1
|
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
|
2
|
Chowdhury KU, Holden ME, Wiley MT, Suppiramaniam V, Reed MN. Effects of Cannabis on Glutamatergic Neurotransmission: The Interplay between Cannabinoids and Glutamate. Cells 2024; 13:1130. [PMID: 38994982 PMCID: PMC11240741 DOI: 10.3390/cells13131130] [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: 04/24/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
There has been a significant increase in the consumption of cannabis for both recreational and medicinal purposes in recent years, and its use can have long-term consequences on cognitive functions, including memory. Here, we review the immediate and long-term effects of cannabis and its derivatives on glutamatergic neurotransmission, with a focus on both the presynaptic and postsynaptic alterations. Several factors can influence cannabinoid-mediated changes in glutamatergic neurotransmission, including dosage, sex, age, and frequency of use. Acute exposure to cannabis typically inhibits glutamate release, whereas chronic use tends to increase glutamate release. Conversely, the postsynaptic alterations are more complicated than the presynaptic effects, as cannabis can affect the glutamate receptor expression and the downstream signaling of glutamate. All these effects ultimately influence cognitive functions, particularly memory. This review will cover the current research on glutamate-cannabis interactions, as well as the future directions of research needed to understand cannabis-related health effects and neurological and psychological aspects of cannabis use.
Collapse
Affiliation(s)
- Kawsar U. Chowdhury
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, GA 30144, USA
| | | | - Miles T. Wiley
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, GA 30144, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
3
|
Pearl-Dowler L, Posa L, Lopez-Canul M, Teggin A, Gobbi G. Anti-allodynic and medullary modulatory effects of a single dose of delta-9-tetrahydrocannabinol (THC) in neuropathic rats tolerant to morphine. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110805. [PMID: 37257771 DOI: 10.1016/j.pnpbp.2023.110805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
Neuropathic pain (NP) is often treated with opioids, the prolonged use of which causes tolerance to their analgesic effect and can potentially cause death by overdose. The phytocannabinoid delta-9-tetrahydrocannabinol (THC) may be an effective alternative analgesic to treat NP in morphine-tolerant subjects. Male Wistar rats developed NP after spared nerve injury, and were then treated with increasing doses of THC (1, 1.5, 2, 2.5, and 5 mg/kg, intraperitoneally), which reduced mechanical allodynia at the dose of 2.5 and 5 mg/kg. Another group of NP rats were treated with morphine (5 mg/kg, twice daily for 7 days, subcutaneously), until tolerance developed, and on day 8 received a single dose of THC (2.5 mg/kg), which significantly reduced mechanical allodynia. To evaluate the modulation of THC in the descending pain pathway, in vivo electrophysiological recordings of pronociceptive ON cells and antinociceptive OFF cells in the rostroventral medulla (RVM) were recorded after intra-PAG microinjection of THC (10 μg/μl). NP rats with morphine tolerance, compared to the control one, showed a tonic reduction of the spontaneous firing rate of ON cells by 44%, but the THC was able to further decrease it (a hallmark of many analgesic drugs acting at supraspinal level). On the other hand, the firing rate, of the antinociceptive OFF cells was increased after morphine tolerance by 133%, but the THC failed to further activate it. Altogether, these findings indicate that a single dose of THC produces antiallodynic effect in individuals with NP who are tolerant to morphine, acting mostly on the ON cells of the descending pain pathways, but not on OFF cells.
Collapse
Affiliation(s)
- Leora Pearl-Dowler
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Luca Posa
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Martha Lopez-Canul
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Alexandra Teggin
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada.
| |
Collapse
|
4
|
Rathod SS, Agrawal YO, Nakhate KT, Meeran MFN, Ojha S, Goyal SN. Neuroinflammation in the Central Nervous System: Exploring the Evolving Influence of Endocannabinoid System. Biomedicines 2023; 11:2642. [PMID: 37893016 PMCID: PMC10604915 DOI: 10.3390/biomedicines11102642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Neuroinflammation is a complex biological process that typically originates as a protective response in the brain. This inflammatory process is triggered by the release of pro-inflammatory substances like cytokines, prostaglandins, and reactive oxygen and nitrogen species from stimulated endothelial and glial cells, including those with pro-inflammatory functions, in the outer regions. While neuronal inflammation is common in various central nervous system disorders, the specific inflammatory pathways linked with different immune-mediated cell types and the various factors influencing the blood-brain barrier significantly contribute to disease-specific characteristics. The endocannabinoid system consists of cannabinoid receptors, endogenous cannabinoids, and enzymes responsible for synthesizing and metabolizing endocannabinoids. The primary cannabinoid receptor is CB1, predominantly found in specific brain regions such as the brainstem, cerebellum, hippocampus, and cortex. The presence of CB2 receptors in certain brain components, like cultured cerebellar granular cells, Purkinje fibers, and microglia, as well as in the areas like the cerebral cortex, hippocampus, and cerebellum is also evidenced by immunoblotting assays, radioligand binding, and autoradiography studies. Both CB1 and CB2 cannabinoid receptors exhibit noteworthy physiological responses and possess diverse neuromodulatory capabilities. This review primarily aims to outline the distribution of CB1 and CB2 receptors across different brain regions and explore their potential roles in regulating neuroinflammatory processes.
Collapse
Affiliation(s)
- Sumit S. Rathod
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.S.R.); (Y.O.A.); (K.T.N.)
- Department of Pharmacy, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, Maharashtra, India
| | - Yogeeta O. Agrawal
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.S.R.); (Y.O.A.); (K.T.N.)
| | - Kartik T. Nakhate
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.S.R.); (Y.O.A.); (K.T.N.)
| | - M. F. Nagoor Meeran
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Abu Dhabi P.O. Box 15551, United Arab Emirates;
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Abu Dhabi P.O. Box 15551, United Arab Emirates;
| | - Sameer N. Goyal
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.S.R.); (Y.O.A.); (K.T.N.)
| |
Collapse
|
5
|
Gajofatto A, Cardobi N, Gobbin F, Calabrese M, Turatti M, Benedetti MD. Resting-state functional connectivity in multiple sclerosis patients receiving nabiximols for spasticity. BMC Neurol 2023; 23:128. [PMID: 36991352 PMCID: PMC10052832 DOI: 10.1186/s12883-023-03171-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Nabiximols (Sativex®) is a cannabinoid approved for multiple sclerosis (MS)-related spasticity. Its mechanism of action is partially understood, and efficacy is variable. OBJECTIVE To conduct an exploratory analysis of brain networks connectivity changes on resting state (RS) functional MRI (fMRI) of MS patients treated with nabiximols. METHODS We identified a group of MS patients treated with Sativex® at Verona University Hospital, who underwent RS brain fMRI in the 4 weeks before (T0) and 4-8 weeks after (T1) treatment start. Sativex® response was defined as ≥ 20% spasticity Numerical Rating Scale score reduction at T1 vs. T0. Connectivity changes on fMRI were compared between T0 and T1 in the whole group and according to response status. ROI-to-ROI and seed-to-voxel connectivity were evaluated. RESULTS Twelve MS patients (7 males) were eligible for the study. Seven patients (58.3%) resulted Sativex® responders at T1. On fMRI analysis, Sativex® exposure was associated with global brain connectivity increase (particularly in responders), decreased connectivity of motor areas, and bidirectional connectivity changes of the left cerebellum with a number of cortical areas. CONCLUSIONS Nabiximols administration is associated with brain connectivity increase of MS patients with spasticity. Modulation of sensorimotor cortical areas and cerebellum connectivity could play a role in nabiximols effect.
Collapse
Affiliation(s)
- Alberto Gajofatto
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, Verona, 37134, Italy.
- Unit of Neurology, Regional Multiple Sclerosis Center, Borgo Roma Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| | - Nicolò Cardobi
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, Verona, 37134, Italy
| | - Francesca Gobbin
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, Verona, 37134, Italy
- Unit of Neurology, Regional Multiple Sclerosis Center, Borgo Roma Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Massimiliano Calabrese
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Piazzale L.A. Scuro 10, Verona, 37134, Italy
- Unit of Neurology, Regional Multiple Sclerosis Center, Borgo Roma Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Marco Turatti
- Unit of Neurology, Regional Multiple Sclerosis Center, Borgo Roma Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Maria Donata Benedetti
- Unit of Neurology, Regional Multiple Sclerosis Center, Borgo Roma Hospital, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
6
|
Sitzia G, Abrahao KP, Liput D, Calandra GM, Lovinger DM. Distinct mechanisms of CB1 and GABA B receptor presynaptic modulation of striatal indirect pathway projections to mouse globus pallidus. J Physiol 2023; 601:195-209. [PMID: 36412169 PMCID: PMC10107704 DOI: 10.1113/jp283614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Presynaptic modulation is a fundamental process regulating synaptic transmission. Striatal indirect pathway projections originate from A2A-expressing spiny projection neurons (iSPNs), targeting the globus pallidus external segment (GPe) and control the firing of the tonically active GPe neurons via GABA release. It is unclear if and how the presynaptic G-protein-coupled receptors (GPCRs), GABAB and CB1 receptors modulate iSPN-GPe projections. Here we used an optogenetic platform to study presynaptic Ca2+ and GABAergic transmission at iSPN projections, using a genetic strategy to express the calcium sensor GCaMP6f or the excitatory channelrhodopsin (hChR2) on iSPNs. We found that P/Q-type calcium channels are the primary voltage-gated Ca2+ channel (VGCC) subtype controlling presynaptic calcium and GABA release at iSPN-GPe projections. N-type and L-type VGCCs also contribute to GABA release at iSPN-GPe synapses. GABAB receptor activation resulted in a reversible inhibition of presynaptic Ca2+ transients (PreCaTs) and an inhibition of GABAergic transmission at iSPN-GPe synapses. CB1 receptor activation did not inhibit PreCaTs but inhibited GABAergic transmission at iSPN-GPe projections. CB1 effects on GABAergic transmission persisted in experiments where NaV and KV 1 were blocked, indicating a VGCC- and KV 1-independent presynaptic mechanism of action of CB1 receptors. Taken together, presynaptic modulation of iSPN-GPe projections by CB1 and GABAB receptors is mediated by distinct mechanisms. KEY POINTS: P/Q-type are the predominant voltage-gated Ca2+ channels controlling presynaptic Ca2+ and GABA release on the striatal indirect pathway projections. GABAB receptors modulate iSPN-GPe projections via a VGCC-dependent mechanism. CB1 receptors modulate iSPN-GPe projections via a VGCC-independent mechanism.
