PMCID: PMC12057448 DOI: 10.1016/j.phrs.2025.107682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
Recreational use of synthetic cannabinoid agonists (i.e., "spice compounds") that target the cannabinoid type 1 receptor (CB1) can cause acute respiratory failure in humans. However, Δ9-tetrahydrocannabinol (Δ9-THC), the major psychoactive phytocannabinoid in cannabis, is not traditionally thought to interact with the brain respiratory system, based largely upon sparse labeling of CB1 receptors in the medulla and relative safety suggested by widespread human use. Here we used whole body plethysmography and RNAscope in situ hybridization in mice to reconcile this conflict between conventional wisdom and human data. We examined the respiratory effects of the synthetic CB1 full agonist CP55,940 and Δ9-THC in male and female mice. CP55,940 and Δ9-THC potently and dose-dependently suppressed minute ventilation and tidal volume, decreasing measures of respiratory effort (i.e., peak inspiratory and expiratory flow). Both cannabinoids reduced respiratory frequency, decreasing inspiratory and expiratory time while markedly increasing inspiratory and expiratory pause. Respiratory suppressive effects were fully blocked by the CB1 antagonist AM251, were minimally impacted by the peripherally-restricted CB1 antagonist AM6545, and occurred at doses lower than those that produce cardinal behavioral signs of CB1 activation. Using RNAscope in situ hybridization, we also demonstrated extensive coexpression of Cnr1 (encoding the CB1 receptor) and Oprm1 (encoding the µ-opioid receptor) mRNA in respiratory cells in the medullary pre-Bötzinger complex, a critical nucleus of respiratory control. Our results show that mRNA for CB1 is present in respiratory cells in a medullary brain region essential for breathing and demonstrate that cannabinoids produce respiratory suppression via activation of central CB1 receptors.
Collapse
MESH Headings
- Animals
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/genetics
- Male
- Female
- Respiratory Insufficiency/chemically induced
- Respiratory Insufficiency/metabolism
- Respiratory Insufficiency/physiopathology
- Dronabinol/pharmacology
- Cannabinoid Receptor Agonists/pharmacology
- Mice, Inbred C57BL
- Cyclohexanols/pharmacology
- Mice
- Respiration/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Wakefulness
Collapse
Affiliation(s)
- Joshua Watkins
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN 47405, United States; Program in Neuroscience, Indiana University Bloomington, Bloomington, IN 47405, United States
| | - Petra Aradi
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN 47405, United States
| | - Rachel Hahn
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN 47405, United States
| | | | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN 47405, United States; Program in Neuroscience, Indiana University Bloomington, Bloomington, IN 47405, United States; Gill Institute for Neuroscience, Indiana University, Bloomington, IN 47405, United States
| | - Istvan Katona
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN 47405, United States; Program in Neuroscience, Indiana University Bloomington, Bloomington, IN 47405, United States; Institute of Experimental Medicine, HUN-REN, Budapest, Hungary
| | - Andrea G Hohmann
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN 47405, United States; Program in Neuroscience, Indiana University Bloomington, Bloomington, IN 47405, United States; Gill Institute for Neuroscience, Indiana University, Bloomington, IN 47405, United States.
| |
Collapse
|
8
|
Farbman E, Anis S, Torres-Russotto D. Hope vs Hype III: Should physicians be more open in endorsing cannabis for Parkinson's disease? (PSG Debate 2024). Parkinsonism Relat Disord 2025:107811. [PMID: 40175169 DOI: 10.1016/j.parkreldis.2025.107811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Affiliation(s)
- Eric Farbman
- Community Neurosciences Institute, Fresno, CA, USA
| | - Saar Anis
- Center for Neurological Restoration, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Diego Torres-Russotto
- Baptist Health Miami Neuroscience Institute, Florida International University, FL, USA
| |
Collapse
|
9
|
Valizadeh A, Veenhuis RT, Bradley BA, Xu K. Transcriptomic Alterations Induced by Tetrahydrocannabinol in SIV/HIV Infection: A Systematic Review. Int J Mol Sci 2025; 26:2598. [PMID: 40141240 PMCID: PMC11942185 DOI: 10.3390/ijms26062598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Given the high prevalence of cannabis use among people with HIV (PWH) and its potential to modulate immune responses and reduce inflammation, this systematic review examines preclinical evidence on how tetrahydrocannabinol (THC), a key compound in cannabis, affects gene and micro-RNA expression in simian immunodeficiency virus (SIV)-infected macaques and HIV-infected human cells. Through a comprehensive search, 19 studies were identified, primarily involving SIV-infected macaques, with a pooled sample size of 176, though methodological quality varied across the studies. Pathway analysis of differentially expressed genes (DEGs) and miRNAs associated with THC revealed enrichment in pathways related to inflammation, epithelial cell proliferation, and adhesion. Notably, some DEGs were targets of the differentially expressed miRNAs, suggesting that epigenetic regulation may contribute to THC's effects on gene function. These findings indicate that THC may help mitigate chronic immune activation in HIV infection by altering gene and miRNA expression, suggesting its potential immunomodulatory role. However, the evidence is constrained by small sample sizes and inconsistencies across studies. Further research employing advanced methodologies and larger cohorts is essential to confirm THC's potential as a complementary therapy for PWH and fully elucidate the underlying mechanisms, which could inform targeted interventions to harness its immunomodulatory effects.
Collapse
Affiliation(s)
- Amir Valizadeh
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA; (A.V.); (B.A.B.)
- VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Rebecca T. Veenhuis
- Department of Molecular and Comparative Pathobiology and Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA;
| | - Brooklyn A. Bradley
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA; (A.V.); (B.A.B.)
- VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Ke Xu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA; (A.V.); (B.A.B.)
- VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
10
|
Sescil J, Havens SM, Wang W. Principles and Design of Molecular Tools for Sensing and Perturbing Cell Surface Receptor Activity. Chem Rev 2025; 125:2665-2702. [PMID: 39999110 PMCID: PMC11934152 DOI: 10.1021/acs.chemrev.4c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Cell-surface receptors are vital for controlling numerous cellular processes with their dysregulation being linked to disease states. Therefore, it is necessary to develop tools to study receptors and the signaling pathways they control. This Review broadly describes molecular approaches that enable 1) the visualization of receptors to determine their localization and distribution; 2) sensing receptor activation with permanent readouts as well as readouts in real time; and 3) perturbing receptor activity and mimicking receptor-controlled processes to learn more about these processes. Together, these tools have provided valuable insight into fundamental receptor biology and helped to characterize therapeutics that target receptors.
Collapse
Affiliation(s)
- Jennifer Sescil
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
| | - Steven M. Havens
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
| | - Wenjing Wang
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
- Neuroscience Graduate Program, University of Michigan, Ann
Arbor, MI, 48109
- Program in Chemical Biology, University of Michigan, Ann
Arbor, MI, 48109
| |
Collapse
|
11
|
DeVuono MV, Venkatesan T, Hillard CJ. Endocannabinoid signaling in stress, nausea, and vomiting. Neurogastroenterol Motil 2025; 37:e14911. [PMID: 39223918 PMCID: PMC11872018 DOI: 10.1111/nmo.14911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Classical antiemetics that target the serotonin system may not be effective in treating certain nausea and vomiting conditions like cyclic vomiting syndrome (CVS) and cannabinoid hyperemesis syndrome (CHS). As a result, there is a need for better therapies to manage the symptoms of these disorders, including nausea, vomiting, and anxiety. Cannabis is often used for its purported antiemetic and anxiolytic effects, given regulation of these processes by the endocannabinoid system (ECS). However, there is considerable evidence that cannabinoids can also produce nausea and vomiting and increase anxiety in certain instances, especially at higher doses. This paradoxical effect of cannabinoids on nausea, vomiting, and anxiety may be due to the dysregulation of the ECS, altering how it maintains these processes and contributing to the pathophysiology of CVS or CHS. PURPOSE The purpose of this review is to highlight the involvement of the ECS in the regulation of stress, nausea, and vomiting. We discuss how prolonged cannabis use, such as in the case of CHS or heightened stress, can dysregulate the ECS and affect its modulation of these functions. The review also examines the evidence for the roles of ECS and stress systems' dysfunction in CVS and CHS to better understand the underlying mechanisms of these conditions.
Collapse
Affiliation(s)
- Marieka V. DeVuono
- Department of Anatomy and Cell BiologySchulich School of Medicine & Dentistry, Western UniversityLondonOntarioCanada
| | - Thangam Venkatesan
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal MedicineThe Ohio State University College of MedicineColumbusOhioUSA
| | - Cecilia J. Hillard
- Department of Pharmacology and Toxicology and Neuroscience Research CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
12
|
Pakkhesal S, Shakouri M, Mosaddeghi-Heris R, Kiani Nasab S, Salehi N, Sharafi A, Ahmadalipour A. Bridging the gap: The endocannabinoid system as a functional fulcrum for benzodiazepines in a novel frontier of anxiety pharmacotherapy. Pharmacol Ther 2025; 267:108799. [PMID: 39862927 DOI: 10.1016/j.pharmthera.2025.108799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/27/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
While benzodiazepines have been a mainstay of the pharmacotherapy of anxiety disorders, their short-term efficacy and risk of abuse have driven the exploration of alternative treatment approaches. The endocannabinoid (eCB) system has emerged as a key modulator of anxiety-related processes, with evidence suggesting dynamic interactions between the eCB system and the GABAergic system, the primary target of benzodiazepines. According to the existing literature, the activation of the cannabinoid receptors has been shown to exert anxiolytic effects, while their blockade or genetic deletion results in heightened anxiety-like responses. Moreover, studies have provided evidence of interactions between the eCB system and benzodiazepines in anxiety modulation. For instance, the attenuation of benzodiazepine-induced anxiolysis by cannabinoid receptor antagonism or genetic variations in the eCB system components in animal studies, have been associated with variations in benzodiazepine response and susceptibility to anxiety disorders. The combined use of cannabinoid-based medications, such as cannabinoid receptor agonists and benzodiazepine co-administration, has shown promise in augmenting anxiolytic effects and reducing benzodiazepine dosage requirements. This article aims to comprehensively review and discuss the current evidence on the involvement of the eCB system as a key modulator of benzodiazepine-related anxiolytic effects, and further, the possible mechanisms by which the region-specific eCB system-GABAergic connectivity modulates the neuro-endocrine/behavioral stress response, providing an inclusive understanding of the complex interplay between the eCB system and benzodiazepines in the context of anxiety regulation, to inform future research and clinical practice.
Collapse
Affiliation(s)
- Sina Pakkhesal
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Shakouri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mosaddeghi-Heris
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Kiani Nasab
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Salehi
- Student Research Committee, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - AmirMohammad Sharafi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Ahmadalipour
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| |
Collapse
|
13
|
Gao Q, Asim M. CB 1 receptor signaling: Linking neuroplasticity, neuronal types, and mental health outcomes. Neurochem Int 2025; 184:105938. [PMID: 39904420 DOI: 10.1016/j.neuint.2025.105938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/18/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025]
Abstract
The endocannabinoid system (ECS) is crucial in the pathophysiology of mental disorders. Historically, cannabis has been utilized for centuries to mitigate symptoms of anxiety and depression; however, the precise role of cannabinoids in these conditions has only recently garnered extensive research attention. Despite the growing body of literature on the ECS and its association with mental health, several critical questions remain unresolved. This review primarily focuses on cannabinoid CB1 receptors (CB1R), providing an examination of their regulatory roles in states related to mental disorders. Evidence suggests that CB1R distribution occurs among various neuronal types, astrocytes, and subcellular membranes across multiple brain regions, potentially exhibiting both analogous and antagonistic effects. Additionally, various forms of stress have been shown to produce divergent impacts on CB1R signaling pathways. Furthermore, numerous CB1R agonists demonstrate biphasic, dose-dependent effects on anxiety and depression; specifically, low doses may exert anxiolytic effects, while higher doses can induce anxiogenic responses, a phenomenon observed in both rodent models and human studies. We also discuss the diverse underlying mechanisms that mediate these effects. We anticipate that this review will yield valuable insights into the role of CB1R in mental disorders and provide a framework for future research endeavors on CB1R and the ECS. This knowledge may ultimately inform therapeutic strategies aimed at alleviating symptoms associated with mental health conditions.
