1
|
Oubraim S, Fauzan M, Studholme K, Gordon C, Glaser ST, Shen RY, Ojima I, Kaczocha M, Haj-Dahmane S. Astrocytic FABP5 mediates retrograde endocannabinoid transport at central synapses. iScience 2025; 28:112342. [PMID: 40292318 PMCID: PMC12033926 DOI: 10.1016/j.isci.2025.112342] [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: 12/11/2024] [Revised: 02/19/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Endocannabinoids (eCBs) regulate synaptic function via cannabinoid receptors. While eCB signaling is well understood, the mechanisms underlying eCB synaptic transport are poorly characterized. Using 2-arachidonoylglycerol (2-AG)-mediated depolarization-induced suppression of inhibition (DSI) in the hippocampus as a readout of retrograde eCB signaling, we demonstrate that the deletion of fatty acid binding protein 5 (FABP5) impairs DSI. In FABP5 KO mice, DSI was rescued by re-expressing wild-type FABP5 but not an FABP5 mutant that does not bind 2-AG. Importantly, the deletion of astrocytic FABP5 blunted DSI, which was rescued by its re-expression in the astrocytes of FABP5 KO mice. Neuronal FABP5 was dispensable for 2-AG signaling. DSI was also rescued by expressing a secreted FABP5 variant but not by FABP7, an astrocytic FABP that does not undergo secretion. Our results demonstrate that extracellular FABP5 of astrocytic origin controls 2-AG transport and that FABP5 is adapted to coordinate intracellular and synaptic eCB transport.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Mohammad Fauzan
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Keith Studholme
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Chris Gordon
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Sherrye T. Glaser
- Department of Biological Sciences, Kingsborough Community College, Brooklyn, NY, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
- Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
2
|
Malhotra S, Donneger F, Farrell JS, Dudok B, Losonczy A, Soltesz I. Integrating endocannabinoid signaling, CCK interneurons, and hippocampal circuit dynamics in behaving animals. Neuron 2025:S0896-6273(25)00188-6. [PMID: 40267911 DOI: 10.1016/j.neuron.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
The brain's endocannabinoid signaling system modulates a diverse range of physiological phenomena and is also involved in various psychiatric and neurological disorders. The basic components of the molecular machinery underlying endocannabinoid-mediated synaptic signaling have been known for decades. However, limitations associated with the short-lived nature of endocannabinoid lipid signals had made it challenging to determine the spatiotemporal specificity and dynamics of endocannabinoid signaling in vivo. Here, we discuss how novel technologies have recently enabled unprecedented insights into endocannabinoid signaling taking place at specific synapses in behaving animals. In this review, we primarily focus on cannabinoid-sensitive inhibition in the hippocampus in relation to place cell properties to illustrate the potential of these novel methodologies. In addition, we highlight implications of these approaches and insights for the unraveling of cannabinoid regulation of synapses in vivo in other brain circuits in both health and disease.
Collapse
Affiliation(s)
- Shreya Malhotra
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
| | - Florian Donneger
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Barna Dudok
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
3
|
Elliott GO, Petrie GN, Kroll SL, Roche DJO, Mayo LM. Changes in peripheral endocannabinoid levels in substance use disorders: a review of clinical evidence. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2025; 51:152-164. [PMID: 40197861 DOI: 10.1080/00952990.2025.2456499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/31/2024] [Accepted: 01/16/2025] [Indexed: 04/10/2025]
Abstract
Background: The endocannabinoid (eCB) system is a key modulator of stress and reward and is impacted by alcohol and drug use. Recently, the eCB system has been highlighted as a potential novel target in the treatment of substance use disorders (SUDs).Objectives: Understanding how chronic substance use impacts the function of the eCB system can provide a mechanistic rationale for targeting this system in the treatment of SUDs.Methods: A comprehensive review of studies assessing concentrations of eCB ligands N-arachidonoyl ethanolamine (anandamide; AEA) and 2-arachidonoylglycerol (2-AG) in individuals with a SUD diagnosis was performed using all EBSCO databases, PubMed, and Google Scholar. Methods and results related to eCB concentrations, diagnosis, and other factors (e.g. treatment status) were extracted from papers written in English and published in peer-reviewed journals before May 22, 2024.Results: Fifteen studies were reviewed; three in alcohol use disorder (AUD), three in cannabis use disorder (CUD), four in cocaine use disorder, one in opioid use disorder (OUD) and four across SUDs. Generally, AEA concentrations were usually, but not always, increased in AUD, CUD, OUD, and cocaine use disorder. 2-AG concentrations were measured less often but were increased in CUD and decreased in cocaine use disorder.Conclusions: Studies generally support the hypothesis that chronic substance use can impact eCB levels, most often with increased AEA and decreased (or not quantified) 2-AG concentrations, though results were often conflicting. Variability in methodology and study design may limit generalizability across studies.
Collapse
Affiliation(s)
- Georgia O Elliott
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
| | - Gavin N Petrie
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
| | - Sara L Kroll
- University Hospital of Psychiatry, Adult Psychiatry and Psychotherapy, University of Zurich, Zurich, Switzerland
| | - Daniel J O Roche
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland Baltimore, Baltimore, MD, USA
| | - Leah M Mayo
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
| |
Collapse
|
4
|
Chong B, Kumar V, Nguyen D, Hopkins M, Ferry F, Spera L, Paul E, Hutson A, Tabuchi M. Neuropeptide-Dependent Spike Time Precision and Plasticity in Circadian Output Neurons. Eur J Neurosci 2025; 61:e70037. [PMID: 40080910 PMCID: PMC11906214 DOI: 10.1111/ejn.70037] [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: 02/19/2024] [Revised: 01/30/2025] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Circadian rhythms influence various physiological and behavioral processes such as sleep-wake cycles, hormone secretion, and metabolism. In Drosophila, an important set of circadian output neurons is called pars intercerebralis (PI) neurons, which receive input from specific clock neurons called DN1. These DN1 neurons can further be subdivided into functionally and anatomically distinctive anterior (DN1a) and posterior (DN1p) clusters. The neuropeptide diuretic hormones 31 (Dh31) and 44 (Dh44) are the insect neuropeptides known to activate PI neurons to control activity rhythms. However, the neurophysiological basis of how Dh31 and Dh44 affect circadian clock neural coding mechanisms underlying sleep in Drosophila is not well understood. Here, we identify Dh31/Dh44-dependent spike time precision and plasticity in PI neurons. We first find that a mixture of Dh31 and Dh44 enhanced the firing of PI neurons, compared to the application of Dh31 alone and Dh44 alone. We next find that the application of synthesized Dh31 and Dh44 affects membrane potential dynamics of PI neurons in the precise timing of the neuronal firing through their synergistic interaction, possibly mediated by calcium-activated potassium channel conductance. Further, we characterize that Dh31/Dh44 enhances postsynaptic potentials in PI neurons. Together, these results suggest multiplexed neuropeptide-dependent spike time precision and plasticity as circadian clock neural coding mechanisms underlying sleep in Drosophila.
Collapse
Affiliation(s)
- Bryan Chong
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Vipin Kumar
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Dieu Linh Nguyen
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Makenzie A. Hopkins
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Faith S. Ferry
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Lucia K. Spera
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Elizabeth M. Paul
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Anelise N. Hutson
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Masashi Tabuchi
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| |
Collapse
|
5
|
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
|
6
|
Dellal S, Zurita H, Valero M, Abad-Perez P, Kruglikov I, Meng J, Prönneke A, Hanson JL, Mir E, Ongaro M, Wang XJ, Buzsáki G, Machold R, Rudy B. Inhibitory and disinhibitory VIP IN-mediated circuits in neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640383. [PMID: 40060562 PMCID: PMC11888407 DOI: 10.1101/2025.02.26.640383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Cortical GABAergic interneurons (INs) are comprised of distinct types that provide tailored inhibition to pyramidal cells (PCs) and other INs, thereby enabling precise control of cortical circuit activity. INs expressing the neuropeptide vasoactive-intestinal peptide (VIP) have attracted attention recently following the discovery that they predominantly function by inhibiting dendritic-targeting somatostatin (SST) expressing INs, thereby disinhibiting PCs. This VIP-SST disinhibitory circuit motif is observed throughout the neocortex from mice to humans, and serves as a key mechanism for top-down (feedback) and context-dependent information processing. Thus, VIP IN-mediated disinhibition has been found to play an important role in sensory processing, control of executive functions, attention, sensorimotor integration and other cortico-cortical and thalamocortical feedback interactions. Furthermore, VIP INs have been implicated in mediating the effects of reinforcement signals, both reward and aversive, via their responsiveness to neuromodulators such as acetylcholine (ACh), and in facilitating synaptic plasticity and learning. While it is evident from transcriptomic analyses that VIP INs are a molecularly heterogeneous group, the physiological significance of this diversity is unclear at present. Here, we have characterized the functional diversity of VIP INs in the primary somatosensory cortex by leveraging intersectional genetic approaches to study distinct VIP IN subtypes. We found that VIP INs can be divided into four different populations: a group that expresses the Ca2+-binding protein calretinin (CR), two distinct groups that express the neuropeptide cholecystokinin (CCK), and a group that does not express either CR or CCK (non-CCK non-CR; or nCCK nCR). VIP neurons in each group exhibit different laminar distributions, axonal and dendritic arbors, intrinsic electrophysiological properties, and efferent connectivity, VIP/CR INs target almost exclusively SST INs, VIP/nCCK nCR INs also mainly target SST INs but also have connections to parvalbumin (PV) expressing INs. These two groups have essentially no connectivity to pyramidal cells (PCs). On the other hand, the two types of VIP/CCK INs target PCs, but differ in the degree to which synaptic release from each type is modulated by endocannabinoids. We also found that long-range inputs differentially recruit distinct VIP IN groups. Intriguingly, we find that distinct VIP IN populations target distinct SST INs subtypes in turn, indicating the presence of specialized VIP-SST disinhibitory subcircuits. Activation of distinct VIP IN subpopulations in vivo results in differential effects on the cortical network, thus providing evidence for modularity in VIP IN-mediated actions during cortical information processing.
Collapse
Affiliation(s)
- Shlomo Dellal
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - Hector Zurita
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - Manuel Valero
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
- Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Pablo Abad-Perez
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
- Universidad Cardenal Herrera-CEU, CEU Universities, Spain
| | - Ilya Kruglikov
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - John Meng
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
- Center for Neural Science, NYU, New York, NY, 10003
| | - Alvar Prönneke
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - Jessica L. Hanson
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - Ema Mir
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - Marina Ongaro
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - Xiao-Jing Wang
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
- Center for Neural Science, NYU, New York, NY, 10003
| | - György Buzsáki
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
- Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, 10016
| | - Robert Machold
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
| | - Bernardo Rudy
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016
- Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, 10016
- Department of Anesthesiology, Perioperative Care and Pain Medicine, NYU School of Medicine, New York, NY, 10016
| |
Collapse
|
7
|
Straub VM, Barti B, Tandar ST, Stevens AF, van Egmond N, van der Wel T, Zhu N, Rüegger J, van der Horst C, Heitman LH, Li Y, Stella N, van Hasselt JGC, Katona I, van der Stelt M. The endocannabinoid 2-arachidonoylglycerol is released and transported on demand via extracellular microvesicles. Proc Natl Acad Sci U S A 2025; 122:e2421717122. [PMID: 39977325 PMCID: PMC11873938 DOI: 10.1073/pnas.2421717122] [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: 10/23/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
While it is known that endocannabinoids (eCB) modulate multiple neuronal functions, the molecular mechanism governing their release and transport remains elusive. Here, we propose an "on-demand release" model, wherein the formation of microvesicles, a specific group of extracellular vesicles (EVs) containing the eCB, 2-arachidonoylglycerol (2-AG), is an important step. A coculture model system that combines a reporter cell line expressing the fluorescent eCB sensor, G protein-coupled receptor-based (GRAB)eCB2.0, and neuronal cells revealed that neurons release EVs containing 2-AG, but not anandamide, in a stimulus-dependent process regulated by protein kinase C, Diacylglycerol lipase, Adenosinediphosphate (ADP) ribosylation factor 6 (Arf6), and which was sensitive to inhibitors of eCB facilitated diffusion. A vesicle contained approximately 2,000 2-AG molecules. Accordingly, hippocampal eCB-mediated synaptic plasticity was modulated by Arf6 and transport inhibitors. The "on-demand release" model, supported by mathematical analysis, offers a cohesive framework for understanding eCB trafficking at the molecular level and suggests that microvesicles carrying signaling lipids in their membrane regulate neuronal functions in parallel to canonical synaptic vesicles.