Collapse
Affiliation(s)
- Giacomo Sitzia
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and AlcoholismUS National Institutes of HealthRockvilleMarylandUSA
- Molecular Neurophysiology LaboratoryDepartment of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
| | - Karina Possa Abrahao
- Departamento de PsicobiologiaUniversidade Federal de São PauloSão PauloSao PauloBrazil
| | - Daniel Liput
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and AlcoholismUS National Institutes of HealthRockvilleMarylandUSA
| | - Gian Marco Calandra
- Institute for Stroke and Dementia ResearchLudwig‐Maximilians‐UniversitätMunichGermany
| | - David M. Lovinger
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and AlcoholismUS National Institutes of HealthRockvilleMarylandUSA
| |
Collapse
|
7
|
Hammad AM, Meknas SJ, Hall FS, Hikmat S, Sari Y, Al-Qirim TM, Alfaraj M, Amawi H. Effects of waterpipe tobacco smoke and ceftriaxone treatment on the expression of endocannabinoid receptors in mesocorticolimbic brain regions. Brain Res Bull 2022; 185:56-63. [PMID: 35490908 DOI: 10.1016/j.brainresbull.2022.04.014] [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: 12/09/2021] [Revised: 04/15/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022]
Abstract
Chronic tobacco exposure can alter the endocannabinoid (eCB) system, consequently leading to an anxiety state. In this study, we investigated the effects of waterpipe tobacco smoke (WTS) on cannabinoid receptor 1 and 2 (CBR1 and CBR2) gene and protein expression in mesocorticolimbic brain regions. Using elevated plus maze (EPM) and open field (OF) tests, the effects of WTS exposure on withdrawal-induced anxiety-like behavior were examined. The effect of ceftriaxone (CEF), a β-lactam known to upregulate glutamate transporter 1 (GLT-1), on anxiety and the expression of cannabinoid receptors was also determined. Male Sprague-Dawley rats were randomly assigned to four groups: 1) the Control group was exposed only to standard room air; 2) the WTS group was exposed to tobacco smoke and treated with saline vehicle; 3) the WTS-CEF group was exposed to WTS and treated with ceftriaxone; and 4) the CEF group was exposed only to standard room air and treated with ceftriaxone. Rats were exposed to WTS (or room air) for two hours per day, five days per week for a period of four weeks. Behavioral tests (EPM and OF) were conducted weekly during acute withdrawal, 24 h following WTS exposure. Rats were given either saline or ceftriaxone (200 mg/kg i.p.) for five days during Week 4, 30 min prior to WTS exposure. Withdrawal-induced anxiety was induced by WTS exposure but was reduced by ceftriaxone treatment. WTS exposure decreased CBR1 mRNA and protein expression in the NAc and VTA, but not PFC, and ceftriaxone treatment attenuated these effects. WTS exposure did not change CBR2 mRNA expression in the NAc, VTA, or PFC. These findings demonstrate that WTS exposure dysregulated the endocannabinoid system and increased anxiety-like behavior, and these effects were reversed by ceftriaxone treatment, which suggest the involvement of glutamate transporter 1 in these effects.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan.
| | - Sara Jamal Meknas
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Suhair Hikmat
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - T M Al-Qirim
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Malek Alfaraj
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Haneen Amawi
- Department of Clinical Pharmacy and Pharmacy Practice, College of Pharmacy, Yarmouk University, Irbid 21163, Jordan
| |
Collapse
|
8
|
Sugaya Y, Kano M. Endocannabinoid-Mediated Control of Neural Circuit Excitability and Epileptic Seizures. Front Neural Circuits 2022; 15:781113. [PMID: 35046779 PMCID: PMC8762319 DOI: 10.3389/fncir.2021.781113] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/29/2021] [Indexed: 01/11/2023] Open
Abstract
Research on endocannabinoid signaling has greatly advanced our understanding of how the excitability of neural circuits is controlled in health and disease. In general, endocannabinoid signaling at excitatory synapses suppresses excitability by inhibiting glutamate release, while that at inhibitory synapses promotes excitability by inhibiting GABA release, although there are some exceptions in genetically epileptic animal models. In the epileptic brain, the physiological distributions of endocannabinoid signaling molecules are disrupted during epileptogenesis, contributing to the occurrence of spontaneous seizures. However, it is still unknown how endocannabinoid signaling changes during seizures and how the redistribution of endocannabinoid signaling molecules proceeds during epileptogenesis. Recent development of cannabinoid sensors has enabled us to investigate endocannabinoid signaling in much greater spatial and temporal details than before. Application of cannabinoid sensors to epilepsy research has elucidated activity-dependent changes in endocannabinoid signaling during seizures. Furthermore, recent endocannabinoid research has paved the way for the clinical use of cannabidiol for the treatment of refractory epilepsy, such as Dravet syndrome, Lennox-Gastaut syndrome and tuberous sclerosis complex. Cannabidiol significantly reduces seizures and is considered to have comparable tolerability to conventional antiepileptic drugs. In this article, we introduce recent advances in research on the roles of endocannabinoid signaling in epileptic seizures and discuss future directions.
Collapse
Affiliation(s)
- Yuki Sugaya
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
- *Correspondence: Masanobu Kano,
| |
Collapse
|
9
|
Boczek T, Zylinska L. Receptor-Dependent and Independent Regulation of Voltage-Gated Ca 2+ Channels and Ca 2+-Permeable Channels by Endocannabinoids in the Brain. Int J Mol Sci 2021; 22:ijms22158168. [PMID: 34360934 PMCID: PMC8348342 DOI: 10.3390/ijms22158168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/27/2022] Open
Abstract
The activity of specific populations of neurons in different brain areas makes decisions regarding proper synaptic transmission, the ability to make adaptations in response to different external signals, as well as the triggering of specific regulatory pathways to sustain neural function. The endocannabinoid system (ECS) appears to be a very important, highly expressed, and active system of control in the central nervous system (CNS). Functionally, it allows the cells to respond quickly to processes that occur during synaptic transmission, but can also induce long-term changes. The endocannabinoids (eCBs) belong to a large family of bioactive lipid mediators that includes amides, esters, and ethers of long-chain polyunsaturated fatty acids. They are produced “on demand” from the precursors located in the membranes, exhibit a short half-life, and play a key role as retrograde messengers. eCBs act mainly through two receptors, CB1R and CB2R, which belong to the G-protein coupled receptor superfamily (GPCRs), but can also exert their action via multiple non-receptor pathways. The action of eCBs depends on Ca2+, but eCBs can also regulate downstream Ca2+ signaling. In this short review, we focus on the regulation of neuronal calcium channels by the most effective members of eCBs-2-arachidonoylglycerol (2-AG), anandamide (AEA) and originating from AEA-N-arachidonoylglycine (NAGly), to better understand the contribution of ECS to brain function under physiological conditions.
Collapse
|
10
|
Davatolhagh MF, Fuccillo MV. Neurexin1⍺ differentially regulates synaptic efficacy within striatal circuits. Cell Rep 2021; 34:108773. [PMID: 33626349 PMCID: PMC8071350 DOI: 10.1016/j.celrep.2021.108773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/18/2020] [Accepted: 01/29/2021] [Indexed: 02/08/2023] Open
Abstract
Mutations in genes essential for synaptic function, such as the presynaptic adhesion molecule Neurexin1α (Nrxn1α), are strongly implicated in neuropsychiatric pathophysiology. As the input nucleus of the basal ganglia, the striatum integrates diverse excitatory projections governing cognitive and motor control, and its impairment may represent a recurrent pathway to disease. Here, we test the functional relevance of Nrxn1α in striatal circuits by employing optogenetic-mediated afferent recruitment of dorsal prefrontal cortical (dPFC) and parafascicular thalamic connections onto dorsomedial striatal (DMS) spiny projection neurons (SPNs). For dPFC-DMS circuits, we find decreased synaptic strength specifically onto indirect pathway SPNs in both Nrxn1α+/- and Nrxn1α-/- mice, driven by reductions in neurotransmitter release. In contrast, thalamic excitatory inputs to DMS exhibit relatively normal excitatory synaptic strength despite changes in synaptic N-methyl-D-aspartate receptor (NMDAR) content. These findings suggest that dysregulation of Nrxn1α modulates striatal function in an input- and target-specific manner.
Collapse
Affiliation(s)
- M Felicia Davatolhagh
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc V Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Yadav-Samudrala BJ, Fitting S. Mini-review: The therapeutic role of cannabinoids in neuroHIV. Neurosci Lett 2021; 750:135717. [PMID: 33587986 DOI: 10.1016/j.neulet.2021.135717] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/25/2022]
Abstract
In the era of combined antiretroviral therapy (cART), human immunodeficiency virus type 1 (HIV-1) is considered a chronic disease with an inflammatory component that specifically targets the brain and causes a high prevalence of HIV-1-associated neurocognitive disorders (HAND). The endocannabinoid (eCB) system has attracted interest as a target for treatment of neurodegenerative disorders, due to the potential anti-inflammatory and neuroprotective properties of cannabinoids, including its potential therapeutic use in HIV-1 neuropathogenesis. In this review, we summarize what is currently known about the structural and functional changes of the eCB system under conditions of HAND. This will be followed by summarizing the current clinical and preclinical findings on the effects of cannabis use and cannabinoids in the context of HIV-1 infection, with specifically focusing on viral load, cognition, inflammation, and neuroprotection. Lastly, we present some potential future directions to better understand the involvement of the eCB system and the role that cannabis use and cannabinoids play in neuroHIV.
Collapse
Affiliation(s)
- Barkha J Yadav-Samudrala
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Sylvia Fitting
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
12
|
De Caro C, Cristiano C, Avagliano C, Cuozzo M, La Rana G, Aviello G, De Sarro G, Calignano A, Russo E, Russo R. Analgesic and Anti-Inflammatory Effects of Perampanel in Acute and Chronic Pain Models in Mice: Interaction With the Cannabinergic System. Front Pharmacol 2021; 11:620221. [PMID: 33597883 PMCID: PMC7883473 DOI: 10.3389/fphar.2020.620221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Pain conditions, such as neuropathic pain (NP) and persistent inflammatory pain are therapeutically difficult to manage. Previous studies have shown the involvement of glutamate receptor in pain modulation and in particular same of these showed the key role of the AMPA ionotropic glutamate receptor subtype. Antiseizure medications (ASMs) are often used to treat this symptom, however the effect of perampanel (PER), an ASM acting as selective, non-competitive inhibitor of the AMPA receptor on the management of pain has not well been investigated yet. Here we tested the potential analgesic and anti-inflammatory effects of PER, in acute and chronic pain models. PER was given orally either in acute (5 mg/kg) or repeated administration (3 mg/kg/d for 4 days). Pain response was assessed using models of nociceptive sensitivity, visceral and inflammatory pain, and mechanical allodynia and hyperalgesia induced by chronic constriction injury to the sciatic nerve. PER significantly reduced pain perception in all behavioral tests as well as CCI-induced mechanical allodynia and hyperalgesia in acute regimen (5 mg/kg). This effect was also observed after repeated treatment using the dose of 3 mg/kg/d. The antinociceptive, antiallodynic and antihyperalgesic effects of PER were attenuated when the CB1 antagonist AM251 (1 mg/kg/i.p.) was administered before PER treatment, suggesting the involvement of the cannabinergic system. Moreover, Ex vivo analyses showed that PER significantly increased CB1 receptor expression and reduced inflammatory cytokines (i.e. TNFα, IL-1β, and IL-6) in the spinal cord. In conclusion, these results extend our knowledge on PER antinociceptive and antiallodynic effects and support the involvement of cannabinergic system on its mode of action.
