1
|
Miske R, Scharf M, Borowski K, Specht I, Corty M, Loritz MJ, Rombach F, Laureys G, Rochow N, Radzimski C, Schnitter L, Ratuszny D, Skripuletz T, Wattjes MP, Hahn S, Denno Y, Guerti K, Oyaert M, Benkhadra F, Bien CI, Nitsch S, Wandinger KP, van Pesch V, Probst C, Teegen B, Komorowski L, Sühs KW. Identification of DAGLA as an autoantibody target in cerebellar ataxia. J Neurol Neurosurg Psychiatry 2024; 95:1064-1076. [PMID: 38663995 PMCID: PMC11503208 DOI: 10.1136/jnnp-2024-333458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/04/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND We aimed to investigate the clinical, imaging and fluid biomarker characteristics in patients with antidiacylglycerol lipase alpha (DAGLA)-autoantibody-associated cerebellitis. METHODS Serum and cerebrospinal fliud (CSF) samples from four index patients were subjected to comprehensive autoantibody screening by indirect immunofluorescence assay (IIFA). Immunoprecipitation, mass spectrometry and recombinant protein assays were used to identify the autoantigen. Sera from 101 patients with various neurological symptoms and a similar tissue staining pattern as the index patient samples, and 102 healthy donors were analysed in recombinant cell-based IIFA (RC-IIFA) with the identified protein. Epitope characterisation of all positive samples was performed via ELISA, immunoblot, immunoprecipitation and RC-IIFA using different DAGLA fragments. RESULTS All index patients were relatively young (age: 18-34) and suffered from pronounced gait ataxia, dysarthria and visual impairments. Paraclinical hallmarks in early-stage disease were inflammatory CSF changes and cerebellar cortex hyperintensity in MRI. Severe cerebellar atrophy developed in three of four patients within 6 months. All patient samples showed the same unclassified IgG reactivity with the cerebellar molecular layer. DAGLA was identified as the target antigen and confirmed by competitive inhibition experiments and DAGLA-specific RC-IIFA. In RC-IIFA, serum reactivity against DAGLA was also found in 17/101 disease controls, including patients with different clinical phenotypes than the one of the index patients, and in 1/102 healthy donors. Epitope characterisation revealed that 17/18 anti-DAGLA-positive control sera reacted with a C-terminal intracellular DAGLA 583-1042 fragment, while the CSF samples of the index patients targeted a conformational epitope between amino acid 1 and 157. CONCLUSIONS We propose that anti-DAGLA autoantibodies detected in CSF, with a characteristic tissue IIFA pattern, represent novel biomarkers for rapidly progressive cerebellitis.
Collapse
Affiliation(s)
- Ramona Miske
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Madeleine Scharf
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Kathrin Borowski
- Clinical Immunological Laboratory Prof. h.c. (RCH) Dr. med. Winfried Stöcker, Lübeck, Germany
| | - Ina Specht
- Agaplesion Diakonieklinikum Rotenburg, Rotenburg, Germany
| | - Merle Corty
- Clinical Immunological Laboratory Prof. h.c. (RCH) Dr. med. Winfried Stöcker, Lübeck, Germany
| | | | | | - Guy Laureys
- Department of Neurology, Ghent University Hospital, Gent, Belgium
| | - Nadine Rochow
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Christiane Radzimski
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Linda Schnitter
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | | | | | - Mike P Wattjes
- Department of Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Stefanie Hahn
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Yvonne Denno
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Khadija Guerti
- Department of Clinical Chemistry, Antwerp University Hospital, Edegem, Belgium
| | - Matthijs Oyaert
- Department of Laboratory Medicine, Ghent University Hospitals, Gent, Belgium
| | - Farid Benkhadra
- Department of Clinical Biology, Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg
| | | | - Sophie Nitsch
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel/Lübeck, Germany
| | - Vincent van Pesch
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Christian Probst
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Bianca Teegen
- Clinical Immunological Laboratory Prof. h.c. (RCH) Dr. med. Winfried Stöcker, Lübeck, Germany
| | - Lars Komorowski
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | | |
Collapse
|
2
|
Boo KJ, Kim DH, Cho E, Kim DH, Jeon SJ, Shin CY. Neonatal dysregulation of 2-arachidonoylglycerol induces impaired brain function in adult mice. Neuropharmacology 2024; 257:110045. [PMID: 38885736 DOI: 10.1016/j.neuropharm.2024.110045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
The endocannabinoid system (ECS) regulates neurotransmission linked to synaptic plasticity, cognition, and emotion. While it has been demonstrated that dysregulation of the ECS in adulthood is relevant not only to central nervous system (CNS) disorders such as autism spectrum disorder, cognitive dysfunction, and depression but also to brain function, there are few studies on how dysregulation of the ECS in the neonatal period affects the manifestation and pathophysiology of CNS disorders later in life. In this study, DO34, a diacylglycerol lipase alpha (DAGLα) inhibitor affecting endocannabinoid 2-AG production, was injected into C57BL/6N male mice from postnatal day (PND) 7 to PND 10, inducing dysregulation of the ECS in the neonatal period. Subsequently, we examined whether it affects neuronal function in adulthood through electrophysiological and behavioral evaluation. DO34-injected mice showed significantly decreased cognitive functions, attributed to impairment of hippocampal synaptic plasticity. The findings suggest that regulation of ECS activity in the neonatal period may induce enduring effects on adult brain function.
Collapse
Affiliation(s)
- Kyung-Jun Boo
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Dae Hyun Kim
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea
| | - Eunbi Cho
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Dong Hyun Kim
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Republic of Korea.
| | - Se Jin Jeon
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon, Gangwon, 24252, Republic of Korea.
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, 05029, Republic of Korea; Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea; Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
3
|
Zhu D, Zhang J, Ma X, Hu M, Gao F, Hashem JB, Lyu J, Wei J, Cui Y, Qiu S, Chen C. Overabundant endocannabinoids in neurons are detrimental to cognitive function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613513. [PMID: 39345517 PMCID: PMC11430108 DOI: 10.1101/2024.09.17.613513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
2-Arachidonoylglycerol (2-AG) is the most prevalent endocannabinoid involved in maintaining brain homeostasis. Previous studies have demonstrated that inactivating monoacylglycerol lipase (MAGL), the primary enzyme responsible for degrading 2-AG in the brain, alleviates neuropathology and prevents synaptic and cognitive decline in animal models of neurodegenerative diseases. However, we show that selectively inhibiting 2-AG metabolism in neurons impairs cognitive function in mice. This cognitive impairment appears to result from decreased expression of synaptic proteins and synapse numbers, impaired long-term synaptic plasticity and cortical circuit functional connectivity, and diminished neurogenesis. Interestingly, the synaptic and cognitive deficits induced by neuronal MAGL inactivation can be counterbalanced by inhibiting astrocytic 2-AG metabolism. Transcriptomic analyses reveal that inhibiting neuronal 2-AG degradation leads to widespread changes in expression of genes associated with synaptic function. These findings suggest that crosstalk in 2-AG signaling between astrocytes and neurons is crucial for maintaining synaptic and cognitive functions and that excessive 2-AG in neurons alone is detrimental to cognitive function.
Collapse
Affiliation(s)
- Dexiao Zhu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jian Zhang
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Xiaokuang Ma
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Mei Hu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Fei Gao
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jack B. Hashem
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jianlu Lyu
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jing Wei
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Yuehua Cui
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Shenfeng Qiu
- Departments of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Chu Chen
- Department of Cellular and Integrative Physiology, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| |
Collapse
|
4
|
Rivi V, Rigillo G, Batabyal A, Lukowiak K, Pani L, Tascedda F, Benatti C, Blom JMC. Different stressors uniquely affect the expression of endocannabinoid-metabolizing enzymes in the central ring ganglia of Lymnaea stagnalis. J Neurochem 2024; 168:2848-2867. [PMID: 38922726 DOI: 10.1111/jnc.16147] [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: 02/17/2024] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024]
Abstract
The endocannabinoid system (ECS) plays an important role in neuroprotection, neuroplasticity, energy balance, modulation of stress, and inflammatory responses, acting as a critical link between the brain and the body's peripheral regions, while also offering promising potential for novel therapeutic strategies. Unfortunately, in humans, pharmacological inhibitors of different ECS enzymes have led to mixed results in both preclinical and clinical studies. As the ECS has been highly conserved throughout the eukaryotic lineage, the use of invertebrate model organisms like the pond snail Lymnaea stagnalis may provide a flexible tool to unravel unexplored functions of the ECS at the cellular, synaptic, and behavioral levels. In this study, starting from the available genome and transcriptome of L. stagnalis, we first identified putative transcripts of all ECS enzymes containing an open reading frame. Each predicted protein possessed a high degree of sequence conservation to known orthologues of other invertebrate and vertebrate organisms. Sequences were confirmed by qualitative PCR and sequencing. Then, we investigated the transcriptional effects induced by different stress conditions (i.e., bacterial LPS injection, predator scent, food deprivation, and acute heat shock) on the expression levels of the enzymes of the ECS in Lymnaea's central ring ganglia. Our results suggest that in Lymnaea as in rodents, the ECS is involved in mediating inflammatory and anxiety-like responses, promoting energy balance, and responding to acute stressors. To our knowledge, this study offers the most comprehensive analysis so far of the ECS in an invertebrate model organism.
Collapse
Affiliation(s)
- Veronica Rivi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanna Rigillo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anuradha Batabyal
- Department of Physical and Natural Sciences, FLAME University, Pune, India
| | - Ken Lukowiak
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Luca Pani
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, Florida, USA
| | - Fabio Tascedda
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- CIB, Consorzio Interuniversitario Biotecnologie, Trieste, Italy
| | - Cristina Benatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Johanna M C Blom
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
5
|
Szente L, Balla GY, Varga ZK, Toth B, Biro L, Balogh Z, Hill MN, Toth M, Mikics E, Aliczki M. Endocannabinoid and neuroplasticity-related changes as susceptibility factors in a rat model of posttraumatic stress disorder. Neurobiol Stress 2024; 32:100662. [PMID: 39183773 PMCID: PMC11341941 DOI: 10.1016/j.ynstr.2024.100662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/18/2024] [Accepted: 07/10/2024] [Indexed: 08/27/2024] Open
Abstract
Traumatic experiences result in the development of posttraumatic stress disorder (PTSD) in 10-25% of exposed individuals. While human clinical studies suggest that susceptibility is potentially linked to endocannabinoid (eCB) signaling, neurobiological PTSD susceptibility factors are poorly understood. Employing a rat model of contextual conditioned fear, we characterized distinct resilient and susceptible subpopulations based on lasting generalized fear, a core symptom of PTSD. In these groups, we assessed i.) eCB levels by mass spectrometry and ii.) expression variations of eCB system- and iii.) neuroplasticity-related genes by real-time quantitative PCR in the circuitry relevant in trauma-induced changes. Furthermore, employing unsupervised and semi-supervised machine learning based statistical analytical models, we assessed iv.) gene expression patterns with the most robust predictive power regarding PTSD susceptibility. According to our findings, in our model, generalized fear responses occurred with sufficient variability to characterize distinct resilient and susceptible subpopulations. Resilient subjects showed elevated prelimbic and lower ventral hippocampal levels of eCB 2-arachidonoyl-glycerol (2-AG) compared to resilient and non-shocked control subjects. Ventral hippocampal 2-AG content positively correlated with the strength of fear generalization. Furthermore, susceptibility was associated with i.) prefrontal, hippocampal and amygdalar neuronal hypoactivity, ii.) marked decrease in the expression of genes of transcription factors modulating neuroplasticity and iii.) an altered expression pattern of eCB-related genes, including enzymes involved in eCB metabolism. Unsupervised and semi-supervised statistical approaches highlighted that hippocampal gene expression patterns possess strong predictive power regarding susceptibility. Taken together, the marked eCB and neuroplasticity changes in susceptible individuals associated with abnormal activity patterns in the fear circuitry possibly contribute to context coding deficits, resulting in generalized fear.