Collapse
Affiliation(s)
- Qianqian Gao
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong, 0000, China; Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon Tong, Hong Kong, 0000, China
| | - Muhammad Asim
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong, 0000, China; Research Centre for Treatments of Brain Disorders, City University of Hong Kong, Kowloon Tong, Hong Kong, 0000, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, 0000, China; Current: Department of Psychiatry and Behavioral Science, Stanford University, California, USA.
| |
Collapse
|
14
|
Ballotari M, Truver MT, Sojin NA, Agliano LA, Parimoo R, Hoyer JL, Goodin AJ, Varma DS, Chronister CW, Roussos-Ross K, Goldberger BA. Quantitative analysis of Δ 8- and Δ 9-tetrahydrocannabinol metabolites and isomers: a rapid assay in urine by LC-MS/MS. Anal Bioanal Chem 2025; 417:1507-1518. [PMID: 39847096 DOI: 10.1007/s00216-025-05738-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/20/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025]
Abstract
An increasing number of cannabis-related products have become available and entered the market, particularly those containing cannabidiol (CBD) and Δ8-tetrahydrocannabinol (Δ8-THC). Analytical methods for cannabinoids in urine have been described extensively in the literature. However, methods providing good resolution for distinguishing interferences from THC positional isomers are needed. The aim of this project was to develop and validate a liquid chromatography with tandem mass spectrometry (LC-MS/MS) method to quantitate a broad panel of cannabinoids in authentic urine specimens. The method was optimized to quantitate Δ8-THC and Δ9-THC, 11-OH-Δ8-THC and 11-OH-Δ9-THC, Δ8-THC-COOH and Δ9-THC-COOH, CBD, 7-COOH-CBD, CBG, and CBN, and validated with the guidance of the American Academy of Forensic Sciences Standards Board (ASB) Standard 036. The validated assay was then used to evaluate urine samples collected over various time points from female patients (N = 69) enrolled in a study assessing prevalence of marijuana/CBD use during pregnancy from November 2022 to May 2024. Δ8- and Δ9- isomers were chromatographically resolved and successfully separated. For all analytes, the lower limit of quantitation (LLOQ) was determined to be 10 ng/mL, and the upper limit of quantitation (ULOQ) was 1000 ng/mL. In the authentic samples, the most frequently detected analyte was Δ9-THC-COOH, with a median concentration of 278 ng/mL (n = 38). Δ9-THC and 11-OH-Δ9-THC were detected with a median concentration of 42.4 ng/mL (n = 5) and 65.7 ng/mL (n = 34), respectively. Δ8-THC-COOH was detected in n = 3 specimens, with a median concentration of 25.5 ng/mL. The study provided a rapid assay for the analysis of cannabinoids in urine.
Collapse
Affiliation(s)
- Marco Ballotari
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA.
| | - Michael T Truver
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Nayana A Sojin
- Department of Obstetrics & Gynecology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lauren A Agliano
- Department of Obstetrics & Gynecology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rhea Parimoo
- Department of Obstetrics & Gynecology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jennifer L Hoyer
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Amie J Goodin
- Pharmaceutical Outcomes & Policy, Center for Drug Evaluation and Safety, Consortium for Medical Marijuana Clinical Outcomes Research, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Deepthi S Varma
- Department of Epidemiology, University of Florida College of Public Health and Health Professions & College of Medicine, Gainesville, FL, USA
| | - Chris W Chronister
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Kay Roussos-Ross
- Department of Obstetrics & Gynecology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Bruce A Goldberger
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
15
|
Pavlic M, Innerhofer C, Pitterl F. Quantification of ∆9-tetrahydrocannabinol, 11-OH-THC, THC-COOH, hexahydrocannabinol, and cannabidiol in human plasma and blood by liquid chromatography-tandem mass spectrometry. J Anal Toxicol 2025; 49:85-95. [PMID: 39656878 PMCID: PMC11829072 DOI: 10.1093/jat/bkae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 12/17/2024] Open
Abstract
Ongoing legalization of cannabis for recreational use contributes to increasing numbers not only of incidents of driving under the influence, but within all forensic fields. In addition, newly emerging cannabinoids such as hexahydrocannabinol (HHC) and the increasing use of cannabidiol (CBD) products have to be addressed. The aims of this study were first to extend laboratory analysis capacity for the "established" cannabinoid ∆9-tetrahydrocannabinol (THC) and its metabolites 11-OH-THC and THC-COOH in human plasma/blood, and second to develop analytical procedures concerning HHC and CBD. An LC-MS-MS method based on the available (low-end) instrumentation was used. Samples (250 µl) were prepared by protein precipitation and solid-phase extraction. Chromatographic separation was achieved on a reversed-phase C18 column within 15 min. Detection was performed on a 3200 QTRAP instrument (Sciex) in positive multiple reaction monitoring (MRM) mode. Matrix-matched six-point calibrations were generated applying deuterated internal standards for all analytes except HHC. The method was fully validated according to GTFCh guidelines. Linear ranges were 0.5-25 µg/l for THC, 11-OH-THC, HHC and CBD, and 2.0-100 µg/l for THC-COOH, respectively. Limits of detection and limits of quantification were 0.5 and 1.0 µg/l (THC, 11-OH-THC, HHC, CBD), and 2.0 and 4.0 µg/l (THC-COOH). Applicability of plasma calibrations to blood samples was demonstrated. Acceptance criteria for intra- and inter-day accuracy, precision, extraction efficiency, and matrix effects were met. No interfering signals were detected for 80 exogenous compounds. The presented method is sensitive, specific, easy to handle, and does not require high-end equipment. Since its implementation and accreditation according to ISO 17025, the method has proven to be fit for purpose not only in driving under the influence of drug cases but also within postmortem samples. Furthermore, the design of the method allows for an uncomplicated extension to further cannabinoids if required.
Collapse
Affiliation(s)
- Marion Pavlic
- Institute of Legal Medicine, Medical University of Innsbruck, Muellerstrasse 44, Innsbruck 6020, Austria
| | - Carolin Innerhofer
- Institute of Legal Medicine, Medical University of Innsbruck, Muellerstrasse 44, Innsbruck 6020, Austria
| | - Florian Pitterl
- Institute of Legal Medicine, Medical University of Innsbruck, Muellerstrasse 44, Innsbruck 6020, Austria
| |
Collapse
|
16
|
Aloisio Caruso E, De Nunzio V, Tutino V, Notarnicola M. The Endocannabinoid System: Implications in Gastrointestinal Physiology and Pathology. Int J Mol Sci 2025; 26:1306. [PMID: 39941074 PMCID: PMC11818434 DOI: 10.3390/ijms26031306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025] Open
Abstract
The endocannabinoid system (ECS), composed of receptors, endocannabinoids, and enzymes that regulate biosynthesis and degradation, plays a fundamental role in the physiology and pathology of the gastrointestinal tract, particularly in the small and large intestine and liver. Specifically, cannabinoid receptor type 1 (CB1R) and cannabinoid receptor type 2 (CB2R), located principally in the nervous system and immune cells, orchestrate processes such as intestinal motility, intestinal and hepatic inflammation, and energy metabolism, respectively. The main endocannabinoids, anandamide (AEA) and 2-arachidonoylglycerol (2-AG), influence appetite, body weight regulation, and inflammatory states and thus have implications in obesity, non-alcoholic fatty liver disease (NAFLD) and irritable bowel syndrome (IBS). Recent studies have highlighted the therapeutic potential of targeting the ECS to modulate gastrointestinal and metabolic diseases. In particular, peripheral CB1R antagonists and CB2R agonists have shown efficacy in treating intestinal inflammation, reducing hepatic steatosis, and controlling IBS symptoms. Moreover, the ECS is emerging as a potential target for the treatment of colorectal cancer, acting on cell proliferation and apoptosis. This review highlights the opportunity to exploit the endocannabinoid system in the search for innovative therapeutic strategies, emphasizing the importance of a targeted approach to optimize treatment efficacy and minimize side effects.
Collapse
Affiliation(s)
- Emanuela Aloisio Caruso
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Bari, Italy; (E.A.C.); (V.D.N.)
| | - Valentina De Nunzio
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Bari, Italy; (E.A.C.); (V.D.N.)
| | - Valeria Tutino
- Laboratory of Clinical Pathology, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Bari, Italy;
| | - Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Bari, Italy; (E.A.C.); (V.D.N.)
| |
Collapse
|
17
|
Huang PJ, Son JJ, Arif Y, John JA, Horne LK, Schantell M, Springer SD, Rempe MP, Okelberry HJ, Killanin AD, Glesinger R, Coutant AT, Ward TW, Willett MP, Johnson HJ, Heinrichs-Graham E, Wilson TW. Chronic cannabis use differentially modulates neural oscillations serving the manipulate versus maintain components of working memory processing. Neurobiol Dis 2025; 205:106792. [PMID: 39765275 PMCID: PMC11798582 DOI: 10.1016/j.nbd.2025.106792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
The legalization of recreational cannabis use has expanded the availability of this psychoactive substance in the United States. Research has shown that chronic cannabis use is associated with altered working memory function, however, the brain areas and neural dynamics underlying these affects remain poorly understood. In this study, we leveraged magnetoencephalography (MEG) to investigate neurophysiological activity in 45 participants (22 heavy cannabis users) during a numerical WM task, whereby participants were asked to either maintain or manipulate (i.e., rearrange in ascending order) a group of visually presented numbers. Significant oscillatory responses were imaged using a beamformer and subjected to whole-brain ANOVAs. Notably, we found that cannabis users exhibited significantly weaker alpha oscillations in superior parietal, occipital, and other regions during the encoding phase relative to nonusers. Interestingly, during the maintenance phase, there was a group-by-condition interaction in the right inferior frontal gyrus, left prefrontal, parietal, and other regions, such that cannabis users exhibited weaker alpha and beta oscillations relative to nonusers during maintain trials. Additionally, chronic cannabis users exhibited stronger alpha and beta maintenance responses in these same brain regions and prolonged reaction times during manipulate relative to maintain trials, while no such differences were found in nonusers. Neurobehavioral relationships were also detected in the prefrontal cortices of nonusers, but not cannabis users. In sum, chronic cannabis users exhibit weaker neural oscillations during working memory encoding but may compensate for these deficiencies through stronger oscillatory responses during memory maintenance, especially during strenuous tasks such as manipulating the to-be remembered items.
Collapse
Affiliation(s)
- Peihan J Huang
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA
| | - Jake J Son
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Yasra Arif
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Jason A John
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Lucy K Horne
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Mikki Schantell
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Seth D Springer
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Maggie P Rempe
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Hannah J Okelberry
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Abraham D Killanin
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE, USA
| | - Ryan Glesinger
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Anna T Coutant
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Thomas W Ward
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA
| | - Madelyn P Willett
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Hallie J Johnson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA
| | - Elizabeth Heinrichs-Graham
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA
| | - Tony W Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, USA; Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA; College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE, USA.
| |
Collapse
|
18
|
McDonald K, Schantell M, John J, Coutant A, Glesinger R, Horne L, Okelberry H, Springer S, Embury C, Arif Y, Wilson T. Altered Functional Connectivity Dynamics Serving Cognitive Flexibility in Regular Cannabis Users. Addict Biol 2025; 30:e70023. [PMID: 40042248 PMCID: PMC11881162 DOI: 10.1111/adb.70023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 12/10/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
Despite its widespread use and popularity, cannabis is known to impact higher order cognitive processes such as attention and executive function. However, far less is known about the impact of chronic cannabis use on cognitive flexibility, a component of executive function, and this is especially true for the underlying functional connectivity dynamics. To address this, we enrolled 25 chronic cannabis users and 30 demographically matched non-users who completed an interview probing current and past substance use, a urinalysis to confirm self-reported substance use and a task-switch cognitive paradigm during magnetoencephalography (MEG). Time-frequency windows of interest were identified using a data-driven statistical approach, and spectrally specific neural oscillatory responses were imaged using a beamformer. The resulting maps were grand-averaged across all participants and conditions, and the peak voxels in these maps of neural oscillatory activity were used as seeds to compute connectivity using a whole-brain cortical-coherence approach. Whole-brain neural switch cost connectivity maps were then computed by subtracting the connectivity map for the no-switch condition from that of the switch condition per participant. These switch cost functional connectivity maps were then correlated with the behavioural switch cost per group and probed for group differences in the neuro-behavioural associations. Our behavioural results indicated that all participants had slower reaction times during switch compared to no-switch trials. Regarding the MEG data, cannabis users exhibited altered associations between functional connectivity switch costs and behavioural switch costs along pathways connecting visual cortices and regions in the ventral attention network, within the theta, alpha and gamma frequency ranges. These results indicate modified multispectral associations between functional connectivity and behavioural switch costs among visual cortices and key brain regions underlying executive function in cannabis users.