Collapse
Affiliation(s)
- Carmen De Caro
- Department of Health Sciences, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Claudia Cristiano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Giovanna La Rana
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gabriella Aviello
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Giovambattista De Sarro
- Department of Health Sciences, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Emilio Russo
- Department of Health Sciences, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Roberto Russo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
13
|
Reid HMO, Lysenko-Martin MR, Snowden TM, Thomas JD, Christie BR. A Systematic Review of the Effects of Perinatal Alcohol Exposure and Perinatal Marijuana Exposure on Adult Neurogenesis in the Dentate Gyrus. Alcohol Clin Exp Res 2020; 44:1164-1174. [PMID: 32246781 PMCID: PMC7905844 DOI: 10.1111/acer.14332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Marijuana and alcohol are both substances that, when used during pregnancy, may have profound effects on the developing fetus. There is evidence to suggest that both drugs have the capacity to affect working memory, one function of the hippocampal formation; however, there is a paucity of data on how perinatal exposure to alcohol or cannabis impacts the process of adult neurogenesis. METHODS This systematic review examines immunohistochemical data from adult rat and mouse models that assess perinatal alcohol or perinatal marijuana exposure. A comprehensive list of search terms was designed and used to search 3 separate databases. All results were imported to Mendeley and screened by 2 authors. Consensus was reached on a set of final papers that met the inclusion criteria, and their results were summarized. RESULTS Twelve papers were identified as relevant, 10 of which pertained to the effects of perinatal alcohol on the adult hippocampus, and 2 pertained to the effects of perinatal marijuana on the adult hippocampus. Cellular proliferation in the dentate gyrus was not affected in adult rats and mice exposed to alcohol perinatally. In general, perinatal alcohol exposure did not have a significant and reliable effect on the maturation and survival of adult born granule neurons in the dentate gyrus. In contrast, interneuron numbers appear to be reduced in the dentate gyrus of adult rats and mice exposed perinatally to alcohol. Perinatal marijuana exposure was also found to reduce inhibitory interneuron numbers in the dentate gyrus. CONCLUSIONS Perinatal alcohol exposure and perinatal marijuana exposure both act on inhibitory interneurons in the hippocampal formation of adult rats. These findings suggest simultaneous perinatal alcohol and marijuana exposure (SAM) may have a dramatic impact on inhibitory processes in the dentate gyrus.
Collapse
Affiliation(s)
- Hannah M O Reid
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Melanie R Lysenko-Martin
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Taylor M Snowden
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Jennifer D Thomas
- Center for Behavioral Teratology, (JDT), San Diego State University, San Diego, California
| | - Brian R Christie
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, (BRC), University of British Columbia, Victoria, British Columbia
| |
Collapse
|
14
|
Bouchet CA, Ingram SL. Cannabinoids in the descending pain modulatory circuit: Role in inflammation. Pharmacol Ther 2020; 209:107495. [PMID: 32004514 PMCID: PMC7183429 DOI: 10.1016/j.pharmthera.2020.107495] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/17/2020] [Indexed: 01/09/2023]
Abstract
The legalization of cannabis in some states has intensified interest in the potential for cannabis and its constituents to lead to novel therapeutics for pain. Our understanding of the cellular mechanisms underlying cannabinoid actions in the brain have lagged behind opioids; however, the current opioid epidemic has also increased attention on the use of cannabinoids as alternatives to opioids for pain, especially chronic pain that requires long-term use. Endogenous cannabinoids are lipid signaling molecules that have complex roles in modulating neuronal function throughout the brain. In this review, we discuss cannabinoid functions in the descending pain modulatory pathway, a brain circuit that integrates cognitive and emotional processing of pain to modulate incoming sensory inputs. In addition, we highlight areas where further studies are necessary to understand cannabinoid regulation of descending pain modulation.
Collapse
Affiliation(s)
- Courtney A Bouchet
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR 97239, United States of America
| | - Susan L Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR 97239, United States of America.
| |
Collapse
|
15
|
CB 1 Activity Drives the Selection of Navigational Strategies: A Behavioral and c-Fos Immunoreactivity Study. Int J Mol Sci 2020; 21:ijms21031072. [PMID: 32041135 PMCID: PMC7036945 DOI: 10.3390/ijms21031072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 11/26/2022] Open
Abstract
To promote efficient explorative behaviors, subjects adaptively select spatial navigational strategies based on landmarks or a cognitive map. The hippocampus works alone or in conjunction with the dorsal striatum, both representing the neuronal underpinnings of the navigational strategies organized on the basis of different systems of spatial coordinate integration. The high expression of cannabinoid type 1 (CB1) receptors in structures related to spatial learning—such as the hippocampus, dorsal striatum and amygdala—renders the endocannabinoid system a critical target to study the balance between landmark- and cognitive map-based navigational strategies. In the present study, mice treated with the CB1-inverse agonist/antagonist AM251 or vehicle were trained on a Circular Hole Board, a task that could be solved through either navigational strategy. At the end of the behavioral testing, c-Fos immunoreactivity was evaluated in specific nuclei of the hippocampus, dorsal striatum and amygdala. AM251 treatment impaired spatial learning and modified the pattern of the performed navigational strategies as well as the c-Fos immunoreactivity in the hippocampus, dorsal striatum and amygdala. The present findings shed light on the involvement of CB1 receptors as part of the selection system of the navigational strategies implemented to efficiently solve the spatial problem.
Collapse
|
16
|
Blumenstock S, Dudanova I. Cortical and Striatal Circuits in Huntington's Disease. Front Neurosci 2020; 14:82. [PMID: 32116525 PMCID: PMC7025546 DOI: 10.3389/fnins.2020.00082] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 12/28/2022] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder that typically manifests in midlife with motor, cognitive, and/or psychiatric symptoms. The disease is caused by a CAG triplet expansion in exon 1 of the huntingtin gene and leads to a severe neurodegeneration in the striatum and cortex. Classical electrophysiological studies in genetic HD mouse models provided important insights into the disbalance of excitatory, inhibitory and neuromodulatory inputs, as well as progressive disconnection between the cortex and striatum. However, the involvement of local cortical and striatal microcircuits still remains largely unexplored. Here we review the progress in understanding HD-related impairments in the cortical and basal ganglia circuits, and outline new opportunities that have opened with the development of modern circuit analysis methods. In particular, in vivo imaging studies in mouse HD models have demonstrated early structural and functional disturbances within the cortical network, and optogenetic manipulations of striatal cell types have started uncovering the causal roles of certain neuronal populations in disease pathogenesis. In addition, the important contribution of astrocytes to HD-related circuit defects has recently been recognized. In parallel, unbiased systems biology studies are providing insights into the possible molecular underpinnings of these functional defects at the level of synaptic signaling and neurotransmitter metabolism. With these approaches, we can now reach a deeper understanding of circuit-based HD mechanisms, which will be crucial for the development of effective and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sonja Blumenstock
- Department of Molecules – Signaling – Development, Max Planck Institute of Neurobiology, Martinsried, Germany
- Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Irina Dudanova
- Molecular Neurodegeneration Group, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
17
|
Melin E, Nanobashvili A, Avdic U, Gøtzsche CR, Andersson M, Woldbye DPD, Kokaia M. Disease Modification by Combinatorial Single Vector Gene Therapy: A Preclinical Translational Study in Epilepsy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:179-193. [PMID: 31660420 PMCID: PMC6807261 DOI: 10.1016/j.omtm.2019.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
Gene therapy has been suggested as a plausible novel approach to achieve seizure control in patients with focal epilepsy that do not adequately respond to pharmacological treatment. We investigated the seizure-suppressant potential of combinatorial neuropeptide Y and Y2 receptor single vector gene therapy based on adeno-associated virus serotype 1 (AAV1) in rats. First, a dose-response study in the systemic kainate-induced acute seizure model was performed, whereby the 1012 genomic particles (gp)/mL titer of the vector was selected as an optimal concentration. Second, an efficacy study was performed in the intrahippocampal kainate chronic model of spontaneous recurrent seizures (SRSs), designed to reflect a likely clinical scenario, with magnetic resonance image (MRI)-guided focal unilateral administration of the vector in the hippocampus during the chronic stage of the disease. The efficacy study demonstrated a favorable outcome of the gene therapy, with a 31% responder rate (more than 50% reduction in SRS frequency) and 13% seizure-freedom rate, whereas no such effects were observed in the control animals. The inter-SRS and SRS cluster intervals were also significantly prolonged in the treated group compared to controls. In addition, the SRS duration was significantly reduced in the treated group but not in the controls. This study establishes the SRS-suppressant ability of the single vector combinatorial neuropeptide Y/Y2 receptor gene therapy in a clinically relevant chronic model of epilepsy.