Collapse
Affiliation(s)
- Laszlo Szente
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Gyula Y. Balla
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Zoltan K. Varga
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Blanka Toth
- Department of Inorganic and Analytical Chemistry, University of Technology and Economics, Budapest, Hungary
| | - Laszlo Biro
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Zoltan Balogh
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Matthew N. Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mate Toth
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Eva Mikics
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| | - Mano Aliczki
- Translational Behavioural Neuroscience Research Group, Institute of Experimental Medicine, Hungarian Research Network, Budapest, Hungary
| |
Collapse
|
6
|
Smith G, McCoy K, Di Prisco GV, Kuklish A, Grant E, Bhat M, Patel S, Mackie K, Atwood B, Kalinovsky A. Deletion of endocannabinoid synthesizing enzyme DAGLα from cerebellar Purkinje cells decreases social preference and elevates anxiety. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607068. [PMID: 39211155 PMCID: PMC11361171 DOI: 10.1101/2024.08.08.607068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The endocannabinoid (eCB) signaling system is robustly expressed in the cerebellum starting from the embryonic developmental stages to adulthood. There it plays a key role in regulating cerebellar synaptic plasticity and excitability, suggesting that impaired eCB signaling will lead to deficits in cerebellar adjustments of ongoing behaviors and cerebellar learning. Indeed, human mutations in DAGLα are associated with neurodevelopmental disorders. In this study, we show that selective deletion of the eCB synthesizing enzyme diacylglycerol lipase alpha (Daglα) from mouse cerebellar Purkinje cells (PCs) alters motor and social behaviors, disrupts short-term synaptic plasticity in both excitatory and inhibitory synapses, and reduces Purkinje cell activity during social exploration. Our results provide the first evidence for cerebellar-specific eCB regulation of social behaviors and implicate eCB regulation of synaptic plasticity and PC activity as the neural substrates contributing to these deficits. Abstract Figure
Collapse
|
7
|
Briand-Mésange F, Gennero I, Salles J, Trudel S, Dahan L, Ausseil J, Payrastre B, Salles JP, Chap H. From Classical to Alternative Pathways of 2-Arachidonoylglycerol Synthesis: AlterAGs at the Crossroad of Endocannabinoid and Lysophospholipid Signaling. Molecules 2024; 29:3694. [PMID: 39125098 PMCID: PMC11314389 DOI: 10.3390/molecules29153694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
2-arachidonoylglycerol (2-AG) is the most abundant endocannabinoid (EC), acting as a full agonist at both CB1 and CB2 cannabinoid receptors. It is synthesized on demand in postsynaptic membranes through the sequential action of phosphoinositide-specific phospholipase Cβ1 (PLCβ1) and diacylglycerol lipase α (DAGLα), contributing to retrograde signaling upon interaction with presynaptic CB1. However, 2-AG production might also involve various combinations of PLC and DAGL isoforms, as well as additional intracellular pathways implying other enzymes and substrates. Three other alternative pathways of 2-AG synthesis rest on the extracellular cleavage of 2-arachidonoyl-lysophospholipids by three different hydrolases: glycerophosphodiesterase 3 (GDE3), lipid phosphate phosphatases (LPPs), and two members of ecto-nucleotide pyrophosphatase/phosphodiesterases (ENPP6-7). We propose the names of AlterAG-1, -2, and -3 for three pathways sharing an ectocellular localization, allowing them to convert extracellular lysophospholipid mediators into 2-AG, thus inducing typical signaling switches between various G-protein-coupled receptors (GPCRs). This implies the critical importance of the regioisomerism of both lysophospholipid (LPLs) and 2-AG, which is the object of deep analysis within this review. The precise functional roles of AlterAGs are still poorly understood and will require gene invalidation approaches, knowing that both 2-AG and its related lysophospholipids are involved in numerous aspects of physiology and pathology, including cancer, inflammation, immune defenses, obesity, bone development, neurodegeneration, or psychiatric disorders.
Collapse
Affiliation(s)
- Fabienne Briand-Mésange
- Infinity-Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, INSERM, CNRS, Paul Sabatier University, 31059 Toulouse, France; (F.B.-M.); (I.G.); (J.S.); (S.T.); (J.A.); (J.-P.S.)
| | - Isabelle Gennero
- Infinity-Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, INSERM, CNRS, Paul Sabatier University, 31059 Toulouse, France; (F.B.-M.); (I.G.); (J.S.); (S.T.); (J.A.); (J.-P.S.)
- Centre Hospitalier Universitaire de Toulouse, Service de Biochimie, Institut Fédératif de Biologie, 31059 Toulouse, France
| | - Juliette Salles
- Infinity-Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, INSERM, CNRS, Paul Sabatier University, 31059 Toulouse, France; (F.B.-M.); (I.G.); (J.S.); (S.T.); (J.A.); (J.-P.S.)
- Centre Hospitalier Universitaire de Toulouse, Service de Psychiatrie D’urgences, de Crise et de Liaison, Institut des Handicaps Neurologiques, Psychiatriques et Sensoriels, 31059 Toulouse, France
| | - Stéphanie Trudel
- Infinity-Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, INSERM, CNRS, Paul Sabatier University, 31059 Toulouse, France; (F.B.-M.); (I.G.); (J.S.); (S.T.); (J.A.); (J.-P.S.)
- Centre Hospitalier Universitaire de Toulouse, Service de Biochimie, Institut Fédératif de Biologie, 31059 Toulouse, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France;
| | - Jérôme Ausseil
- Infinity-Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, INSERM, CNRS, Paul Sabatier University, 31059 Toulouse, France; (F.B.-M.); (I.G.); (J.S.); (S.T.); (J.A.); (J.-P.S.)
- Centre Hospitalier Universitaire de Toulouse, Service de Biochimie, Institut Fédératif de Biologie, 31059 Toulouse, France
| | - Bernard Payrastre
- I2MC-Institute of Metabolic and Cardiovascular Diseases, INSERM UMR1297 and University of Toulouse III, 31400 Toulouse, France;
- Centre Hospitalier Universitaire de Toulouse, Laboratoire d’Hématologie, 31400 Toulouse, France
| | - Jean-Pierre Salles
- Infinity-Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, INSERM, CNRS, Paul Sabatier University, 31059 Toulouse, France; (F.B.-M.); (I.G.); (J.S.); (S.T.); (J.A.); (J.-P.S.)
- Centre Hospitalier Universitaire de Toulouse, Unité d’Endocrinologie et Maladies Osseuses, Hôpital des Enfants, 31059 Toulouse, France
| | - Hugues Chap
- Infinity-Toulouse Institute for Infectious and Inflammatory Diseases, University of Toulouse, INSERM, CNRS, Paul Sabatier University, 31059 Toulouse, France; (F.B.-M.); (I.G.); (J.S.); (S.T.); (J.A.); (J.-P.S.)
- Académie des Sciences, Inscriptions et Belles Lettres de Toulouse, Hôtel d’Assézat, 31000 Toulouse, France
| |
Collapse
|
8
|
Yoneda T, Kameyama K, Gotou T, Terata K, Takagi M, Yoshimura Y, Sakimura K, Kano M, Hata Y. Layer specific regulation of critical period timing and maturation of mouse visual cortex by endocannabinoids. iScience 2024; 27:110145. [PMID: 38952682 PMCID: PMC11215304 DOI: 10.1016/j.isci.2024.110145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 04/15/2024] [Accepted: 05/27/2024] [Indexed: 07/03/2024] Open
Abstract
Plasticity during the critical period is important for the functional maturation of cortical neurons. While characteristics of plasticity are diverse among cortical layers, it is unknown whether critical period timing is controlled by a common or unique molecular mechanism among them. We here clarified layer-specific regulation of the critical period timing of ocular dominance plasticity in the primary visual cortex. Mice lacking the endocannabinoid synthesis enzyme diacylglycerol lipase-α exhibited precocious critical period timing, earlier maturation of inhibitory synaptic function in layers 2/3 and 4, and impaired development of the binocular matching of orientation selectivity exclusively in layer 2/3. Activation of cannabinoid receptor restored ocular dominance plasticity at the normal critical period in layer 2/3. Suppression of GABAA receptor rescued precocious ocular dominance plasticity in layer 4. Therefore, endocannabinoids regulate critical period timing and maturation of visual function partly through the development of inhibitory synaptic functions in a layer-dependent manner.
Collapse
Affiliation(s)
- Taisuke Yoneda
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki 444-8585, Japan
| | - Katsuro Kameyama
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Takahiro Gotou
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Keiko Terata
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Masahiro Takagi
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Yumiko Yoshimura
- Division of Visual Information Processing, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki 444-8585, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo 113-0033, Japan
- Advanced Comprehensive Research Organization (ACRO), Teikyo University, Tokyo 173-0003, Japan
| | - Yoshio Hata
- Division of Neuroscience, School of Life Science, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| |
Collapse
|
9
|
Barti B, Dudok B, Kenesei K, Zöldi M, Miczán V, Balla GY, Zala D, Tasso M, Sagheddu C, Kisfali M, Tóth B, Ledri M, Vizi ES, Melis M, Barna L, Lenkei Z, Soltész I, Katona I. Presynaptic nanoscale components of retrograde synaptic signaling. SCIENCE ADVANCES 2024; 10:eado0077. [PMID: 38809980 PMCID: PMC11135421 DOI: 10.1126/sciadv.ado0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
While our understanding of the nanoscale architecture of anterograde synaptic transmission is rapidly expanding, the qualitative and quantitative molecular principles underlying distinct mechanisms of retrograde synaptic communication remain elusive. We show that a particular form of tonic cannabinoid signaling is essential for setting target cell-dependent synaptic variability. It does not require the activity of the two major endocannabinoid-producing enzymes. Instead, by developing a workflow for physiological, anatomical, and molecular measurements at the same unitary synapse, we demonstrate that the nanoscale stoichiometric ratio of type 1 cannabinoid receptors (CB1Rs) to the release machinery is sufficient to predict synapse-specific release probability. Accordingly, selective decrease of extrasynaptic CB1Rs does not affect synaptic transmission, whereas in vivo exposure to the phytocannabinoid Δ9-tetrahydrocannabinol disrupts the intrasynaptic nanoscale stoichiometry and reduces synaptic variability. These findings imply that synapses leverage the nanoscale stoichiometry of presynaptic receptor coupling to the release machinery to establish synaptic strength in a target cell-dependent manner.