Collapse
Affiliation(s)
- Kellen M. McDonald
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
- Department of Pharmacology & NeuroscienceCreighton UniversityOmahaNebraskaUSA
| | - Mikki Schantell
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
- College of MedicineUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Jason A. John
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
| | - Anna T. Coutant
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
| | - Ryan Glesinger
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
| | - Lucy K. Horne
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
| | - Hannah J. Okelberry
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
| | - Seth D. Springer
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
- College of MedicineUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Christine M. Embury
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
| | - Yasra Arif
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
| | - Tony W. Wilson
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
- Department of Pharmacology & NeuroscienceCreighton UniversityOmahaNebraskaUSA
- College of MedicineUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| |
Collapse
|
19
|
Niepokny TD, Frey-Burkart H, Mintz EM. Temporal and spatial layout of endocannabinoid system components in the mouse suprachiasmatic nucleus. Neuroscience 2025; 564:179-193. [PMID: 39571963 DOI: 10.1016/j.neuroscience.2024.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/27/2024]
Abstract
Environmental light serves as the main entraining signal for the central circadian pacemaker, the suprachiasmatic nucleus of the hypothalamus (SCN). To shift clock timing with the changing environment, minute adjustments are necessary and the endocannabinoid system (ECS) acts as a neuromodulatory signaling mechanism in the SCN. These systems exert bidirectional effects on one another, still, limited knowledge exists about the role of endocannabinoids in circadian rhythm regulation. Therefore, we investigated the temporal and spatial molecular layouts of the ECS in the SCN of male and female C57BL/6J mice. We utilized laser capture microdissection and quantitative RT-PCR to investigate the ECS temporal layout in the SCN, detected 13 of 19 examined ECS components, and followed up with two 24-hour time course experiments, one under 12:12 light/dark and one under constant dark conditions. All enzymatic machinery related to endocannabinoid synthesis and degradation investigated were found present; however, only cannabinoid receptor 1 (Cnr1) was detected from the 6 ECS related receptors investigated. Cosinor analysis revealed circadian rhythms in many components in both sexes and lighting conditions. Next, we investigated the spatial localization of ECS components in the SCN with RNAscope in situ hybridization. Some genes, such as Cnr1, were more highly expressed in neurons with others, such as Fabp7, were elevated in astrocytes. Cnr1 levels were highest in neurons that do not express the neuropeptides Avp or Vip, and lowest in Vip neurons. Our results support the idea that locally regulated ECS signaling through neuronal CB1 modulates circadian clock function.
Collapse
Affiliation(s)
- Timothy D Niepokny
- School of Biomedical Sciences, Kent State University, 1275 University Esplanade, Kent, OH 44242, USA; Brain Health Research Institute, Kent State University, 1275 University Esplanade, Kent, OH 44242, USA
| | - Hunter Frey-Burkart
- Department of Biological Sciences, Kent State University, 1275 University Esplanade, Kent, OH 44242, USA; Brain Health Research Institute, Kent State University, 1275 University Esplanade, Kent, OH 44242, USA
| | - Eric M Mintz
- Department of Biological Sciences, Kent State University, 1275 University Esplanade, Kent, OH 44242, USA; School of Biomedical Sciences, Kent State University, 1275 University Esplanade, Kent, OH 44242, USA; Brain Health Research Institute, Kent State University, 1275 University Esplanade, Kent, OH 44242, USA.
| |
Collapse
|
20
|
Marcus DJ, English AE, Chun G, Seth EF, Oomen R, Hwang S, Wells B, Piantadosi SC, Suko A, Li Y, Zweifel LS, Land BB, Stella N, Bruchas MR. Endocannabinoids facilitate transitory reward engagement through retrograde gain-control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.630792. [PMID: 39829909 PMCID: PMC11741309 DOI: 10.1101/2025.01.06.630792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Neuromodulatory signaling is poised to serve as a neural mechanism for gain control, acting as a crucial tuning factor to influence neuronal activity by dynamically shaping excitatory and inhibitory fast neurotransmission. The endocannabinoid (eCB) signaling system, the most widely expressed neuromodulatory system in the mammalian brain, is known to filter excitatory and inhibitory inputs through retrograde, pre-synaptic action. However, whether eCBs exert retrograde gain control to ultimately facilitate reward-seeking behaviors in freely moving mammals is not established. Using a suite of in vivo physiological, imaging, genetic and machine learning-based approaches, we report a fundamental role for eCBs in controlling behavioral engagement in reward-seeking behavior through a defined thalamo-striatal circuit.
Collapse
|
21
|
Estudillo E, Castillo-Arellano JI, Martínez E, Rangel-López E, López-Ornelas A, Magaña-Maldonado R, Adalid-Peralta L, Velasco I, Escobedo-Ávila I. Modeling the Effect of Cannabinoid Exposure During Human Neurodevelopment Using Bidimensional and Tridimensional Cultures. Cells 2025; 14:70. [PMID: 39851498 PMCID: PMC11763397 DOI: 10.3390/cells14020070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
Our knowledge about the consumption of cannabinoids during pregnancy lacks consistent evidence to determine whether it compromises neurodevelopment. Addressing this task is challenging and complex since pregnant women display multiple confounding factors that make it difficult to identify the real effect of cannabinoids' consumption. Recent studies shed light on this issue by using pluripotent stem cells of human origin, which can recapitulate human neurodevelopment. These revolutionary platforms allow studying how exogenous cannabinoids could alter human neurodevelopment without ethical concerns and confounding factors. Here, we review the information to date on the clinical studies about the impact of exogenous cannabinoid consumption on human brain development and how exogenous cannabinoids alter nervous system development in humans using cultured pluripotent stem cells as 2D and 3D platforms to recapitulate brain development.
Collapse
Affiliation(s)
- Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Jorge Iván Castillo-Arellano
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Emilio Martínez
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Edgar Rangel-López
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico;
- Hospital Nacional Homeopático, Hospitales Federales de Referencia, Mexico City 06800, Mexico
| | - Roxana Magaña-Maldonado
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Laura Adalid-Peralta
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Iván Velasco
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Itzel Escobedo-Ávila
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
22
|
Patel C, Patel R, Kesharwani A, Rao L, Jain NS. Central cholinergic transmission modulates endocannabinoid-induced marble-burying behavior in mice. Behav Brain Res 2025; 476:115252. [PMID: 39278464 DOI: 10.1016/j.bbr.2024.115252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Central cholinergic system and endocannabinoid, anandamide exhibits anti-compulsive-like behavior in mice. However, the role of the central cholinergic system in the anandamide-induced anti-compulsive-like behavior is still unexplored. Therefore, the present study assessed the role of central cholinergic transmission in the anandamide-induced anti-compulsive activity using a marble-burying behavior (MBB) model in mice. The modulation in the anandamide-induced effect on MBB was evaluated using mice with altered central cholinergic transmission achieved by pretreatment (i.c.v.) with various cholinergic agents like acetylcholine (ACh), acetylcholinesterase inhibitor (AChEI), neostigmine, nicotine, mAChR antagonist, atropine, and nAChR antagonist, mecamylamine. The influence of anandamide treatment on the brain AChE activity was also evaluated. The results revealed that i.c.v. injection of anandamide (10, 20 µg/mouse, i.c.v.) dose-dependently reduced MBB in mice. Moreover, anandamide in all the tested doses inhibited the brain AChE activity indicating the role of an enhanced central cholinergic transmission in its anti-compulsive-like effect . Furthermore, the anti-compulsive-like effect of anandamide (20 µg/mouse, i.c.v.) was found to be enhanced in mice centrally pre-treated with, ACh (0.1 µg/mouse, i.c.v.) or AChEI, neostigmine (0.3 µg/mouse, i.c.v.). In addition, the anandamide-induced anti-compulsive-like effect was significantly increased in mice pre-treated with a low dose of nicotine (0.1 µg/mouse, i.c.v.) while, it was attenuated by the higher dose of nicotine (2 µg/mouse, i.c.v.). On the other hand, the anandamide (20 µg/mouse, i.c.v.) induced anti-compulsive-like effect was found to be diminished in mice pre-treated with mAChR antagonist, atropine (0.1, 0.5 µg/mouse, i.c.v.) and pre-injection of nAChR antagonist, mecamylamine (0.1, 0.5 µg/mouse, i.c.v.) potentiated the anandamide induced anti-compulsive-like response in mice. Thus, the present investigation delineates the modulatory role of an enhanced central cholinergic transmission in the anandamide-induced anti-compulsive-like behavior in mice by inhibition of brain AChE or via muscarinic and nicotinic receptors mediated mechanism.
Collapse
Affiliation(s)
- Chhatrapal Patel
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Richa Patel
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Anuradha Kesharwani
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Laxmi Rao
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Nishant Sudhir Jain
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India.
| |
Collapse
|
23
|
Ajalin R, Al‐Abdulrasul H, Tuisku JM, Hirvonen J, Lahdenpohja S, Rinne JO, Brück A. Impaired Gait, Postural Instability, and Rigidity in Relation to CB1 Receptor Availability in Parkinson's Disease. Mov Disord 2025; 40:163-167. [PMID: 39435606 PMCID: PMC11752992 DOI: 10.1002/mds.30042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND In Parkinson's disease (PD), postural instability and gait disorder (PIGD) symptoms are associated with a worse prognosis for an unknown reason. OBJECTIVE The objective was to explore the relationship between cannabinoid receptor type 1 (CB1R) availability and motor symptoms in PD with [18F]FMPEP-d2 positron emission tomography (PET). METHODS Fifteen individuals with PD underwent [18F]FMPEP-d2 PET to measure cerebral CB1R availability. The Unified Parkinson's Disease Rating Scale motor part (UPDRS-III) was used to evaluate the motor symptoms. RESULTS A negative correlation was observed between [18F]FMPEP-d2 VT and PIGD score (P = 0.002) as well as rigidity subscore (P < 0.001). Both clusters covered widespread areas of both hemispheres. In contrast, tremor or bradykinesia did not correlate to [18F]FMPEP-d2 VT. CONCLUSIONS Gait, postural instability, and rigidity in PD are associated with decreased CB1R availability, unlike tremor or bradykinesia, suggesting that the endocannabinoid system has a role in the pathophysiology of different motor symptoms in PD. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Riikka Ajalin
- Turku PET CentreTurku University and Turku University HospitalTurkuFinland
- Neurocenter, Turku University Hospital and Clinical NeurosciencesUniversity of TurkuTurkuFinland
| | - Haidar Al‐Abdulrasul
- Turku PET CentreTurku University and Turku University HospitalTurkuFinland
- Department of Neurology, Clinical Neurosciences (Neurology), Helsinki University HospitalUniversity of HelsinkiHelsinkiFinland
| | - Jouni M. Tuisku
- Turku PET CentreTurku University and Turku University HospitalTurkuFinland
| | - Jussi Hirvonen
- Turku PET CentreTurku University and Turku University HospitalTurkuFinland
- Department of RadiologyUniversity of Turku and Turku University HospitalTurkuFinland
| | - Salla Lahdenpohja
- Turku PET CentreTurku University and Turku University HospitalTurkuFinland
| | - Juha O. Rinne
- Turku PET CentreTurku University and Turku University HospitalTurkuFinland
- Neurocenter, Turku University Hospital and Clinical NeurosciencesUniversity of TurkuTurkuFinland
| | - Anna Brück
- Turku PET CentreTurku University and Turku University HospitalTurkuFinland
- Neurocenter, Turku University Hospital and Clinical NeurosciencesUniversity of TurkuTurkuFinland
| |
Collapse
|
24
|
Nadler E, Jacobus J, Rabin RA. Prenatal Cannabis and Tobacco Co-Exposure and Its Association with Behavioural Outcomes in Middle Childhood: Co-exposition prénatale au cannabis et au tabac et son association avec les résultats comportementaux au cours de l'enfance intermédiaire. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2025; 70:41-53. [PMID: 39140868 PMCID: PMC11572036 DOI: 10.1177/07067437241271696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
OBJECTIVES Cannabis legalization has triggered an increase in prenatal cannabis use. Given that tobacco is commonly co-used with cannabis, determining outcomes associated with prenatal cannabis and tobacco co-exposure is crucial. While literature exists regarding the individual effects of prenatal cannabis and tobacco exposure on childhood behaviour, there is a gap regarding their combined use, which may have interactive effects. Therefore, we investigated whether prenatal cannabis and tobacco co-exposure was associated with greater externalizing and internalizing problems in middle childhood compared to prenatal exposure to either substance alone or no exposure. METHODS Baseline data from the Adolescent Brain Cognitive Development (ABCD) Study (collected in children ages 9-11) were used to explore differences in externalizing and internalizing scores derived from the Childhood Behavior Checklist across four groups: children with prenatal cannabis and tobacco co-exposure (CT, n = 290), children with prenatal cannabis-only exposure (CAN, n = 225), children with prenatal tobacco-only exposure (TOB, n = 966), and unexposed children (CTL, n = 8,311). We also examined if the daily quantity of tobacco exposure modulated the effect of cannabis exposure on outcomes. RESULTS Adjusting for covariates, a 2 × 2 ANCOVA revealed significant main effects for prenatal cannabis (p = 0.03) and tobacco exposure (p < 0.001), and a significant interaction effect on externalizing scores (p = 0.032); no significant main effects or interactions were found for internalizing scores. However, interactions between daily quantity of cannabis and tobacco exposure significantly predicted both externalizing and internalizing scores (p < 0.01). CONCLUSIONS These findings indicate that co-exposure is associated with greater externalizing problems than exposure to either substance alone, which did not differ from each other. Further, greater tobacco exposure may amplify the negative effect of cannabis exposure on both externalizing and internalizing behaviours in children. These findings underscore the need for interventions that target cannabis and tobacco co-use in pregnant women to circumvent their adverse impact on middle childhood behaviour. PLAIN LANGUAGE SUMMARY TITLE Prenatal Cannabis and Tobacco Co-exposure and its Association with Middle Childhood Behaviours.