Collapse
Affiliation(s)
- Verena M. Straub
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Benjamin Barti
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN47405-2204
| | - Sebastian T. Tandar
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden2333 CC, The Netherlands
| | - A. Floor Stevens
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Noëlle van Egmond
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Na Zhu
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Joel Rüegger
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
| | - Cas van der Horst
- Department of Medicinal Chemistry, Leiden University, Leiden2333 CC, The Netherlands
| | - Laura H. Heitman
- Department of Medicinal Chemistry, Leiden University, Leiden2333 CC, The Netherlands
- Oncode Institute, Leiden2333 CC, The Netherlands
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Beijing100871, China
| | - Nephi Stella
- Department of Pharmacology, School of Medicine, University of Washington, Seattle, WA98195
| | - J. G. Coen van Hasselt
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden2333 CC, The Netherlands
| | - István Katona
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN47405-2204
- Molecular Neurobiology Research Group, Hungarian Research Network, Institute of Experimental Medicine, BudapestH-1083, Hungary
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden University, Leiden2333 CC, The Netherlands
- Oncode Institute, Leiden2333 CC, The Netherlands
| |
Collapse
|
8
|
Dvorakova M, Mackie K, Straiker A. Muscarinic cannabinoid suppression of excitation, a novel form of coincidence detection. Pharmacol Res 2025; 212:107606. [PMID: 39824373 DOI: 10.1016/j.phrs.2025.107606] [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: 11/19/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Δ9-tetrahydrocannabinol (THC), the chief psychoactive ingredient of cannabis, acts in the brain primarily via cannabinoid CB1 receptors. These receptors are implicated in several forms of synaptic plasticity - depolarization-induced suppression of excitation (DSE), metabotropic suppression of excitation (MSE), long term depression (LTD) and activation-dependent desensitization. Cultured autaptic hippocampal neurons express all of these, illustrating the rich functional and temporal heterogeneity of CB1 at a single set of synapses. Here we report that coincident activation of muscarinic acetylcholine receptors and elicitation of DSE in autaptic hippocampal neurons results in a substantial (∼40 %) and temporally precise inhibition of excitatory transmission lasting ∼10 minutes. Its induction is blocked by CB1 and muscarinic M3/M5 receptor antagonists and is absent in CB1 receptor knockout neurons. Notably, once it is established, inhibition is reversed by a CB1, but not a muscarinic, antagonist, suggesting that the inhibition occurs via persistent activation of CB1 receptors. We refer to this inhibition as muscarinic cannabinoid suppression of excitation (MCSE). MCSE can be mimicked by coapplication of muscarinic and cannabinoid agonists and requires Ca2+-release from internal stores. As such, MCSE represents a novel and targeted form of coincidence detection - important for many modes of learning and memory -- between cannabinoid and muscarinic signaling systems that elicits a medium-duration depression of synaptic signaling. Given the known roles of muscarinic and cannabinoid receptors in the hippocampus, MCSE may be important in the modulation of hippocampal signaling at the site of septal inputs, with potential implications for learning and memory, epilepsy and addiction.
Collapse
Affiliation(s)
- Michaela Dvorakova
- Gill Institute for Neuroscience, United States; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States
| | - Ken Mackie
- Gill Institute for Neuroscience, United States; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States
| | - Alex Straiker
- Gill Institute for Neuroscience, United States; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, United States.
| |
Collapse
|
9
|
Caccavano AP, Vlachos A, McLean N, Kimmel S, Kim JH, Vargish G, Mahadevan V, Hewitt L, Rossi AM, Spineux I, Wu SJ, Furlanis E, Dai M, Garcia BL, Wang Y, Chittajallu R, London E, Yuan X, Hunt S, Abebe D, Eldridge MAG, Cummins AC, Hines BE, Plotnikova A, Mohanty A, Averbeck BB, Zaghloul K, Dimidschstein J, Fishell G, Pelkey KA, McBain CJ. Divergent opioid-mediated suppression of inhibition between hippocampus and neocortex across species and development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.20.576455. [PMID: 38313283 PMCID: PMC10836073 DOI: 10.1101/2024.01.20.576455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Within the adult rodent hippocampus, opioids suppress inhibitory parvalbumin-expressing interneurons (PV-INs), thus disinhibiting local micro-circuits. However, it is unknown if this disinhibitory motif is conserved in other cortical regions, species, or across development. We observed that PV-IN mediated inhibition is robustly suppressed by opioids in hippocampus proper but not primary neocortex in mice and nonhuman primates, with spontaneous inhibitory tone in resected human tissue also following a consistent dichotomy. This hippocampal disinhibitory motif was established in early development when PV-INs and opioids were found to regulate early population activity. Acute opioid-mediated modulation was partially occluded with morphine pretreatment, with implications for the effects of opioids on hippocampal network activity important for learning and memory. Together, these findings demonstrate that PV-INs exhibit a divergence in opioid sensitivity across brain regions that is remarkably conserved across evolution and highlights the underappreciated role of opioids acting through immature PV-INs in shaping hippocampal development.
Collapse
Affiliation(s)
- Adam P Caccavano
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anna Vlachos
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nadiya McLean
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sarah Kimmel
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - June Hoan Kim
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Geoffrey Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Vivek Mahadevan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Lauren Hewitt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anthony M Rossi
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ilona Spineux
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sherry Jingjing Wu
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elisabetta Furlanis
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Min Dai
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brenda Leyva Garcia
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yating Wang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Edra London
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Daniel Abebe
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mark A G Eldridge
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Alex C Cummins
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Brendan E Hines
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Anya Plotnikova
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Arya Mohanty
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Bruno B Averbeck
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Kareem Zaghloul
- National Institute of Neurological Disorders and Stroke (NINDS) Intramural Research Program, NIH, Bethesda, MD 20892, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Gonçalves de Queiroz BF, Cristina de Sousa Fonseca F, Pinto Barra WC, Viana GB, Irie AL, de Castro Perez A, Lima Romero TR, Gama Duarte ID. Interaction between the dopaminergic and endocannabinoid systems promotes peripheral antinociception. Eur J Pharmacol 2025; 987:177195. [PMID: 39662656 DOI: 10.1016/j.ejphar.2024.177195] [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: 08/29/2024] [Revised: 11/11/2024] [Accepted: 12/09/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Dopamine has been widely related to pain modulation, at central and peripheral levels. In this study we aimed to investigate the mechanisms involved in peripheral antinociception, evaluating the interaction between the dopaminergic and endocannabinoid systems in this event. METHODS Male Swiss mice (30-40 g) were pre-sensitized by administration of the hyperalgesic PGE2 (2 μg/paw). The nociceptive threshold was measured using the paw withdrawal test. RESULTS Dopamine (80 ng/paw) promoted antinociception. This effect was reversed by the CB1 and CB2 cannabinoid receptor antagonists AM251 (20, 40, and 80 μg/paw) and AM630 (25, 50, and 100 μg/paw). JZL (4 μg/paw), an inhibitor of the degradation of the 2-arachidonylglycerol (2-AG), potentiated the antinociceptive action of the submaximal dose of dopamine (5 ng/paw). While anandamide degradation and reuptake inhibitors (MAFP 0.5 μg/paw and VDM11 2.5 μg/paw) did not promote changes in intermediate antinociception induced by dopamine. Anandamide at a submaximal dose (12.5 ng/paw) promoted intermediate antinociception that was not potentiated by the administration of the dopamine reuptake inhibitor GBR 12783 (16 μg/paw). In contrast, the administration of GBR potentiated the intermediate antinociception induced by a submaximal dose of 2-AG (10 μg/paw). Furthermore, the dopaminergic receptor antagonists D2 Remoxipride (4 μg/paw) and D3 U99194 (16 μg/paw) reversed the antinociception mediated by the maximum dose of this endocannabinoid (20 μg/paw). In contrast, the D4 receptor antagonist L-745,870 (16 μg/paw) did not change the nociceptive threshold. CONCLUSIONS In this way, we demonstrate the interaction between the dopaminergic and endocannabinoid systems to promote analgesia peripherally.
Collapse
Affiliation(s)
- Bárbara Formiga Gonçalves de Queiroz
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Flávia Cristina de Sousa Fonseca
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Walace Cassio Pinto Barra
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Giovanna Bauer Viana
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Audrey Lopes Irie
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Andrea de Castro Perez
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Thiago Roberto Lima Romero
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Igor Dimitri Gama Duarte
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
11
|
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
|
12
|
Wang S, Yu L, Guo H, Zuo W, Guo Y, Liu H, Wang J, Wang J, Li X, Hou W, Wang M. Gastrodin Ameliorates Post-Stroke Depressive-Like Behaviors Through Cannabinoid-1 Receptor-Dependent PKA/RhoA Signaling Pathway. Mol Neurobiol 2025; 62:366-385. [PMID: 38856794 DOI: 10.1007/s12035-024-04267-5] [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: 11/25/2023] [Accepted: 05/26/2024] [Indexed: 06/11/2024]
Abstract
Post-stroke depression (PSD) is a significant complication in stroke patients, increases long-term mortality, and exaggerates ischemia-induced brain injury. However, the underlying molecular mechanisms and effective therapeutic targets related to PSD have remained elusive. Here, we employed an animal behavioral model of PSD by combining the use of middle cerebral artery occlusion (MCAO) followed by spatial restraint stress to study the molecular underpinnings and potential therapies of PSD. Interestingly, we found that sub-chronic application of gastrodin (Gas), a traditional Chinese medicinal herb Gastrodia elata extraction, relieved depression-related behavioral deficits, increased the impaired expression of synaptic transmission-associated proteins, and restored the altered spine density in hippocampal CA1 of PSD animals. Furthermore, our results indicated that the anti-PSD effect of Gas was dependent on membrane cannabinoid-1 receptor (CB1R) expression. The contents of phosphorated protein kinase A (p-PKA) and phosphorated Ras homolog gene family member A (p(ser188)-RhoA) were decreased in the hippocampus of PSD-mice, which was reversed by Gas treatment, and CB1R depletion caused a diminished efficacy of Gas on p-PKA and p-RhoA expression. In addition, the anti-PSD effect of Gas was partially blocked by PKA inhibition or RhoA activation, indicating that the anti-PSD effect of Gas is associated with the CB1R-mediated PKA/RhoA signaling pathway. Together, our findings revealed that Gas treatment possesses protective effects against the post-stroke depressive-like state; the CB1R-involved PKA/RhoA signaling pathway is critical in mediating Gas's anti-PSD potency, suggesting that Gas application may be beneficial in the prevention and adjunctive treatment of PSD.
Collapse
Affiliation(s)
- Shiquan Wang
- College of Life Sciences, Northwest University, Xi'an, 710127, Shaanxi, China
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Liang Yu
- Department of Information, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Wenqiang Zuo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yaru Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Huiqing Liu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiajia Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jin Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Minghui Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
13
|
Somogyi P, Horie S, Lukacs I, Hunter E, Sarkany B, Viney T, Livermore J, Plaha P, Stacey R, Ansorge O, El Mestikawy S, Zhao Q. Synaptic Targets and Cellular Sources of CB1 Cannabinoid Receptor and Vesicular Glutamate Transporter-3 Expressing Nerve Terminals in Relation to GABAergic Neurons in the Human Cerebral Cortex. Eur J Neurosci 2025; 61:e16652. [PMID: 39810425 PMCID: PMC11733414 DOI: 10.1111/ejn.16652] [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: 10/11/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025]
Abstract
Cannabinoid receptor 1 (CB1) regulates synaptic transmission through presynaptic receptors in nerve terminals, and its physiological roles are of clinical relevance. The cellular sources and synaptic targets of CB1-expressing terminals in the human cerebral cortex are undefined. We demonstrate a variable laminar pattern of CB1-immunoreactive axons and electron microscopically show that CB1-positive GABAergic terminals make type-2 synapses innervating dendritic shafts (69%), dendritic spines (20%) and somata (11%) in neocortical layers 2-3. Of the CB1-immunopositive GABAergic terminals, 25% were vesicular-glutamate-transporter-3 (VGLUT3)-immunoreactive, suggesting GABAergic/glutamatergic co-transmission on dendritic shafts. In vitro recorded and labelled VGLUT3 or CB1-positive GABAergic interneurons expressed cholecystokinin, vasoactive-intestinal-polypeptide and calretinin, had diverse firing, axons and dendrites, and included rosehip, neurogliaform and basket cells, but not double bouquet or axo-axonic cells. CB1-positive interneurons innervated pyramidal cells and GABAergic interneurons. Glutamatergic synaptic terminals formed type-1 synapses and some were positive for CB1 receptor with a distribution that appeared different from that in GABAergic terminals. From the sampled VGLUT3-positive terminals, 60% formed type-1 synapses with dendritic spines (80%) or shafts (20%) and 52% were also positive for VGLUT1, suggesting intracortical origin. Some VGLUT3-positive terminals were immunopositive for vesicular-monoamine-transporter-2, suggesting 5-HT/glutamate co-transmission. Overall, the results show that CB1 regulates GABA release mainly to dendritic shafts of both pyramidal cells and interneurons and predict CB1-regulated co-release of GABA and glutamate from single cortical interneurons. We also demonstrate the co-existence of multiple vesicular glutamate transporters in a select population of terminals probably originating from cortical neurons and innervating dendritic spines in the human cerebral cortex.