Collapse
Affiliation(s)
- Benjámin Barti
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Barna Dudok
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Departments of Neurology and Neuroscience, Baylor College of Medicine, 1 Baylor Plz, Houston, TX 77030, USA
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - Kata Kenesei
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miklós Zöldi
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Vivien Miczán
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Center, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Gyula Y. Balla
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Diana Zala
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Mariana Tasso
- Institute of Nanosystems, School of Bio and Nanotechnologies, National University of San Martín - CONICET, 25 de Mayo Ave., 1021 San Martín, Argentina
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - Máté Kisfali
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- BiTrial Ltd., Tállya st 23, H-1121 Budapest, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Szt. Gellért square 4, H-1111 Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Marco Ledri
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 17, BMC A11, 221 84 Lund, Sweden
| | - E. Sylvester Vizi
- Molecular Pharmacology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - László Barna
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
| | - Zsolt Lenkei
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Iván Soltész
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - István Katona
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| |
Collapse
|
10
|
Ramos-Medina L, Rosas-Vidal LE, Patel S. Pharmacological diacylglycerol lipase inhibition impairs contextual fear extinction in mice. Psychopharmacology (Berl) 2024; 241:569-584. [PMID: 38182791 PMCID: PMC10884152 DOI: 10.1007/s00213-023-06523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 12/21/2023] [Indexed: 01/07/2024]
Abstract
Acquisition and extinction of associative fear memories are critical for guiding adaptive behavioral responses to environmental threats, and dysregulation of these processes is thought to represent important neurobehavioral substrates of trauma and stress-related disorders including posttraumatic stress disorder (PTSD). Endogenous cannabinoid (eCB) signaling has been heavily implicated in the extinction of aversive fear memories and we have recently shown that pharmacological inhibition of 2-arachidonoylglycerol (2-AG) synthesis, a major eCB regulating synaptic suppression, impairs fear extinction in an auditory cue conditioning paradigm. Despite these data, the role of 2-AG signaling in contextual fear conditioning is not well understood. Here, we show that systemic pharmacological blockade of diacylglycerol lipase, the rate-limiting enzyme catalyzing in the synthesis of 2-AG, enhances contextual fear learning and impairs within-session extinction. In sham-conditioned mice, 2-AG synthesis inhibition causes a small increase in unconditioned freezing behavior. No effects of 2-AG synthesis inhibition were noted in the Elevated Plus Maze in mice tested after fear extinction. These data provide support for 2-AG signaling in the suppression of contextual fear learning and the expression of within-session extinction of contextual fear memories.
Collapse
Affiliation(s)
| | - Luis E Rosas-Vidal
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sachin Patel
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
11
|
Levine AA, Liktor-Busa E, Balasubramanian S, Palomino SM, Burtman AM, Couture SA, Lipinski AA, Langlais PR, Largent-Milnes TM. Depletion of Endothelial-Derived 2-AG Reduces Blood-Endothelial Barrier Integrity via Alteration of VE-Cadherin and the Phospho-Proteome. Int J Mol Sci 2023; 25:531. [PMID: 38203706 PMCID: PMC10778805 DOI: 10.3390/ijms25010531] [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: 12/11/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Mounting evidence supports the role of the endocannabinoid system in neurophysiology, including blood-brain barrier (BBB) function. Recent work has demonstrated that activation of endocannabinoid receptors can mitigate insults to the BBB during neurological disorders like traumatic brain injury, cortical spreading depression, and stroke. As alterations to the BBB are associated with worsening clinical outcomes in these conditions, studies herein sought to examine the impact of endocannabinoid depletion on BBB integrity. Barrier integrity was investigated in vitro via bEnd.3 cell monolayers to assess endocannabinoid synthesis, barrier function, calcium influx, junctional protein expression, and proteome-wide changes. Inhibition of 2-AG synthesis using DAGLα inhibition and siRNA inhibition of DAGLα led to loss of barrier integrity via altered expression of VE-cadherin, which could be partially rescued by exogenous application of 2-AG. Moreover, the deleterious effects of DAGLα inhibition on BBB integrity showed both calcium and PKC (protein kinase C)-dependency. These data indicate that disruption of 2-AG homeostasis in brain endothelial cells, in the absence of insult, is sufficient to disrupt BBB integrity thus supporting the role of the endocannabinoid system in neurovascular disorders.
Collapse
Affiliation(s)
- Aidan A. Levine
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Erika Liktor-Busa
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Shreya Balasubramanian
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Seph M. Palomino
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Anya M. Burtman
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Sarah A. Couture
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| | - Austin A. Lipinski
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (P.R.L.)
| | - Paul R. Langlais
- Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (P.R.L.)
| | - Tally M. Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (A.A.L.); (E.L.-B.); (S.B.); (S.M.P.); (A.M.B.); (S.A.C.)
| |
Collapse
|
12
|
Chen M, Shin M, Ware TB, Donvito G, Muchhala KH, Mischel R, Mustafa MA, Serbulea V, Upchurch CM, Leitinger N, Akbarali HI, Lichtman AH, Hsu KL. Endocannabinoid biosynthetic enzymes regulate pain response via LKB1-AMPK signaling. Proc Natl Acad Sci U S A 2023; 120:e2304900120. [PMID: 38109529 PMCID: PMC10756258 DOI: 10.1073/pnas.2304900120] [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: 03/24/2023] [Accepted: 11/08/2023] [Indexed: 12/20/2023] Open
Abstract
Diacylglycerol lipase-beta (DAGLβ) serves as a principal 2-arachidonoylglycerol (2-AG) biosynthetic enzyme regulating endocannabinoid and eicosanoid metabolism in immune cells including macrophages and dendritic cells. Genetic or pharmacological inactivation of DAGLβ ameliorates inflammation and hyper-nociception in preclinical models of pathogenic pain. These beneficial effects have been assigned principally to reductions in downstream proinflammatory lipid signaling, leaving alternative mechanisms of regulation largely underexplored. Here, we apply quantitative chemical- and phospho-proteomics to find that disruption of DAGLβ in primary macrophages leads to LKB1-AMPK signaling activation, resulting in reprogramming of the phosphoproteome and bioenergetics. Notably, AMPK inhibition reversed the antinociceptive effects of DAGLβ blockade, thereby directly supporting DAGLβ-AMPK crosstalk in vivo. Our findings uncover signaling between endocannabinoid biosynthetic enzymes and ancient energy-sensing kinases to mediate cell biological and pain responses.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | - Myungsun Shin
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | - Timothy B. Ware
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
| | - Giulia Donvito
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Karan H. Muchhala
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Ryan Mischel
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Mohammed A. Mustafa
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Vlad Serbulea
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
| | - Clint M. Upchurch
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
| | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA23298
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298
| | - Ku-Lung Hsu
- Department of Chemistry, University of Virginia, Charlottesville, VA22904
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA22908
- University of Virginia Cancer Center, Cancer Biology Program, University of Virginia, Charlottesville, VA22903
| |
Collapse
|
13
|
Kouchaeknejad A, Van Der Walt G, De Donato MH, Puighermanal E. Imaging and Genetic Tools for the Investigation of the Endocannabinoid System in the CNS. Int J Mol Sci 2023; 24:15829. [PMID: 37958825 PMCID: PMC10648052 DOI: 10.3390/ijms242115829] [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: 09/15/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
As central nervous system (CNS)-related disorders present an increasing cause of global morbidity, mortality, and high pressure on our healthcare system, there is an urgent need for new insights and treatment options. The endocannabinoid system (ECS) is a critical network of endogenous compounds, receptors, and enzymes that contribute to CNS development and regulation. Given its multifaceted involvement in neurobiology and its significance in various CNS disorders, the ECS as a whole is considered a promising therapeutic target. Despite significant advances in our understanding of the ECS's role in the CNS, its complex architecture and extensive crosstalk with other biological systems present challenges for research and clinical advancements. To bridge these knowledge gaps and unlock the full therapeutic potential of ECS interventions in CNS-related disorders, a plethora of molecular-genetic tools have been developed in recent years. Here, we review some of the most impactful tools for investigating the neurological aspects of the ECS. We first provide a brief introduction to the ECS components, including cannabinoid receptors, endocannabinoids, and metabolic enzymes, emphasizing their complexity. This is followed by an exploration of cutting-edge imaging tools and genetic models aimed at elucidating the roles of these principal ECS components. Special emphasis is placed on their relevance in the context of CNS and its associated disorders.
Collapse
Affiliation(s)
| | | | | | - Emma Puighermanal
- Neuroscience Institute, Autonomous University of Barcelona, 08193 Bellaterra, Spain; (A.K.); (G.V.D.W.); (M.H.D.D.)
| |
Collapse
|
14
|
Maccarrone M, Di Marzo V, Gertsch J, Grether U, Howlett AC, Hua T, Makriyannis A, Piomelli D, Ueda N, van der Stelt M. Goods and Bads of the Endocannabinoid System as a Therapeutic Target: Lessons Learned after 30 Years. Pharmacol Rev 2023; 75:885-958. [PMID: 37164640 PMCID: PMC10441647 DOI: 10.1124/pharmrev.122.000600] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023] Open
Abstract
The cannabis derivative marijuana is the most widely used recreational drug in the Western world and is consumed by an estimated 83 million individuals (∼3% of the world population). In recent years, there has been a marked transformation in society regarding the risk perception of cannabis, driven by its legalization and medical use in many states in the United States and worldwide. Compelling research evidence and the Food and Drug Administration cannabis-derived cannabidiol approval for severe childhood epilepsy have confirmed the large therapeutic potential of cannabidiol itself, Δ9-tetrahydrocannabinol and other plant-derived cannabinoids (phytocannabinoids). Of note, our body has a complex endocannabinoid system (ECS)-made of receptors, metabolic enzymes, and transporters-that is also regulated by phytocannabinoids. The first endocannabinoid to be discovered 30 years ago was anandamide (N-arachidonoyl-ethanolamine); since then, distinct elements of the ECS have been the target of drug design programs aimed at curing (or at least slowing down) a number of human diseases, both in the central nervous system and at the periphery. Here a critical review of our knowledge of the goods and bads of the ECS as a therapeutic target is presented to define the benefits of ECS-active phytocannabinoids and ECS-oriented synthetic drugs for human health. SIGNIFICANCE STATEMENT: The endocannabinoid system plays important roles virtually everywhere in our body and is either involved in mediating key processes of central and peripheral diseases or represents a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of the components of this complex system, and in particular of key receptors (like cannabinoid receptors 1 and 2) and metabolic enzymes (like fatty acid amide hydrolase and monoacylglycerol lipase), will advance our understanding of endocannabinoid signaling and activity at molecular, cellular, and system levels, providing new opportunities to treat patients.
Collapse
Affiliation(s)
- Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Vincenzo Di Marzo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Jürg Gertsch
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Uwe Grether
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Allyn C Howlett
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Tian Hua
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Alexandros Makriyannis
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Daniele Piomelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Natsuo Ueda
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Mario van der Stelt
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| |
Collapse
|
15
|
Yan YC, Meng GX, Yang CC, Yang YF, Tan SY, Yan LJ, Ding ZN, Ma YL, Dong ZR, Li T. Diacylglycerol lipase alpha promotes hepatocellular carcinoma progression and induces lenvatinib resistance by enhancing YAP activity. Cell Death Dis 2023; 14:404. [PMID: 37414748 PMCID: PMC10325985 DOI: 10.1038/s41419-023-05919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/08/2023]
Abstract
As an important hydrolytic enzyme that yields 2-AG and free fatty acids, diacylglycerol lipase alpha (DAGLA) is involved in exacerbating malignant phenotypes and cancer progression, but the role of the DAGLA/2-AG axis in HCC progression remains unclear. Here, we found that the upregulation of components of the DAGLA/2-AG axis in HCC samples is correlated with tumour stage and patient prognosis. In vitro and in vivo experiments demonstrated that the DAGLA/2-AG axis promoted HCC progression by regulating cell proliferation, invasion and metastasis. Mechanistically, the DAGLA/2AG axis significantly inhibited LATS1 and YAP phosphorylation, promoted YAP nuclear translocation and activity, and ultimately led to TEAD2 upregulation and increased PHLDA2 expression, which could be enhanced by DAGLA/2AG-induced activation of the PI3K/AKT pathway. More importantly, DAGLA induced resistance to lenvatinib therapy during HCC treatment. Our study demonstrates that inhibiting the DAGLA/2-AG axis could be a novel therapeutic strategy to inhibit HCC progression and enhance the therapeutic effects of TKIs, which warrant further clinical studies.