Collapse
Affiliation(s)
- Esbjörn Melin
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Avtandil Nanobashvili
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden.,CombiGene AB, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden
| | - Una Avdic
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Casper R Gøtzsche
- CombiGene AB, Medicon Village, Scheelevägen 2, 223 81 Lund, Sweden.,Laboratory of Neural Plasticity, Center for Neuroscience, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - My Andersson
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - David P D Woldbye
- Laboratory of Neural Plasticity, Center for Neuroscience, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Merab Kokaia
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| |
Collapse
|
18
|
Jacobs IR, Xu C, Hermes DJ, League AF, Xu C, Nath B, Jiang W, Niphakis MJ, Cravatt BF, Mackie K, Mukhopadhyay S, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Inhibitory Control Deficits Associated with Upregulation of CB 1R in the HIV-1 Tat Transgenic Mouse Model of Hand. J Neuroimmune Pharmacol 2019; 14:661-678. [PMID: 31372820 PMCID: PMC6898753 DOI: 10.1007/s11481-019-09867-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 07/11/2019] [Indexed: 12/14/2022]
Abstract
In the era of combined antiretroviral therapy, HIV-1 infected individuals are living longer lives; however, longevity is met with an increasing number of HIV-1 associated neurocognitive disorders (HAND) diagnoses. The transactivator of transcription (Tat) is known to mediate the neurotoxic effects in HAND by acting directly on neurons and also indirectly via its actions on glia. The Go/No-Go (GNG) task was used to examine HAND in the Tat transgenic mouse model. The GNG task involves subjects discriminating between two stimuli sets in order to determine whether or not to inhibit a previously trained response. Data reveal inhibitory control deficits in female Tat(+) mice (p = .048) and an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group (p < .05). A significant negative correlation was noted between inhibitory control and IL CB1R expression (r = −.543, p = .045), with CB1R expression predicting 30% of the variance of inhibitory control (R2 = .295, p = .045). Furthermore, there was a significant increase in spontaneous excitatory postsynaptic current (sEPSC) frequencies in Tat(+) compared to Tat(−) mice (p = .008, across sexes). The increase in sEPSC frequency was significantly attenuated by bath application of PF3845, a fatty acid amide hydrolase (FAAH) enzyme inhibitor (p < .001). Overall, the GNG task is a viable measure to assess inhibitory control deficits in Tat transgenic mice and results suggest a potential therapeutic treatment for the observed deficits with drugs which modulate endocannabinoid enzyme activity. Results of the Go/No-Go operant conditioning task reveal inhibitory control deficits in female transgenic Tat(+) mice without significantly affecting males. The demonstrated inhibitory control deficits appear to be associated with an upregulation of cannabinoid type 1 receptors (CB1R) in the infralimbic (IL) cortex in the same female Tat(+) group. ![]()
Collapse
MESH Headings
- AIDS Dementia Complex/genetics
- AIDS Dementia Complex/metabolism
- AIDS Dementia Complex/psychology
- Animals
- Disease Models, Animal
- Female
- HIV-1
- Inhibition, Psychological
- Limbic Lobe/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neurocognitive Disorders/genetics
- Neurocognitive Disorders/metabolism
- Psychomotor Performance/physiology
- Receptor, Cannabinoid, CB1/biosynthesis
- Receptor, Cannabinoid, CB1/genetics
- Up-Regulation/physiology
- tat Gene Products, Human Immunodeficiency Virus/biosynthesis
- tat Gene Products, Human Immunodeficiency Virus/genetics
Collapse
Affiliation(s)
- Ian R Jacobs
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Changqing Xu
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Douglas J Hermes
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexis F League
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Callie Xu
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Bhupendra Nath
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC, 27707, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Micah J Niphakis
- The Skaggs Institute for Chemical Biology, Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology, Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ken Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Somnath Mukhopadhyay
- Department of Chemistry & Biochemistry, North Carolina Central University, Durham, NC, 27707, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | | | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
19
|
Bassir Nia A, Bender R, Harpaz-Rotem I. Endocannabinoid System Alterations in Posttraumatic Stress Disorder: A Review of Developmental and Accumulative Effects of Trauma. ACTA ACUST UNITED AC 2019; 3. [PMID: 31660473 PMCID: PMC6816276 DOI: 10.1177/2470547019864096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The role of the endocannabinoid system in stress-related psychiatric symptoms has
been investigated in many animal and human studies. Although most of these
studies consistently report long-lasting effects of prolonged stress and trauma
on the endocannabinoid system, the nature and direction of these changes are
controversial. We reviewed the available preclinical and clinical studies
investigating the endocannabinoid system alterations long after chronic stress
and trauma. We propose that the effects of prolonged stress or trauma on the
endocannabinoid system are different based on the developmental age of subjects
at the time of experiencing the trauma and its repetitiveness and accumulative
effects. The current literature consistently demonstrates decreased levels of
endocannabinoid ligands and receptors if the trauma occurs in childhood, whereas
decreased levels of endocannabinoid ligands and increased levels of cannabinoid
receptors are reported when trauma has happened in adulthood. It is important to
note that these changes are region-specific in the brain and also there are
important sex differences, which are beyond the scope of this review.
Collapse
Affiliation(s)
| | - Ricci Bender
- Yale School of Medicine, Yale University, New Haven, CT, USA
| | | |
Collapse
|
20
|
Smith‐Dijak AI, Sepers MD, Raymond LA. Alterations in synaptic function and plasticity in Huntington disease. J Neurochem 2019; 150:346-365. [DOI: 10.1111/jnc.14723] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/28/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Amy I. Smith‐Dijak
- Graduate Program in Neuroscience the University of British Columbia Vancouver British Columbia Canada
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| | - Marja D. Sepers
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| | - Lynn A. Raymond
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health the University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
21
|
Burke KJ, Bender KJ. Modulation of Ion Channels in the Axon: Mechanisms and Function. Front Cell Neurosci 2019; 13:221. [PMID: 31156397 PMCID: PMC6533529 DOI: 10.3389/fncel.2019.00221] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/01/2019] [Indexed: 12/11/2022] Open
Abstract
The axon is responsible for integrating synaptic signals, generating action potentials (APs), propagating those APs to downstream synapses and converting them into patterns of neurotransmitter vesicle release. This process is mediated by a rich assortment of voltage-gated ion channels whose function can be affected on short and long time scales by activity. Moreover, neuromodulators control the activity of these proteins through G-protein coupled receptor signaling cascades. Here, we review cellular mechanisms and signaling pathways involved in axonal ion channel modulation and examine how changes to ion channel function affect AP initiation, AP propagation, and the release of neurotransmitter. We then examine how these mechanisms could modulate synaptic function by focusing on three key features of synaptic information transmission: synaptic strength, synaptic variability, and short-term plasticity. Viewing these cellular mechanisms of neuromodulation from a functional perspective may assist in extending these findings to theories of neural circuit function and its neuromodulation.
Collapse
Affiliation(s)
| | - Kevin J. Bender
- Neuroscience Graduate Program and Department of Neurology, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
22
|
Zurawski Z, Yim YY, Alford S, Hamm HE. The expanding roles and mechanisms of G protein-mediated presynaptic inhibition. J Biol Chem 2019; 294:1661-1670. [PMID: 30710014 PMCID: PMC6364771 DOI: 10.1074/jbc.tm118.004163] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Throughout the past five decades, tremendous advancements have been made in our understanding of G protein signaling and presynaptic inhibition, many of which were published in the Journal of Biological Chemistry under the tenure of Herb Tabor as Editor-in-Chief. Here, we identify these critical advances, including the formulation of the ternary complex model of G protein-coupled receptor signaling and the discovery of Gβγ as a critical signaling component of the heterotrimeric G protein, along with the nature of presynaptic inhibition and its physiological role. We provide an overview for the discovery and physiological relevance of the two known Gβγ-mediated mechanisms for presynaptic inhibition: first, the action of Gβγ on voltage-gated calcium channels to inhibit calcium influx to the presynaptic active zone and, second, the direct binding of Gβγ to the SNARE complex to displace synaptotagmin downstream of calcium entry, which has been demonstrated to be important in neurons and secretory cells. These two mechanisms act in tandem with each other in a synergistic manner to provide more complete spatiotemporal control over neurotransmitter release.
Collapse
Affiliation(s)
- Zack Zurawski
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600; Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois 60612-7308
| | - Yun Young Yim
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois 60612-7308
| | - Heidi E Hamm
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600.
| |
Collapse
|
23
|
Hakami AY, Alshehri FS, Sari Y. β-lactams modulate astroglial glutamate transporters and attenuate dependence to CP 55,940, a CB1 receptor agonist, in rat model. Behav Brain Res 2019; 359:709-718. [PMID: 30257184 DOI: 10.1016/j.bbr.2018.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
Abstract
Studies on cannabinoids have reported contradictory findings, showing both aversion and rewarding outcomes in conditioned place preference (CPP). Various possibilities have been suggested to explain the aversive properties of cannabinoids, including the pharmacokinetics profile and dose selection. In this study, we have established a CPP method to investigate the effects of modulating astroglial glutamate transporters in cannabinoid dependence using a cannabinoid receptor 1 (CB1R) agonist, CP 55,940 (CP). Previous reports using CPP paradigm demonstrated the involvement of glutamatergic system in seeking behavior of several drugs of abuse such as cocaine, heroin and nicotine. Glutamate homeostasis is maintained by several astroglial glutamate transporters, such as glutamate transporter 1 (GLT-1), cystine/glutamate transporter (xCT) and glutamate aspartate transporter (GLAST). In this study, we investigated the effects of Ampicillin/Sulbactam, β-lactam compounds known to upregulate GLT-1 and xCT, on cannabinoid seeking behavior using CP. We found first that one prime dose of CP induced CP reinstatement; this effect was associated, in part, with significant downregulation of xCT expression in the nucleus accumbens, dorsomedial prefrontal cortex and amygdala. Moreover, GLT-1 expression was downregulated in the amygdala. Importantly, Ampicillin/Sulbactam treatment during the extinction phase attenuated CP-induced reinstatement and restored the expression of GLT-1 and xCT in mesocorticolimbic brain regions. These findings suggest that β-lactams may play a potential therapeutic role in attenuating dependence to cannabinoids, in part, through upregulation of GLT-1 and xCT.
Collapse
Affiliation(s)
- Alqassem Y Hakami
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Fahad S Alshehri
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
24
|
Endocannabinoid System and Alcohol Abuse Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1162:89-127. [PMID: 31332736 DOI: 10.1007/978-3-030-21737-2_6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Δ9-tetrahydrocannabinol (Δ9-THC), the primary active component in Cannabis sativa preparations such as hashish and marijuana, signals by binding to cell surface receptors. Two types of receptors have been cloned and characterized as cannabinoid (CB) receptors. CB1 receptors (CB1R) are ubiquitously present in the central nervous system (CNS) and are present in both inhibitory interneurons and excitatory neurons at the presynaptic terminal. CB2 receptors (CB2R) are demonstrated in microglial cells, astrocytes, and several neuron subpopulations and are present in both pre- and postsynaptic terminals. The majority of studies on these receptors have been conducted in the past two and half decades after the identification of the molecular constituents of the endocannabinoid (eCB) system that started with the characterization of CB1R. Subsequently, the seminal discovery was made, which suggested that alcohol (ethanol) alters the eCB system, thus establishing the contribution of the eCB system in the motivation to consume ethanol. Several preclinical studies have provided evidence that CB1R significantly contributes to the motivational and reinforcing properties of ethanol and that the chronic consumption of ethanol alters eCB transmitters and CB1R expression in the brain nuclei associated with addiction pathways. Additionally, recent seminal studies have further established the role of the eCB system in the development of ethanol-induced developmental disorders, such as fetal alcohol spectrum disorders (FASD). These results are augmented by in vitro and ex vivo studies, showing that acute and chronic treatment with ethanol produces physiologically relevant alterations in the function of the eCB system during development and in the adult stage. This chapter provides a current and comprehensive review of the literature concerning the role of the eCB system in alcohol abuse disorders (AUD).
Collapse
|
25
|
Molecular Imaging in Huntington's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 142:289-333. [PMID: 30409256 DOI: 10.1016/bs.irn.2018.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huntington's disease (HD) is a rare monogenic neurodegenerative disorder caused by a trinucleotide CAG repeat expansion in the huntingtin gene resulting in the formation of intranuclear inclusions of mutated huntingtin. The accumulation of mutated huntingtin leads to loss of GABAergic medium spiny neurons (MSNs); subsequently resulting in the development of chorea, cognitive dysfunction and psychiatric symptoms. Premanifest HD gene expansion carriers, provide a unique cohort to examine very early molecular changes, occurring before the development of overt symptoms, to elucidate disease pathophysiology and identify reliable biomarkers of HD progression. Positron emission tomography (PET) is a non-invasive molecular imaging technique allowing the evaluation of specific molecular targets in vivo. Selective PET radioligands provide invaluable tools to investigate the role of the dopaminergic system, brain metabolism, microglial activation, phosphodiesterase 10A, and cannabinoid, GABA, adenosine and opioid receptors in HD. PET has been employed to monitor disease progression aiming to identify a reliable biomarker to predict phenoconversion from premanifest to manifest HD.