Collapse
Affiliation(s)
- Peter Somogyi
- Department of PharmacologyUniversity of OxfordOxfordUK
| | - Sawa Horie
- Department of PharmacologyUniversity of OxfordOxfordUK
- Kawasaki Medical SchoolOkayamaJapan
- Department of Anatomy and NeurobiologyNational Defense Medical CollegeSaitamaJapan
| | - Istvan Lukacs
- Department of PharmacologyUniversity of OxfordOxfordUK
- Institute of Experimental MedicineBudapestHungary
| | - Emily Hunter
- Department of PharmacologyUniversity of OxfordOxfordUK
| | | | | | - James Livermore
- Department of Neurosurgery, John Radcliffe HospitalOUH NHS Foundation TrustOxfordUK
- Department of NeurosurgeryLeeds General InfirmaryLeedsUK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe HospitalOUH NHS Foundation TrustOxfordUK
| | - Richard Stacey
- Department of Neurosurgery, John Radcliffe HospitalOUH NHS Foundation TrustOxfordUK
| | - Olaf Ansorge
- Nuffield Department of Clinical NeurosciencesUniv. OxfordOxfordUK
| | - Salah El Mestikawy
- Douglas Research CentreMcGill University and the Montreal West Island IUHSSCMontréalCanada
| | - Qianru Zhao
- Department of PharmacologyUniversity of OxfordOxfordUK
- Department of Chemical Biology, School of Pharmaceutical SciencesSouth‐Central Minzu UniversityWuhanChina
| |
Collapse
|
14
|
Kumar U. Cannabinoids: Role in Neurological Diseases and Psychiatric Disorders. Int J Mol Sci 2024; 26:152. [PMID: 39796008 PMCID: PMC11720483 DOI: 10.3390/ijms26010152] [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: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
An impact of legalization and decriminalization of marijuana is the gradual increase in the use of cannabis for recreational purposes, which poses a potential threat to society and healthcare systems worldwide. However, the discovery of receptor subtypes, endogenous endocannabinoids, and enzymes involved in synthesis and degradation, as well as pharmacological characterization of receptors, has led to exploration of the use of cannabis in multiple peripheral and central pathological conditions. The role of cannabis in the modulation of crucial events involving perturbed physiological functions and disease progression, including apoptosis, inflammation, oxidative stress, perturbed mitochondrial function, and the impaired immune system, indicates medicinal values. These events are involved in most neurological diseases and prompt the gradual progression of the disease. At present, several synthetic agonists and antagonists, in addition to more than 70 phytocannabinoids, are available with distinct efficacy as a therapeutic alternative in different pathological conditions. The present review aims to describe the use of cannabis in neurological diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
15
|
Chong B, Kumar V, Nguyen DL, Hopkins MA, Ferry FS, Spera LK, Paul EM, Hutson AN, Tabuchi M. Neuropeptide-dependent spike time precision and plasticity in circadian output neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.06.616871. [PMID: 39411164 PMCID: PMC11476009 DOI: 10.1101/2024.10.06.616871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Circadian rhythms influence various physiological and behavioral processes such as sleep-wake cycles, hormone secretion, and metabolism. In Drosophila, an important set of circadian output neurons are called pars intercerebralis (PI) neurons, which receive input from specific clock neurons called DN1. These DN1 neurons can further be subdivided into functionally and anatomically distinctive anterior (DN1a) and posterior (DN1p) clusters. The neuropeptide diuretic hormones 31 (Dh31) and 44 (Dh44) are the insect neuropeptides known to activate PI neurons to control activity rhythms. However, the neurophysiological basis of how Dh31 and Dh44 affect circadian clock neural coding mechanisms underlying sleep in Drosophila is not well understood. Here, we identify Dh31/Dh44-dependent spike time precision and plasticity in PI neurons. We first find that a mixture of Dh31 and Dh44 enhanced the firing of PI neurons, compared to the application of Dh31 alone and Dh44 alone. We next find that the application of synthesized Dh31 and Dh44 affects membrane potential dynamics of PI neurons in the precise timing of the neuronal firing through their synergistic interaction, possibly mediated by calcium-activated potassium channel conductance. Further, we characterize that Dh31/Dh44 enhances postsynaptic potentials in PI neurons. Together, these results suggest multiplexed neuropeptide-dependent spike time precision and plasticity as circadian clock neural coding mechanisms underlying sleep in Drosophila.
Collapse
Affiliation(s)
- Bryan Chong
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Vipin Kumar
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Dieu Linh Nguyen
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Makenzie A. Hopkins
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Faith S. Ferry
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Lucia K. Spera
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Elizabeth M. Paul
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Anelise N. Hutson
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Masashi Tabuchi
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
16
|
Monory K, de Azua IR, Lutz B. Genetic Tools in Rodents to Study Cannabinoid Functions. Curr Top Behav Neurosci 2024. [PMID: 39680319 DOI: 10.1007/7854_2024_550] [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: 12/17/2024]
Abstract
During the past 30 years, the endocannabinoid system (ECS) has emerged as a major signalling system in the mammalian brain regulating neurotransmission in numerous brain regions and in various cell populations. Endocannabinoids are able to regulate specific physiological functions and thus modify their behavioural manifestations and allostatic alterations of the ECS linked to different pathological conditions. As discussed in detail in other chapters of this book, endocannabinoids are involved in learning and memory, stress, and anxiety, feeding, energy balance, development, and ageing. Likewise, many CNS disorders (e.g. schizophrenia, epilepsy, substance use disorders, and multiple sclerosis) are associated with dysregulation of the ECS. Discerning the physiological functions of the synthetic and degrading enzymes of endocannabinoids and their receptors is a challenging task because of their distinct and complex expression patterns. Techniques of genetic engineering have been able to shed light on a number of complex ECS-related tasks during the past years. In this chapter, first, we take a critical look at the toolbox available to researchers who would like to investigate cannabinoid effects using genetic engineering techniques, then we comprehensively discuss genetically modified rodent models in various neuronal and non-neuronal cell populations, both within and outside the nervous system.
Collapse
Affiliation(s)
- Krisztina Monory
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
- Leibniz Institute for Resilience Research (LIR) gGmbH, Mainz, Germany.
| |
Collapse
|
17
|
Camp CR, Banke TG, Xing H, Yu K, Perszyk RE, Epplin MP, Akins NS, Zhang J, Benke TA, Yuan H, Liotta DC, Traynelis SF. Selective Enhancement of the Interneuron Network and Gamma-Band Power via GluN2C/GluN2D NMDA Receptor Potentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622179. [PMID: 39574703 PMCID: PMC11580944 DOI: 10.1101/2024.11.05.622179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
N-methyl-D-aspartate receptors (NMDARs) comprise a family of ligand-gated ionotropic glutamate receptors that mediate a slow, calcium-permeable component to excitatory neurotransmission. The GluN2D subunit is enriched in GABAergic inhibitory interneurons in cortical tissue. Diminished levels of GABAergic inhibition contribute to multiple neuropsychiatric conditions, suggesting that enhancing inhibition may have therapeutic utility, thus making GluN2D modulation an attractive drug target. Here, we describe the actions of a GluN2C/GluN2D-selective positive allosteric modulator (PAM), (+)-EU1180-453, which has improved drug-like properties such as increased aqueous solubility compared to the first-in-class GluN2C/GluN2D-selective prototypical PAM (+)-CIQ. (+)-EU1180-453 doubles the NMDAR response at lower concentrations (< 10 μM) compared to (+)-CIQ, and produces a greater degree of maximal potentiation at 30 μM. Using in vitro electrophysiological recordings, we show that (+)-EU1180-453 potentiates triheteromeric NMDARs containing at least one GluN2C or GluN2D subunit, and is active at both exon5-lacking and exon5-containing GluN1 splice variants. (+)-EU1180-453 increases glutamate efficacy for GluN2C/GluN2D-containing NMDARs by both prolonging the deactivation time and potentiating the peak response amplitude. We show that (+)-EU1180-453 selectively increases synaptic NMDAR-mediated charge transfer onto P11-15 CA1 stratum radiatum hippocampal interneurons, but is without effect on CA1 pyramidal cells. This increased charge transfer enhances inhibitory output from GABAergic interneurons onto CA1 pyramidal cells in a GluN2D-dependent manner. (+)-EU1180-453 also shifts excitatory-to-inhibitory coupling towards increased inhibition and produces enhanced gamma band power from carbachol-induced field potential oscillations in hippocampal slices. Thus, (+)-EU1180-453 can enhance overall circuit inhibition, which could prove therapeutically useful for the treatment of anxiety, depression, schizophrenia, and other neuropsychiatric disorders. Significance Statement Interneuron dysfunction and diminished GABAergic inhibition in neocortical and hippocampal circuits remains a prominent molecular hypothesis for neuropsychiatric diseases including anxiety, depression, and schizophrenia. Pharmacological agents that boost GABA receptor function have shown utility in various forms of depression and treating symptoms of schizophrenia. Cortical GABAergic interneurons, unlike their excitatory pyramidal cell counterparts, are enriched for the GluN2D subunit of the NMDA receptor. Thus, GluN2D subunit-selective modulation could be a useful therapeutic tool to enhance local inhibition, improving the prognosis for neuropsychiatric diseases for which interneuron dysfunction is prominent and causal to circuit aberration.
Collapse
Affiliation(s)
- Chad R. Camp
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tue G. Banke
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hao Xing
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kuai Yu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Riley E. Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Matthew P. Epplin
- Department of Chemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nicholas S. Akins
- Department of Chemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tim A. Benke
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dennis C. Liotta
- Department of Chemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stephen F. Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Fekete Z, Weisz F, Karlócai MR, Veres JM, Andrási T, Hájos N. Synaptic communication within the microcircuits of pyramidal neurons and basket cells in the mouse prefrontal cortex. J Physiol 2024. [PMID: 39418315 DOI: 10.1113/jp286284] [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: 01/16/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Basket cells are inhibitory interneurons in cortical structures with the potential to efficiently control the activity of their postsynaptic partners. Although their contribution to higher order cognitive functions associated with the medial prefrontal cortex (mPFC) relies on the characteristics of their synaptic connections, the way that they are embedded into local circuits is still not fully uncovered. Here, we determined the synaptic properties of excitatory and inhibitory connections between pyramidal neurons (PNs), cholecystokinin-containing basket cells (CCKBCs) and parvalbumin-containing basket cells (PVBCs) in the mouse mPFC. By performing paired recordings, we revealed that PVBCs receive larger unitary excitatory postsynaptic currents from PNs with shorter latency and faster kinetic properties compared to events evoked in CCKBCs. Also, unitary inhibitory postsynaptic currents in PNs were more reliably evoked by PVBCs than by CCKBCs, yet the former connections showed profound short-term depression. Moreover, we demonstrated that CCKBCs and PVBCs in the mPFC are connected with each other. Because alterations in PVBC function have been linked to neurological and psychiatric conditions such as Alzheimer's disease and schizophrenia and CCKBC vulnerability might play a role in mood disorders, a deeper understanding of the general features of basket cell synapses could serve as a reference point for future investigations with therapeutic objectives. KEY POINTS: Cholecystokinin- (CCKBCs) and parvalbumin-expressing basket cells (PVBCs) have distinct passive and active membrane properties. Pyramidal neurons give rise to larger unitary excitatory postsynaptic currents in PVBCs compared to events in CCKBCs. Unitary inhibitory postsynaptic currents in pyramidal neurons are more reliably evoked by PVBCs than by CCKBCs. Basket cells form chemical synapses and gap junctions with their own cell type. The two basket cell types are connected with each other.