Collapse
Affiliation(s)
- Yu-Chuan Yan
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Guang-Xiao Meng
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Chun-Cheng Yang
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Ya-Fei Yang
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Si-Yu Tan
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, 250012, Jinan, China
| | - Lun-Jie Yan
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Zi-Niu Ding
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Yun-Long Ma
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Zhao-Ru Dong
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China.
| | - Tao Li
- Department of General Surgery, Qilu Hospital of Shandong University, 250012, Jinan, China.
| |
Collapse
|
16
|
Oddi S, Fiorenza MT, Maccarrone M. Endocannabinoid signaling in adult hippocampal neurogenesis: A mechanistic and integrated perspective. Prog Lipid Res 2023; 91:101239. [PMID: 37385352 DOI: 10.1016/j.plipres.2023.101239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/01/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
Dentate gyrus of the hippocampus continuously gives rise to new neurons, namely, adult-born granule cells, which contribute to conferring plasticity to the mature brain throughout life. Within this neurogenic region, the fate and behavior of neural stem cells (NSCs) and their progeny result from a complex balance and integration of a variety of cell-autonomous and cell-to-cell-interaction signals and underlying pathways. Among these structurally and functionally diverse signals, there are endocannabinoids (eCBs), the main brain retrograde messengers. These pleiotropic bioactive lipids can directly and/or indirectly influence adult hippocampal neurogenesis (AHN) by modulating, both positively and negatively, multiple molecular and cellular processes in the hippocampal niche, depending on the cell type or stage of differentiation. Firstly, eCBs act directly as cell-intrinsic factors, cell-autonomously produced by NSCs following their stimulation. Secondly, in many, if not all, niche-associated cells, including some local neuronal and nonneuronal elements, the eCB system indirectly modulates the neurogenesis, linking neuronal and glial activity to regulating distinct stages of AHN. Herein, we discuss the crosstalk of the eCB system with other neurogenesis-relevant signal pathways and speculate how the hippocampus-dependent neurobehavioral effects elicited by (endo)cannabinergic medications are interpretable in light of the key regulatory role that eCBs play on AHN.
Collapse
Affiliation(s)
- Sergio Oddi
- Department of Veterinary Medicine, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy; European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy.
| | - Maria Teresa Fiorenza
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Department of Psychology, Division of Neuroscience and "Daniel Bovet" Neurobiology Research Center, Sapienza University of Rome, Via dei Sardi 70, 00185 Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio Snc, 67100 L'Aquila, Italy
| |
Collapse
|
17
|
Oubraim S, Shen RY, Haj-Dahmane S. Oxytocin excites dorsal raphe serotonin neurons and bidirectionally gates their glutamate synapses. iScience 2023; 26:106707. [PMID: 37250336 PMCID: PMC10214716 DOI: 10.1016/j.isci.2023.106707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Oxytocin (OXT) modulates wide spectrum of social and emotional behaviors via modulation of numerous neurotransmitter systems, including serotonin (5-HT). However, how OXT controls the function of dorsal raphe nucleus (DRN) 5-HT neurons remains unknown. Here, we reveal that OXT excites and alters the firing pattern of 5-HT neurons via activation of postsynaptic OXT receptors (OXTRs). In addition, OXT induces cell-type-specific depression and potentiation of DRN glutamate synapses by two retrograde lipid messengers, 2-arachidonoylglycerol (2-AG) and arachidonic acid (AA), respectively. Neuronal mapping demonstrates that OXT preferentially potentiates glutamate synapses of 5-HT neurons projecting to medial prefrontal cortex (mPFC) and depresses glutamatergic inputs to 5-HT neurons projecting to lateral habenula (LHb) and central amygdala (CeA). Thus, by engaging distinct retrograde lipid messengers, OXT exerts a target-specific gating of glutamate synapses on the DRN. As such, our data uncovers the neuronal mechanisms by which OXT modulates the function of DRN 5-HT neurons.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| |
Collapse
|
18
|
Chen C. Inhibiting degradation of 2-arachidonoylglycerol as a therapeutic strategy for neurodegenerative diseases. Pharmacol Ther 2023; 244:108394. [PMID: 36966972 PMCID: PMC10123871 DOI: 10.1016/j.pharmthera.2023.108394] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Endocannabinoids are endogenous lipid signaling mediators that participate in a variety of physiological and pathological processes. 2-Arachidonoylglycerol (2-AG) is the most abundant endocannabinoid and is a full agonist of G-protein-coupled cannabinoid receptors (CB1R and CB2R), which are targets of Δ9-tetrahydrocannabinol (Δ9-THC), the main psychoactive ingredient in cannabis. While 2-AG has been well recognized as a retrograde messenger modulating synaptic transmission and plasticity at both inhibitory GABAergic and excitatory glutamatergic synapses in the brain, growing evidence suggests that 2-AG also functions as an endogenous terminator of neuroinflammation in response to harmful insults, thus maintaining brain homeostasis. Monoacylglycerol lipase (MAGL) is the key enzyme that degrades 2-AG in the brain. The immediate metabolite of 2-AG is arachidonic acid (AA), a precursor of prostaglandins (PGs) and leukotrienes. Several lines of evidence indicate that pharmacological or genetic inactivation of MAGL, which boosts 2-AG levels and reduces its hydrolytic metabolites, resolves neuroinflammation, mitigates neuropathology, and improves synaptic and cognitive functions in animal models of neurodegenerative diseases, including Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and traumatic brain injury (TBI)-induced neurodegenerative disease. Thus, it has been proposed that MAGL is a potential therapeutic target for treatment of neurodegenerative diseases. As the main enzyme hydrolyzing 2-AG, several MAGL inhibitors have been identified and developed. However, our understanding of the mechanisms by which inactivation of MAGL produces neuroprotective effects in neurodegenerative diseases remains limited. A recent finding that inhibition of 2-AG metabolism in astrocytes, but not in neurons, protects the brain from TBI-induced neuropathology might shed some light on this unsolved issue. This review provides an overview of MAGL as a potential therapeutic target for neurodegenerative diseases and discusses possible mechanisms underlying the neuroprotective effects of restraining degradation of 2-AG in the brain.
Collapse
|
19
|
Steiger LJ, Tsintsadze T, Mattheisen GB, Smith SM. Somatic and terminal CB1 receptors are differentially coupled to voltage-gated sodium channels in neocortical neurons. Cell Rep 2023; 42:112247. [PMID: 36933217 PMCID: PMC10106091 DOI: 10.1016/j.celrep.2023.112247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/13/2023] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
Endogenous cannabinoid signaling is vital for important brain functions, and the same pathways can be modified pharmacologically to treat pain, epilepsy, and posttraumatic stress disorder. Endocannabinoid-mediated changes to excitability are predominantly attributed to 2-arachidonoylglycerol (2-AG) acting presynaptically via the canonical cannabinoid receptor, CB1. Here, we identify a mechanism in the neocortex by which anandamide (AEA), another major endocannabinoid, but not 2-AG, powerfully inhibits somatically recorded voltage-gated sodium channel (VGSC) currents in the majority of neurons. This pathway involves intracellular CB1 that, when activated by anandamide, decreases the likelihood of recurrent action potential generation. WIN 55,212-2 similarly activates CB1 and inhibits VGSC currents, indicating that this pathway is also positioned to mediate the actions of exogenous cannabinoids on neuronal excitability. The coupling between CB1 and VGSCs is absent at nerve terminals, and 2-AG does not block somatic VGSC currents, indicating functional compartmentalization of the actions of two endocannabinoids.
Collapse
Affiliation(s)
- Luke J Steiger
- Section of Pulmonary and Critical Care Medicine, VA Portland Health Care System, Portland, OR, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Timur Tsintsadze
- Section of Pulmonary and Critical Care Medicine, VA Portland Health Care System, Portland, OR, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Glynis B Mattheisen
- Section of Pulmonary and Critical Care Medicine, VA Portland Health Care System, Portland, OR, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephen M Smith
- Section of Pulmonary and Critical Care Medicine, VA Portland Health Care System, Portland, OR, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR 97239, USA; Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
20
|
Salinas-Abarca AB, Martínez-Lorenzana G, Condés-Lara M, González-Hernández A. The role of the endocannabinoid 2-arachidonoylglycerol in the in vivo spinal oxytocin-induced antinociception in male rats. Exp Neurol 2023; 363:114383. [PMID: 36921751 DOI: 10.1016/j.expneurol.2023.114383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/17/2023] [Accepted: 03/12/2023] [Indexed: 03/14/2023]
Abstract
Oxytocin receptor (OTR) activation at the spinal level produces antinociception. Some data suggest that central OTR activation enhances social interaction via an increase of endocannabinoids (eCB), but we do not know if this could occur at the spinal level, modulating pain transmission. Considering that oxytocin via OTR stimulates diacylglycerol formation, a key intermediate in synthesizing 2-arachidonylglycerol (2-AG), an eCB molecule, we sought to test the role of the eCB system on the spinal oxytocin-induced antinociception. Behavioral and electrophysiological experiments were conducted in naïve and formalin-treated (to induce long-term mechanical hypersensitivity) male Wistar rats. Intrathecal RHC 80267 injections, an inhibitor of the enzyme diacylglycerol lipase (thus, decreasing 2-AG formation), produces transient mechanical hypersensitivity, an effect unaltered by oxytocin but reversed by gabapentin. Similarly, in in vivo extracellular recordings of naïve spinal wide dynamic range cells, juxtacellular picoinjection of RHC 80267 increases the firing of nociceptive Aδ-, C-fibers, and post-discharge, an effect unaltered by oxytocin. Interestingly, in sensitized rats, oxytocin picoinjection reverses the RHC 80627-induced hyperactivity of Aδ-fibers (but not C- or post-discharge activity). In contrast, a sub-effective dose of JZL184 (a monoacylglycerol lipase inhibitor, thus favoring 2-AG levels), which does not have per se an antinociceptive effect in the formalin-induced hypernociception, the oxytocin-induced antinociception is boosted. Similarly, electrophysiological experiments suggest that juxtacellular JZL184 diminishes the neuronal firing of nociceptive fibers, and co-injection with oxytocin prolongs and enhances the antinociceptive effect. These data may imply that 2-AG formation may play a role in the spinal antinociception induced by oxytocin.