Collapse
Affiliation(s)
| | - Joanna Jacobus
- Department of Psychiatry, University of California San Diego, San Diego, California, USA
| | - Rachel A Rabin
- Department of Psychiatry, McGill University and The Douglas Mental Health University Institute, Montreal, Canada
| |
Collapse
|
25
|
Saenz C, Fernandez G, Llovera R, Tolosa MJ, Cantel S, Fehrentz JA, Mackie K, Leggio L, Zigman J, De Francesco PN, Perello M. Growth hormone secretagogue receptor and cannabinoid receptor type 1 intersection in the mouse brain. Brain Struct Funct 2024; 230:15. [PMID: 39702649 PMCID: PMC11659360 DOI: 10.1007/s00429-024-02876-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 12/21/2024]
Abstract
The growth hormone secretagogue receptor (GHSR) and the cannabinoid receptor type 1 (CB1R) are G-protein coupled receptors highly expressed in the brain and involved in critical regulatory processes, such as energy homeostasis, appetite control, reward, and stress responses. GHSR mediates the effects of both ghrelin and liver-expressed antimicrobial peptide 2, while CB1R is targeted by cannabinoids. Strikingly, both receptors mediate their effects by acting on common brain areas and their individual roles have been well characterized. However, the potential for their co-expression in the same neuronal subsets remains largely unexplored. Here, we aim to map the cell populations where GHSR and CB1R might converge, hypothesizing that their co-expression in specific brain circuits could mediate integrated physiological responses. By utilizing two complementary labeling techniques-GHSR-eGFP mice and Fr-ghrelin labeling of GHSR+ cells-along with specific CB1R immunostaining, we sought to visualize and quantify potential areas of overlap. Also, we analyzed several cell RNA sequencing datasets to estimate the fraction of brain cells expressing both GPCRs and their phenotype. Our neuroanatomical studies revealed evident overlap of GHSR+ and CB1R+ signals in specific neuronal subsets mainly located in the cerebral cortex, hippocampus and the amygdala. Transcriptomic analysis revealed specific subsets of Ghsr+/Cnr1+ glutamatergic neurons in the hippocampus and amygdala, as well as different subtypes of Ghsr+/Cnr1+ neurons in the midbrain, hypothalamus, pons, and medulla. Thus, we revealed that GHSR and CB1R interact differentially across specific regions of the mouse brain, providing new insights into how these receptors' actions are integrated. Current findings may open new avenues for dual therapeutic interventions in metabolic disorders, obesity, and psychiatric conditions.
Collapse
Affiliation(s)
- Camila Saenz
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Ramiro Llovera
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - María J Tolosa
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Kenneth Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington Indiana, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Jeffrey Zigman
- Center for Hypothalamic Research and Division of Endocrinology & Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pablo N De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina.
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
26
|
Simei JLQ, Souza JDR, Pedrazzi JF, Guimarães FS, Campos AC, Zuardi A, Hallak JEC, Crippa JAS. Research and Clinical Practice Involving the Use of Cannabis Products, with Emphasis on Cannabidiol: A Narrative Review. Pharmaceuticals (Basel) 2024; 17:1644. [PMID: 39770486 PMCID: PMC11677192 DOI: 10.3390/ph17121644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Emerging evidence supports cannabidiol (CBD) as a promising therapeutic compound for various health conditions, despite its approval as a medication (product for medical purposes) remaining restricted to a limited range of clinical indications. Simultaneously, the regulation of cannabis-derived products for medicinal and recreational use has expanded their global market availability to meet local community demands. This scenario presents a complex challenge for clinicians, researchers, and industry, as the global appeal of therapeutic uses of CBD is growing more rapidly than the scientific evidence supporting its safety and effectiveness. OUTCOMES A narrative review was conducted to discuss the best evidence regarding the pharmacological profile of CBD, its efficacy, and safety within the context of regulation and perspectives on the development of new cannabinoid-based drugs. Key articles addressing the various facets of this issue were selected for comprehensive analysis. CONCLUSIONS Clinicians and researchers may face unique challenges in understanding the pharmacological profile of CBD and the prospects for developing its clinical indications, given the heterogeneity of clinical terminologies and the quality and composition of cannabis-based medical products available on the market. More basic and clinical research that complies with regulatory agencies' testing guidelines, such as good manufacturing practices (GMPs), good laboratory practices (GLPs), and good clinical practices (GCPs), is needed to obtain approval for CBD or any other cannabinoid as a therapeutic for broader clinical indications.
Collapse
Affiliation(s)
- João Luís Q. Simei
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (J.L.Q.S.); (J.D.R.S.); (J.F.P.); (A.Z.); (J.E.C.H.)
| | - José Diogo R. Souza
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (J.L.Q.S.); (J.D.R.S.); (J.F.P.); (A.Z.); (J.E.C.H.)
| | - João Francisco Pedrazzi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (J.L.Q.S.); (J.D.R.S.); (J.F.P.); (A.Z.); (J.E.C.H.)
| | - Francisco S. Guimarães
- National Institute for Science and Technology-Translational Medicine, Ribeirão Preto 14049-900, São Paulo, Brazil;
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil;
| | - Alline Cristina Campos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil;
| | - Antônio Zuardi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (J.L.Q.S.); (J.D.R.S.); (J.F.P.); (A.Z.); (J.E.C.H.)
- National Institute for Science and Technology-Translational Medicine, Ribeirão Preto 14049-900, São Paulo, Brazil;
| | - Jaime Eduardo C. Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (J.L.Q.S.); (J.D.R.S.); (J.F.P.); (A.Z.); (J.E.C.H.)
- National Institute for Science and Technology-Translational Medicine, Ribeirão Preto 14049-900, São Paulo, Brazil;
| | - José Alexandre S. Crippa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil; (J.L.Q.S.); (J.D.R.S.); (J.F.P.); (A.Z.); (J.E.C.H.)
- National Institute for Science and Technology-Translational Medicine, Ribeirão Preto 14049-900, São Paulo, Brazil;
| |
Collapse
|
27
|
Hossain MK, Chae HJ. Medical cannabis: From research breakthroughs to shifting public perceptions and ensuring safe use. Integr Med Res 2024; 13:101094. [PMID: 39640076 PMCID: PMC11617882 DOI: 10.1016/j.imr.2024.101094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 12/07/2024] Open
Abstract
The landscape of medical cannabis has evolved dramatically over the past few decades. Once stigmatized and illegal in most parts of the world, cannabis is now recognized for its potential therapeutic benefits, supported by an expanding body of scientific research. However, the transition from prohibition to medical recognition is shaped by complex interactions among scientific advancements, public perception and regulatory frameworks for its legalization. This review examines the recent breakthroughs in medical cannabis research, explores the shifting public perceptions and the stigma associated with its use and discusses strategies for enhancing the safety of medical cannabis. We also synthesize the connections between scientific research, public perception and safety considerations in the uses of medical cannabis, providing a comprehensive understanding of how these elements influence each other and shape the future of medical cannabis use for patient adherence.
Collapse
Affiliation(s)
| | - Han Jung Chae
- School of Pharmacy, Jeonbuk National University, Jeonju, Republic of Korea
- Korea Medical Cannabis Research Association, Jeonbuk National University, Jeonju, Republic of Korea
- Non-Clinical Evaluation Center (NCEC), Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Republic of Korea
| |
Collapse
|
28
|
Soontornvipart K, Wongsirichatchai P, Phongphuwanan A, Chatdarong K, Vimolmangkang S. Cannabidiol plus krill oil supplementation improves chronic stifle osteoarthritis in dogs: A double-blind randomized controlled trial. Vet J 2024; 308:106227. [PMID: 39179145 DOI: 10.1016/j.tvjl.2024.106227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Osteoarthritis (OA) is the most common orthopedic disorder characterized by chronic inflammation and pain in dogs and cats. Cannabis spp. contains cannabidiol (CBD), a substance with pain relief and anti-inflammatory properties in different animals including dogs with OA. The use of CBD supplements has been increasingly intertwining in veterinary medicine. This study aimed to evaluate the clinical efficacy of CBD + krill oil-supplemented biscuit against canine OA. In total, 30 dogs with stifle OA were randomized and divided into the placebo, krill oil, and CBD + krill oil groups. The Canine Brief Pain Inventory questionnaire was used to evaluate the efficacy of each treatment against pain. Stifle temperature was monitored to identify degrees of stifle inflammation. Two and one dogs in the placebo group were excluded from the study due to worsening lameness and increased pain interference score (PIS) and pain severity score (PSS) at days 14 and 28, respectively. The PIS and PSS scores of the krill oil and CBD + krill oil groups gradually and significantly improved after two weeks of treatment. The CBD + krill oil group had better PIS and PSS scores than the placebo and krill oil groups. However, there was no statistically significant difference in the PIS and PSS scores between the krill oil and CBD + krill oil groups. The stifle temperature of the three groups at different periods did not significantly differ. In conclusion, CBD + krill oil supplements are safe against canine OA. CBD can reduce pain and inflammation.
Collapse
Affiliation(s)
- K Soontornvipart
- Department of Surgery, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - P Wongsirichatchai
- Department of Surgery, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - A Phongphuwanan
- Department of Surgery, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - K Chatdarong
- Department of Obstetrics, Gynaecology, and Reproduction. Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - S Vimolmangkang
- Research Cluster for Cannabis and its Natural Substances, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence in Plant-Produced Pharmaceuticals, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
29
|
Islam J, Rahman MT, Ali M, Kc E, Park YS. Potential hypothalamic mechanisms in trigeminal neuropathic pain: a comparative analysis with migraine and cluster headache. J Headache Pain 2024; 25:205. [PMID: 39587517 PMCID: PMC11587712 DOI: 10.1186/s10194-024-01914-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Trigeminal neuropathic pain (TNP), migraine, and cluster headache (CH) profoundly impact the quality of life and present significant clinical challenges due to their complex neurobiological underpinnings. This review delves into the pivotal role of the hypothalamus in the pathophysiology of these facial pain syndromes, highlighting its distinctive functions and potential as a primary target for research, diagnosis, and therapy. While the involvement of the hypothalamus in migraine and CH has been increasingly supported by imaging and clinical studies, the precise mechanisms of its role remain under active investigation. The role of the hypothalamus in TNP, in contrast, is less explored and represents a critical gap in our understanding. The hypothalamus's involvement varies significantly across these conditions, orchestrating a unique interplay of neural circuits and neurotransmitter systems that underlie the distinct characteristics of each pain type. We have explored advanced neuromodulation techniques, such as deep brain stimulation (DBS) and optogenetics, which show promise in targeting hypothalamic dysfunction to alleviate pain symptoms. Furthermore, we discuss the neuroplastic changes within the hypothalamus that contribute to the chronicity of these pains and the implications of these findings for developing targeted therapies. By offering a comprehensive examination of the hypothalamus's roles, this paper aims to bridge existing knowledge gaps and propel forward the understanding and management of facial neuralgias, underscoring the hypothalamus's critical position in future neurological research.