Collapse
Affiliation(s)
- Zsuzsanna Fekete
- Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Filippo Weisz
- Institute of Experimental Medicine, Budapest, Hungary
| | | | - Judit M Veres
- Institute of Experimental Medicine, Budapest, Hungary
| | - Tibor Andrási
- Institute of Experimental Medicine, Budapest, Hungary
| | - Norbert Hájos
- Institute of Experimental Medicine, Budapest, Hungary
- The Linda and Jack Gill Center for Molecular Bioscience, Indiana University Bloomington, Indiana, USA
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, Indiana, USA
| |
Collapse
|
19
|
Briânis RC, Andreotti JP, Moreira FA, Iglesias LP. Interplay between endocannabinoid and endovanilloid mechanisms in fear conditioning. Acta Neuropsychiatr 2024; 36:255-264. [PMID: 37982167 DOI: 10.1017/neu.2023.54] [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] [Indexed: 11/21/2023]
Abstract
OBJECTIVE The transient receptor potential cation channel, subfamily V (vanilloid), member 1 (TRPV1) mediates pain perception to thermal and chemical stimuli in peripheral neurons. The cannabinoid receptor type 1 (CB1), on the other hand, promotes analgesia in both the periphery and the brain. TRPV1 and CB1 have also been implicated in learned fear, which involves the association of a previously neutral stimulus with an aversive event. In this review, we elaborate on the interplay between CB1 receptors and TRPV1 channels in learned fear processing. METHODS We conducted a PubMed search for a narrative review on endocannabinoid and endovanilloid mechanisms on fear conditioning. RESULTS TRPV1 and CB1 receptors are activated by a common endogenous agonist, arachidonoyl ethanolamide (anandamide), Moreover, they are expressed in common neuroanatomical structures and recruit converging cellular pathways, acting in concert to modulate fear learning. However, evidence suggests that TRPV1 exerts a facilitatory role, whereas CB1 restrains fear responses. CONCLUSION TRPV1 and CB1 seem to mediate protective and aversive roles of anandamide, respectively. However, more research is needed to achieve a better understanding of how these receptors interact to modulate fear learning.
Collapse
Affiliation(s)
- Rayssa C Briânis
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabrício A Moreira
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lia P Iglesias
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
20
|
Lord MN, Noble EE. Hypothalamic cannabinoid signaling: Consequences for eating behavior. Pharmacol Res Perspect 2024; 12:e1251. [PMID: 39155548 PMCID: PMC11331011 DOI: 10.1002/prp2.1251] [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: 11/29/2023] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024] Open
Abstract
In parallel to the legalization of cannabis for both medicinal and recreational purposes, cannabinoid use has steadily increased over the last decade in the United States. Cannabinoids, such as tetrahydrocannabinol and anandamide, bind to the central cannabinoid-1 (CB1) receptor to impact several physiological processes relevant for body weight regulation, including appetite and energy expenditure. The hypothalamus integrates peripheral signals related to energy balance, houses several nuclei that orchestrate eating, and expresses the CB1 receptor. Herein we review literature to date concerning cannabinergic action in the hypothalamus with a specific focus on eating behaviors. We highlight hypothalamic areas wherein researchers have focused their attention, including the lateral, arcuate, paraventricular, and ventromedial hypothalamic nuclei, and interactions with the hormone leptin. This review serves as a comprehensive analysis of what is known about cannabinoid signaling in the hypothalamus, highlights gaps in the literature, and suggests future directions.
Collapse
Affiliation(s)
- Magen N. Lord
- Department of Nutritional SciencesUniversity of GeorgiaAthensGeorgiaUSA
| | - Emily E. Noble
- Department of Nutritional SciencesUniversity of GeorgiaAthensGeorgiaUSA
| |
Collapse
|
21
|
Gertsch J, Chicca A. CNS Drug Discovery in Academia: Where Basic Research Meets Innovation. Chembiochem 2024; 25:e202400397. [PMID: 38958639 DOI: 10.1002/cbic.202400397] [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] [Revised: 05/27/2024] [Indexed: 07/04/2024]
Abstract
The involvement of academic research in drug discovery is consistently growing. However, academic projects seldom advance to clinical trials. Here, we assess the landscape of drug discovery within the National Centre of Competence in Research (NCCR) TransCure launched by the Swiss National Science Foundation to foster basic research and early-stage drug discovery on membrane transporters. This included transporters in central nervous system (CNS) disorders, which represent a huge unmet medical need. While idea championship, sustainable funding, collaborations between disciplines at the interface of academia and industry are important for translational research, Popperian falsifiability, strong intellectual property and a motivated startup team are key elements for innovation. This is exemplified by the NCCR TransCure spin-off company Synendos Therapeutics, a clinical stage biotech company developing the first selective endocannabinoid reuptake inhibitors (SERIs) as novel treatment for neuropsychiatric disorders. We provide a perspective on the challenges related to entering an uncharted druggable space and bridging the often mentioned "valley of death". The high attrition rate of drug discovery projects in the CNS field within academia is often due to the lack of meaningful animal models that can provide pharmacological proof-of-concept for potentially disruptive technologies at the earliest stages, and the absence of solid intellectual property.
Collapse
Affiliation(s)
- Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
- Synendos Therapeutics, Barfüsserplatz, 3, 4051, Basel, Switzerland
| |
Collapse
|
22
|
Zhu D, Zhang J, Ma X, Hu M, Gao F, Hashem JB, Lyu J, Wei J, Cui Y, Qiu S, Chen C. Overabundant endocannabinoids in neurons are detrimental to cognitive function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613513. [PMID: 39345517 PMCID: PMC11430108 DOI: 10.1101/2024.09.17.613513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
2-Arachidonoylglycerol (2-AG) is the most prevalent endocannabinoid involved in maintaining brain homeostasis. Previous studies have demonstrated that inactivating monoacylglycerol lipase (MAGL), the primary enzyme responsible for degrading 2-AG in the brain, alleviates neuropathology and prevents synaptic and cognitive decline in animal models of neurodegenerative diseases. However, we show that selectively inhibiting 2-AG metabolism in neurons impairs cognitive function in mice. This cognitive impairment appears to result from decreased expression of synaptic proteins and synapse numbers, impaired long-term synaptic plasticity and cortical circuit functional connectivity, and diminished neurogenesis. Interestingly, the synaptic and cognitive deficits induced by neuronal MAGL inactivation can be counterbalanced by inhibiting astrocytic 2-AG metabolism. Transcriptomic analyses reveal that inhibiting neuronal 2-AG degradation leads to widespread changes in expression of genes associated with synaptic function. These findings suggest that crosstalk in 2-AG signaling between astrocytes and neurons is crucial for maintaining synaptic and cognitive functions and that excessive 2-AG in neurons alone is detrimental to cognitive function.
Collapse
Affiliation(s)
- Dexiao Zhu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jian Zhang
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Xiaokuang Ma
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Mei Hu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Fei Gao
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jack B. Hashem
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jianlu Lyu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jing Wei
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Yuehua Cui
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Shenfeng Qiu
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Chu Chen
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| |
Collapse
|
23
|
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
|
24
|
Spicarova D, Palecek J. Anandamide-Mediated Modulation of Nociceptive Transmission at the Spinal Cord Level. Physiol Res 2024; 73:S435-S448. [PMID: 38957948 PMCID: PMC11412359 DOI: 10.33549/physiolres.935371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Three decades ago, the first endocannabinoid, anandamide (AEA), was identified, and its analgesic effect was recognized in humans and preclinical models. However, clinical trial failures pointed out the complexity of the AEA-induced analgesia. The first synapses in the superficial laminae of the spinal cord dorsal horn represent an important modulatory site in nociceptive transmission and subsequent pain perception. The glutamatergic synaptic transmission at these synapses is strongly modulated by two primary AEA-activated receptors, cannabinoid receptor 1 (CB1) and transient receptor potential vanilloid 1 (TRPV1), both highly expressed on the presynaptic side formed by the endings of primary nociceptive neurons. Activation of these receptors can have predominantly inhibitory (CB1) and excitatory (TRPV1) effects that are further modulated under pathological conditions. In addition, dual AEA-mediated signaling and action may occur in primary sensory neurons and dorsal horn synapses. AEA application causes balanced inhibition and excitation of primary afferent synaptic input on superficial dorsal horn neurons in normal conditions, whereas peripheral inflammation promotes AEA-mediated inhibition. This review focuses mainly on the modulation of synaptic transmission at the spinal cord level and signaling in primary nociceptive neurons by AEA via CB1 and TRPV1 receptors. Furthermore, the spinal analgesic effect in preclinical studies and clinical aspects of AEA-mediated analgesia are considered.
Collapse
Affiliation(s)
- D Spicarova
- Laboratory of Pain Research, Institute of Physiology CAS, Praha 4, Czech Republic.
| | | |
Collapse
|
25
|
Machold R, Rudy B. Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity. Front Cell Neurosci 2024; 18:1414955. [PMID: 39113758 PMCID: PMC11303334 DOI: 10.3389/fncel.2024.1414955] [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: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
26
|
Botea MO, Andereggen L, Urman RD, Luedi MM, Romero CS. Cannabinoids for Acute Pain Management: Approaches and Rationale. Curr Pain Headache Rep 2024; 28:681-689. [PMID: 38607548 PMCID: PMC11271357 DOI: 10.1007/s11916-024-01252-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF THE REVIEW Acute pain management remains a challenge and postoperative pain is often undermanaged despite many available treatment options, also including cannabinoids. RECENT FINDINGS In the light of the opioid epidemic, there has been growing interest in alternative care bundles for pain management, including cannabinoids as potential treatment to decrease opioid prescribing. Despite the lack of solid evidence on the efficacy of cannabinoids, their use among patients with pain, including those using opioids, is currently increasing. This use is supported by data suggesting that cannabinoids could potentially contribute to a better pain management and to a reduction in opioid doses while maintaining effective analgesia with minimum side effects. The scientific basis for supporting the use of cannabis is extensive, although it does not necessarily translate into relevant clinical outcomes. The use of cannabinoids in acute pain did not always consistently show statistically significant results in improving acute pain. Large randomized, controlled trials evaluating diverse cannabis extracts are needed in different clinical pain populations to determine safety and efficacy.
Collapse
Affiliation(s)
- Mihai O Botea
- Department of Anaesthesiology and Critical Care, Medicover Pelican Clinic Hospital, Oradea, Romania
| | - Lukas Andereggen
- Department of Neurosurgery, Cantonal Hospital of Aarau, Aarau, Switzerland
- Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Richard D Urman
- Department of Anaesthesiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Markus M Luedi
- Department of Anaesthesiology, Rescue- and Pain Medicine, Cantonal Hospital of St, Gallen, St. Gallen, Switzerland.
- Department of Anaesthesiology and Pain Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Carolina S Romero
- Department of Anaesthesiology and Critical Care, Hospital General Universitario De Valencia, Valencia, Spain
- Research Methods Department, Universidad Europea de Valencia, Valencia, Spain
| |
Collapse
|
27
|
Fu X, Tasker JG. Neuromodulation of inhibitory synaptic transmission in the basolateral amygdala during fear and anxiety. Front Cell Neurosci 2024; 18:1421617. [PMID: 38994327 PMCID: PMC11236696 DOI: 10.3389/fncel.2024.1421617] [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: 04/22/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
The basolateral amygdala plays pivotal roles in the regulation of fear and anxiety and these processes are profoundly modulated by different neuromodulatory systems that are recruited during emotional arousal. Recent studies suggest activities of BLA interneurons and inhibitory synaptic transmission in BLA principal cells are regulated by neuromodulators to influence the output and oscillatory network states of the BLA, and ultimately the behavioral expression of fear and anxiety. In this review, we first summarize a cellular mechanism of stress-induced anxiogenesis mediated by the interaction of glucocorticoid and endocannabinoid signaling at inhibitory synapses in the BLA. Then we discuss cell type-specific activity patterns induced by neuromodulators converging on the Gq signaling pathway in BLA perisomatic parvalbumin-expressing (PV) and cholecystokinin-expressing (CCK) basket cells and their effects on BLA network oscillations and fear learning.