Collapse
Affiliation(s)
- Ana B Salinas-Abarca
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro, QRO 76230, Mexico; Department of Neural and Pain Sciences, University of Maryland Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA.
| | - Guadalupe Martínez-Lorenzana
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro, QRO 76230, Mexico.
| | - Miguel Condés-Lara
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro, QRO 76230, Mexico.
| | - Abimael González-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Querétaro, QRO 76230, Mexico.
| |
Collapse
|
21
|
The phospholipase A 2 superfamily as a central hub of bioactive lipids and beyond. Pharmacol Ther 2023; 244:108382. [PMID: 36918102 DOI: 10.1016/j.pharmthera.2023.108382] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
In essence, "phospholipase A2" (PLA2) means a group of enzymes that release fatty acids and lysophospholipids by hydrolyzing the sn-2 position of glycerophospholipids. To date, more than 50 enzymes possessing PLA2 or related lipid-metabolizing activities have been identified in mammals, and these are subdivided into several families in terms of their structures, catalytic mechanisms, tissue/cellular localizations, and evolutionary relationships. From a general viewpoint, the PLA2 superfamily has mainly been implicated in signal transduction, driving the production of a wide variety of bioactive lipid mediators. However, a growing body of evidence indicates that PLA2s also contribute to phospholipid remodeling or recycling for membrane homeostasis, fatty acid β-oxidation for energy production, and barrier lipid formation on the body surface. Accordingly, PLA2 enzymes are considered one of the key regulators of a broad range of lipid metabolism, and perturbation of specific PLA2-driven lipid pathways often disrupts tissue and cellular homeostasis and may be associated with a variety of diseases. This review covers current understanding of the physiological functions of the PLA2 superfamily, focusing particularly on the two major intracellular PLA2 families (Ca2+-dependent cytosolic PLA2s and Ca2+-independent patatin-like PLA2s) as well as other PLA2 families, based on studies using gene-manipulated mice and human diseases in combination with comprehensive lipidomics.
Collapse
|
22
|
Patthy Á, Hanics J, Zachar G, Kovács GG, Harkany T, Alpár A. Regional redistribution of CB1 cannabinoid receptors in human foetal brains with Down's syndrome and their functional modifications in Ts65Dn +/+ mice. Neuropathol Appl Neurobiol 2023; 49:e12887. [PMID: 36716771 DOI: 10.1111/nan.12887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023]
Abstract
AIMS The endocannabinoid system with its type 1 cannabinoid receptor (CB1 R) expressed in postmitotic neuroblasts is a critical chemotropic guidance module with its actions cascading across neurogenic commitment, neuronal polarisation and synaptogenesis in vertebrates. Here, we present the systematic analysis of regional CB1 R expression in the developing human brain from gestational week 14 until birth. In parallel, we diagrammed differences in CB1 R development in Down syndrome foetuses and identified altered CB1 R signalling. METHODS Foetal brains with normal development or with Down's syndrome were analysed using standard immunohistochemistry, digitalised light microscopy and image analysis (NanoZoomer). CB1 R function was investigated by in vitro neuropharmacology from neonatal Ts65Dn transgenic mice brains carrying an additional copy of ~90 conserved protein-coding gene orthologues of the human chromosome 21. RESULTS We detected a meshwork of fine-calibre, often varicose processes between the subventricular and intermediate zones of the cortical plate in the late first trimester, when telencephalic fibre tracts develop. The density of CB1 Rs gradually decreased during the second and third trimesters in the neocortex. In contrast, CB1 R density was maintained, or even increased, in the hippocampus. We found the onset of CB1 R expression being delayed by ≥1 month in age-matched foetal brains with Down's syndrome. In vitro, CB1 R excitation induced excess microtubule stabilisation and, consequently, reduced neurite outgrowth. CONCLUSIONS We suggest that neuroarchitectural impairments in Down's syndrome brains involve the delayed development and errant functions of the endocannabinoid system, with a particular impact on endocannabinoids modulating axonal wiring.
Collapse
Affiliation(s)
- Ágoston Patthy
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - János Hanics
- Department of Anatomy, Semmelweis University, Budapest, Hungary.,SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Gergely Zachar
- Department of Anatomy, Semmelweis University, Budapest, Hungary
| | - Gábor G Kovács
- Institute of Neurology, Medical University of Vienna, Vienna, Austria.,Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Canada
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.,Department of Neuroscience, Biomedicum, Karolinska Institutet, Solna, Sweden
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, Budapest, Hungary.,SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
23
|
2-AG-Mediated Control of GABAergic Signaling Is Impaired in a Model of Epilepsy. J Neurosci 2023; 43:571-583. [PMID: 36460464 PMCID: PMC9888507 DOI: 10.1523/jneurosci.0541-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/21/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Repeated seizures result in a persistent maladaptation of endocannabinoid (eCB) signaling, mediated part by anandamide signaling deficiency in the basolateral amygdala (BLA) that manifests as aberrant synaptic function and altered emotional behavior. Here, we determined the effect of repeated seizures (kindling) on 2-arachidonoylglycerol (2-AG) signaling on GABA transmission by directly measuring tonic and phasic eCB-mediated retrograde signaling in an in vitro BLA slice preparation from male rats. We report that both activity-dependent and muscarinic acetylcholine receptor (mAChR)-mediated depression of GABA synaptic transmission was reduced following repeated seizure activity. These effects were recapitulated in sham rats by preincubating slices with the 2-AG synthesizing enzyme inhibitor DO34. Conversely, preincubating slices with the 2-AG degrading enzyme inhibitor KML29 rescued activity-dependent 2-AG signaling, but not mAChR-mediated synaptic depression, over GABA transmission in kindled rats. These effects were not attributable to a change in cannabinoid type 1 (CB1) receptor sensitivity or altered 2-AG tonic signaling since the application of the highly selective CB1 receptor agonist CP55,940 provoked a similar reduction in GABA synaptic activity in both sham and kindled rats, while no effect of either DO34 or of the CB1 inverse agonist AM251 was observed on frequency and amplitude of spontaneous IPSCs in either sham or kindled rats. Collectively, these data provide evidence that repeated amygdala seizures persistently alter phasic 2-AG-mediated retrograde signaling at BLA GABAergic synapses, probably by impairing stimulus-dependent 2-AG synthesis/release, which contributes to the enduring aberrant synaptic plasticity associated with seizure activity.SIGNIFICANCE STATEMENT The plastic reorganization of endocannabinoid (eCB) signaling after seizures and during epileptogenesis may contribute to the negative neurobiological consequences associated with seizure activity. Therefore, a deeper understanding of the molecular basis underlying the pathologic long-term eCB signaling remodeling following seizure activity will be crucial to the development of novel therapies for epilepsy that not only target seizure activity, but, most importantly, the epileptogenesis and the comorbid conditions associated with epilepsy.
Collapse
|
24
|
Bernal‐Chico A, Tepavcevic V, Manterola A, Utrilla C, Matute C, Mato S. Endocannabinoid signaling in brain diseases: Emerging relevance of glial cells. Glia 2023; 71:103-126. [PMID: 35353392 PMCID: PMC9790551 DOI: 10.1002/glia.24172] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
The discovery of cannabinoid receptors as the primary molecular targets of psychotropic cannabinoid Δ9 -tetrahydrocannabinol (Δ9 -THC) in late 1980s paved the way for investigations on the effects of cannabis-based therapeutics in brain pathology. Ever since, a wealth of results obtained from studies on human tissue samples and animal models have highlighted a promising therapeutic potential of cannabinoids and endocannabinoids in a variety of neurological disorders. However, clinical success has been limited and major questions concerning endocannabinoid signaling need to be satisfactorily addressed, particularly with regard to their role as modulators of glial cells in neurodegenerative diseases. Indeed, recent studies have brought into the limelight diverse, often unexpected functions of astrocytes, oligodendrocytes, and microglia in brain injury and disease, thus providing scientific basis for targeting glial cells to treat brain disorders. This Review summarizes the current knowledge on the molecular and cellular hallmarks of endocannabinoid signaling in glial cells and its clinical relevance in neurodegenerative and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Ana Bernal‐Chico
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Neuroimmunology UnitBiocruces BizkaiaBarakaldoSpain
| | | | - Andrea Manterola
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Present address:
Parque Científico y Tecnológico de GuipuzkoaViralgenSan SebastianSpain
| | | | - Carlos Matute
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Susana Mato
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Neuroimmunology UnitBiocruces BizkaiaBarakaldoSpain
| |
Collapse
|
25
|
Martinez Ramirez CE, Ruiz-Pérez G, Stollenwerk TM, Behlke C, Doherty A, Hillard CJ. Endocannabinoid signaling in the central nervous system. Glia 2023; 71:5-35. [PMID: 36308424 PMCID: PMC10167744 DOI: 10.1002/glia.24280] [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: 02/01/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
It is hard to overestimate the influence of the endocannabinoid signaling (ECS) system on central nervous system (CNS) function. In the 40 years since cannabinoids were found to trigger specific cell signaling cascades, studies of the ECS system continue to cause amazement, surprise, and confusion! CB1 cannabinoid receptors are expressed widely in the CNS and regulate cell-cell communication via effects on the release of both neurotransmitters and gliotransmitters. CB2 cannabinoid receptors are difficult to detect in the CNS but seem to "punch above their weight" as compounds targeting these receptors have significant effects on inflammatory state and behavior. Positive and negative allosteric modulators for both receptors have been identified and examined in preclinical studies. Concentrations of the endocannabinoid ligands, N-arachidonoylethanolamine and 2-arachidonoylglycerol (2-AG), are regulated by a combination of enzymatic synthesis and degradation and inhibitors of these processes are available and making their way into clinical trials. Importantly, ECS regulates many essential brain functions, including regulation of reward, anxiety, inflammation, motor control, and cellular development. While the field is on the cusp of preclinical discoveries providing impactful clinical and therapeutic insights into many CNS disorders, there is still much to be learned about this remarkable and versatile modulatory system.
Collapse
Affiliation(s)
- César E Martinez Ramirez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Gonzalo Ruiz-Pérez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Todd M Stollenwerk
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christina Behlke
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ashley Doherty
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cecilia J Hillard
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
26
|
Bisogno T. Assay of DAGLα/β Activity. Methods Mol Biol 2023; 2576:275-283. [PMID: 36152195 DOI: 10.1007/978-1-0716-2728-0_23] [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] [Indexed: 06/16/2023]
Abstract
The endocannabinoid 2-arachidonoylglycerol (2-AG) exerts its physiological action by binding to and functionally activating type-1 (CB1) and type-2 (CB2) cannabinoid receptors. It is thought to be produced through the action of sn-1 selective diacylglycerol lipase (DAGL) that catalyzes 2-AG biosynthesis from sn-2-arachidonate-containing diacylglycerols. Different methodological approaches for measuring DAGL activity in biological samples are now available. Here, a highly sensitive radiometric assay to assess DAGL activity, by using 1-oleoyl[1-14C]-2-arachidonoylglycerol as the substrate, is reported. All the steps required to perform lipid extraction, fractionation by thin-layer chromatography (TLC), and quantification of radiolabeled [14C]-oleic acid via scintillation counting are described in detail.