Collapse
Affiliation(s)
- Jaisan Islam
- Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Md Taufiqur Rahman
- Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Department of Neurosurgery, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Muhammad Ali
- Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Elina Kc
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Young Seok Park
- Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, Cheongju, Republic of Korea.
| |
Collapse
|
30
|
Vera-López KJ, Davila-Del-Carpio G, Nieto-Montesinos R. Macamides as Potential Therapeutic Agents in Neurological Disorders. Neurol Int 2024; 16:1611-1625. [PMID: 39585076 PMCID: PMC11587492 DOI: 10.3390/neurolint16060117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024] Open
Abstract
Therapeutic treatment of nervous system disorders has represented one of the significant challenges in medicine for the past several decades. Technological and medical advances have made it possible to recognize different neurological disorders, which has led to more precise identification of potential therapeutic targets, in turn leading to research into developing drugs aimed at these disorders. In this sense, recent years have seen an increase in exploration of the therapeutic effects of various metabolites extracted from Maca (Lepidium meyenii), a plant native to the central alpine region of Peru. Among the most important secondary metabolites contained in this plant are macamides, molecules derived from N-benzylamides of long-chain fatty acids. Macamides have been proposed as active drugs to treat some neurological disorders. Their excellent human tolerance and low toxicity along with neuroprotective, immune-enhancing, and and antioxidant properties make them ideal for exploration as therapeutic agents. In this review, we have compiled information from various studies on macamides, along with theories about the metabolic pathways on which they act.
Collapse
Affiliation(s)
| | | | - Rita Nieto-Montesinos
- Escuela Profesional de Farmacía y Bioquímica, Universidad Católica de Santa María, Urb. San José s/n—Umacollo, Arequipa 04000, Peru; (K.J.V.-L.); (G.D.-D.-C.)
| |
Collapse
|
31
|
Westerhuis JAW, Dudink J, Wijnands BECA, De Zeeuw CI, Canto CB. Impact of Intrauterine Insults on Fetal and Postnatal Cerebellar Development in Humans and Rodents. Cells 2024; 13:1911. [PMID: 39594658 PMCID: PMC11592629 DOI: 10.3390/cells13221911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Many children suffer from neurodevelopmental aberrations that have long-term effects. To understand the consequences of pathological processes during particular periods in neurodevelopment, one has to understand the differences in the developmental timelines of brain regions. The cerebellum is one of the first brain structures to differentiate during development but one of the last to achieve maturity. This relatively long period of development underscores its vulnerability to detrimental environmental exposures throughout gestation. Moreover, as postnatal functionality of the cerebellum is multifaceted, enveloping sensorimotor, cognitive, and emotional domains, prenatal disruptions in cerebellar development can result in a large variety of neurological and mental health disorders. Here, we review major intrauterine insults that affect cerebellar development in both humans and rodents, ranging from abuse of toxic chemical agents, such as alcohol, nicotine, cannabis, and opioids, to stress, malnutrition, and infections. Understanding these pathological mechanisms in the context of the different stages of cerebellar development in humans and rodents can help us to identify critical and vulnerable periods and thereby prevent the risk of associated prenatal and early postnatal damage that can lead to lifelong neurological and cognitive disabilities. The aim of the review is to raise awareness and to provide information for obstetricians and other healthcare professionals to eventually design strategies for preventing or rescuing related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Judith A. W. Westerhuis
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands; (J.A.W.W.); (C.I.D.Z.)
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children’s Hospital, University Medical Centre Utrecht, 3584 EA Utrecht, The Netherlands; (J.D.); (B.E.C.A.W.)
| | - Bente E. C. A. Wijnands
- Department of Neonatology, Wilhelmina Children’s Hospital, University Medical Centre Utrecht, 3584 EA Utrecht, The Netherlands; (J.D.); (B.E.C.A.W.)
| | - Chris I. De Zeeuw
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands; (J.A.W.W.); (C.I.D.Z.)
- Department of Neuroscience, Erasmus Medical Center, 3015 AA Rotterdam, The Netherlands
| | - Cathrin B. Canto
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands; (J.A.W.W.); (C.I.D.Z.)
- Department of Neuroscience, Erasmus Medical Center, 3015 AA Rotterdam, The Netherlands
| |
Collapse
|
32
|
Chiu DN, Carter BC. Extracellular glutamate is not modulated by cannabinoid receptor activity. Sci Rep 2024; 14:26889. [PMID: 39505963 PMCID: PMC11541540 DOI: 10.1038/s41598-024-75962-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Cannabinoid receptor activation has been proposed to trigger glutamate release from astrocytes located in cortical layer 2/3. Here, we measure the basal concentration of extracellular glutamate in layer 2/3 of mouse somatosensory cortex and find it to be 20-30 nM. We further examine the effect of cannabinoid receptor signaling on extracellular glutamate, and find no evidence for increased extracellular glutamate upon cannabinoid receptor agonist application.
Collapse
Affiliation(s)
- Delia N Chiu
- ENI-G, a Joint Initiative of the University Medical Center Göttingen and the Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Brett C Carter
- ENI-G, a Joint Initiative of the University Medical Center Göttingen and the Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany.
| |
Collapse
|
33
|
Murataeva N, Mattox S, Lemieux J, Griffis J, Yust K, Du W, Heinbockel T, Straiker A. Cannabinoid regulation of sex-dependent murine odorant-stimulated salivation. Sci Rep 2024; 14:26720. [PMID: 39496764 PMCID: PMC11535013 DOI: 10.1038/s41598-024-77761-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/24/2024] [Indexed: 11/06/2024] Open
Abstract
Salivation is easily taken for granted, but without normal salivation, simple essential tasks such as chewing and swallowing become difficult, with consequences for quality of life, nutrition and oral health. Many important drug classes cause dry mouth as a side effect, contributing substantially to patient non-compliance. Available treatments are mostly palliative. Cannabis user complaints of dry mouth prompted a study that showed that basal salivation is likely regulated by cannabinoid CB1 receptors on neurons that innervate the submandibular gland. But what about stimulated salivation? The adjoining parotid gland releases saliva in response to olfactory or other cues and contributes a large portion of the net salivation in humans. We investigated cannabinoid regulation of stimulated salivation using functional and protein-expression studies in mice. In developing a model of stimulated salivary responses to food-related odorants in mice, we noted sex-dependent responses to food-related cues. Only male mice learned to salivate in response to the odor of peanut butter while only female mice responded to a chocolate hazelnut spread. Both males and females responded to sugar or marmite. Testing peanut butter, we found that the cannabinoid receptor agonist CP55940 (0.5 mg/kg, IP) lowered baseline salivation, as shown previously, but also prevented the odorant-induced increase in salivation. CB1 receptors are expressed in axons innervating the parotid gland, paralleling our findings in the submandibular gland. Notably, we also found that CB1 deletion impaired some responses (those to peanut butter and chocolate hazelnut spread) but not others (sugar or marmite). In mice, the CB1 antagonist SR141716 (4 mg/kg, IP) prevented a previously learned salivary response to peanut butter. We find that CB1 receptors are expressed in a subset of glomeruli in coronal sections of olfactory bulb that may serve as a site of action for scent-specific effects of CB1 receptors. We additionally observe CB1 expression in accessory olfactory bulb. In summary, we find a novel sex-dependence in responses to a subset of food-related odorant cues and that cannabinoid receptors regulate some of these responses. We propose that CB1 receptors act at the parotid gland to inhibit stimulated salivation but also in the olfactory system, where functional CB1 receptors are required for salivary responses to specific appetitive odors.
Collapse
Affiliation(s)
- Natalia Murataeva
- Gill Institute for Neuroscience, Indiana University, 1101 E 10th St, Bloomington, IN, 47401, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Sam Mattox
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Josh Lemieux
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - John Griffis
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Kyle Yust
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Wenwen Du
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | | | - Alex Straiker
- Gill Institute for Neuroscience, Indiana University, 1101 E 10th St, Bloomington, IN, 47401, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
34
|
De Felice M, Szkudlarek HJ, Uzuneser TC, Rodríguez-Ruiz M, Sarikahya MH, Pusparajah M, Galindo Lazo JP, Whitehead SN, Yeung KKC, Rushlow WJ, Laviolette SR. The Impacts of Adolescent Cannabinoid Exposure on Striatal Anxiety- and Depressive-Like Pathophysiology Are Prevented by the Antioxidant N-Acetylcysteine. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100361. [PMID: 39257692 PMCID: PMC11381987 DOI: 10.1016/j.bpsgos.2024.100361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024] Open
Abstract
Background Exposure to Δ9-tetrahydrocannabinol (THC) is an established risk factor for later-life neuropsychiatric vulnerability, including mood- and anxiety-related symptoms. The psychotropic effects of THC on affect and anxiogenic behavioral phenomena are known to target the striatal network, particularly the nucleus accumbens, a neural region linked to mood and anxiety disorder pathophysiology. THC may increase neuroinflammatory responses via the redox system and dysregulate inhibitory and excitatory neural balance in various brain circuits, including the striatum. Thus, interventions that can induce antioxidant effects may counteract the neurodevelopmental impacts of THC exposure. Methods In the current study, we used an established preclinical adolescent rat model to examine the impacts of adolescent THC exposure on various behavioral, molecular, and neuronal biomarkers associated with increased mood and anxiety disorder vulnerability. Moreover, we investigated the protective properties of the antioxidant N-acetylcysteine against THC-related pathology. Results We demonstrated that adolescent THC exposure induced long-lasting anxiety- and depressive-like phenotypes concomitant with differential neuronal and molecular abnormalities in the two subregions of the nucleus accumbens, the shell and the core. In addition, we report for the first time that N-acetylcysteine can prevent THC-induced accumbal pathophysiology and associated behavioral abnormalities. Conclusions The preventive effects of this antioxidant intervention highlight the critical role of redox mechanisms underlying cannabinoid-induced neurodevelopmental pathology and identify a potential intervention strategy for the prevention and/or reversal of these pathophysiological sequelae.
Collapse
Affiliation(s)
- Marta De Felice
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Taygun C Uzuneser
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mar Rodríguez-Ruiz
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | | | | | - Shawn N Whitehead
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Ken K-C Yeung
- Department of Chemistry, Western University, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), London, Ontario, Canada
| |
Collapse
|
35
|
Javanshir R, Sedghi M, Esmaeili M, Charsouei S, Anvar LH, Ahmadalipour A. Automatic classification of fatty acid amide hydrolase polymorphism genotype based on EEG signal. Soft comput 2024; 28:12575-12585. [DOI: 10.1007/s00500-024-10306-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 02/18/2025]
|
36
|
Valentino RJ, Volkow ND. Cannabis and Cannabinoid Signaling: Research Gaps and Opportunities. J Pharmacol Exp Ther 2024; 391:154-158. [PMID: 39060161 PMCID: PMC11493439 DOI: 10.1124/jpet.124.002331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024] Open
Abstract
Cannabis and its products have been used for centuries for both medicinal and recreational purposes. The recent widespread legalization of cannabis has vastly expanded its use in the United States across all demographics except for adolescents. Meanwhile, decades of research have advanced our knowledge of cannabis pharmacology and particularly of the endocannabinoid system with which the components of cannabis interact. This research has revealed multiple targets and approaches for manipulating the system for therapeutic use and to ameliorate cannabis toxicity or cannabis use disorder. Research has also led to new questions that underscore the potential risks of its widespread use, particularly the enduring consequences of exposure during critical windows of brain development or for consumption of large daily doses of cannabis with high content Δ 9-tetrahydrocannabinol. This article highlights current neuroscience research on cannabis that has shed light on therapeutic opportunities and potential adverse consequences of misuse and points to gaps in knowledge that can guide future research. SIGNIFICANCE STATEMENT: Cannabis use has escalated with its increased availability. Here, the authors highlight the challenges of cannabis research and the gaps in our knowledge of cannabis pharmacology and of the endocannabinoid system that it targets. Future research that addresses these gaps is needed so that the endocannabinoid system can be leveraged for safe and effective use.