Collapse
Affiliation(s)
- Xin Fu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Jeffrey G. Tasker
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
28
|
Yoneda T, Kameyama K, Gotou T, Terata K, Takagi M, Yoshimura Y, Sakimura K, Kano M, Hata Y. Layer specific regulation of critical period timing and maturation of mouse visual cortex by endocannabinoids. iScience 2024; 27:110145. [PMID: 38952682 PMCID: PMC11215304 DOI: 10.1016/j.isci.2024.110145] [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: 05/25/2023] [Revised: 04/15/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024] Open
Abstract
Plasticity during the critical period is important for the functional maturation of cortical neurons. While characteristics of plasticity are diverse among cortical layers, it is unknown whether critical period timing is controlled by a common or unique molecular mechanism among them. We here clarified layer-specific regulation of the critical period timing of ocular dominance plasticity in the primary visual cortex. Mice lacking the endocannabinoid synthesis enzyme diacylglycerol lipase-α exhibited precocious critical period timing, earlier maturation of inhibitory synaptic function in layers 2/3 and 4, and impaired development of the binocular matching of orientation selectivity exclusively in layer 2/3. Activation of cannabinoid receptor restored ocular dominance plasticity at the normal critical period in layer 2/3. Suppression of GABAA receptor rescued precocious ocular dominance plasticity in layer 4. Therefore, endocannabinoids regulate critical period timing and maturation of visual function partly through the development of inhibitory synaptic functions in a layer-dependent manner.
Collapse
Affiliation(s)
- Taisuke Yoneda
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki 444-8585, Japan
| | - Katsuro Kameyama
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Takahiro Gotou
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Keiko Terata
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Masahiro Takagi
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Yumiko Yoshimura
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki 444-8585, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo 113-0033, Japan
- Advanced Comprehensive Research Organization (ACRO), Teikyo University, Tokyo 173-0003, Japan
| | - Yoshio Hata
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| |
Collapse
|
29
|
Carrascosa AJ, Navarrete F, Saldaña R, García-Gutiérrez MS, Montalbán B, Navarro D, Gómez-Guijarro FM, Gasparyan A, Murcia-Sánchez E, Torregrosa AB, Pérez-Doblado P, Gutiérrez L, Manzanares J. Cannabinoid Analgesia in Postoperative Pain Management: From Molecular Mechanisms to Clinical Reality. Int J Mol Sci 2024; 25:6268. [PMID: 38892456 PMCID: PMC11172912 DOI: 10.3390/ijms25116268] [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: 04/26/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Postoperative pain (POP) is a challenging clinical phenomenon that affects the majority of surgical patients and demands effective management to mitigate adverse outcomes such as persistent pain. The primary goal of POP management is to alleviate suffering and facilitate a seamless return to normal function for the patient. Despite compelling evidence of its drawbacks, opioid analgesia remains the basis of POP treatment. Novel therapeutic approaches rely on multimodal analgesia, integrating different pharmacological strategies to optimize efficacy while minimizing adverse effects. The recognition of the imperative role of the endocannabinoid system in pain regulation has prompted the investigation of cannabinoid compounds as a new therapeutic avenue. Cannabinoids may serve as adjuvants, enhancing the analgesic effects of other drugs and potentially replacing or at least reducing the dependence on other long-term analgesics in pain management. This narrative review succinctly summarizes pertinent information on the molecular mechanisms, clinical therapeutic benefits, and considerations associated with the plausible use of various cannabinoid compounds in treating POP. According to the available evidence, cannabinoid compounds modulate specific molecular mechanisms intimately involved in POP. However, only two of the eleven clinical trials that evaluated the efficacy of different cannabinoid interventions showed positive results.
Collapse
Affiliation(s)
- Antonio J. Carrascosa
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Raquel Saldaña
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - María S. García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Belinda Montalbán
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Fernando M. Gómez-Guijarro
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Elena Murcia-Sánchez
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Paloma Pérez-Doblado
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Luisa Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
30
|
Haller J. Herbal Cannabis and Depression: A Review of Findings Published over the Last Three Years. Pharmaceuticals (Basel) 2024; 17:689. [PMID: 38931356 PMCID: PMC11206863 DOI: 10.3390/ph17060689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Public perception contrasts scientific findings on the depression-related effects of cannabis. However, earlier studies were performed when cannabis was predominantly illegal, its production was mostly uncontrolled, and the idea of medical cannabis was incipient only. We hypothesized that recent changes in attitudes and legislations may have favorably affected research. In addition, publication bias against cannabis may have also decreased. To investigate this hypothesis, we conducted a review of research studies published over the last three years. We found 156 relevant research articles. In most cross-sectional studies, depression was higher in those who consumed cannabis than in those who did not. An increase in cannabis consumption was typically followed by an increase in depression, whereas withdrawal from cannabis ameliorated depression in most cases. Although medical cannabis reduced depression in most studies, none of these were placebo-controlled. In clinical studies published in the same period, the placebo also ameliorated depression and, in addition, the average effect size of the placebo was larger than the average effect size of medical cannabis. We also investigated the plausibility of the antidepressant effects of cannabis by reviewing molecular and pharmacological studies. Taken together, the reviewed findings do not support the antidepressant effects of herbal cannabis.
Collapse
Affiliation(s)
- Jozsef Haller
- Drug Research Institute, 1137 Budapest, Hungary;
- Department of Criminal Psychology, Faculty of Law Enforcement, Ludovika University of Public Service, 1083 Budapest, Hungary
| |
Collapse
|
31
|
Zhao Z, Covelo A, Couderc Y, Mitra A, Varilh M, Wu Y, Jacky D, Fayad R, Cannich A, Bellocchio L, Marsicano G, Beyeler A. Cannabinoids regulate an insula circuit controlling water intake. Curr Biol 2024; 34:1918-1929.e5. [PMID: 38636514 DOI: 10.1016/j.cub.2024.03.053] [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: 03/11/2022] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
The insular cortex, or insula, is a large brain region involved in the detection of thirst and the regulation of water intake. However, our understanding of the topographical, circuit, and molecular mechanisms for controlling water intake within the insula remains parcellated. We found that type-1 cannabinoid (CB1) receptors in the insular cortex cells participate in the regulation of water intake and deconstructed the circuit mechanisms of this control. Topographically, we revealed that the activity of excitatory neurons in both the anterior insula (aIC) and posterior insula (pIC) increases in response to water intake, yet only the specific removal of CB1 receptors in the pIC decreases water intake. Interestingly, we found that CB1 receptors are highly expressed in insula projections to the basolateral amygdala (BLA), while undetectable in the neighboring central part of the amygdala. Thus, we recorded the neurons of the aIC or pIC targeting the BLA (aIC-BLA and pIC-BLA) and found that they decreased their activity upon water drinking. Additionally, chemogenetic activation of pIC-BLA projection neurons decreased water intake. Finally, we uncovered CB1-dependent short-term synaptic plasticity (depolarization-induced suppression of excitation [DSE]) selectively in pIC-BLA, compared with aIC-BLA synapses. Altogether, our results support a model where CB1 receptor signaling promotes water intake by inhibiting the pIC-BLA pathway, thereby contributing to the fine top-down control of thirst responses.
Collapse
Affiliation(s)
- Zhe Zhao
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France; Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Ana Covelo
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Yoni Couderc
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Arojit Mitra
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Marjorie Varilh
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Yifan Wu
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Débora Jacky
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Rim Fayad
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Astrid Cannich
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Luigi Bellocchio
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France
| | - Giovanni Marsicano
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France.
| | - Anna Beyeler
- INSERM 1215, Neurocentre Magendie, University of Bordeaux, 146 rue Léo Saignat, 33000 Bordeaux, France.
| |
Collapse
|
32
|
Gupta S, Bharatha A, Cohall D, Rahman S, Haque M, Azim Majumder MA. Aerobic Exercise and Endocannabinoids: A Narrative Review of Stress Regulation and Brain Reward Systems. Cureus 2024; 16:e55468. [PMID: 38440201 PMCID: PMC10910469 DOI: 10.7759/cureus.55468] [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] [Accepted: 03/04/2024] [Indexed: 03/06/2024] Open
Abstract
Aerobic exercise is a widely adopted practice, not solely for enhancing fitness and reducing the risk of various diseases but also for its ability to uplift mood and aid in addressing depression and anxiety disorders. Within the scope of this narrative review, we seek to consolidate current insights into the endocannabinoid-mediated regulation of stress and the brain's reward mechanism resulting from engaging in aerobic exercise. A comprehensive search was conducted across Medline, SPORTDiscus, Pubmed, and Scopus, encompassing data available until November 30, 2023. This review indicates that a bout of aerobic exercise, particularly of moderate intensity, markedly augments circulating levels of endocannabinoids - N-arachidonoyl-ethanolamine (AEA) and 2-acylglycerol (2-AG), that significantly contributes to mood elevation and reducing stress in healthy individuals. The current understanding of how aerobic exercise impacts mental health and mood improvement is still unclear. Moderate and high-intensity aerobic exercise modulates stress through a negative feedback mechanism targeting both the hypothalamus-pituitary-adrenal (HPA) axis and the sympathetic nervous system, thereby facilitating stress regulation crucial role in endocannabinoid synthesis, ultimately culminating in the orchestration of negative feedback across multiple tiers of the HPA axis, coupled with its influence over cortical and subcortical brain structures. The endocannabinoid has been observed to govern the release of neurotransmitters from diverse neuronal populations, implying a universal mechanism that fine-tunes neuronal activity and consequently modulates both emotional and stress-related responses. Endocannabinoids further assume a pivotal function within brain reward mechanisms, primarily mediated by CB1 receptors distributed across diverse cerebral centers. Notably, these endocannabinoids partake in natural reward processes, as exemplified in aerobic exercise, by synergizing with the dopaminergic reward system. The genesis of this reward pathway can be traced to the ventral tegmental area, with dopamine neurons predominantly projecting to the nucleus accumbens, thereby inciting dopamine release in response to rewarding stimuli.
Collapse
Affiliation(s)
- Subir Gupta
- Physiology, Faculty of Medical Sciences, The University of the West Indies, Cave Hill Campus, Bridgetown, BRB
| | - Ambadasu Bharatha
- Pharmacology, Faculty of Medical Sciences, The University of the West Indies, Cave Hill Campus, Bridgetown, BRB
| | - Damian Cohall
- Pharmacology, Faculty of Medical Sciences, The University of the West Indies, Cave Hill Campus, Bridgetown, BRB
| | - Sayeeda Rahman
- Pharmacology, School of Medicine, American University of Integrative Sciences, Bridgetown, BRB
| | - Mainul Haque
- Pharmacology and Therapeutics, Karnavati Scientific Research Center (KSRC) School of Dentistry, Karnavati University, Gandhinagar, IND
- Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
| | - Md Anwarul Azim Majumder
- Medical Education, Faculty of Medical Sciences, The University of the West Indies, Cave Hill Campus, Bridgetown, BRB
| |
Collapse
|
33
|
Dudok B, Fan LZ, Farrell JS, Malhotra S, Homidan J, Kim DK, Wenardy C, Ramakrishnan C, Li Y, Deisseroth K, Soltesz I. Retrograde endocannabinoid signaling at inhibitory synapses in vivo. Science 2024; 383:967-970. [PMID: 38422134 PMCID: PMC10921710 DOI: 10.1126/science.adk3863] [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: 08/18/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024]
Abstract
Endocannabinoid (eCB)-mediated suppression of inhibitory synapses has been hypothesized, but this has not yet been demonstrated to occur in vivo because of the difficulty in tracking eCB dynamics and synaptic plasticity during behavior. In mice navigating a linear track, we observed location-specific eCB signaling in hippocampal CA1 place cells, and this was detected both in the postsynaptic membrane and the presynaptic inhibitory axons. All-optical in vivo investigation of synaptic responses revealed that postsynaptic depolarization was followed by a suppression of inhibitory synaptic potentials. Furthermore, interneuron-specific cannabinoid receptor deletion altered place cell tuning. Therefore, rapid, postsynaptic, activity-dependent eCB signaling modulates inhibitory synapses on a timescale of seconds during behavior.