Collapse
Affiliation(s)
- Tiziana Bisogno
- Endocannabinoid Research Group, Istituto di Farmacologia Traslazionale, Consiglio Nazionale Delle Ricerche, Roma, Italy.
| |
Collapse
|
27
|
Anand U, Pacchetti B, Anand P, Sodergren MH. The Endocannabinoid Analgesic Entourage Effect: Investigations in Cultured DRG Neurons. J Pain Res 2022; 15:3493-3507. [PMID: 36394060 PMCID: PMC9642605 DOI: 10.2147/jpr.s378876] [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: 06/30/2022] [Accepted: 10/08/2022] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The endocannabinoid 2-Arachidonyl glycerol (2-AG) exerts dose-related anti-nociceptive effects, which are potentiated by the related but inactive 2-palmitoyl glycerol (2-PG) and 2-linoleoyl glycerol (2-LG). This potentiation of analgesia and other in vivo measures was described as the "entourage effect". We investigated this effect on TRPV1 signalling in cultured dorsal root ganglion (DRG) nociceptors. METHODS Adult rat DRG neurons were cultured in medium containing NGF and GDNF at 37°C. 48 h later cultures were loaded with 2 µM Fura2AM for calcium imaging, and treated with 2-AG, 2-PG and 2-LG, individually or combined, for 5 min, followed by 1 µMol capsaicin. The amplitude and latency of capsaicin responses were measured (N=3-7 rats, controls N=16), and analysed. RESULTS In controls, 1 µMol capsaicin elicited immediate calcium influx in a subset of neurons, with average latency of 1.27 ± 0.2 s and amplitude of 0.15 ± 0.01 Units. 2-AG (10-100 µMol) elicited calcium influx in some neurons. In the presence of 2-AG (0.001-100 µMol), capsaicin responses were markedly delayed in 64% neurons by up to 320 s (P<0.001). 2-PG increased capsaicin response latency at 0.1 nMol-100 µMol (P<0.001), in 60% neurons, as did 2-LG at 0.1-100 µMol (P<0.001), in 76% neurons. Increased capsaicin response latency due to 2-AG and 2-PG was sensitive to the CB2 but not to the CB1 receptor antagonist. Combined application of 1 µMol 2-AG, 5 µMol 2-PG and 10 µMol 2-LG, also resulted in significantly increased capsaicin response latency up to 281.5 ± 41.5 s (P<0.001), in 96% neurons, that was partially restored by the CB2, but not the CB1 antagonist. CONCLUSION 2-AG, 2-LG and 2-PG significantly delayed TRPV1 signalling in the majority of capsaicin-sensitive DRG neurons, that was markedly increased following combined application. Further studies of these endocannabinoids are required to identify the underlying mechanisms.
Collapse
Affiliation(s)
- Uma Anand
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, London, W12 0HS, UK
| | | | - Praveen Anand
- Professor of Clinical Neurology, Department of Brain Sciences, Imperial College London, London, W12 0HS, UK
| | - Mikael Hans Sodergren
- Medical Cannabis Research Group, Department of Surgery and Cancer, Imperial College London, London, W12 0HS, UK
- Curaleaf International Limited, London, EC2A 2EW, UK
| |
Collapse
|
28
|
Echeazarra L, Barrondo S, García del Caño G, Bonilla-Del Río I, Egaña-Huguet J, Puente N, Aretxabala X, Montaña M, López de Jesús M, González-Burguera I, Saumell-Esnaola M, Goicolea MA, Grandes P, Sallés J. Up-regulation of CB1 cannabinoid receptors located at glutamatergic terminals in the medial prefrontal cortex of the obese Zucker rat. Front Neuroanat 2022; 16:1004702. [PMID: 36329829 PMCID: PMC9623818 DOI: 10.3389/fnana.2022.1004702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
The present study describes a detailed neuroanatomical distribution map of the cannabinoid type 1 (CB1) receptor, along with the biochemical characterization of the expression and functional coupling to their cognate Gi/o proteins in the medial prefrontal cortex (mPCx) of the obese Zucker rats. The CB1 receptor density was higher in the prelimbic (PL) and infralimbic (IL) subregions of the mPCx of obese Zucker rats relative to their lean littermates which was associated with a higher percentage of CB1 receptor immunopositive excitatory presynaptic terminals in PL and IL. Also, a higher expression of CB1 receptors and WIN55,212-2-stimulated [35S]GTPγS binding was observed in the mPCx but not in the neocortex (NCx) and hippocampus of obese rats. Low-frequency stimulation in layers II/III of the mPCx induced CB1 receptor-dependent long-term synaptic plasticity in IL of area obese Zucker but not lean rats. Overall, the elevated 2-AG levels, up-regulation of CB1 receptors, and increased agonist-stimulated [35S]GTPγS binding strongly suggest that hyperactivity of the endocannabinoid signaling takes place at the glutamatergic terminals of the mPCx in the obese Zucker rat. These findings could endorse the importance of the CB1 receptors located in the mPCx in the development of obesity in Zucker rats.
Collapse
Affiliation(s)
- Leyre Echeazarra
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, Dispositivos Móviles para el Control de Enfermedades Crónicas, Vitoria-Gasteiz, Spain
| | - Sergio Barrondo
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
| | - Gontzal García del Caño
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Itziar Bonilla-Del Río
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jon Egaña-Huguet
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Nagore Puente
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Xabier Aretxabala
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Mario Montaña
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Maider López de Jesús
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
| | - Imanol González-Burguera
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Miquel Saumell-Esnaola
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
| | - María Aránzazu Goicolea
- Department of Analytical Chemistry, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country (UPV/EHU), Leioa, Spain
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Joan Sallés
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, Vitoria-Gasteiz, Spain
- *Correspondence: Joan Sallés,
| |
Collapse
|
29
|
Itami C, Uesaka N, Huang JY, Lu HC, Sakimura K, Kano M, Kimura F. Endocannabinoid-dependent formation of columnar axonal projection in the mouse cerebral cortex. Proc Natl Acad Sci U S A 2022; 119:e2122700119. [PMID: 36067295 PMCID: PMC9477236 DOI: 10.1073/pnas.2122700119] [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: 12/16/2021] [Accepted: 07/19/2022] [Indexed: 11/18/2022] Open
Abstract
Columnar structure is one of the most fundamental morphological features of the cerebral cortex and is thought to be the basis of information processing in higher animals. Yet, how such a topographically precise structure is formed is largely unknown. Formation of columnar projection of layer 4 (L4) axons is preceded by thalamocortical formation, in which type 1 cannabinoid receptors (CB1R) play an important role in shaping barrel-specific targeted projection by operating spike timing-dependent plasticity during development (Itami et al., J. Neurosci. 36, 7039-7054 [2016]; Kimura & Itami, J. Neurosci. 39, 3784-3791 [2019]). Right after the formation of thalamocortical projections, CB1Rs start to function at L4 axon terminals (Itami & Kimura, J. Neurosci. 32, 15000-15011 [2012]), which coincides with the timing of columnar shaping of L4 axons. Here, we show that the endocannabinoid 2-arachidonoylglycerol (2-AG) plays a crucial role in columnar shaping. We found that L4 axon projections were less organized until P12 and then became columnar after CB1Rs became functional. By contrast, the columnar organization of L4 axons was collapsed in mice genetically lacking diacylglycerol lipase α, the major enzyme for 2-AG synthesis. Intraperitoneally administered CB1R agonists shortened axon length, whereas knockout of CB1R in L4 neurons impaired columnar projection of their axons. Our results suggest that endocannabinoid signaling is crucial for shaping columnar axonal projection in the cerebral cortex.
Collapse
Affiliation(s)
- Chiaki Itami
- Department of Physiology, Faculty of Medicine, Saitama Medical University, Moroyama, Saitama 350-0495, Japan
- The Linda and Jack Gill Center for Biomolecular Sciences, Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405
| | - Naofumi Uesaka
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- Present address, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Jui-Yen Huang
- The Linda and Jack Gill Center for Biomolecular Sciences, Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, 113-0033, Japan
| | - Fumitaka Kimura
- Department of Molecular Neuroscience, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Laboratory of Brain Neuroscience, Faculty of Medical Sciences, Jikei University of Health Care and Sciences, Osaka, 532-0003, Japan
| |
Collapse
|
30
|
Kaczocha M, Haj-Dahmane S. Mechanisms of endocannabinoid transport in the brain. Br J Pharmacol 2022; 179:4300-4310. [PMID: 33786823 PMCID: PMC8481389 DOI: 10.1111/bph.15469] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The endocannabinoids 2-arachidonoylglycerol (2-AG) and anandamide are among the best studied lipid messengers in the brain. By activating cannabinoid receptors in the CNS, endocannabinoids tune synaptic function, thereby influencing a variety of physiological and behavioural processes. Extensive research conducted over the last few decades has considerably enhanced our understanding of the molecular mechanisms and physiological functions of the endocannabinoid system. It is now well-established that endocannabinoids are synthesized by postsynaptic neurons and serve as retrograde messengers that suppress neurotransmitter release at central synapses. While the detailed mechanisms by which endocannabinoids gate synaptic function and behavioural processes are relatively well characterized, the mechanisms governing endocannabinoid transport at central synapses remain ill defined. Recently, several studies have begun to unravel the mechanisms governing intracellular and intercellular endocannabinoid transport. In this review, we will focus on new advances in the mechanisms of intracellular and synaptic endocannabinoid transport in the CNS. LINKED ARTICLES: This article is part of a themed issue on New discoveries and perspectives in mental and pain disorders. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.17/issuetoc.
Collapse
Affiliation(s)
- Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
- Neuroscience Program, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
31
|
Martin SC, Gay SM, Armstrong ML, Pazhayam NM, Reisdorph N, Diering GH. Tonic endocannabinoid signaling supports sleep through development in both sexes. Sleep 2022; 45:6565640. [PMID: 35395682 PMCID: PMC9366650 DOI: 10.1093/sleep/zsac083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Sleep is an essential behavior that supports brain function and cognition throughout life, in part by acting on neuronal synapses. The synaptic signaling pathways that mediate the restorative benefits of sleep are not fully understood, particularly in the context of development. Endocannabinoids (eCBs) including 2-arachidonyl glycerol (2-AG) and anandamide (AEA), are bioactive lipids that activate cannabinoid receptor, CB1, to regulate synaptic transmission and mediate cognitive functions and many behaviors, including sleep. We used targeted mass spectrometry to measure changes in forebrain synaptic eCBs during the sleep/wake cycle in juvenile and adolescent mice of both sexes. We find that eCBs lack a daily rhythm in juvenile mice, while in adolescents AEA and related oleoyl ethanolamide are increased during the sleep phase in a circadian manner. Next, we manipulated the eCB system using selective pharmacology and measured the effects on sleep behavior in developing and adult mice of both sexes using a noninvasive piezoelectric home-cage recording apparatus. Enhancement of eCB signaling through inhibition of 2-AG or AEA degradation, increased dark-phase sleep amount and bout length in developing and adult males, but not in females. Inhibition of CB1 by injection of the antagonist AM251 reduced sleep time and caused sleep fragmentation in developing and adult males and females. Our data suggest that males are more sensitive to the sleep-promoting effects of enhanced eCBs but that tonic eCB signaling supports sleep behavior through multiple stages of development in both sexes. This work informs the further development of cannabinoid-based therapeutics for sleep disruption.
Collapse
Affiliation(s)
- Shenée C Martin
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Sean M Gay
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Michael L Armstrong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, CO , USA
| | - Nila M Pazhayam
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, CO , USA
| | - Graham H Diering
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill , Chapel Hill, NC , USA
- Carolina Institute for Developmental Disabilities , Carrboro, NC , USA
| |
Collapse
|
32
|
Murakami M, Takamiya R, Miki Y, Sugimoto N, Nagasaki Y, Suzuki-Yamamoto T, Taketomi Y. Segregated functions of two cytosolic phospholipase A 2 isoforms (cPLA 2α and cPLA 2ε) in lipid mediator generation. Biochem Pharmacol 2022; 203:115176. [PMID: 35841927 DOI: 10.1016/j.bcp.2022.115176] [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: 06/01/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/16/2022]
Abstract
Among the phospholipase A2 (PLA2) superfamily, group IVA cytosolic PLA2 (cPLA2α) is currently attracting much attention as a central regulator of arachidonic acid (AA) metabolism linked to eicosanoid biosynthesis. Following cell activation, cPLA2α selectively releases AA, a precursor of a variety of eicosanoids, from phospholipids in perinuclear membrane compartments. cPLA2α-null mice display various phenotypes that could be largely explained by reduced eicosanoid signaling. In contrast, group IVE cPLA2ε, another member of the cPLA2 family, acts as a Ca2+-dependent N-acyltransferase rather than a PLA2, thereby regulating the biosynthesis of N-acylethanolamines (NAEs), a unique class of lipid mediators with an anti-inflammatory effect. In response to Ca2+ signaling, cPLA2ε translocates to phosphatidylserine-rich organelle membranes in the endocytic/recycling pathway. In vivo, cPLA2ε is induced in keratinocytes of psoriatic skin, and its genetic deletion exacerbates psoriatic inflammation due to a marked reduction of NAE-related lipids. cPLA2ε also contributes to NAE generation in several if not all mouse tissues. Thus, the two members of the cPLA2 family, cPLA2α and cPLA2ε, catalyze distinct enzymatic reactions to mobilize distinct sets of lipid mediators, thereby differently regulating pathophysiological events in health and disease. Such segregation of the cPLA2α-eicosanoid and cPLA2ε-NAE pathways represents a new paradigm of research on PLA2s and lipid mediators.