Collapse
Affiliation(s)
| | - Nora D Volkow
- National Institute on Drug Abuse, North Bethesda, Maryland
| |
Collapse
|
37
|
Zheng X, Ehrlich B, Finlay D, Glass M. No Evidence for Endocannabinoid-Induced G Protein Subtype Selectivity at Human and Rodent Cannabinoid CB 1 Receptors. Cannabis Cannabinoid Res 2024. [PMID: 39373143 DOI: 10.1089/can.2024.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024] Open
Abstract
Introduction: The endocannabinoid system (ECS) is a widespread neurotransmitter system. A key characteristic of the ECS is that there are multiple endogenous ligands (endocannabinoids). Of these, the most extensively studied are arachidonoyl ethanolamide (AEA) and 2-arachidonoyl-glycerol (2-AG), both act as agonists at the cannabinoid CB1 receptor. In humans, three CB1 variants have been identified: hCB1, considered the most abundant G protein-coupled receptor in the brain, alongside the less abundant and studied variants, hCB1a and hCB1b. CB1 exhibits a preference for coupling with inhibitory Gi/o proteins, although its interactions with specific members of the Gi/o family remain poorly characterized. This study aimed to compare the AEA and 2-AG-induced activation of various G protein subtypes at CB1. Furthermore, we compared the response of human CB1 (hCB1, hCB1a, hCB1b) and explored species differences by examining rodent receptors (mCB1, rCB1). Materials and Methods: Activation of individual G protein subtypes in HEK293 cells transiently expressing CB1 was measured with G protein dissociation assay utilizing TRUPATH biosensors. The performance of the TRUPATH biosensors was evaluated using Z-factor analysis. Pathway potencies and efficacies were analyzed using the operational analysis of bias to determine G protein subtype selectivity for AEA and 2-AG. Results: Initial screening of TRUPATH biosensors performance revealed variable sensitivities within our system. Based on the biosensor performance, the G protein subtypes pursued for further characterization were Gi1, Gi3, GoA, GoB, GZ, G12, and G13. Across all pathways, AEA demonstrated partial agonism, whereas 2-AG exhibited full or high-efficacy agonism. Notably, we provide direct evidence that the hCB1 receptor couples to G12 and G13 proteins. Our findings do not indicate any evidence of G protein subtype selectivity. Similar observations were made across the human receptor variants (hCB1, hCB1a, hCB1b), as well as at mCB1 and rCB1. Discussion: There was no evidence suggesting G protein subtype selectivity for AEA and 2-AG at CB1, and this finding remained consistent across human receptor variants and different species.
Collapse
Affiliation(s)
- Xiaoxi Zheng
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Beth Ehrlich
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - David Finlay
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Michelle Glass
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
38
|
Guardiola-Ripoll M, Sotero-Moreno A, Chaumette B, Kebir O, Hostalet N, Almodóvar-Payá C, Moreira M, Giralt-López M, Krebs MO, Fatjó-Vilas M. Genetic and Neurodevelopmental Markers in Schizophrenia-Spectrum Disorders: Analysis of the Combined Role of the CNR1 Gene and Dermatoglyphics. Biomedicines 2024; 12:2270. [PMID: 39457583 PMCID: PMC11505170 DOI: 10.3390/biomedicines12102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Dermatoglyphic pattern deviances have been associated with schizophrenia-spectrum disorders (SSD) and are considered neurodevelopment vulnerability markers based on the shared ectodermal origin of the epidermis and the central nervous system. The endocannabinoid system participates in epidermal differentiation, is sensitive to prenatal insults and is associated with SSD. Objective: We aimed to investigate whether the Cannabinoid Receptor 1 gene (CNR1) modulates the dermatoglyphics-SSD association. Methods: In a sample of 112 controls and 97 patients with SSD, three dermatoglyphic markers were assessed: the total palmar a-b ridge count (TABRC), the a-b ridge count fluctuating asymmetry (ABRC-FA), and the pattern intensity index (PII). Two CNR1 polymorphisms were genotyped: rs2023239-T/C and rs806379-A/T. We tested: (i) the CNR1 association with SSD and dermatoglyphic variability within groups; and (ii) the CNR1 × dermatoglyphic measures interaction on SSD susceptibility. Results: Both polymorphisms were associated with SSD. The polymorphism rs2023239 modulated the relationship between PII and SSD: a high PII score was associated with a lower SSD risk within C-allele carriers and a higher SSD risk within TT-homozygotes. This result indicates an inverse relationship between the PII and the SSD predicted probability conditional to the rs2023239 genotype. Conclusions: These novel findings suggest the endocannabinoid system's role in the development and variability of dermatoglyphic patterns. The identified interaction encourages combining genetic and dermatoglyphics to assess neurodevelopmental alterations predisposing to SSD in future studies.
Collapse
Affiliation(s)
- Maria Guardiola-Ripoll
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Spain
- CIBERER (Biomedical Research Network in Rare Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alejandro Sotero-Moreno
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Boris Chaumette
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (INSERM U1266), GHU-Paris Psychiatrie et Neurosciences, 75014 Paris, France
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Oussama Kebir
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (INSERM U1266), GHU-Paris Psychiatrie et Neurosciences, 75014 Paris, France
| | - Noemí Hostalet
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Carmen Almodóvar-Payá
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Mónica Moreira
- Servei de Psiquiatria Infantil i de l’Adolescència, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain
| | - Maria Giralt-López
- Servei de Psiquiatria Infantil i de l’Adolescència, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain
| | - Marie-Odile Krebs
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (INSERM U1266), GHU-Paris Psychiatrie et Neurosciences, 75014 Paris, France
| | - Mar Fatjó-Vilas
- FIDMAG Germanes Hospitalàries Research Foundation, 08830 Sant Boi de Llobregat, Spain
- CIBERSAM (Biomedical Research Network in Mental Health), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
39
|
Lim MJH, Iyyalol R, Lee JWY, Martin-Iverson MT. Multi-modal and bi-directional effects of a synthetic Δ9-Tetrahydrocannabinol (THC) analogue, Nabilone, on spatio-temporal binding windows: Evidence from the projected hand illusion. PLoS One 2024; 19:e0309614. [PMID: 39250476 PMCID: PMC11383222 DOI: 10.1371/journal.pone.0309614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Abnormally widened spatial and temporal binding windows (SBW/TBWs; length of space/time whereby stimuli are considered part of the same percept) are observed in schizophrenia. TBW alterations have been associated with altered sense of agency (hereafter referred to as agency), and an associative relationship between embodiment (body ownership) and agency has been proposed. SBWs/TBWs are investigated separately, but no evidence exists of these being separate in mechanism, system or function. The underlying neural substrate of schizophrenia remains unclear. The literature claims either pro-psychotic or anti-psychotic effects of Δ9-Tetrahydrocannabinol (THC) in patients and healthy individuals, but major support for cannabis in the aetiology of schizophrenia is associative, not causal. To clarify if THC is pro- or anti-psychotic, this single-blind, placebo-controlled within-subjects cross-over study tested several hypotheses. 1) Competing hypotheses that a synthetic THC analogue, Nabilone (NAB, 1-2 mg), would alter measures of agency and embodiment in healthy volunteers (n = 32) similarly, or opposite, to that of in patients with schizophrenia. 2) That there would be significant associations between any NAB-induced alterations in individual agency and embodiment measures in the Projected Hand Illusion (PHI). 3) That there is a unitary spatio-temporal binding window (STBW). A large proportion of individuals did not experience the PHI. Multimodal and bi-directional effects of NAB on the PHI were observed. Evidence of a unitary spatio-temporal binding window (STBW) was observed. NAB widened the STBW in some but narrowed it in others as a function of space and delay. No associations were found between agency and embodiment.
Collapse
Affiliation(s)
- Mark J H Lim
- Pharmacology, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Rajan Iyyalol
- Psychiatry, School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Joseph W Y Lee
- Psychiatry, School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Mathew T Martin-Iverson
- Pharmacology, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
40
|
Meza C, Stefan C, Staines WR, Feinstein A. A preliminary investigation of sex differences in cognitive and fMRI changes following 28 days of cannabis abstinence. Mult Scler Relat Disord 2024; 89:105759. [PMID: 39024968 DOI: 10.1016/j.msard.2024.105759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Previous studies have investigated the influence of cannabis on cognition among people with MS (pwMS), yet the influence of sex in the context of cannabis use remains unexplored. We aim to fill this gap by investigating cannabis-sex related differences in verbal learning, memory and processing speed in association with fMRI (resting state, and task-based) metrics. METHOD Our sample consisted of 19 long-term, frequent cannabis users (8 males, 11 females). Assessments were conducted at baseline and after 28 days of cannabis abstinence. The tests included measures of verbal memory (Selective Reminding Test (SRT)), working memory (n-back), information processing speed (Symbol Digit Modalities Test (SDMT)) and the resting state DMN. To evaluate the effects of cannabis abstinence, we performed a group x time interaction analysis using repeated measures ANCOVA. This analysis controlled for several covariates, including the level of disability (EDSS), baseline cannabis THC metabolite levels, and cannabis withdrawal symptoms. By controlling for these variables, we aimed to isolate the impact of cannabis abstinence on cognitive performance over time. Statistical significance was set at p < 0.05. RESULTS There were no baseline cognitive differences between the sexes. After 28 days of cannabis abstinence, females performed better on the Selective Reminding Test (SRT) (p = 0.04), with a large effect size (η² = 0.286). The mean correct response improved over time for females, but there was no statistically significant group x time interaction on the Symbol Digit Modalities Test (SDMT) and the n-back task. Resting state default mode network data showed overall increased activation in females relative to males at day 28, which meshed with lower brain activation during task-based fMRI paradigms. CONCLUSION Cannabis negated sex-based cognitive differences. Functional MRI task-based paradigms revealed less cerebral activation in females compared to males, which was associated with comparable or better cognitive performance in females, particularly after cannabis abstinence.
Collapse
Affiliation(s)
- Cecilia Meza
- Sunnybrook Research Institute, Division of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Cristiana Stefan
- Clinical Laboratory and Diagnostic Services, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - W Richard Staines
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Anthony Feinstein
- Sunnybrook Research Institute, Division of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Szente L, Balla GY, Varga ZK, Toth B, Biro L, Balogh Z, Hill MN, Toth M, Mikics E, Aliczki M. Endocannabinoid and neuroplasticity-related changes as susceptibility factors in a rat model of posttraumatic stress disorder. Neurobiol Stress 2024; 32:100662. [PMID: 39183773 PMCID: PMC11341941 DOI: 10.1016/j.ynstr.2024.100662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/18/2024] [Accepted: 07/10/2024] [Indexed: 08/27/2024] Open
Abstract
Traumatic experiences result in the development of posttraumatic stress disorder (PTSD) in 10-25% of exposed individuals. While human clinical studies suggest that susceptibility is potentially linked to endocannabinoid (eCB) signaling, neurobiological PTSD susceptibility factors are poorly understood. Employing a rat model of contextual conditioned fear, we characterized distinct resilient and susceptible subpopulations based on lasting generalized fear, a core symptom of PTSD. In these groups, we assessed i.) eCB levels by mass spectrometry and ii.) expression variations of eCB system- and iii.) neuroplasticity-related genes by real-time quantitative PCR in the circuitry relevant in trauma-induced changes. Furthermore, employing unsupervised and semi-supervised machine learning based statistical analytical models, we assessed iv.) gene expression patterns with the most robust predictive power regarding PTSD susceptibility. According to our findings, in our model, generalized fear responses occurred with sufficient variability to characterize distinct resilient and susceptible subpopulations. Resilient subjects showed elevated prelimbic and lower ventral hippocampal levels of eCB 2-arachidonoyl-glycerol (2-AG) compared to resilient and non-shocked control subjects. Ventral hippocampal 2-AG content positively correlated with the strength of fear generalization. Furthermore, susceptibility was associated with i.) prefrontal, hippocampal and amygdalar neuronal hypoactivity, ii.) marked decrease in the expression of genes of transcription factors modulating neuroplasticity and iii.) an altered expression pattern of eCB-related genes, including enzymes involved in eCB metabolism. Unsupervised and semi-supervised statistical approaches highlighted that hippocampal gene expression patterns possess strong predictive power regarding susceptibility. Taken together, the marked eCB and neuroplasticity changes in susceptible individuals associated with abnormal activity patterns in the fear circuitry possibly contribute to context coding deficits, resulting in generalized fear.