Collapse
|
26
|
Derouiche L, Massotte D. G protein-coupled receptor heteromers are key players in substance use disorder. Neurosci Biobehav Rev 2018; 106:73-90. [PMID: 30278192 DOI: 10.1016/j.neubiorev.2018.09.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
G protein-coupled receptors (GPCR) represent the largest family of membrane proteins in the human genome. Physical association between two different GPCRs is linked to functional interactions which generates a novel entity, called heteromer, with specific ligand binding and signaling properties. Heteromerization is increasingly recognized to take place in the mesocorticolimbic pathway and to contribute to various aspects related to substance use disorder. This review focuses on heteromers identified in brain areas relevant to drug addiction. We report changes at the molecular and cellular levels that establish specific functional impact and highlight behavioral outcome in preclinical models. Finally, we briefly discuss selective targeting of native heteromers as an innovative therapeutic option.
Collapse
Affiliation(s)
- Lyes Derouiche
- Institut des Neurosciences Cellulaires et Integratives, UPR 3212, 5 rue Blaise Pascal, F-67000 Strasbourg, France
| | - Dominique Massotte
- Institut des Neurosciences Cellulaires et Integratives, UPR 3212, 5 rue Blaise Pascal, F-67000 Strasbourg, France.
| |
Collapse
|
27
|
Sugaya Y, Kano M. Control of excessive neural circuit excitability and prevention of epileptic seizures by endocannabinoid signaling. Cell Mol Life Sci 2018; 75:2793-2811. [PMID: 29737364 PMCID: PMC11105219 DOI: 10.1007/s00018-018-2834-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022]
Abstract
Progress in research on endocannabinoid signaling has greatly advanced our understanding of how it controls neural circuit excitability in health and disease. In general, endocannabinoid signaling at excitatory synapses suppresses seizures by inhibiting glutamate release. In contrast, endocannabinoid signaling promotes seizures by inhibiting GABA release at inhibitory synapses. The physiological distribution of endocannabinoid signaling molecules becomes disrupted with the development of epileptic focus in patients with mesial temporal lobe epilepsy and in animal models of experimentally induced epilepsy. Augmentation of endocannabinoid signaling can promote the development of epileptic focus at initial stages. However, at later stages, increased endocannabinoid signaling delays it and suppresses spontaneous seizures. Thus, the regulation of endocannabinoid signaling at specific synapses that cause hyperexcitability during particular stages of disease development may be effective for treating epilepsy and epileptogenesis.
Collapse
Affiliation(s)
- Yuki Sugaya
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, 113-0033, Japan.
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
28
|
Parallel, but Dissociable, Processing in Discrete Corticostriatal Inputs Encodes Skill Learning. Neuron 2017; 96:476-489.e5. [PMID: 29024667 DOI: 10.1016/j.neuron.2017.09.040] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 07/20/2017] [Accepted: 09/22/2017] [Indexed: 12/24/2022]
Abstract
Changes in cortical and striatal function underlie the transition from novel actions to refined motor skills. How discrete, anatomically defined corticostriatal projections function in vivo to encode skill learning remains unclear. Using novel fiber photometry approaches to assess real-time activity of associative inputs from medial prefrontal cortex to dorsomedial striatum and sensorimotor inputs from motor cortex to dorsolateral striatum, we show that associative and sensorimotor inputs co-engage early in action learning and disengage in a dissociable manner as actions are refined. Disengagement of associative, but not sensorimotor, inputs predicts individual differences in subsequent skill learning. Divergent somatic and presynaptic engagement in both projections during early action learning suggests potential learning-related in vivo modulation of presynaptic corticostriatal function. These findings reveal parallel processing within associative and sensorimotor circuits that challenges and refines existing views of corticostriatal function and expose neuronal projection- and compartment-specific activity dynamics that encode and predict action learning.
Collapse
|
29
|
Martella G, Meringolo M, Trobiani L, De Jaco A, Pisani A, Bonsi P. The neurobiological bases of autism spectrum disorders: the R451C-neuroligin 3 mutation hampers the expression of long-term synaptic depression in the dorsal striatum. Eur J Neurosci 2017; 47:701-708. [PMID: 28921757 DOI: 10.1111/ejn.13705] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 09/07/2017] [Accepted: 09/11/2017] [Indexed: 02/03/2023]
Abstract
Autism spectrum disorders (ASDs) comprise a heterogeneous group of disorders with a complex genetic etiology. Current theories on the pathogenesis of ASDs suggest that they might arise from an aberrant synaptic transmission affecting specific brain circuits and synapses. The striatum, which is part of the basal ganglia circuit, is one of the brain regions involved in ASDs. Mouse models of ASDs have provided evidence for an imbalance between excitatory and inhibitory neurotransmission. Here, we investigated the expression of long-term synaptic plasticity at corticostriatal glutamatergic synapses in the dorsal striatum of the R451C-NL3 phenotypic mouse model of autism. This mouse model carries the human R451C mutation in the neuroligin 3 (NL3) gene that has been associated with highly penetrant autism in a Swedish family. The R451C-NL3 mouse has been shown to exhibit autistic-like behaviors and alterations of synaptic transmission in different brain areas. However, excitatory glutamatergic transmission and its long-term plasticity have not been investigated in the dorsal striatum so far. Our results indicate that the expression of long-term synaptic depression (LTD) at corticostriatal glutamatergic synapses in the dorsal striatum is impaired by the R451C-NL3 mutation. A partial rescue of LTD was obtained by exogenous activation of cannabinoid CB1 receptors or enhancement of the endocannabinoid tone, suggesting that an altered cannabinoid drive might underlie the deficit of synaptic plasticity in the dorsal striatum of R451C-NL3 mice.
Collapse
Affiliation(s)
- Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Laura Trobiani
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy
| | - Antonella De Jaco
- Department of Biology and Biotechnologies 'Charles Darwin', Sapienza University of Rome, Rome, Italy
| | - Antonio Pisani
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
30
|
Stanslowsky N, Jahn K, Venneri A, Naujock M, Haase A, Martin U, Frieling H, Wegner F. Functional effects of cannabinoids during dopaminergic specification of human neural precursors derived from induced pluripotent stem cells. Addict Biol 2017; 22:1329-1342. [PMID: 27027565 DOI: 10.1111/adb.12394] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/22/2016] [Accepted: 02/22/2016] [Indexed: 01/28/2023]
Abstract
Among adolescents cannabis is one of the most widely used illicit drugs. In adolescence brain development continues, characterized by neuronal maturation and synaptic plasticity. The endocannabinoid system plays an important role during brain development by modulating neuronal function and neurogenesis. Changes in endocannabinoid signaling by Δ9 -tetrahydrocannabinol (THC), the psychoactive component of cannabis, might therefore lead to neurobiological changes influencing brain function and behavior. We investigated the functional maturation and dopaminergic specification of human cord blood-derived induced pluripotent stem cell (hCBiPSC)-derived small molecule neural precursor cells (smNPCs) after cultivation with the endogenous cannabinoid anandamide (AEA) and the exogenous THC, both potent agonists at the cannabinoid 1 receptor (CB1 R). Higher dosages of 10-μM AEA or THC significantly decreased functionality of neurons, indicated by reduced ion currents and synaptic activity. A lower concentration of 1-μM THC had no marked effect on neuronal and dopaminergic maturation, while 1-μM AEA significantly enhanced the frequency of synaptic activity. As there were no significant effects on DNA methylation in promotor regions of genes important for neuronal function, these cannabinoid actions seem to be mediated by another than this epigenetic mechanism. Our data suggest that there are concentration-dependent actions of cannabinoids on neuronal function in vitro indicating neurotoxic, dysfunctional effects of 10-μM AEA and THC during human neurogenesis.
Collapse
Affiliation(s)
| | - Kirsten Jahn
- Center for Addiction Research, Department of Psychiatry, Social Psychiatry and Psychotherapy; Hannover Medical School; Hannover Germany
| | - Anna Venneri
- Department of Neurology; Hannover Medical School; Hannover Germany
| | - Maximilian Naujock
- Department of Neurology; Hannover Medical School; Hannover Germany
- Center for Systems Neuroscience; Hannover Germany
| | - Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation and Vascular Surgery; Hannover Medical School; Hannover Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation and Vascular Surgery; Hannover Medical School; Hannover Germany
- REBIRTH-Cluster of Excellence; Hannover Germany
| | - Helge Frieling
- Center for Addiction Research, Department of Psychiatry, Social Psychiatry and Psychotherapy; Hannover Medical School; Hannover Germany
- Center for Systems Neuroscience; Hannover Germany
| | - Florian Wegner
- Department of Neurology; Hannover Medical School; Hannover Germany
- Center for Systems Neuroscience; Hannover Germany
| |
Collapse
|
31
|
Musella A, Fresegna D, Rizzo FR, Gentile A, Bullitta S, De Vito F, Guadalupi L, Centonze D, Mandolesi G. A novel crosstalk within the endocannabinoid system controls GABA transmission in the striatum. Sci Rep 2017; 7:7363. [PMID: 28779174 PMCID: PMC5544685 DOI: 10.1038/s41598-017-07519-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/28/2017] [Indexed: 11/20/2022] Open
Abstract
The N-palmitoylethanolamine (PEA) is an endogenous member of the endocannabinoid system (ECS) with several biological functions, including a neuromodulatory activity in the central nervous system. To shed light on the neuronal function of PEA, we investigated its involvement in the control of both excitatory and inhibitory transmission in the murine striatum, a brain region strongly modulated by the ECS. By means of electrophysiological recordings, we showed that PEA modulates inhibitory synaptic transmission, through activation of GPR55 receptors, promoting a transient increase of GABAergic spontaneous inhibitory postsynaptic current (sIPSC) frequency. The subsequently rundown effect on sIPSC frequency was secondary to the delayed stimulation of presynaptic cannabinoid CB1 receptors (CB1Rs) by the endocannabinoid 2-AG, whose synthesis was stimulated by PEA on postsynaptic neurons. Our results indicate that PEA, acting on GPR55, enhances GABA transmission in the striatum, and triggers a parallel synthesis of 2-AG at the postsynaptic site, that in turn acts in a retrograde manner to inhibit GABA release through the stimulation of presynaptic CB1Rs. This electrophysiological study identifies a previously unrecognized function of PEA and of GPR55, demonstrating that GABAergic transmission is under the control of this compound and revealing that PEA modulates the release of the endocannabinoid 2-AG.
Collapse
Affiliation(s)
- A Musella
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143, Rome, Italy
| | - D Fresegna
- Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli (IS), Italy
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - F R Rizzo
- Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli (IS), Italy
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - A Gentile
- Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli (IS), Italy
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - S Bullitta
- Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli (IS), Italy
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - F De Vito
- Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli (IS), Italy
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - L Guadalupi
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143, Rome, Italy
| | - D Centonze
- Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli (IS), Italy.