Collapse
Affiliation(s)
- Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Rina Takamiya
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshimi Miki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nao Sugimoto
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Nagasaki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Nutritional Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan
| | - Toshiko Suzuki-Yamamoto
- Department of Nutritional Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
33
|
Dhakal P, Chaudhry SI, Signorelli R, Collins KM. Serotonin signals through postsynaptic Gαq, Trio RhoGEF, and diacylglycerol to promote Caenorhabditis elegans egg-laying circuit activity and behavior. Genetics 2022; 221:iyac084. [PMID: 35579369 PMCID: PMC9252285 DOI: 10.1093/genetics/iyac084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/26/2022] [Indexed: 11/12/2022] Open
Abstract
Activated Gαq signals through phospholipase-Cβ and Trio, a Rho GTPase exchange factor (RhoGEF), but how these distinct effector pathways promote cellular responses to neurotransmitters like serotonin remains poorly understood. We used the egg-laying behavior circuit of Caenorhabditis elegans to determine whether phospholipase-Cβ and Trio mediate serotonin and Gαq signaling through independent or related biochemical pathways. Our genetic rescue experiments suggest that phospholipase-Cβ functions in neurons while Trio Rho GTPase exchange factor functions in both neurons and the postsynaptic vulval muscles. While Gαq, phospholipase-Cβ, and Trio Rho GTPase exchange factor mutants fail to lay eggs in response to serotonin, optogenetic stimulation of the serotonin-releasing HSN neurons restores egg laying only in phospholipase-Cβ mutants. Phospholipase-Cβ mutants showed vulval muscle Ca2+ transients while strong Gαq and Trio Rho GTPase exchange factor mutants had little or no vulval muscle Ca2+ activity. Treatment with phorbol 12-myristate 13-acetate that mimics 1,2-diacylglycerol, a product of PIP2 hydrolysis, rescued egg-laying circuit activity and behavior defects of Gαq signaling mutants, suggesting both phospholipase-C and Rho signaling promote synaptic transmission and egg laying via modulation of 1,2-diacylglycerol levels. 1,2-Diacylglycerol activates effectors including UNC-13; however, we find that phorbol esters, but not serotonin, stimulate egg laying in unc-13 and phospholipase-Cβ mutants. These results support a model where serotonin signaling through Gαq, phospholipase-Cβ, and UNC-13 promotes neurotransmitter release, and that serotonin also signals through Gαq, Trio Rho GTPase exchange factor, and an unidentified, phorbol 12-myristate 13-acetate-responsive effector to promote postsynaptic muscle excitability. Thus, the same neuromodulator serotonin can signal in distinct cells and effector pathways to coordinate activation of a motor behavior circuit.
Collapse
Affiliation(s)
- Pravat Dhakal
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | - Sana I Chaudhry
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | | | - Kevin M Collins
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
34
|
Vicente-Acosta A, Ceprian M, Sobrino P, Pazos MR, Loría F. Cannabinoids as Glial Cell Modulators in Ischemic Stroke: Implications for Neuroprotection. Front Pharmacol 2022; 13:888222. [PMID: 35721207 PMCID: PMC9199389 DOI: 10.3389/fphar.2022.888222] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the second leading cause of death worldwide following coronary heart disease. Despite significant efforts to find effective treatments to reduce neurological damage, many patients suffer from sequelae that impair their quality of life. For this reason, the search for new therapeutic options for the treatment of these patients is a priority. Glial cells, including microglia, astrocytes and oligodendrocytes, participate in crucial processes that allow the correct functioning of the neural tissue, being actively involved in the pathophysiological mechanisms of ischemic stroke. Although the exact mechanisms by which glial cells contribute in the pathophysiological context of stroke are not yet completely understood, they have emerged as potentially therapeutic targets to improve brain recovery. The endocannabinoid system has interesting immunomodulatory and protective effects in glial cells, and the pharmacological modulation of this signaling pathway has revealed potential neuroprotective effects in different neurological diseases. Therefore, here we recapitulate current findings on the potential promising contribution of the endocannabinoid system pharmacological manipulation in glial cells for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Andrés Vicente-Acosta
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria Ceprian
- ERC Team, PGNM, INSERM U1315, CNRS UMR5261, University of Lyon 1, University of Lyon, Lyon, France
| | - Pilar Sobrino
- Departamento de Neurología, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Maria Ruth Pazos
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Frida Loría
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| |
Collapse
|
35
|
Liu Z, Yang N, Dong J, Tian W, Chang L, Ma J, Guo J, Tan J, Dong A, He K, Zhou J, Cinar R, Wu J, Salinas AG, Sun L, Kumar M, Sullivan BT, Oldham BB, Pitz V, Makarious MB, Ding J, Kung J, Xie C, Hawes SL, Wang L, Wang T, Chan P, Zhang Z, Le W, Chen S, Lovinger DM, Blauwendraat C, Singleton AB, Cui G, Li Y, Cai H, Tang B. Deficiency in endocannabinoid synthase DAGLB contributes to early onset Parkinsonism and murine nigral dopaminergic neuron dysfunction. Nat Commun 2022; 13:3490. [PMID: 35715418 PMCID: PMC9205912 DOI: 10.1038/s41467-022-31168-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
Endocannabinoid (eCB), 2-arachidonoyl-glycerol (2-AG), the most abundant eCB in the brain, regulates diverse neural functions. Here we linked multiple homozygous loss-of-function mutations in 2-AG synthase diacylglycerol lipase β (DAGLB) to an early onset autosomal recessive Parkinsonism. DAGLB is the main 2-AG synthase in human and mouse substantia nigra (SN) dopaminergic neurons (DANs). In mice, the SN 2-AG levels were markedly correlated with motor performance during locomotor skill acquisition. Genetic knockdown of Daglb in nigral DANs substantially reduced SN 2-AG levels and impaired locomotor skill learning, particularly the across-session learning. Conversely, pharmacological inhibition of 2-AG degradation increased nigral 2-AG levels, DAN activity and dopamine release and rescued the locomotor skill learning deficits. Together, we demonstrate that DAGLB-deficiency contributes to the pathogenesis of Parkinsonism, reveal the importance of DAGLB-mediated 2-AG biosynthesis in nigral DANs in regulating neuronal activity and dopamine release, and suggest potential benefits of 2-AG augmentation in alleviating Parkinsonism.
Collapse
Affiliation(s)
- Zhenhua Liu
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Nannan Yang
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Jie Dong
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Clinical Research Center on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, 116011, Dalian, Liaoning, China
| | - Wotu Tian
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 20025, Shanghai, China
| | - Lisa Chang
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jinghong Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, 100053, Beijing, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Jieqiong Tan
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, 410008, Changsha, Hunan, China
| | - Ao Dong
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
| | - Kaikai He
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, 100871, Beijing, China
| | - Jingheng Zhou
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Junbing Wu
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Armando G Salinas
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mantosh Kumar
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Breanna T Sullivan
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Braden B Oldham
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Vanessa Pitz
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mary B Makarious
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jinhui Ding
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Justin Kung
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chengsong Xie
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sarah L Hawes
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lupeng Wang
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, Hubei, China
| | - Piu Chan
- Department of Neurology, Xuanwu Hospital of Capital Medical University, 100053, Beijing, China
| | - Zhuohua Zhang
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, 410008, Changsha, Hunan, China
- Department of Neurosciences, University of South China Medical School, 421200, Hengyang, Hunan, China
| | - Weidong Le
- Clinical Research Center on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, 116011, Dalian, Liaoning, China
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Medical School of University of Electronics & Technology of China, 610045, Chengdu, Sichuan, China
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 20025, Shanghai, China
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20852, USA
| | - Cornelis Blauwendraat
- Integrative Neurogenomics Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew B Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guohong Cui
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, 100871, Beijing, China
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Centre for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, 410008, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
36
|
Molecular Alterations of the Endocannabinoid System in Psychiatric Disorders. Int J Mol Sci 2022; 23:ijms23094764. [PMID: 35563156 PMCID: PMC9104141 DOI: 10.3390/ijms23094764] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/07/2023] Open
Abstract
The therapeutic benefits of the current medications for patients with psychiatric disorders contrast with a great variety of adverse effects. The endocannabinoid system (ECS) components have gained high interest as potential new targets for treating psychiatry diseases because of their neuromodulator role, which is essential to understanding the regulation of many brain functions. This article reviewed the molecular alterations in ECS occurring in different psychiatric conditions. The methods used to identify alterations in the ECS were also described. We used a translational approach. The animal models reproducing some behavioral and/or neurochemical aspects of psychiatric disorders and the molecular alterations in clinical studies in post-mortem brain tissue or peripheral tissues were analyzed. This article reviewed the most relevant ECS changes in prevalent psychiatric diseases such as mood disorders, schizophrenia, autism, attentional deficit, eating disorders (ED), and addiction. The review concludes that clinical research studies are urgently needed for two different purposes: (1) To identify alterations of the ECS components potentially useful as new biomarkers relating to a specific disease or condition, and (2) to design new therapeutic targets based on the specific alterations found to improve the pharmacological treatment in psychiatry.
Collapse
|
37
|
Davies AK, Alecu JE, Ziegler M, Vasilopoulou CG, Merciai F, Jumo H, Afshar-Saber W, Sahin M, Ebrahimi-Fakhari D, Borner GHH. AP-4-mediated axonal transport controls endocannabinoid production in neurons. Nat Commun 2022; 13:1058. [PMID: 35217685 PMCID: PMC8881493 DOI: 10.1038/s41467-022-28609-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/08/2022] [Indexed: 01/20/2023] Open
Abstract
The adaptor protein complex AP-4 mediates anterograde axonal transport and is essential for axon health. AP-4-deficient patients suffer from a severe neurodevelopmental and neurodegenerative disorder. Here we identify DAGLB (diacylglycerol lipase-beta), a key enzyme for generation of the endocannabinoid 2-AG (2-arachidonoylglycerol), as a cargo of AP-4 vesicles. During normal development, DAGLB is targeted to the axon, where 2-AG signalling drives axonal growth. We show that DAGLB accumulates at the trans-Golgi network of AP-4-deficient cells, that axonal DAGLB levels are reduced in neurons from a patient with AP-4 deficiency, and that 2-AG levels are reduced in the brains of AP-4 knockout mice. Importantly, we demonstrate that neurite growth defects of AP-4-deficient neurons are rescued by inhibition of MGLL (monoacylglycerol lipase), the enzyme responsible for 2-AG hydrolysis. Our study supports a new model for AP-4 deficiency syndrome in which axon growth defects arise through spatial dysregulation of endocannabinoid signalling. Davies et al. identify a putative mechanism underlying the childhood neurological disorder AP-4 deficiency syndrome. In the absence of AP-4, an enzyme that makes 2-AG is not transported to the axon, leading to axonal growth defects, which can be rescued by inhibition of 2-AG breakdown.