Collapse
Affiliation(s)
- Laszlo Szente
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Gyula Y. Balla
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Zoltan K. Varga
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Blanka Toth
- Department of Inorganic and Analytical Chemistry, University of Technology and Economics, Budapest, Hungary
| | - Laszlo Biro
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Zoltan Balogh
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Matthew N. Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mate Toth
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Eva Mikics
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Mano Aliczki
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| |
Collapse
|
42
|
Lissitsa D, Hovers M, Shamuilova M, Ezrapour T, Peled-Avron L. Update on cannabis in human sexuality. Psychopharmacology (Berl) 2024; 241:1721-1730. [PMID: 38977465 PMCID: PMC11339138 DOI: 10.1007/s00213-024-06643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
RATIONALE Sexuality is a central aspect of being human that encompasses many facets. Cannabis, a widely used psychoactive substance, has been associated with various effects on sexuality. The relationship between cannabis and sexuality is complex and multifaceted, involving physiological, psychological, and social factors. OBJECTIVES This review aims to provide an overview of the current literature on the effects of cannabis on several sexual functions, including sexual desire, arousal, orgasm, and sexual satisfaction. It also discusses the potential mechanisms underlying these effects, as well as the impact of dose and frequency of use. RESULTS This review has revealed a complex relationship between cannabis dosage and its influence on sexuality. It appears that the frequency of cannabis use in humans has been associated with the frequency of sexual activities. Individuals who use cannabis more frequently tend to report higher levels of sexual activity. Moreover, there is a notable gender difference in how cannabis affects sexuality. In addition, we found lower doses of cannabis to be linked to heightened sexual desire and enjoyment, whereas higher doses may lead to a decrease in sexual desire and performance. CONCLUSIONS Overall, the association between cannabis and sexuality is complex and warrants further research to better understand the psychological and neurological mechanisms that underlie the effect of cannabis on these sexuality functions and its implications for sexual health. To advance in this endeavor, a crucial step is establishing a precise measurement of dosage in human studies.
Collapse
Affiliation(s)
- Denis Lissitsa
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, 5290002, Ramat Gan, Israel.
- Department of Psychology, Bar-Ilan University, 5290002, Ramat Gan, Israel.
| | - May Hovers
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, 5290002, Ramat Gan, Israel
| | - Michal Shamuilova
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, 5290002, Ramat Gan, Israel
| | - Tal Ezrapour
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, 5290002, Ramat Gan, Israel
| | - Leehe Peled-Avron
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, 5290002, Ramat Gan, Israel
- Department of Psychology, Bar-Ilan University, 5290002, Ramat Gan, Israel
| |
Collapse
|
43
|
Bortolato M, Braccagni G, Pederson CA, Floris G, Fite PJ. "Weeding out" violence? Translational perspectives on the neuropsychobiological links between cannabis and aggression. AGGRESSION AND VIOLENT BEHAVIOR 2024; 78:101948. [PMID: 38828012 PMCID: PMC11141739 DOI: 10.1016/j.avb.2024.101948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Recent shifts in societal attitudes towards cannabis have led to a dramatic increase in consumption rates in many Western countries, particularly among young people. This trend has shed light on a significant link between cannabis use disorder (CUD) and pathological reactive aggression, a condition involving disproportionate aggressive and violent reactions to minor provocations. The discourse on the connection between cannabis use and aggression is frequently enmeshed in political and legal discussions, leading to a polarized understanding of the causative relationship between cannabis use and aggression. However, integrative analyses from both human and animal research indicate a complex, bidirectional interplay between cannabis misuse and pathological aggression. On the one hand, emerging research reveals a shared genetic and environmental predisposition for both cannabis use and aggression, suggesting a common underlying biological mechanism. On the other hand, there is evidence that cannabis consumption can lead to violent behaviors while also being used as a self-medication strategy to mitigate the negative emotions associated with pathological reactive aggression. This suggests that the coexistence of pathological aggression and CUD may result from overlapping vulnerabilities, potentially creating a self-perpetuating cycle where each condition exacerbates the other, escalating into externalizing and violent behaviors. This article aims to synthesize existing research on the intricate connections between these issues and propose a theoretical model to explain the neurobiological mechanisms underpinning this complex relationship.
Collapse
Affiliation(s)
- Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
- Consortium for Translational Research on Aggression and Drug Abuse (ConTRADA), University of Kansas, Lawrence, KS, USA
| | - Giulia Braccagni
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Casey A. Pederson
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gabriele Floris
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
- Center for Substance Abuse Research, Temple University, Philadelphia, PA, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, USA
| | - Paula J. Fite
- Consortium for Translational Research on Aggression and Drug Abuse (ConTRADA), University of Kansas, Lawrence, KS, USA
- Clinical Child Psychology Program, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
44
|
McFadden MH, Emeritt MB, Xu H, Cui Y, Leterrier C, Zala D, Venance L, Lenkei Z. Actomyosin-mediated inhibition of synaptic vesicle release under CB 1R activation. Transl Psychiatry 2024; 14:335. [PMID: 39168993 PMCID: PMC11339458 DOI: 10.1038/s41398-024-03017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/16/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Long-term synaptic plasticity is critical for adaptive function of the brain, but presynaptic mechanisms of functional plasticity remain poorly understood. Here, we show that changes in synaptic efficacy induced by activation of the cannabinoid type-1 receptor (CB1R), one of the most widespread G-protein coupled receptors in the brain, requires contractility of the neuronal actomyosin cytoskeleton. Specifically, using a synaptophysin-pHluorin probe (sypH2), we show that inhibitors of non-muscle myosin II (NMII) ATPase as well as one of its upstream effectors Rho-associated kinase (ROCK) prevent the reduction of synaptic vesicle release induced by CB1R activation. Using 3D STORM super-resolution microscopy, we find that activation of CB1R induces a redistribution of synaptic vesicles within presynaptic boutons in an actomyosin dependent manner, leading to vesicle clustering within the bouton and depletion of synaptic vesicles from the active zone. We further show, using sypH2, that inhibitors of NMII and ROCK specifically restore the release of the readily releasable pool of synaptic vesicles from the inhibition induced by CB1R activation. Finally, using slice electrophysiology, we find that activation of both NMII and ROCK is necessary for the long-term, but not the short-term, form of CB1R induced synaptic plasticity at excitatory cortico-striatal synapses. We thus propose a novel mechanism underlying CB1R-induced plasticity, whereby CB1R activation leads to a contraction of the actomyosin cytoskeleton inducing a reorganization of the functional presynaptic vesicle pool, preventing vesicle release and inducing long-term depression.
Collapse
Affiliation(s)
- Maureen H McFadden
- Institut Pasteur, Université Paris Cité, Synapse and Circuit Dynamics Laboratory, CNRS UMR 3571, Paris, France
- Brain Plasticity Unit, ESPCI Paris, PSL Research University, CNRS, Paris, France
| | - Michel-Boris Emeritt
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, Paris, France
| | - Hao Xu
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Yihui Cui
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | | | - Diana Zala
- Brain Plasticity Unit, ESPCI Paris, PSL Research University, CNRS, Paris, France
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, Paris, France
| | - Laurent Venance
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Zsolt Lenkei
- Brain Plasticity Unit, ESPCI Paris, PSL Research University, CNRS, Paris, France.
- Institute of Psychiatry and Neuroscience of Paris (IPNP), Université Paris Cité, INSERM U1266, Paris, France.
| |
Collapse
|
45
|
Wen J, Tanaka M, Zhang Y. Inhibition of 2-AG hydrolysis alleviates posttraumatic headache attributed to mild traumatic brain injury. J Headache Pain 2024; 25:115. [PMID: 39014318 PMCID: PMC11253377 DOI: 10.1186/s10194-024-01817-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Posttraumatic headache (PTH) is a common and debilitating symptom following repetitive mild traumatic brain injury (rmTBI), and it mainly resembles a migraine-like phenotype. While modulation of the endocannabinoid system (ECS) is effective in treating TBI and various types of pain including migraine, the role of augmentation of endocannabinoids in treating PTH has not been investigated. METHODS Repetitive mild TBI was induced in male C57BL/6J mice using the non-invasive close-head impact model of engineered rotational acceleration (CHIMERA). Periorbital allodynia was assessed using von Frey filaments and determined by the "Up-Down" method. Immunofluorescence staining was employed to investigate glial cell activation and calcitonin gene-related peptide (CGRP) expression in the trigeminal ganglion (TG) and trigeminal nucleus caudalis (TNC) of the rmTBI mice. Levels of 2-arachidonoyl glycerol (2-AG), anandamide (AEA), and arachidonic acid (AA) in the TG, medulla (including TNC), and periaqueductal gray (PAG) were measured by mass spectrometry. The therapeutic effect of endocannabinoid modulation on PTH was also assessed. RESULTS The rmTBI mice exhibited significantly increased cephalic pain hypersensitivity compared to the sham controls. MJN110, a potent and selective inhibitor of the 2-AG hydrolytic enzyme monoacylglycerol lipase (MAGL), dose-dependently attenuated periorbital allodynia in the rmTBI animals. Administration of CGRP at 0.01 mg/kg reinstated periorbital allodynia in the rmTBI animals on days 33 and 45 post-injury but had no effect in the sham and MJN110 treatment groups. Activation of glial cells along with increased production of CGRP in the TG and TNC at 7 and 14 days post-rmTBI were attenuated by MJN110 treatment. The anti-inflammatory and anti-nociceptive effects of MJN110 were partially mediated by cannabinoid receptor activation, and the pain-suppressive effect of MJN110 was completely blocked by co-administration of DO34, an inhibitor of 2-AG synthase. The levels of 2-AG in TG, TNC and PAG were decreased in TBI animals, significantly elevated and further reduced by the selective inhibitors of 2-AG hydrolytic and synthetic enzymes, respectively. CONCLUSION Enhancing endogenous levels of 2-AG appears to be an effective strategy for the treatment of PTH by attenuating pain initiation and transmission in the trigeminal pathway and facilitating descending pain inhibitory modulation.
Collapse
Affiliation(s)
- Jie Wen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
46
|
Göntér K, Dombi Á, Kormos V, Pintér E, Pozsgai G. Examination of the Effect of Dimethyl Trisulfide in Acute Stress Mouse Model with the Potential Involvement of the TRPA1 Ion Channel. Int J Mol Sci 2024; 25:7701. [PMID: 39062944 PMCID: PMC11277546 DOI: 10.3390/ijms25147701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Polysulfides are endogenously produced in mammals and generally associated with protective functions. Our aim was to investigate the effect of dimethyl trisulfide (DMTS) in a mouse model of acute stress. DMTS activates transient receptor potential ankyrin 1 (TRPA1) channels and leads to neuropeptide release, potentially that of substance P (SP). We hypothesize that DMTS might inhibit the degrading enzymes of endocannabinoids, so this system was also investigated as another possible pathway for mediating the effects of DMTS. Trpa1 gene wild-type (WT) and knockout (KO) mice were used to confirm the role of the TRPA1 ion channel in mediating the effects of DMTS. C57BL/6J, NK1 gene KO, and Tac1 gene KO mice were used to evaluate the effect of DMTS on the release and expression of SP. Some C57BL/6J animals were treated with AM251, an inhibitor of the cannabinoid CB1 receptor, to elucidate the role of the endocannabinoid system in these processes. Open field test (OFT) and forced swim test (FST) were performed in each mouse strain. A tail suspension test (TST) was performed in Trpa1 WT and KO animals. C-FOS immunohistochemistry was carried out on Trpa1 WT and KO animals. The DMTS treatment increased the number of highly active periods and decreased immobility time in the FST in WT animals, but had no effect on the Trpa1 KO mice. The DMTS administration induced neuronal activation in the Trpa1 WT mice in the stress-related brain areas, such as the locus coeruleus, dorsal raphe nucleus, lateral septum, paraventricular nucleus of the thalamus, and paraventricular nucleus of the hypothalamus. DMTS may have a potential role in the regulation of stress-related processes, and the TRPA1 ion channel may also be involved in mediating the effects of DMTS. DMTS can be an ideal candidate for further study as a potential remedy for stress-related disorders.
Collapse
Affiliation(s)
- Kitti Göntér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (K.G.); (V.K.); (E.P.)
| | - Ágnes Dombi
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary;
| | - Viktória Kormos
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (K.G.); (V.K.); (E.P.)