Collapse
Affiliation(s)
- Barna Dudok
- Departments of Neurology and Neuroscience, Baylor College of Medicine; Houston, TX, 77030, USA
- Department of Neurosurgery, Stanford University; Stanford, CA, 94305, USA
| | - Linlin Z. Fan
- Department of Bioengineering, Stanford University; Stanford, CA, 94305, USA
| | - Jordan S. Farrell
- Department of Neurosurgery, Stanford University; Stanford, CA, 94305, USA
- F.M. Kirby Neurobiology Center and Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital; Boston, MA, 02115, USA
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Shreya Malhotra
- Department of Neurosurgery, Stanford University; Stanford, CA, 94305, USA
| | - Jesslyn Homidan
- Department of Neurosurgery, Stanford University; Stanford, CA, 94305, USA
| | - Doo Kyung Kim
- Department of Bioengineering, Stanford University; Stanford, CA, 94305, USA
| | - Celestine Wenardy
- Department of Bioengineering, Stanford University; Stanford, CA, 94305, USA
| | - Charu Ramakrishnan
- Cracking the Neural Code (CNC) Program, Stanford University; Stanford, CA, 94305, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University; Beijing, 100871, China
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University; Stanford, CA, 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University; Stanford, CA, 94305, USA
- Howard Hughes Medical Institute; Stanford, CA, 94305, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University; Stanford, CA, 94305, USA
| |
Collapse
|
34
|
Nguyen R, Sivakumaran S, Lambe EK, Kim JC. Ventral hippocampal cholecystokinin interneurons gate contextual reward memory. iScience 2024; 27:108824. [PMID: 38303709 PMCID: PMC10831933 DOI: 10.1016/j.isci.2024.108824] [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: 04/21/2023] [Revised: 11/06/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Associating contexts with rewards depends on hippocampal circuits, with local inhibitory interneurons positioned to play an important role in shaping activity. Here, we demonstrate that the encoding of context-reward memory requires a ventral hippocampus (vHPC) to nucleus accumbens (NAc) circuit that is gated by cholecystokinin (CCK) interneurons. In a sucrose conditioned place preference (CPP) task, optogenetically inhibiting vHPC-NAc terminals impaired the acquisition of place preference. Transsynaptic rabies tracing revealed vHPC-NAc neurons were monosynaptically innervated by CCK interneurons. Using intersectional genetic targeting of CCK interneurons, ex vivo optogenetic activation of CCK interneurons increased GABAergic transmission onto vHPC-NAc neurons, while in vivo optogenetic inhibition of CCK interneurons increased cFos in these projection neurons. Notably, CCK interneuron inhibition during sucrose CPP learning increased time spent in the sucrose-associated location, suggesting enhanced place-reward memory. Our findings reveal a previously unknown hippocampal microcircuit crucial for modulating the strength of contextual reward learning.
Collapse
Affiliation(s)
- Robin Nguyen
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of OBGYN, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
Cherry AL, Wheeler MJ, Mathisova K, Di Miceli M. In silico analyses of the involvement of GPR55, CB1R and TRPV1: response to THC, contribution to temporal lobe epilepsy, structural modeling and updated evolution. Front Neuroinform 2024; 18:1294939. [PMID: 38404644 PMCID: PMC10894036 DOI: 10.3389/fninf.2024.1294939] [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: 09/28/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction The endocannabinoid (eCB) system is named after the discovery that endogenous cannabinoids bind to the same receptors as the phytochemical compounds found in Cannabis. While endogenous cannabinoids include anandamide (AEA) and 2-arachidonoylglycerol (2-AG), exogenous phytocannabinoids include Δ-9 tetrahydrocannabinol (THC) and cannabidiol (CBD). These compounds finely tune neurotransmission following synapse activation, via retrograde signaling that activates cannabinoid receptor 1 (CB1R) and/or transient receptor potential cation channel subfamily V member 1 (TRPV1). Recently, the eCB system has been linked to several neurological diseases, such as neuro-ocular abnormalities, pain insensitivity, migraine, epilepsy, addiction and neurodevelopmental disorders. In the current study, we aim to: (i) highlight a potential link between the eCB system and neurological disorders, (ii) assess if THC exposure alters the expression of eCB-related genes, and (iii) identify evolutionary-conserved residues in CB1R or TRPV1 in light of their function. Methods To address this, we used several bioinformatic approaches, such as transcriptomic (Gene Expression Omnibus), protein-protein (STRING), phylogenic (BLASTP, MEGA) and structural (Phyre2, AutoDock, Vina, PyMol) analyzes. Results Using RNA sequencing datasets, we did not observe any dysregulation of eCB-related transcripts in major depressive disorders, bipolar disorder or schizophrenia in the anterior cingulate cortex, nucleus accumbens or dorsolateral striatum. Following in vivo THC exposure in adolescent mice, GPR55 was significantly upregulated in neurons from the ventral tegmental area, while other transcripts involved in the eCB system were not affected by THC exposure. Our results also suggest that THC likely induces neuroinflammation following in vitro application on mice microglia. Significant downregulation of TPRV1 occurred in the hippocampi of mice in which a model of temporal lobe epilepsy was induced, confirming previous observations. In addition, several transcriptomic dysregulations were observed in neurons of both epileptic mice and humans, which included transcripts involved in neuronal death. When scanning known interactions for transcripts involved in the eCB system (n = 12), we observed branching between the eCB system and neurophysiology, including proteins involved in the dopaminergic system. Our protein phylogenic analyzes revealed that CB1R forms a clade with CB2R, which is distinct from related paralogues such as sphingosine-1-phosphate, receptors, lysophosphatidic acid receptors and melanocortin receptors. As expected, several conserved residues were identified, which are crucial for CB1R receptor function. The anandamide-binding pocket seems to have appeared later in evolution. Similar results were observed for TRPV1, with conserved residues involved in receptor activation. Conclusion The current study found that GPR55 is upregulated in neurons following THC exposure, while TRPV1 is downregulated in temporal lobe epilepsy. Caution is advised when interpreting the present results, as we have employed secondary analyzes. Common ancestors for CB1R and TRPV1 diverged from jawless vertebrates during the late Ordovician, 450 million years ago. Conserved residues are identified, which mediate crucial receptor functions.
Collapse
Affiliation(s)
- Amy L. Cherry
- Worcester Biomedical Research Group, School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| | - Michael J. Wheeler
- Sustainable Environments Research Group, School of Science and the Environment University of Worcester, Worcester, United Kingdom
| | - Karolina Mathisova
- School of Science and the Environment University of Worcester, Worcester, United Kingdom
| | - Mathieu Di Miceli
- Worcester Biomedical Research Group, School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| |
Collapse
|
36
|
Wei M, Yang L, Su F, Liu Y, Zhao X, Luo L, Sun X, Liu S, Dong Z, Zhang Y, Shi YS, Liang J, Zhang C. ABHD6 drives endocytosis of AMPA receptors to regulate synaptic plasticity and learning flexibility. Prog Neurobiol 2024; 233:102559. [PMID: 38159878 DOI: 10.1016/j.pneurobio.2023.102559] [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: 06/23/2023] [Revised: 10/26/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Trafficking of α-Amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors (AMPARs), mediated by AMPAR interacting proteins, enabled neurons to maintain tuning capabilities at rest or active state. α/β-Hydrolase domain-containing 6 (ABHD6), an endocannabinoid hydrolase, was an AMPAR auxiliary subunit found to negatively regulate the surface delivery of AMPARs. While ABHD6 was found to prevent AMPAR tetramerization in endoplasmic reticulum, ABHD6 was also reported to localize at postsynaptic site. Yet, the role of ABHD6 interacting with AMPAR at postsynaptic site, and the physiological significance of ABHD6 regulating AMPAR trafficking remains elusive. Here, we generated the ABHD6 knockout (ABHD6KO) mice and found that deletion of ABHD6 selectively enhanced AMPAR-mediated basal synaptic responses and the surface expression of postsynaptic AMPARs. Furthermore, we found that loss of ABHD6 impaired hippocampal long-term depression (LTD) and synaptic downscaling in hippocampal synapses. AMPAR internalization assays revealed that ABHD6 was essential for neuronal activity-dependent endocytosis of surface AMPARs, which is independent of ABHD6's hydrolase activity. The defects of AMPAR endocytosis and LTD are expressed as deficits in learning flexibility in ABHD6KO mice. Collectively, we demonstrated that ABHD6 is an endocytic accessory protein promoting AMPAR endocytosis, thereby contributes to the formation of LTD, synaptic downscaling and reversal learning.
Collapse
Affiliation(s)
- Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China.
| | - Lei Yang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Feng Su
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ying Liu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xinyi Zhao
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Lin Luo
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Xinyue Sun
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Sen Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Zhaoqi Dong
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Yong Zhang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission of the PR China, IDG/McGovern Institute for Brain Research at PKU, Peking University, Beijing 100083, China
| | - Yun Stone Shi
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School, Nanjing University, Nanjing 210032, China
| | - Jing Liang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China.
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
37
|
Forte N, Nicois A, Marfella B, Mavaro I, D'Angelo L, Piscitelli F, Scandurra A, De Girolamo P, Baldelli P, Benfenati F, Di Marzo V, Cristino L. Early endocannabinoid-mediated depolarization-induced suppression of excitation delays the appearance of the epileptic phenotype in synapsin II knockout mice. Cell Mol Life Sci 2024; 81:37. [PMID: 38214769 PMCID: PMC11072294 DOI: 10.1007/s00018-023-05029-7] [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: 06/13/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 01/13/2024]
Abstract
The mechanism underlying the transition from the pre-symptomatic to the symptomatic state is a crucial aspect of epileptogenesis. SYN2 is a member of a multigene family of synaptic vesicle phosphoproteins playing a fundamental role in controlling neurotransmitter release. Human SYN2 gene mutations are associated with epilepsy and autism spectrum disorder. Mice knocked out for synapsin II (SynII KO) are prone to epileptic seizures that appear after 2 months of age. However, the involvement of the endocannabinoid system, known to regulate seizure development and propagation, in the modulation of the excitatory/inhibitory balance in the epileptic hippocampal network of SynII KO mice has not been explored. In this study, we investigated the impact of endocannabinoids on glutamatergic and GABAergic synapses at hippocampal dentate gyrus granule cells in young pre-symptomatic (1-2 months old) and adult symptomatic (5-8 months old) SynII KO mice. We observed an increase in endocannabinoid-mediated depolarization-induced suppression of excitation in young SynII KO mice, compared to age-matched wild-type controls. In contrast, the endocannabinoid-mediated depolarization-induced suppression of inhibition remained unchanged in SynII KO mice at both ages. This selective alteration of excitatory synaptic transmission was accompanied by changes in hippocampal endocannabinoid levels and cannabinoid receptor type 1 distribution among glutamatergic and GABAergic synaptic terminals contacting the granule cells of the dentate gyrus. Finally, inhibition of type-1 cannabinoid receptors in young pre-symptomatic SynII KO mice induced seizures during a tail suspension test. Our results suggest that endocannabinoids contribute to maintaining network stability in a genetic mouse model of human epilepsy.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Alessandro Nicois
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Brenda Marfella
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Isabella Mavaro
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy
| | - Anna Scandurra
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Paolo De Girolamo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy.
- Faculty of Medicine and Faculty of Agricultural and Food Sciences, Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QC, Canada.
- Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada.
- Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli (NA), Italy.
| |
Collapse
|
38
|
Hall RR, Cohall DH. The Relationship between Muscarinic and Cannabinoid Receptors in Neuronal Excitability and Epilepsy: A Review. Med Cannabis Cannabinoids 2024; 7:91-98. [PMID: 39015608 PMCID: PMC11250071 DOI: 10.1159/000538297] [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: 07/02/2023] [Accepted: 03/06/2024] [Indexed: 07/18/2024] Open
Abstract
Background Of the seventy million people who suffer from epilepsy, 40 percent of them become resistant to more than one antiepileptic medication and have a higher chance of death. While the classical definition of epilepsy was due to the imbalance between excitatory glutamatergic and inhibitory γ-aminobutyric acid (GABA)-ergic signalling, substantial evidence implicates muscarinic receptors in the regulation of neural excitability. Summary Cannabinoids have shown to reduce seizure activity and neuronal excitability in several epileptic models through the activation of muscarinic receptors with drugs which modulate their activity. Cannabinoids also have been effective in reducing antiepileptic activity in pharmaco-resistant individuals; however, the mechanism of its effects in temporal lobe epilepsy is not clear. Key Messages This review seeks to elucidate the relationship between muscarinic and cannabinoid receptors in epilepsy and neural excitability.