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy.
| | - G Mandolesi
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143, Rome, Italy
| |
Collapse
|
32
|
Mandolesi G, Bullitta S, Fresegna D, Gentile A, De Vito F, Dolcetti E, Rizzo FR, Strimpakos G, Centonze D, Musella A. Interferon-γ causes mood abnormalities by altering cannabinoid CB1 receptor function in the mouse striatum. Neurobiol Dis 2017; 108:45-53. [PMID: 28757328 DOI: 10.1016/j.nbd.2017.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/21/2017] [Accepted: 07/26/2017] [Indexed: 01/10/2023] Open
Abstract
Interferon-γ (IFN-γ) has been implicated in the pathogenesis of multiple sclerosis (MS) and in its animal model, experimental autoimmune encephalomyelitis (EAE). The type-1 cannabinoid receptors (CB1Rs) are heavily involved in MS pathophysiology, and a growing body of evidence suggests that mood disturbances reflect specific effects of proinflammatory cytokines on neuronal activity. Here, we investigated whether IFN-γ could exert a role in the anxiety- and depressive-like behavior observed in mice with EAE, and in the modulation of CB1Rs. Anxiety and depression in fact are often diagnosed in MS, and have already been shown to depend on cannabinoid system. We performed biochemical, behavioral and electrophysiological experiments to assess the role of IFN-γ on mood control and on synaptic transmission in mice. Intracerebroventricular delivery of IFN-γ caused a depressive- and anxiety-like behavior in mice, associated with the selective dysfunction of CB1Rs controlling GABA transmission in the striatum. EAE induction was associated with increased striatal expression of IFN-γ, and with CB1R transmission deficits, which were rescued by pharmacological blockade of IFN-γ. IFN-γ was unable to replicate the effects of EAE on excitatory and inhibitory transmission in the striatum, but mimicked the effects of EAE on CB1R function in this brain area. Overall these results indicate that IFN-γ exerts a relevant control on mood, through the modulation of CB1R function. A better understanding of the biological pathways underling the psychological disorders during neuroinflammatory conditions is crucial for developing effective therapeutic strategies.
Collapse
Affiliation(s)
- Georgia Mandolesi
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| | - Silvia Bullitta
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Diego Fresegna
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Antonietta Gentile
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Francesca De Vito
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Ettore Dolcetti
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143 Rome, Italy; Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | - Francesca R Rizzo
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - Georgios Strimpakos
- Institute of Cell Biology and Neurobiology CNR, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology and of Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy.
| | - Alessandra Musella
- Centro Europeo per la Ricerca sul Cervello (CERC), IRCCS Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
33
|
Komaki H, Saadat F, Shahidi S, Sarihi A, Hasanein P, Komaki A. The interactive role of CB1 receptors and L-type calcium channels in hippocampal long-term potentiation in rats. Brain Res Bull 2017; 131:168-175. [PMID: 28442324 DOI: 10.1016/j.brainresbull.2017.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 02/03/2017] [Accepted: 04/19/2017] [Indexed: 01/22/2023]
Abstract
Long-term potentiation (LTP) of synaptic responses is a widely researched model of synaptic plasticity that occurs during learning and memory. The cannabinoid system is an endogenous system that modulate this kind of synaptic plasticity. In addition, voltage dependent calcium channels is essential for induction of LTP at some synapses in the hippocampus. However, there is currently debate over the interaction between L-type calcium channels and cannabinoid system on the synaptic plasticity. In this study, we examined the effects of an acute administration of the cannabinoid antagonist AM251 following a chronic administration of the Ca2+ channel blocker verapamil on LTP induction in the hippocampal dentate gyrus(DG) of rats. Male Wistar rats were administered verapamil(10,25,50mg/kg) or saline intraperitoneally(IP) daily for 13days(n=10/group). After this treatment period, animals were anesthetized with an IP injection of urethane; the recording and stimulating electrodes were positioned in the DG and the perforant pathway. After obtaining a steady state baseline response, a single IP injection of saline or AM251(1 or 5mg/kg) was administered. LTP was induced by high-frequency stimulation(HFS). The population spike(PS) amplitude and the slope of excitatory postsynaptic potentials(EPSP) were compared between the experimental groups. The acute administration of the CB1 antagonist AM251 increased LTP induction. The EPSP slopes and PS amplitude in the verapamil and AM251 groups differed after HFS, such that AM251 increased LTP, whereas verapamil decreased LTP induction. These findings suggest that there are functional interactions between the L-type calcium channels and cannabinoid system in this model of synaptic plasticity in the hippocampus.
Collapse
Affiliation(s)
- Hamidreza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fargol Saadat
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parisa Hasanein
- Department of Biology, School of Basic Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
34
|
Wilson H, De Micco R, Niccolini F, Politis M. Molecular Imaging Markers to Track Huntington's Disease Pathology. Front Neurol 2017; 8:11. [PMID: 28194132 PMCID: PMC5278260 DOI: 10.3389/fneur.2017.00011] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/09/2017] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is a progressive, monogenic dominant neurodegenerative disorder caused by repeat expansion mutation in the huntingtin gene. The accumulation of mutant huntingtin protein, forming intranuclear inclusions, subsequently leads to degeneration of medium spiny neurons in the striatum and cortical areas. Genetic testing can identify HD gene carriers before individuals develop overt cognitive, psychiatric, and chorea symptoms. Thus, HD gene carriers can be studied in premanifest stages to understand and track the evolution of HD pathology. While advances have been made, the precise pathophysiological mechanisms underlying HD are unclear. Magnetic resonance imaging (MRI) and positron emission tomography (PET) have been employed to understand HD pathology in presymptomatic and symptomatic disease stages. PET imaging uses radioactive tracers to detect specific changes, at a molecular level, which could be used as markers of HD progression and to monitor response to therapeutic treatments for HD gene expansion carriers (HDGECs). This review focuses on available PET techniques, employed in cross-sectional and longitudinal human studies, as biomarkers for HD, and highlights future potential PET targets. PET studies have assessed changes in postsynaptic dopaminergic receptors, brain metabolism, microglial activation, and recently phosphodiesterase 10A (PDE10A) as markers to track HD progression. Alterations in PDE10A expression are the earliest biochemical change identified in HD gene carriers up to 43 years before predicted symptomatic onset. Thus, PDE10A expression could be a promising marker to track HD progression from early premanifest disease stages. Other PET targets which have been less well investigated as biomarkers include cannabinoid, adenosine, and GABA receptors. Future longitudinal studies are required to fully validate these PET biomarkers for use to track disease progression from far-onset premanifest to manifest HD stages. PET imaging is a crucial neuroimaging tool, with the potential to detect early changes and validate sensitivity of biomarkers for tracking HD pathology. Moreover, continued development of novel PET tracers provides exciting opportunities to investigate new molecular targets, such as histamine and serotonin receptors, to further understand the mechanisms underlying HD pathology.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| | - Rosa De Micco
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| | - Flavia Niccolini
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, King's College London , London , UK
| |
Collapse
|
35
|
Wilkerson JL, Ghosh S, Mustafa M, Abdullah RA, Niphakis MJ, Cabrera R, Maldonado RL, Cravatt BF, Lichtman AH. The endocannabinoid hydrolysis inhibitor SA-57: Intrinsic antinociceptive effects, augmented morphine-induced antinociception, and attenuated heroin seeking behavior in mice. Neuropharmacology 2016; 114:156-167. [PMID: 27890602 DOI: 10.1016/j.neuropharm.2016.11.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/11/2016] [Accepted: 11/21/2016] [Indexed: 12/18/2022]
Abstract
Although opioids are highly efficacious analgesics, their abuse potential and other untoward side effects diminish their therapeutic utility. The addition of non-opioid analgesics offers a promising strategy to reduce required antinociceptive opioid doses that concomitantly reduce opioid-related side effects. Inhibitors of the primary endocannabinoid catabolic enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) show opioid-sparing effects in preclinical models of pain. As simultaneous inhibition of these enzymes elicits enhanced antinociceptive effects compared with single enzyme inhibition, the present study tested whether the dual FAAH-MAGL inhibitor SA-57 [4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester] produces morphine-sparing antinociceptive effects, without major side effects associated with either drug class. SA-57 dose-dependently reversed mechanical allodynia in the constriction injury (CCI) of the sciatic nerve model of neuropathic pain and carrageenan inflammatory pain model. As previously reported, SA-57 was considerably more potent in elevating anandamide (AEA) than 2-arachidonyl glycerol (2-AG) in brain. Its anti-allodynic effects required cannabinoid (CB)1 and CB2 receptors; however, only CB2 receptors were necessary for the anti-edematous effects in the carrageenan assay. Although high doses of SA-57 alone were required to produce antinociception, low doses of this compound, which elevated AEA and did not affect 2-AG brain levels, augmented the antinociceptive effects of morphine, but lacked cannabimimetic side effects. Because of the high abuse liability of opioids and implication of the endocannabinoid system in the reinforcing effects of opioids, the final experiment tested whether SA-57 would alter heroin seeking behavior. Strikingly, SA-57 reduced heroin-reinforced nose poke behavior and the progressive ratio break point for heroin. In conclusion, the results of the present study suggest that inhibition of endocannabinoid degradative enzymes represents a promising therapeutic approach to decrease effective doses of opioids needed for clinical pain control, and may also possess therapeutic potential to reduce opioid abuse.
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
| | - Sudeshna Ghosh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Mohammed Mustafa
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Rehab A Abdullah
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Micah J Niphakis
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Roberto Cabrera
- Laboratory of Neuropharmacology. Department de Ciencies Experimentals i de la Salut, Pompeu Fabra University, PRBB, C/ Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Rafael L Maldonado
- Laboratory of Neuropharmacology. Department de Ciencies Experimentals i de la Salut, Pompeu Fabra University, PRBB, C/ Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
36
|
Citraro R, Russo E, Leo A, Russo R, Avagliano C, Navarra M, Calignano A, De Sarro G. Pharmacokinetic-pharmacodynamic influence of N-palmitoylethanolamine, arachidonyl-2'-chloroethylamide and WIN 55,212-2 on the anticonvulsant activity of antiepileptic drugs against audiogenic seizures in DBA/2 mice. Eur J Pharmacol 2016; 791:523-534. [PMID: 27663280 DOI: 10.1016/j.ejphar.2016.09.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 09/16/2016] [Accepted: 09/19/2016] [Indexed: 12/31/2022]
Abstract
We evaluated the effects of ACEA (selective cannabinoid (CB)1 receptor agonist), WIN 55,212-2 mesylate (WIN; non-selective CB1 and CB2 receptor agonist) and N-palmitoylethanolamine (PEA; an endogenous fatty acid of ethanolamide) in DBA/2 mice, a genetic model of reflex audiogenic epilepsy. PEA, ACEA or WIN intraperitoneal (i.p.) administration decreased the severity of tonic-clonic seizures. We also studied the effects of PEA, WIN or ACEA after co-administration with NIDA-41020 (CB1 receptor antagonist) or GW6471 (PPAR-α antagonist) and compared the effects of WIN, ACEA and PEA in order to clarify their mechanisms of action. PEA has anticonvulsant features in DBA/2 mice mainly through PPAR-α and likely indirectly on CB1 receptors, whereas ACEA and WIN act through CB1 receptors. The co-administration of ineffective doses of ACEA, PEA and WIN with some antiepileptic drugs (AEDs) was examined in order to identify potential pharmacological interactions in DBA/2 mice. We found that PEA, ACEA and WIN co-administration potentiated the efficacy of carbamazepine, diazepam, felbamate, gabapentin, phenobarbital, topiramate and valproate and PEA only also that of oxcarbazepine and lamotrigine whereas, their co-administration with levetiracetam and phenytoin did not have effects. PEA, ACEA or WIN administration did not significantly influence the total plasma and brain levels of AEDs; therefore, it can be concluded that the observed potentiation was only of pharmacodynamic nature. In conclusion, PEA, ACEA and WIN show anticonvulsant effects in DBA/2 mice and potentiate the effects several AEDs suggesting a possible therapeutic relevance of these drugs and their mechanisms of action.