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, H-7624 Pécs, Hungary; (K.G.); (V.K.); (E.P.)
| | - Gábor Pozsgai
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary;
| |
Collapse
|
47
|
Chesworth R, Yim HCH, Watt G, El-Omar E, Karl T. Cannabidiol (CBD) facilitates cocaine extinction and ameliorates cocaine-induced changes to the gut microbiome in male C57BL/6JArc mice. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111014. [PMID: 38649130 DOI: 10.1016/j.pnpbp.2024.111014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Cocaine use disorder (CUD) is a global health problem with no approved medications. One potential treatment target is the gut microbiome, but it is unknown if cocaine induces long-lasting effects on gut microbes. A novel therapeutic candidate for CUD, cannabidiol (CBD), can improve gut function in rodent models. It is possible that protective effects of CBD against cocaine use are mediated by improving gut health. We examined this question in this experiment. Cocaine conditioned place preference (CPP) was conducted in adult male C57BL/6JArc mice. Mice were treated with vehicle or 20 mg/kg CBD prior to all cocaine CPP sessions (N = 11-13/group). Mice were tested drug free 1, 14 and 28 days after cessation of cocaine and CBD treatment. Fecal samples were collected prior to drug treatment and after each test session. Gut microbiome analyses were conducted using 16 s rRNA sequencing and correlated with behavioural parameters. We found a persistent preference for a cocaine-environment in mice, and long-lasting changes to gut microbe alpha diversity. Cocaine caused persistent changes to beta diversity which lasted for 4 weeks. CBD treatment reduced cocaine-environment preference during abstinence from cocaine and returned gut beta diversity measures to control levels. CBD treatment increased the relative abundance of Firmicutes phyla and Oscillospira genus, but decreased Bacteroidetes phyla and Bacteroides acidifaciens species. Preference score in cocaine-treated mice was positively correlated with abundance of Actinobacteria, whereas in mice treated with CBD and cocaine, the preference score was negatively correlated with Tenericutes abundance. Here we show that CBD facilitates cocaine extinction memory and reverses persistent cocaine-induced changes to gut microbe diversity. Furthermore, CBD increases the abundance of gut microbes which have anti-inflammatory properties. This suggests that CBD may act via the gut to reduce the memory of cocaine reward. Our data suggest that improving gut health and using CBD could limit cocaine abuse.
Collapse
Affiliation(s)
- Rose Chesworth
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.
| | - Howard Chi-Ho Yim
- St George and Sutherland Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW, Sydney, Australia; UNSW Microbiome Research Centre, St George and Sutherland Clinical School, UNSW, Sydney, Australia; Department of Gastroenterology and Hepatology, St George Hospital, Sydney, Australia
| | - Georgia Watt
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Emad El-Omar
- St George and Sutherland Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW, Sydney, Australia; UNSW Microbiome Research Centre, St George and Sutherland Clinical School, UNSW, Sydney, Australia; Department of Gastroenterology and Hepatology, St George Hospital, Sydney, Australia
| | - Tim Karl
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
48
|
Grether U, Foxton RH, Gruener S, Korn C, Kimbara A, Osterwald A, Zirwes E, Uhles S, Thoele J, Colé N, Rogers-Evans M, Röver S, Nettekoven M, Martin RE, Adam JM, Fingerle J, Bissantz C, Guba W, Alker A, Szczesniak AM, Porter RF, Toguri TJ, Revelant F, Poirier A, Perret C, Winther L, Caruso A, Fezza F, Maccarrone M, Kelly MEM, Fauser S, Ullmer C. RG7774 (Vicasinabin), an orally bioavailable cannabinoid receptor 2 (CB2R) agonist, decreases retinal vascular permeability, leukocyte adhesion, and ocular inflammation in animal models. Front Pharmacol 2024; 15:1426446. [PMID: 39070793 PMCID: PMC11272598 DOI: 10.3389/fphar.2024.1426446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/19/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Preclinical studies suggest that cannabinoid receptor type 2 (CB2R) activation has a therapeutic effect in animal models on chronic inflammation and vascular permeability, which are key pathological features of diabetic retinopathy (DR). A novel CB2R agonist, triazolopyrimidine RG7774, was generated through lead optimization of a high-throughput screening hit. The aim of this study was to characterize the pharmacology, absorption, distribution, metabolism, elimination, and toxicity (ADMET) profile of RG7774, and to explore its potential for managing the key pathological features associated with retinal disease in rodents. Methods The in vitro pharmacology of RG7774 was investigated for CB2R binding and receptor activation using recombinant human and mouse CB2R expression in Chinese hamster ovary cells, and endogenous CB2R expression in human Jurkat cells, and rat and mouse spleen cells. The ADMET profile was evaluated and the effects of RG7774 on retinal permeability, leukocyte adhesion, and choroidal neovascularization (CNV) were investigated in rodent models of retinal disease. Pharmacokinetic (PK) parameters and the exposure-response relationship were characterized in healthy animals and in animals with laser-induced CNV. Results RG7774 was found to be a potent (EC50: 2.8 nM and Ki: 51.3 nM), selective, and full CB2R agonist with no signs of cannabinoid receptor type 1 (CB1R) binding or activation. The ligand showed a favorable ADMET profile and exhibited systemic and ocular exposure after oral delivery. Functional potency in vitro translated from recombinant to endogenous expression systems. In vivo, orally administered RG7774 reduced retinal permeability and leukocyte adhesion in rodents with lipopolysaccharide (LPS)-induced uveitis and streptozotocin (STZ)-induced DR, and reduced lesion areas in rats with laser-induced CNV with an ED50 of 0.32 mg/kg. Anatomically, RG7774 reduced the migration of retinal microglia to retinal lesions. Discussion RG7774 is a novel, highly selective, and orally bioavailable CB2R agonist, with an acceptable systemic and ocular PK profile, and beneficial effects on retinal vascular permeability, leukocyte adhesion, and ocular inflammation in rodent animal models. Results support the development of RG7774 as a potential treatment for retinal diseases with similar pathophysiologies as addressed by the animal models.
Collapse
Affiliation(s)
- Uwe Grether
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Richard H. Foxton
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Sabine Gruener
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Claudia Korn
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Atsushi Kimbara
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Anja Osterwald
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Elisabeth Zirwes
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Sabine Uhles
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Janina Thoele
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Nadine Colé
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Mark Rogers-Evans
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Stephan Röver
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Matthias Nettekoven
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Rainer E. Martin
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Jean-Michel Adam
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Jürgen Fingerle
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Caterina Bissantz
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Wolfgang Guba
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - André Alker
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Anna M. Szczesniak
- Departments of Pharmacology, Anesthesia, Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Ross F. Porter
- Departments of Pharmacology, Anesthesia, Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Tom J. Toguri
- Departments of Pharmacology, Anesthesia, Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Franco Revelant
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Agnès Poirier
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Camille Perret
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Lotte Winther
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Antonello Caruso
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Filomena Fezza
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
- European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
| | - Melanie E. M. Kelly
- Departments of Pharmacology, Anesthesia, Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Sascha Fauser
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Christoph Ullmer
- F. Hoffmann-La Roche Ltd, Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
49
|
Simei JLQ, de Souza JDR, Lisboa JR, Guimarães FS, Crippa JADS. Cannabidiol in anxiety disorders: Current and future perspectives. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 177:205-234. [PMID: 39029985 DOI: 10.1016/bs.irn.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
Anxiety disorders are highly prevalent psychiatric disorders, characterized by a chronic course and often accompanied by comorbid symptoms that impair functionality and decrease quality of life. Despite advances in basic and clinical research in our understanding of these disorders, currently available pharmacological options are associated with limited clinical benefits and side effects that frequently lead to treatment discontinuation. Importantly, a significant number of patients do not achieve remission and live with lifelong residual symptoms that limit daily functioning. Since the 1970s, basic and clinical research on cannabidiol (CBD), a non-psychotomimetic compound found in the Cannabis sativa plant, has indicated relevant anxiolytic effects, garnering attention for its therapeutic potential as an option in anxiety disorder treatment. This chapter aims to review the history of these studies on the anxiolytic effects of CBD within the current understanding of anxiety disorders. It highlights the most compelling current evidence supporting its anxiolytic effects and explores future perspectives for its clinical use in anxiety disorders.
Collapse
Affiliation(s)
- João Luís Queiroz Simei
- Department of Neuroscience and Behavior Sciences, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - José Diogo Ribeiro de Souza
- Department of Neuroscience and Behavior Sciences, Ribeirão Preto Medical School, University of São Paulo, Brazil.
| | - João Roberto Lisboa
- Department of Neuroscience and Behavior Sciences, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Francisco Silveira Guimarães
- National Institute for Science and Technology, Translational Medicine, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - José Alexandre de Souza Crippa
- Department of Neuroscience and Behavior Sciences, Ribeirão Preto Medical School, University of São Paulo, Brazil; National Institute for Science and Technology, Translational Medicine, Brazil
| |
Collapse
|
50
|
Macedo I, Paiva TO, Pasion R, Daedelow L, Heinz A, Magalhães A, Banaschewski T, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Orfanos DP, Paus T, Poustka L, Hohmann S, Holz N, Fröhner JH, Smolka MN, Vaidya N, Walter H, Whelan R, Schumann G, Barbosa F. Light Cannabis Use and the Adolescent Brain: An 8-years Longitudinal Assessment of Mental Health, Cognition, and Reward Processing. Psychopharmacology (Berl) 2024; 241:1447-1461. [PMID: 38532040 PMCID: PMC11199211 DOI: 10.1007/s00213-024-06575-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024]
Abstract
RATIONALE For decades, cannabis has been the most widely used illicit substance in the world, particularly among youth. Research suggests that mental health problems associated with cannabis use may result from its effect on reward brain circuit, emotional processes, and cognition. However, findings are mostly derived from correlational studies and inconsistent, particularly in adolescents. OBJECTIVES AND METHODS Using data from the IMAGEN study, participants (non-users, persistent users, abstinent users) were classified according to their cannabis use at 19 and 22 years-old. All participants were cannabis-naïve at baseline (14 years-old). Psychopathological symptoms, cognitive performance, and brain activity while performing a Monetary Incentive Delay task were used as predictors of substance use and to analyze group differences over time. RESULTS Higher scores on conduct problems and lower on peer problems at 14 years-old (n = 318) predicted a greater likelihood of transitioning to cannabis use within 5 years. At 19 years of age, individuals who consistently engaged in low-frequency (i.e., light) cannabis use (n = 57) exhibited greater conduct problems and hyperactivity/inattention symptoms compared to non-users (n = 52) but did not differ in emotional symptoms, cognitive functioning, or brain activity during the MID task. At 22 years, those who used cannabis at both 19 and 22 years-old n = 17), but not individuals that had been abstinent for ≥ 1 month (n = 19), reported higher conduct problems than non-users (n = 17). CONCLUSIONS Impairments in reward-related brain activity and cognitive functioning do not appear to precede or succeed cannabis use (i.e., weekly, or monthly use). Cannabis-naïve adolescents with conduct problems and more socially engaged with their peers may be at a greater risk for lighter yet persistent cannabis use in the future.
Collapse
Affiliation(s)
- Inês Macedo
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Educational Sciences (Laboratory of Neuropsychophysiology), University of Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal.
- Addiction Biology Group, i3S-Instituto de Investigação E Inovação Em Saúde, Porto, Portugal.
| | | | - Rita Pasion
- HEI-LAB, Lusófona University, Porto, Portugal
| | - Laura Daedelow
- Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin, Institute of Health, Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin, Institute of Health, Berlin, Germany
| | - Ana Magalhães
- Addiction Biology Group, i3S-Instituto de Investigação E Inovação Em Saúde, Porto, Portugal
- Instituto de Biologia Molecular E Celular (IBMC), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, 68131, Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, 91191, Gif-Sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT, 05405, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, UK
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt, Braunschweig und Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de La Santé Et de La Recherche Médicale, INSERM U 1299 Trajectoires Développementales & Psychiatrie, CNRS; EcoleNormaleSupérieure Paris-Saclay, Centre Borelli, University Paris-Saclay, Gif-Sur-Yvette, France
| | - Marie-Laure Paillère Martinot
- Institut National de La Santé Et de La Recherche Médicale, INSERM U 1299 Trajectoires Développementales & Psychiatrie, University Paris-Saclay, CNRS; Ecole Normale Supérieure Paris-Saclay, Centre Borelli; Gif-Sur-Yvette, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, and AP-HP. Sorbonne University, Paris, France
| | - Eric Artiges
- Institut National de La Santé Et de La Recherche Médicale, INSERM U 1299 Trajectoires Développementales & Psychiatrie, CNRS; EcoleNormaleSupérieure Paris-Saclay, Centre Borelli; Gif-Sur-Yvette; and Psychiatry Department, EPS Barthélémy Durand, University Paris-Saclay, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany
| | | | - Tomáš Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hosptalier, Universitaire Sainte-Justine, University of Montreal, Montreal, QC, Canada
- Departments of Psychiatry and Psychology, University of Toronto, Toronto, ON, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry Psychotherapy and Psychosomatics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Holz
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159, Mannheim, Germany
| | - Juliane H Fröhner
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Neuroimaging Center, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Neuroscience, Charité Universitätsmedizin, Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin, Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Neuroscience, Charité Universitätsmedizin, Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology of Brain-Inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | - Fernando Barbosa
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Educational Sciences (Laboratory of Neuropsychophysiology), University of Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| |
Collapse
|