Collapse
Affiliation(s)
- Ryan Renaldo Hall
- Faculty of Medical Sciences, University of the West Indies, Cave Hill, Barbados
| | - Damian Hugh Cohall
- Faculty of Medical Sciences, University of the West Indies, Cave Hill, Barbados
| |
Collapse
|
39
|
Zhu C, Lan X, Wei Z, Yu J, Zhang J. Allosteric modulation of G protein-coupled receptors as a novel therapeutic strategy in neuropathic pain. Acta Pharm Sin B 2024; 14:67-86. [PMID: 38239234 PMCID: PMC10792987 DOI: 10.1016/j.apsb.2023.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/09/2023] [Accepted: 07/12/2023] [Indexed: 01/22/2024] Open
Abstract
Neuropathic pain is a debilitating pathological condition that presents significant therapeutic challenges in clinical practice. Unfortunately, current pharmacological treatments for neuropathic pain lack clinical efficacy and often lead to harmful adverse reactions. As G protein-coupled receptors (GPCRs) are widely distributed throughout the body, including the pain transmission pathway and descending inhibition pathway, the development of novel neuropathic pain treatments based on GPCRs allosteric modulation theory is gaining momentum. Extensive research has shown that allosteric modulators targeting GPCRs on the pain pathway can effectively alleviate symptoms of neuropathic pain while reducing or eliminating adverse effects. This review aims to provide a comprehensive summary of the progress made in GPCRs allosteric modulators in the treatment of neuropathic pain, and discuss the potential benefits and adverse factors of this treatment. We will also concentrate on the development of biased agonists of GPCRs, and based on important examples of biased agonist development in recent years, we will describe universal strategies for designing structure-based biased agonists. It is foreseeable that, with the continuous improvement of GPCRs allosteric modulation and biased agonist theory, effective GPCRs allosteric drugs will eventually be available for the treatment of neuropathic pain with acceptable safety.
Collapse
Affiliation(s)
- Chunhao Zhu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaobing Lan
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Zhiqiang Wei
- Medicinal Chemistry and Bioinformatics Center, Ocean University of China, Qingdao 266100, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jian Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
40
|
Samanta A, Aleman-Zapata A, Agarwal K, Özsezer P, Alonso A, van der Meij J, Rayan A, Navarro-Lobato I, Genzel L. CBD lengthens sleep but shortens ripples and leads to intact simple but worse cumulative memory. iScience 2023; 26:108327. [PMID: 38026151 PMCID: PMC10656268 DOI: 10.1016/j.isci.2023.108327] [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: 04/21/2023] [Revised: 09/21/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Cannabidiol (CBD) is on the rise as over-the-counter medication to treat sleep disturbances, anxiety, pain, and epilepsy due to its action on the excitatory/inhibitory balance in the brain. However, it remains unclear if CBD also leads to adverse effects on memory via changes of sleep macro- and microarchitecture. To investigate the effect of CBD on sleep and memory consolidation, we performed two experiments using the object space task testing for both simple and cumulative memory in rats. We show that oral CBD administration extended the sleep period but changed the properties of rest and non-REM sleep oscillations (delta, spindle, ripples). Specifically, CBD also led to less long (>100 ms) ripples and, consequently, worse cumulative memory consolidation. In contrast, simple memories were not affected. In sum, we can confirm the beneficial effect of CBD on sleep; however, this comes with changes in oscillations that negatively impact memory consolidation.
Collapse
Affiliation(s)
- Anumita Samanta
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Adrian Aleman-Zapata
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Kopal Agarwal
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Pelin Özsezer
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Alejandra Alonso
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Jacqueline van der Meij
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Abdelrahman Rayan
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Irene Navarro-Lobato
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| | - Lisa Genzel
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Postbus 9010, 6500 GL, Nijmegen
| |
Collapse
|
41
|
Yano H, Chitsazi R, Lucaj C, Tran P, Hoffman AF, Baumann MH, Lupica CR, Shi L. Subtle Structural Modification of a Synthetic Cannabinoid Receptor Agonist Drastically Increases its Efficacy at the CB1 Receptor. ACS Chem Neurosci 2023; 14:3928-3940. [PMID: 37847546 PMCID: PMC10623572 DOI: 10.1021/acschemneuro.3c00530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/26/2023] [Indexed: 10/18/2023] Open
Abstract
The emergence of synthetic cannabinoid receptor agonists (SCRAs) as illicit psychoactive substances has posed considerable public health risks, including fatalities. Many SCRAs exhibit much higher efficacy and potency compared with the phytocannabinoid Δ9-tetrahydrocannabinol (THC) at the cannabinoid receptor 1 (CB1R), leading to dramatic differences in signaling levels that can be toxic. In this study, we investigated the structure-activity relationships of aminoalkylindole SCRAs at CB1Rs, focusing on 5F-pentylindoles containing an amide linker attached to different head moieties. Using in vitro bioluminescence resonance energy transfer assays, we identified a few SCRAs exhibiting significantly higher efficacy in engaging the Gi protein and recruiting β-arrestin than the reference CB1R full agonist CP55940. Importantly, the extra methyl group on the head moiety of 5F-MDMB-PICA, as compared to that of 5F-MMB-PICA, led to a large increase in efficacy and potency at the CB1R. This pharmacological observation was supported by the functional effects of these SCRAs on glutamate field potentials recorded in hippocampal slices. Molecular modeling and simulations of the CB1R models bound with both of the SCRAs revealed critical structural determinants contributing to the higher efficacy of 5F-MDMB-PICA and how these subtle differences propagated to the receptor-G protein interface. Thus, we find that apparently minor structural changes in the head moiety of SCRAs can cause major changes in efficacy. Our results highlight the need for close monitoring of the structural modifications of newly emerging SCRAs and their potential for toxic drug responses in humans.
Collapse
Affiliation(s)
- Hideaki Yano
- Department
of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical
Sciences, Bouvé College of Health Sciences, Center for Drug
Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Rezvan Chitsazi
- Computational
Chemistry and Molecular Biophysics Section, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Christopher Lucaj
- Department
of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical
Sciences, Bouvé College of Health Sciences, Center for Drug
Discovery, Northeastern University, Boston, Massachusetts 02115, United States
| | - Phuong Tran
- Computational
Chemistry and Molecular Biophysics Section, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Alexander F. Hoffman
- Electrophysiology
Research Section, National Institutes of
Health, Baltimore, Maryland 21224, United States
| | - Michael H. Baumann
- Designer
Drug Research Unit, Intramural Research Program, National Institute
on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Carl R. Lupica
- Electrophysiology
Research Section, National Institutes of
Health, Baltimore, Maryland 21224, United States
| | - Lei Shi
- Computational
Chemistry and Molecular Biophysics Section, National Institutes of Health, Baltimore, Maryland 21224, United States
| |
Collapse
|
42
|
Haller J. Anxiety Modulation by Cannabinoids-The Role of Stress Responses and Coping. Int J Mol Sci 2023; 24:15777. [PMID: 37958761 PMCID: PMC10650718 DOI: 10.3390/ijms242115777] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Endocannabinoids were implicated in a variety of pathological conditions including anxiety and are considered promising new targets for anxiolytic drug development. The optimism concerning the potentials of this system for anxiolysis is probably justified. However, the complexity of the mechanisms affected by endocannabinoids, and discrepant findings obtained with various experimental approaches makes the interpretation of research results difficult. Here, we review the anxiety-related effects of the three main interventions used to study the endocannabinoid system: pharmacological agents active at endocannabinoid-binding sites present on both the cell membrane and in the cytoplasm, genetic manipulations targeting cannabinoid receptors, and function-enhancers represented by inhibitors of endocannabinoid degradation and transport. Binding-site ligands provide inconsistent findings probably because they activate a multitude of mechanisms concomitantly. More robust findings were obtained with genetic manipulations and particularly with function enhancers, which heighten ongoing endocannabinoid activation rather than affecting all mechanisms indiscriminately. The enhancement of ongoing activity appears to ameliorate stress-induced anxiety without consistent effects on anxiety in general. Limited evidence suggests that this effect is achieved by promoting active coping styles in critical situations. These findings suggest that the functional enhancement of endocannabinoid signaling is a promising drug development target for stress-related anxiety disorders.
Collapse
Affiliation(s)
- József Haller
- Drug Research Institute, 1137 Budapest, Hungary;
- Department of Criminal Psychology, University of Public Service, 1082 Budapest, Hungary
| |
Collapse
|
43
|
Reece AS, Hulse GK. Perturbation of 3D nuclear architecture, epigenomic aging and dysregulation, and cannabinoid synaptopathy reconfigures conceptualization of cannabinoid pathophysiology: part 2-Metabolome, immunome, synaptome. Front Psychiatry 2023; 14:1182536. [PMID: 37854446 PMCID: PMC10579598 DOI: 10.3389/fpsyt.2023.1182536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023] Open
Abstract
The second part of this paper builds upon and expands the epigenomic-aging perspective presented in Part 1 to describe the metabolomic and immunomic bases of the epigenomic-aging changes and then considers in some detail the application of these insights to neurotoxicity, neuronal epigenotoxicity, and synaptopathy. Cannabinoids are well-known to have bidirectional immunomodulatory activities on numerous parts of the immune system. Immune perturbations are well-known to impact the aging process, the epigenome, and intermediate metabolism. Cannabinoids also impact metabolism via many pathways. Metabolism directly impacts immune, genetic, and epigenetic processes. Synaptic activity, synaptic pruning, and, thus, the sculpting of neural circuits are based upon metabolic, immune, and epigenomic networks at the synapse, around the synapse, and in the cell body. Many neuropsychiatric disorders including depression, anxiety, schizophrenia, bipolar affective disorder, and autistic spectrum disorder have been linked with cannabis. Therefore, it is important to consider these features and their complex interrelationships in reaching a comprehensive understanding of cannabinoid dependence. Together these findings indicate that cannabinoid perturbations of the immunome and metabolome are important to consider alongside the well-recognized genomic and epigenomic perturbations and it is important to understand their interdependence and interconnectedness in reaching a comprehensive appreciation of the true nature of cannabinoid pathophysiology. For these reasons, a comprehensive appreciation of cannabinoid pathophysiology necessitates a coordinated multiomics investigation of cannabinoid genome-epigenome-transcriptome-metabolome-immunome, chromatin conformation, and 3D nuclear architecture which therefore form the proper mechanistic underpinning for major new and concerning epidemiological findings relating to cannabis exposure.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia, Crawley, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia, Crawley, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
44
|
Veres JM, Fekete Z, Müller K, Andrasi T, Rovira-Esteban L, Barabas B, Papp OI, Hajos N. Fear learning and aversive stimuli differentially change excitatory synaptic transmission in perisomatic inhibitory cells of the basal amygdala. Front Cell Neurosci 2023; 17:1120338. [PMID: 37731462 PMCID: PMC10507864 DOI: 10.3389/fncel.2023.1120338] [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: 12/09/2022] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
Inhibitory circuits in the basal amygdala (BA) have been shown to play a crucial role in associative fear learning. How the excitatory synaptic inputs received by BA GABAergic interneurons are influenced by memory formation, a network parameter that may contribute to learning processes, is still largely unknown. Here, we investigated the features of excitatory synaptic transmission received by the three types of perisomatic inhibitory interneurons upon cue-dependent fear conditioning and aversive stimulus and tone presentations without association. Acute slices were prepared from transgenic mice: one group received tone presentation only (conditioned stimulus, CS group), the second group was challenged by mild electrical shocks unpaired with the CS (unsigned unconditioned stimulus, unsigned US group) and the third group was presented with the CS paired with the US (signed US group). We found that excitatory synaptic inputs (miniature excitatory postsynaptic currents, mEPSCs) recorded in distinct interneuron types in the BA showed plastic changes with different patterns. Parvalbumin (PV) basket cells in the unsigned US and signed US group received mEPSCs with reduced amplitude and rate in comparison to the only CS group. Coupling the US and CS in the signed US group caused a slight increase in the amplitude of the events in comparison to the unsigned US group, where the association of CS and US does not take place. Excitatory synaptic inputs onto cholecystokinin (CCK) basket cells showed a markedly different change from PV basket cells in these behavioral paradigms: only the decay time was significantly faster in the unsigned US group compared to the only CS group, whereas the amplitude of mEPSCs increased in the signed US group compared to the only CS group. Excitatory synaptic inputs received by PV axo-axonic cells showed the least difference in the three behavioral paradigm: the only significant change was that the rate of mEPSCs increased in the signed US group when compared to the only CS group. These results collectively show that associative learning and aversive stimuli unpaired with CS cause different changes in excitatory synaptic transmission in BA perisomatic interneuron types, supporting the hypothesis that they play distinct roles in the BA network operations upon pain information processing and fear memory formation.