Collapse
Affiliation(s)
- Rita Citraro
- Science of Health Department, Clinical Pharmacology Unit, School of Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Emilio Russo
- Science of Health Department, Clinical Pharmacology Unit, School of Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Antonio Leo
- Science of Health Department, Clinical Pharmacology Unit, School of Medicine, University "Magna Graecia" of Catanzaro, Italy
| | - Roberto Russo
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Michele Navarra
- Department of Experimental Pharmacology, University of Messina, Messina, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Giovambattista De Sarro
- Science of Health Department, Clinical Pharmacology Unit, School of Medicine, University "Magna Graecia" of Catanzaro, Italy.
| |
Collapse
|
37
|
Zhang X, Yao N, Chergui K. The GABAA receptor agonist muscimol induces an age- and region-dependent form of long-term depression in the mouse striatum. ACTA ACUST UNITED AC 2016; 23:479-85. [PMID: 27531838 PMCID: PMC4986858 DOI: 10.1101/lm.043190.116] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 07/11/2016] [Indexed: 01/21/2023]
Abstract
Several forms of long-term depression (LTD) of glutamatergic synaptic transmission have been identified in the dorsal striatum and in the nucleus accumbens (NAc). Such experience-dependent synaptic plasticity might play important roles in reward-related learning. The GABAA receptor agonist muscimol was recently found to trigger a long-lasting depression of glutamatergic synaptic transmission in the NAc of adolescent mice, but the mechanisms that underlie this novel form of LTD were not studied. Here we examined the effect of muscimol applied in the perfusion solution on the amplitude of field excitatory postsynaptic potentials/population spikes (fEPSP/PSs) in mouse brain slices. We found that muscimol depressed the fEPSP/PS in the NAc of adolescent mice but not adult mice, through both postsynaptic and presynaptic mechanisms. Indeed, muscimol altered the fEPSP/PS paired-pulse ratio, depolarized the membrane of projection neurons, and decreased the frequency, but not amplitude, of spontaneous excitatory postsynaptic currents in the NAc of adolescent mice. The LTD induced by muscimol likely involved endocannabinoids, metabotropic glutamate receptors (mGluRs), but not TRPV1 receptors. Muscimol-LTD was occluded by prior induction of LTD through low-frequency stimulation (LFS) of the slice, demonstrating a common pathway in the induction of LFS-LTD and muscimol-LTD. We also found that muscimol induced a form of LTD in the dorsolateral striatum of adult but not adolescent mice. This LTD was mediated by endocannabinoids but did not involve mGluRs or TRPV1 receptors. These results identify a novel form of synaptic plasticity, and its mechanisms of induction, which is age and region dependent. These findings may contribute to a better understanding of the increased susceptibility of the adolescent brain to long-term synaptic changes in regions associated with reward mechanisms.
Collapse
Affiliation(s)
- Xiaoqun Zhang
- The Karolinska Institute, Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, 171 77 Stockholm, Sweden
| | - Ning Yao
- The Karolinska Institute, Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, 171 77 Stockholm, Sweden
| | - Karima Chergui
- The Karolinska Institute, Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, 171 77 Stockholm, Sweden
| |
Collapse
|
38
|
Andrzejewski K, Barbano R, Mink J. Cannabinoids in the treatment of movement disorders: A systematic review of case series and clinical trials. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.baga.2016.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
39
|
Wilkerson JL, Niphakis MJ, Grim TW, Mustafa MA, Abdullah RA, Poklis JL, Dewey WL, Akbarali H, Banks ML, Wise LE, Cravatt BF, Lichtman AH. The Selective Monoacylglycerol Lipase Inhibitor MJN110 Produces Opioid-Sparing Effects in a Mouse Neuropathic Pain Model. J Pharmacol Exp Ther 2016; 357:145-56. [PMID: 26791602 PMCID: PMC4809319 DOI: 10.1124/jpet.115.229971] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 01/19/2016] [Indexed: 12/28/2022] Open
Abstract
Serious clinical liabilities associated with the prescription of opiates for pain control include constipation, respiratory depression, pruritus, tolerance, abuse, and addiction. A recognized strategy to circumvent these side effects is to combine opioids with other antinociceptive agents. The combination of opiates with the primary active constituent of cannabis (Δ(9)-tetrahydrocannabinol) produces enhanced antinociceptive actions, suggesting that cannabinoid receptor agonists can be opioid sparing. Here, we tested whether elevating the endogenous cannabinoid 2-arachidonoylglycerol through the inhibition of its primary hydrolytic enzyme monoacylglycerol lipase (MAGL), will produce opioid-sparing effects in the mouse chronic constriction injury (CCI) of the sciatic nerve model of neuropathic pain. The dose-response relationships of i.p. administration of morphine and the selective MAGL inhibitor 2,5-dioxopyrrolidin-1-yl 4-(bis(4-chlorophenyl)methyl)piperazine-1-carboxylate (MJN110) were tested alone and in combination at equieffective doses for reversal of CCI-induced mechanical allodynia and thermal hyperalgesia. The respective ED50 doses (95% confidence interval) of morphine and MJN110 were 2.4 (1.9-3.0) mg/kg and 0.43 (0.23-0.79) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. Acute antinociceptive effects of the combination of morphine and MJN110 required μ-opioid, CB1, and CB2 receptors. This combination did not reduce gastric motility or produce subjective cannabimimetic effects in the drug discrimination assay. Importantly, combinations of MJN110 and morphine given repeatedly (i.e., twice a day for 6 days) continued to produce antiallodynic effects with no evidence of tolerance. Taken together, these findings suggest that MAGL inhibition produces opiate-sparing events with diminished tolerance, constipation, and cannabimimetic side effects.
Collapse
MESH Headings
- Analgesics, Opioid/therapeutic use
- Animals
- Arachidonic Acids/metabolism
- Behavior, Animal/drug effects
- Carbamates/pharmacology
- Constriction, Pathologic/complications
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Synergism
- Endocannabinoids/metabolism
- Enzyme Inhibitors/pharmacology
- Glycerides/metabolism
- Hyperalgesia/chemically induced
- Hyperalgesia/drug therapy
- Male
- Mice
- Mice, Inbred C57BL
- Monoacylglycerol Lipases/antagonists & inhibitors
- Morphine/administration & dosage
- Morphine/therapeutic use
- Neuralgia/chemically induced
- Neuralgia/drug therapy
- Neuralgia/psychology
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB2/drug effects
- Receptors, Opioid, mu/drug effects
- Succinimides/pharmacology
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Micah J Niphakis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Travis W Grim
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Mohammed A Mustafa
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Rehab A Abdullah
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Justin L Poklis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - William L Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Hamid Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Laura E Wise
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Benjamin F Cravatt
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (J.L.W., T.W.G., M.A.M., R.A.A., J.L.P., W.L.D., H.A., M.L.B., L.E.W., A.H.L.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| |
Collapse
|
40
|
Keyshams N, Zendehdel M, Babapour V, Baghbanzadeh A. Cannabinoid–glutamate interactions in the regulation of food intake in neonatal layer- type chicks: role of glutamate NMDA and AMPA receptors. Vet Res Commun 2016; 40:63-71. [DOI: 10.1007/s11259-016-9655-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 03/15/2016] [Indexed: 10/22/2022]
|
41
|
Xu C, Hermes DJ, Mackie K, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. Cannabinoids Occlude the HIV-1 Tat-Induced Decrease in GABAergic Neurotransmission in Prefrontal Cortex Slices. J Neuroimmune Pharmacol 2016; 11:316-31. [PMID: 26993829 DOI: 10.1007/s11481-016-9664-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/10/2016] [Indexed: 01/03/2023]
Abstract
In the era of combined antiretroviral therapy (cART), human immunodeficiency virus type 1 (HIV-1) is now considered a chronic disease that specifically targets the brain and causes HIV-1-associated neurocognitive disorders (HAND). Endocannabinoids exhibit neuroprotective and anti-inflammatory properties in several central nervous system (CNS) disease models, but their effects in HAND are poorly understood. To address this issue, whole-cell recordings were performed on young (14-24 day old) C57BL/6J mice. We investigated the actions of the synthetic cannabinoid WIN55,212-2 (1 μM) and the endocannabinoid N-arachidonoyl ethanolamine (anandamide; AEA, 1 μM) in the presence of HIV-1 Tat on GABAergic neurotransmission in mouse prefrontal cortex (PFC) slices. We found a Tat concentration-dependent (5-50 nM) decrease in the frequency and amplitude of miniature inhibitory postsynaptic currents (mIPSCs). The cannabinoid 1 receptor (CB1R) antagonist rimonabant (1 μM) and zero extracellular calcium prevented the significant Tat-induced decrease in mIPSCs. Further, bath-applied WIN55,212-2 or AEA by itself, significantly decreased the frequency, but not amplitude of mIPSCs and/or spontaneous IPSCs (sIPSCs), and occluded a further downregulation of IPSCs by Tat. Pretreatment with rimonabant but not the CB2R antagonist AM630 (1 μM) prevented the WIN55,212-2- and AEA-induced decrease in IPSCs frequency without any further Tat effect. Results indicated a Tat-induced decrease in GABAergic neurotransmission, which was occluded by cannabinoids via a CB1R-related mechanism. Understanding the relationship between Tat toxicity and endocannabinoid signaling has the potential to identify novel therapeutic interventions to benefit individuals suffering from HAND and other cognitive impairments.