Collapse
Affiliation(s)
- Judit M. Veres
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Zsuzsanna Fekete
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Kinga Müller
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Tibor Andrasi
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Laura Rovira-Esteban
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Bence Barabas
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Orsolya I. Papp
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Norbert Hajos
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- The Linda and Jack Gill Center for Molecular Bioscience, Indiana University Bloomington, Bloomington, IN, United States
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN, United States
| |
Collapse
|
45
|
Machold R, Dellal S, Valero M, Zurita H, Kruglikov I, Meng JH, Hanson JL, Hashikawa Y, Schuman B, Buzsáki G, Rudy B. Id2 GABAergic interneurons comprise a neglected fourth major group of cortical inhibitory cells. eLife 2023; 12:e85893. [PMID: 37665123 PMCID: PMC10581691 DOI: 10.7554/elife.85893] [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: 12/30/2022] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Cortical GABAergic interneurons (INs) represent a diverse population of mainly locally projecting cells that provide specialized forms of inhibition to pyramidal neurons and other INs. Most recent work on INs has focused on subtypes distinguished by expression of Parvalbumin (PV), Somatostatin (SST), or Vasoactive Intestinal Peptide (VIP). However, a fourth group that includes neurogliaform cells (NGFCs) has been less well characterized due to a lack of genetic tools. Here, we show that these INs can be accessed experimentally using intersectional genetics with the gene Id2. We find that outside of layer 1 (L1), the majority of Id2 INs are NGFCs that express high levels of neuropeptide Y (NPY) and exhibit a late-spiking firing pattern, with extensive local connectivity. While much sparser, non-NGFC Id2 INs had more variable properties, with most cells corresponding to a diverse group of INs that strongly expresses the neuropeptide CCK. In vivo, using silicon probe recordings, we observed several distinguishing aspects of NGFC activity, including a strong rebound in activity immediately following the cortical down state during NREM sleep. Our study provides insights into IN diversity and NGFC distribution and properties, and outlines an intersectional genetics approach for further study of this underappreciated group of INs.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Shlomo Dellal
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Manuel Valero
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Hector Zurita
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Ilya Kruglikov
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - John Hongyu Meng
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Center for Neural Science, New York UniversityNew YorkUnited States
| | - Jessica L Hanson
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Yoshiko Hashikawa
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Benjamin Schuman
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - György Buzsáki
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
46
|
Shang H, Li P, Lin X, Cai Q, Li Z, Deng L, Song Y, Chen JF, Zhou J. Neuronal and astrocytic CB1R signaling differentially modulates goal-directed behavior and working memory by distinct temporal mechanisms. Neuropsychopharmacology 2023; 48:1520-1531. [PMID: 36694040 PMCID: PMC10425374 DOI: 10.1038/s41386-023-01533-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/26/2023]
Abstract
Several cognitive processes, including instrumental behavior and working memory, are controlled by endocannabinoids acting on cannabinoid receptor 1 (CB1R) in the brain through retrograde and presynaptic inhibition of GABA or glutamate release. However, the temporal mechanisms underlying the control of these cognitive processes by CB1Rs remain largely unknown. Here, we have developed a light-sensitive CB1R chimera (optoCB1R) by replacing the intracellular domains of bovine rhodopsin with those of human CB1R. We demonstrated that light stimulation of optoCB1R triggered canonical CB1R signaling by inhibiting cAMP (but not cGMP or IP1) signaling and activating the MAPK pathway in vitro or in vivo. Moreover, light stimulation of optoCB1R in corticostriatal glutamatergic neurons could temporally inhibit excitatory postsynaptic currents (EPSCs) at the level of seconds. Importantly, transient (3 s) and "time-locked", but not random, activation of optoCB1R signaling in corticostriatal neurons at the time of reward affected animal sensitivity to outcome devaluation and inhibited goal-directed behavior. However, prolonged (~30 min) but not transient (10 or 30 s) activation of astrocytic CB1R signaling in the hippocampus impaired working memory. Consequently, neuronal and astrocytic CB1R signaling differentially regulate working memory and goal-directed behavior through distinct temporal and cellular mechanisms. Ultimately, the pharmacological blockade of adenosine A2AR improved the neuronal and astrocytic CB1R-induced impairments in goal-directed behavior and working memory, possibly through modulation of EPSCs and c-Fos, respectively. Therefore, A2AR may represent a promising target for managing cognitive dysfunction resulting from the use of CB1R drugs.
Collapse
Affiliation(s)
- Huiping Shang
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peijun Li
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangxiang Lin
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qionghui Cai
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhihui Li
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lu Deng
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue Song
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiang-Fan Chen
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Jianhong Zhou
- Molecular Neuropharmacology Laboratory, School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
47
|
Zhuo Y, Luo B, Yi X, Dong H, Wan J, Cai R, Williams JT, Qian T, Campbell MG, Miao X, Li B, Wei Y, Li G, Wang H, Zheng Y, Watabe-Uchida M, Li Y. Improved dual-color GRAB sensors for monitoring dopaminergic activity in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554559. [PMID: 37662187 PMCID: PMC10473776 DOI: 10.1101/2023.08.24.554559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Dopamine (DA) plays multiple roles in a wide range of physiological and pathological processes via a vast network of dopaminergic projections. To fully dissect the spatiotemporal dynamics of DA release in both dense and sparsely innervated brain regions, we developed a series of green and red fluorescent GPCR activation-based DA (GRABDA) sensors using a variety of DA receptor subtypes. These sensors have high sensitivity, selectivity, and signal-to-noise properties with subsecond response kinetics and the ability to detect a wide range of DA concentrations. We then used these sensors in freely moving mice to measure both optogenetically evoked and behaviorally relevant DA release while measuring neurochemical signaling in the nucleus accumbens, amygdala, and cortex. Using these sensors, we also detected spatially resolved heterogeneous cortical DA release in mice performing various behaviors. These next-generation GRABDA sensors provide a robust set of tools for imaging dopaminergic activity under a variety of physiological and pathological conditions.
Collapse
Affiliation(s)
- Yizhou Zhuo
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- These authors contributed equally
| | - Bin Luo
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
- These authors contributed equally
| | - Xinyang Yi
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Hui Dong
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Ruyi Cai
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - John T. Williams
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Tongrui Qian
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Malcolm G. Campbell
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - Xiaolei Miao
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Bozhi Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing 100853, China
| | - Yu Wei
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Guochuan Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Yu Zheng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Mitsuko Watabe-Uchida
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing 102206, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518055, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
48
|
Harbour K, Cappel Z, Baccei ML. Effects of Corticosterone on the Excitability of Glutamatergic and GABAergic Neurons of the Adolescent Mouse Superficial Dorsal Horn. Neuroscience 2023; 526:290-304. [PMID: 37437798 PMCID: PMC10530204 DOI: 10.1016/j.neuroscience.2023.07.009] [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: 04/04/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Stress evokes age-dependent effects on pain sensitivity and commonly occurs during adolescence. However, the mechanisms linking adolescent stress and pain remain poorly understood, in part due to a lack of information regarding how stress hormones modulate the function of nociceptive circuits in the adolescent CNS. Here we investigate the short- and long-term effects of corticosterone (CORT) on the excitability of GABAergic and presumed glutamatergic neurons of the spinal superficial dorsal horn (SDH) in Gad1-GFP mice at postnatal days (P)21-P34. In situ hybridization revealed that glutamatergic SDH neurons expressed significantly higher mRNA levels of both glucocorticoid receptors (GR) and mineralocorticoid receptors (MR) compared to adjacent GABAergic neurons. The incubation of spinal cord slices with CORT (90 min) evoked select long-term changes in spontaneous synaptic transmission across both cell types in a sex-dependent manner, without altering the intrinsic firing of either Gad1-GFP+ or GFP- neurons. Meanwhile, the acute bath application of CORT significantly decreased the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs), as well as the frequency of miniature inhibitory postsynaptic currents (mIPSCs), in both cell types leading to a net reduction in the balance of spontaneous excitation vs. inhibition (E:I ratio). This CORT-induced reduction in the E:I ratio was not prevented by selective antagonists of either GR (mifepristone) or MR (eplerenone), although eplerenone blocked the effect on mEPSC amplitude. Collectively, these data suggest that corticosterone modulates synaptic function within the adolescent SDH which could influence the overall excitability and output of the spinal nociceptive network.
Collapse
Affiliation(s)
- Kyle Harbour
- Molecular, Cellular and Biochemical Pharmacology Graduate Program, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Zoe Cappel
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; American Society for Pharmacology and Experimental Therapeutics Summer Research Program, Department of Pharmacology and Systems Physiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Mark L Baccei
- Molecular, Cellular and Biochemical Pharmacology Graduate Program, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; American Society for Pharmacology and Experimental Therapeutics Summer Research Program, Department of Pharmacology and Systems Physiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA.
| |
Collapse
|
49
|
Xue Z, Ye L, Ge J, Lan Z, Zou X, Mao C, Bao X, Yu L, Xu Y, Zhu X. Wwl70-induced ABHD6 inhibition attenuates memory deficits and pathological phenotypes in APPswe/PS1dE9 mice. Pharmacol Res 2023; 194:106864. [PMID: 37480972 DOI: 10.1016/j.phrs.2023.106864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 07/04/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Synaptic dysfunction plays a crucial role in the pathogenesis of Alzheimer's disease (AD). α/β-hydrolase domain-containing 6 (ABHD6) contributes to synaptic dysfunctions, and ABHD6 inhibition has shown potential therapeutic value in neurological disorders. However, the role of ABHD6 in AD has not been fully defined. In this study, we demonstrated that adeno-associated virus (AAV) mediated shRNA targeting ABHD6 in hippocampal neurons attenuated synaptic dysfunction and memory impairment of APPswe/PS1dE9 (APP/PS1) mice, while it didn't affect the amyloid-beta (Aβ) levels and neuroinflammation in the brains. In addition, intraperitoneal injection of wwl70, a specific inhibitor of ABHD6, improved synaptic plasticity and memory function in APP/PS1 mice, which might attribute to the activation of endogenous cannabinoid signaling. Furthermore, wwl70 significantly decreased the Aβ levels and neuroinflammation in the hippocampus of AD mice, and enhanced Aβ phagocytized by microglia. In conclusion, for the first time our data have shown that ABHD6 inhibition might be a promising strategy for AD treatment, and wwl70 is a potential candidate for AD drug development pipeline.
Collapse
Affiliation(s)
- Zhiwei Xue
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Lei Ye
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Jianwei Ge
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Zhen Lan
- Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinxin Zou
- Department of Neurology, Drum Tower Hospital of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - Chenglu Mao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Linjie Yu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China; Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurology, Drum Tower Hospital of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China; Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China; Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
50
|
Piscura MK, Henderson-Redmond AN, Barnes RC, Mitra S, Guindon J, Morgan DJ. Mechanisms of cannabinoid tolerance. Biochem Pharmacol 2023; 214:115665. [PMID: 37348821 PMCID: PMC10528043 DOI: 10.1016/j.bcp.2023.115665] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
Cannabis has been used recreationally and medically for centuries, yet research into understanding the mechanisms of its therapeutic effects has only recently garnered more attention. There is evidence to support the use of cannabinoids for the treatment of chronic pain, muscle spasticity, nausea and vomiting due to chemotherapy, improving weight gain in HIV-related cachexia, emesis, sleep disorders, managing symptoms in Tourette syndrome, and patient-reported muscle spasticity from multiple sclerosis. However, tolerance and the risk for cannabis use disorder are two significant disadvantages for cannabinoid-based therapies in humans. Recent work has revealed prominent sex differences in the acute response and tolerance to cannabinoids in both humans and animal models. This review will discuss evidence demonstrating cannabinoid tolerance in rodents, non-human primates, and humans and our current understanding of the neuroadaptations occurring at the cannabinoid type 1 receptor (CB1R) that are responsible tolerance. CB1R expression is downregulated in tolerant animals and humans while there is strong evidence of CB1R desensitization in cannabinoid tolerant rodent models. Throughout the review, critical knowledge gaps are indicated and discussed, such as the lack of a neuroimaging probe to assess CB1R desensitization in humans. The review discusses the intracellular signaling pathways that are responsible for mediating CB1R desensitization and downregulation including the action of G protein-coupled receptor kinases, β-arrestin2 recruitment, c-Jun N-terminal kinases, protein kinase A, and the intracellular trafficking of CB1R. Finally, the review discusses approaches to reduce cannabinoid tolerance in humans based on our current understanding of the neuroadaptations and mechanisms responsible for this process.
Collapse
Affiliation(s)
- Mary K Piscura
- Department of Biomedical Sciences, Marshall University, Huntington, WV 25755, USA; Department of Biomedical Sciences, Edward Via College of Osteopathic Medicine, Auburn, AL 36832, USA
| | | | - Robert C Barnes
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Swarup Mitra
- Department of Biomedical Sciences, Marshall University, Huntington, WV 25755, USA
| | - Josée Guindon
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Daniel J Morgan
- Department of Biomedical Sciences, Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|