Collapse
Affiliation(s)
- Changqing Xu
- Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Douglas J Hermes
- Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ken Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | | | - Sylvia Fitting
- Department of Psychology & Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
42
|
Dopamine-dependent CB1 receptor dysfunction at corticostriatal synapses in homozygous PINK1 knockout mice. Neuropharmacology 2016; 101:460-70. [DOI: 10.1016/j.neuropharm.2015.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 10/07/2015] [Accepted: 10/16/2015] [Indexed: 11/18/2022]
|
43
|
Nazario LR, Antonioli RJ, Capiotti KM, Hallak JEC, Zuardi AW, Crippa JAS, Bonan CD, da Silva RS. Reprint of "Caffeine protects against memory loss induced by high and non-anxiolytic dose of cannabidiol in adult zebrafish (Danio rerio)". Pharmacol Biochem Behav 2015; 139 Pt B:134-40. [PMID: 26569549 DOI: 10.1016/j.pbb.2015.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 06/03/2015] [Accepted: 06/13/2015] [Indexed: 12/18/2022]
Abstract
Cannabidiol (CBD) has been investigated in a wide spectrum of clinical approaches due to its psychopharmacological properties. CBD has low affinity for cannabinoid neuroreceptors and agonistic properties to 5-HT receptors. An interaction between cannabinoid and purinergic receptor systems has been proposed. The purpose of this study is to evaluate CBD properties on memory behavioral and locomotor parameters and the effects of pre-treatment of adenosine receptor blockers on CBD impacts on memory using adult zebrafish. CBD (0.1, 0.5, 5, and 10mg/kg) was tested in the avoidance inhibitory paradigm and anxiety task. We analyzed the effect of a long-term caffeine pre-treatment (~20mg/L - four months). Also, acute block of adenosine receptors was performed in co-administration with CBD exposure in the memory assessment. CBD promoted an inverted U-shaped dose-response curve in the anxiety task; in the memory assessment, CBD in the dose of 5mg/Kg promoted the strongest effects without interfering with social and aggressive behavior. Caffeine treatment was able to prevent CBD (5mg/kg) effects on memory when CBD was given after the training session. CBD effects on memory were partially prevented by co-treatment with a specific A2A adenosine receptor antagonist when given prior to or after the training session, while CBD effects after the training session were fully prevented by adenosine A1 receptor antagonist. These results indicated that zebrafish have responses to CBD anxiolytic properties that are comparable to other animal models, and high doses changed memory retention in a way dependent on adenosine.
Collapse
Affiliation(s)
- Luiza Reali Nazario
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil
| | - Régis Junior Antonioli
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil
| | - Katiucia Marques Capiotti
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil
| | - Jaime Eduardo Cecílio Hallak
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Antonio Waldo Zuardi
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - José Alexandre S Crippa
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil
| | - Rosane Souza da Silva
- Laboratório de Neuroquímica e Psicofarmacologia, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Caixa Postal 1429, 90619-900 Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
44
|
Fetal Alcohol Spectrum Disorder: Potential Role of Endocannabinoids Signaling. Brain Sci 2015; 5:456-93. [PMID: 26529026 PMCID: PMC4701023 DOI: 10.3390/brainsci5040456] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/19/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022] Open
Abstract
One of the unique features of prenatal alcohol exposure in humans is impaired cognitive and behavioral function resulting from damage to the central nervous system (CNS), which leads to a spectrum of impairments referred to as fetal alcohol spectrum disorder (FASD). Human FASD phenotypes can be reproduced in the rodent CNS following prenatal ethanol exposure. Several mechanisms are expected to contribute to the detrimental effects of prenatal alcohol exposure on the developing fetus, particularly in the developing CNS. These mechanisms may act simultaneously or consecutively and differ among a variety of cell types at specific developmental stages in particular brain regions. Studies have identified numerous potential mechanisms through which alcohol can act on the fetus. Among these mechanisms are increased oxidative stress, mitochondrial damage, interference with the activity of growth factors, glia cells, cell adhesion molecules, gene expression during CNS development and impaired function of signaling molecules involved in neuronal communication and circuit formation. These alcohol-induced deficits result in long-lasting abnormalities in neuronal plasticity and learning and memory and can explain many of the neurobehavioral abnormalities found in FASD. In this review, the author discusses the mechanisms that are associated with FASD and provides a current status on the endocannabinoid system in the development of FASD.
Collapse
|
45
|
Méndez-Díaz M, Amancio-Belmont O, Hernández-Vázquez E, Ruiz-Contreras AE, Hernández-Luis F, Prospéro-García O. ENP11, a potential CB1R antagonist, induces anorexia in rats. Pharmacol Biochem Behav 2015; 135:177-81. [DOI: 10.1016/j.pbb.2015.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/12/2015] [Accepted: 06/10/2015] [Indexed: 11/29/2022]
|
46
|
Caffeine protects against memory loss induced by high and non-anxiolytic dose of cannabidiol in adult zebrafish (Danio rerio). Pharmacol Biochem Behav 2015; 135:210-6. [DOI: 10.1016/j.pbb.2015.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 06/03/2015] [Accepted: 06/13/2015] [Indexed: 02/02/2023]
|
47
|
Lovelace JW, Corches A, Vieira PA, Hiroto AS, Mackie K, Korzus E. An animal model of female adolescent cannabinoid exposure elicits a long-lasting deficit in presynaptic long-term plasticity. Neuropharmacology 2015; 99:242-55. [PMID: 25979486 DOI: 10.1016/j.neuropharm.2015.04.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 04/16/2015] [Accepted: 04/29/2015] [Indexed: 11/29/2022]
Abstract
Cannabis continues to be the most accessible and popular illicit recreational drug. Whereas current data link adolescence cannabinoid exposure to increased risk for dependence on other drugs, depression, anxiety disorders and psychosis, the mechanism(s) underlying these adverse effects remains controversial. Here we show in a mouse model of female adolescent cannabinoid exposure deficient endocannabinoid (eCB)-mediated signaling and presynaptic forms of long-term depression at adult central glutamatergic synapses in the prefrontal cortex. Increasing endocannabinoid levels by blockade of monoacylglycerol lipase, the primary enzyme responsible for degrading the endocannabinoid 2-arachidonoylglycerol (2-AG), with the specific inhibitor JZL 184 ameliorates eCB-LTD deficits. The observed deficit in cortical presynaptic signaling may represent a neural maladaptation underlying network instability and abnormal cognitive functioning. Our study suggests that adolescent cannabinoid exposure may permanently impair brain functions, including the brain's intrinsic ability to appropriately adapt to external influences.
Collapse
Affiliation(s)
- Jonathan W Lovelace
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA
| | - Alex Corches
- Biomedical Sciences Program, University of California Riverside, CA 92521, USA
| | - Philip A Vieira
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA
| | - Alex S Hiroto
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA
| | - Ken Mackie
- Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Edward Korzus
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA; Biomedical Sciences Program, University of California Riverside, CA 92521, USA.
| |
Collapse
|
48
|
Kupferschmidt DA, Lovinger DM. Inhibition of presynaptic calcium transients in cortical inputs to the dorsolateral striatum by metabotropic GABA(B) and mGlu2/3 receptors. J Physiol 2015; 593:2295-310. [PMID: 25781000 DOI: 10.1113/jp270045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/06/2015] [Indexed: 01/30/2023] Open
Abstract
Cortical inputs to the dorsolateral striatum (DLS) are dynamically regulated during skill learning and habit formation, and are dysregulated in disorders characterized by impaired action control. Therefore, a mechanistic investigation of the processes regulating corticostriatal transmission is key to understanding DLS-associated circuit function, behaviour and pathology. Presynaptic GABA(B) and group II metabotropic glutamate (mGlu2/3) receptors exert marked inhibitory control over corticostriatal glutamate release in the DLS, yet the signalling pathways through which they do so are unclear. We developed a novel approach using the genetically encoded calcium (Ca(2+) ) indicator GCaMP6 to assess presynaptic Ca(2+) in corticostriatal projections to the DLS. Using simultaneous photometric presynaptic Ca(2+) and striatal field potential recordings, we report that relative to P/Q-type Ca(2+) channels, N-type channels preferentially contributed to evoked presynaptic Ca(2+) influx in motor cortex projections to, and excitatory transmission in, the DLS. Activation of GABA(B) or mGlu2/3 receptors inhibited both evoked presynaptic Ca(2+) transients and striatal field potentials. mGlu2/3 receptor-mediated depression did not require functional N-type Ca(2+) channels, but was attenuated by blockade of P/Q-type channels. These findings reveal presynaptic mechanisms of inhibitory modulation of corticostriatal function that probably contribute to the selection and shaping of behavioural repertoires.
Collapse
Affiliation(s)
- David A Kupferschmidt
- Section on Synaptic Pharmacology & In Vivo Neural Function, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD, USA
| | - David M Lovinger
- Section on Synaptic Pharmacology & In Vivo Neural Function, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
49
|
Neurobiology of l-DOPA induced dyskinesia and the novel therapeutic strategies. Biomed Pharmacother 2015; 70:283-93. [DOI: 10.1016/j.biopha.2015.01.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 01/23/2015] [Indexed: 12/27/2022] Open
|
50
|
Zhang X, Feng ZJ, Chergui K. Induction of cannabinoid- and N-methyl-D-aspartate receptor-mediated long-term depression in the nucleus accumbens and dorsolateral striatum is region and age dependent. Int J Neuropsychopharmacol 2015; 18:pyu052. [PMID: 25618403 PMCID: PMC4360221 DOI: 10.1093/ijnp/pyu052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The adolescent brain is sensitive to experience-dependent plasticity and might be more vulnerable than the adult brain to the effects of some drugs of abuse. The factors that contribute to these differences are not fully identified. We have examined the ability of cannabinoids to induce a form of synaptic plasticity, long-term depression, in the nucleus accumbens and dorsolateral striatum of adolescent and adult mice. METHODS We measured field excitatory postsynaptic potentials/population spikes in brain slices. RESULTS We found that the cannabinoid receptor agonist WIN 55,212-2 (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone mesylate) induced long-term depression in the nucleus accumbens of adolescent but not adult mice and failed to induce long-term depression in the dorsolateral striatum of adolescent or adult mice. Similar results were obtained with the group I metabotropic glutamate receptor agonist (S)-3,5- dihydroxyphenylglycine, which has previously been shown to promote the release of endocannabinoids. These age-related differences were associated with reduced protein levels of the cannabinoid type 1 receptor and metabotropic glutamate receptor 1 in adult nucleus accumbens and dorsolateral striatum and with an increased tone of endocannabinoids in the dorsolateral striatum of adult mice. We also found that N-methyl-D-aspartate receptor-dependent long-term depression, which was induced in the nucleus accumbens of adolescent mice, was blunted in adult mice, possibly because of decreased levels of GluN1, the obligatory subunit of N-methyl-D-aspartate receptors. CONCLUSIONS This study identifies region- and age-specific differences in the ability of endogenous and exogenous cannabinoids, and of N-methyl-D-aspartate receptors, to induce long-term depression in the striatal complex. These observations might contribute to a better understanding of the increased sensitivity of the adolescent brain to drug induced-plasticity.
Collapse
Affiliation(s)
| | | | - Karima Chergui
- The Karolinska Institute, Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, Von Eulers väg 8, 171 77 Stockholm, Sweden (Drs Zhang, Feng, and Chergui).
| |
Collapse
|