Collapse
Affiliation(s)
- Alexandra K Davies
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| | - Julian E Alecu
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marvin Ziegler
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Functional Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, INF 307, Heidelberg, 69120, Germany
| | - Catherine G Vasilopoulou
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Fabrizio Merciai
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.,Department of Pharmacy and PhD Program in Drug Discovery and Development, University of Salerno, 84084, Fisciano, SA, Italy
| | - Hellen Jumo
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wardiya Afshar-Saber
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Darius Ebrahimi-Fakhari
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Georg H H Borner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.
| |
Collapse
|
38
|
Nakano T, Goto K. Diacylglycerol Kinase ε in Adipose Tissues: A Crosstalk Between Signal Transduction and Energy Metabolism. Front Physiol 2022; 13:815085. [PMID: 35153836 PMCID: PMC8829450 DOI: 10.3389/fphys.2022.815085] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Diacylglycerol (DG) is unique in lipid metabolism because it serves not only as an intermediate product for triglyceride synthesis, but also as a signaling molecule that activates proteins containing DG-responsive elements, such as protein kinase C. Consequently, DG acts as a hub between energy metabolism and intracellular signaling. Of DG metabolizing pathways, DG kinase (DGK) phosphorylates DG to produce phosphatidic acid, which also serves as a second messenger. Several lines of evidence suggest that DGK is deeply involved in metabolic diseases such as obesity and insulin resistance. Of DGK isozymes, DGKε is simplest in terms of structure, but it is characterized by substrate specificity toward arachidonoyl-DG. Recently, we have reported that DGKε deficiency promotes adipose tissue remodeling in mice during the course of high fat diet (HFD) feeding regimen including obesity, insulin resistance, and beige adipogenesis. DGKε ablation engenders altered expression of other lipid metabolizing enzymes, including adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), and diacylglycerol acyltransferase (DGAT). Subcellular localization of DGKε in the endoplasmic reticulum suggests involvement of this isozyme in lipid energy homeostasis. This review presents current findings of DGKε in lipid-orchestrated pathophysiology, especially unique phenotypes of DGKε-knockout mice in the early and late stages of obesogenic conditions.
Collapse
|
39
|
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: 12] [Impact Index Per Article: 6.0] [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
|
40
|
Selvaraj P, Tanaka M, Wen J, Zhang Y. The Novel Monoacylglycerol Lipase Inhibitor MJN110 Suppresses Neuroinflammation, Normalizes Synaptic Composition and Improves Behavioral Performance in the Repetitive Traumatic Brain Injury Mouse Model. Cells 2021; 10:cells10123454. [PMID: 34943962 PMCID: PMC8700188 DOI: 10.3390/cells10123454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
Modulation of the endocannabinoid system has emerged as an effective approach for the treatment of many neurodegenerative and neuropsychological diseases. However, the underlying mechanisms are still uncertain. Using a repetitive mild traumatic brain injury (mTBI) mouse model, we found that there was an impairment in locomotor function and working memory within two weeks post-injury, and that treatment with MJN110, a novel inhibitor of the principal 2-arachidononyl glycerol (2-AG) hydrolytic enzyme monoacylglycerol lipase dose-dependently ameliorated those behavioral changes. Spatial learning and memory deficits examined by Morris water maze between three and four weeks post-TBI were also reversed in the drug treated animals. Administration of MJN110 selectively elevated the levels of 2-AG and reduced the production of arachidonic acid (AA) and prostaglandin E2 (PGE2) in the TBI mouse brain. The increased production of proinflammatory cytokines, accumulation of astrocytes and microglia in the TBI mouse ipsilateral cerebral cortex and hippocampus were significantly reduced by MJN110 treatment. Neuronal cell death was also attenuated in the drug treated animals. MJN110 treatment normalized the expression of the NMDA receptor subunits NR2A and NR2B, the AMPA receptor subunits GluR1 and GluR2, and the GABAA receptor subunits α1, β2,3 and γ2, which were all reduced at 1, 2 and 4 weeks post-injury. The reduced inflammatory response and restored glutamate and GABA receptor expression likely contribute to the improved motor function, learning and memory in the MJN110 treated animals. The therapeutic effects of MJN110 were partially mediated by activation of CB1 and CB2 cannabinoid receptors and were eliminated when it was co-administered with DO34, a novel inhibitor of the 2-AG biosynthetic enzymes. Our results suggest that augmentation of the endogenous levels of 2-AG can be therapeutically useful in the treatment of TBI by suppressing neuroinflammation and maintaining the balance between excitatory and inhibitory neurotransmission.
Collapse
Affiliation(s)
- Prabhuanand Selvaraj
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
| | - Jie Wen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (P.S.); (M.T.); (J.W.)
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +1-301-295-3212
| |
Collapse
|
41
|
Phospholipase Cγ2 regulates endocannabinoid and eicosanoid networks in innate immune cells. Proc Natl Acad Sci U S A 2021; 118:2112971118. [PMID: 34607960 DOI: 10.1073/pnas.2112971118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
Human genetic studies have pointed to a prominent role for innate immunity and lipid pathways in immunological and neurodegenerative disorders. Our understanding of the composition and function of immunomodulatory lipid networks in innate immune cells, however, remains incomplete. Here, we show that phospholipase Cγ2 (PLCγ2 or PLCG2)-mutations in which are associated with autoinflammatory disorders and Alzheimer's disease-serves as a principal source of diacylglycerol (DAG) pools that are converted into a cascade of bioactive endocannabinoid and eicosanoid lipids by DAG lipase (DAGL) and monoacylglycerol lipase (MGLL) enzymes in innate immune cells. We show that this lipid network is tonically stimulated by disease-relevant human mutations in PLCγ2, as well as Fc receptor activation in primary human and mouse macrophages. Genetic disruption of PLCγ2 in mouse microglia suppressed DAGL/MGLL-mediated endocannabinoid-eicosanoid cross-talk and also caused widespread transcriptional and proteomic changes, including the reorganization of immune-relevant lipid pathways reflected in reductions in DAGLB and elevations in PLA2G4A. Despite these changes, Plcg2 -/- mice showed generally normal proinflammatory cytokine and chemokine responses to lipopolysaccharide treatment, instead displaying a more restricted deficit in microglial activation that included impairments in prostaglandin production and CD68 expression. Our findings enhance the understanding of PLCγ2 function in innate immune cells, delineating a role in cross-talk with endocannabinoid/eicosanoid pathways and modulation of subsets of cellular responses to inflammatory stimuli.
Collapse
|
42
|
Lee J, Kwag J. Activation of PLCβ1 enhances endocannabinoid mobilization to restore hippocampal spike-timing-dependent potentiation and contextual fear memory impaired by Alzheimer's amyloidosis. ALZHEIMERS RESEARCH & THERAPY 2021; 13:165. [PMID: 34625112 PMCID: PMC8501622 DOI: 10.1186/s13195-021-00901-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/12/2021] [Indexed: 11/11/2022]
Abstract
Background Accumulation of amyloid beta oligomers (AβO) in Alzheimer’s disease (AD) impairs hippocampal long-term potentiation (LTP), leading to memory deficits. Thus, identifying the molecular targets of AβO involved in LTP inhibition is critical for developing therapeutics for AD. Endocannabinoid (eCB) synthesis and release, a process collectively called eCB mobilization by hippocampal CA1 pyramidal cells, is known to facilitate LTP induction. eCB can be mobilized either by postsynaptic depolarization in an intracellular Ca2+ concentration ([Ca2+]i)-dependent pathway or by group 1 metabotropic glutamate receptor (mGluR) activation in a phospholipase Cβ (PLCβ)-dependent pathway. Moreover, group 1 mGluR activation during postsynaptic depolarization, which is likely to occur in vivo during memory processing, can cause synergistic enhancement of eCB (S-eCB) mobilization in a PLCβ-dependent pathway. Although AβO has been shown to disrupt [Ca2+]i-dependent eCB mobilization, the effect of AβO on PLCβ-dependent S-eCB mobilization and its association with LTP and hippocampus-dependent memory impairments in AD is unknown. Methods We used in vitro whole-cell patch-clamp recordings and western blot analyses to investigate the effect of AβO on PLCβ protein levels, PLCβ-dependent S-eCB mobilization, and spike-timing-dependent potentiation (tLTP) in AβO-treated rat hippocampal slices in vitro. In addition, we assessed the relationship between PLCβ protein levels and hippocampus-dependent memory impairment by performing a contextual fear memory task in vivo in the 5XFAD mouse model of AD. Results We found that AβO treatment in rat hippocampal slices in vitro decreased hippocampal PLCβ1 protein levels and disrupted S-eCB mobilization, as measured by western blot analysis and in vitro whole-cell patch-clamp recordings. This consequently led to the impairment of NMDA receptor (NMDAR)-mediated tLTP at CA3-CA1 excitatory synapses in AβO-treated rat hippocampal slices in vitro. Application of the PLCβ activator, m-3M3FBS, in rat hippocampal slices reinstated PLCβ1 protein levels to fully restore S-eCB mobilization and NMDAR-mediated tLTP. In addition, direct hippocampal injection of m-3M3FBS in 5XFAD mice reinstated PLCβ1 protein levels to those observed in wild type control mice and fully restored hippocampus-dependent contextual fear memory in vivo in 5XFAD mice. Conclusion We suggest that these results might be the consequence of memory impairment in AD by disrupting S-eCB mobilization. Therefore, we propose that PLCβ-dependent S-eCB mobilization could provide a new therapeutic strategy for treating memory deficits in AD.
Collapse
Affiliation(s)
- Jaedong Lee
- Department of Brain and Cognitive Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, South Korea
| | - Jeehyun Kwag
- Department of Brain and Cognitive Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, South Korea.
| |
Collapse
|
43
|
Zanfirescu A, Ungurianu A, Mihai DP, Radulescu D, Nitulescu GM. Targeting Monoacylglycerol Lipase in Pursuit of Therapies for Neurological and Neurodegenerative Diseases. Molecules 2021; 26:5668. [PMID: 34577139 PMCID: PMC8468992 DOI: 10.3390/molecules26185668] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Neurological and neurodegenerative diseases are debilitating conditions, and frequently lack an effective treatment. Monoacylglycerol lipase (MAGL) is a key enzyme involved in the metabolism of 2-AG (2-arachidonoylglycerol), a neuroprotective endocannabinoid intimately linked to the generation of pro- and anti-inflammatory molecules. Consequently, synthesizing selective MAGL inhibitors has become a focus point in drug design and development. The purpose of this review was to summarize the diverse synthetic scaffolds of MAGL inhibitors concerning their potency, mechanisms of action and potential therapeutic applications, focusing on the results of studies published in the past five years. The main irreversible inhibitors identified were derivatives of hexafluoroisopropyl alcohol carbamates, glycol carbamates, azetidone triazole ureas and benzisothiazolinone, whereas the most promising reversible inhibitors were derivatives of salicylketoxime, piperidine, pyrrolidone and azetidinyl amides. We reviewed the results of in-depth chemical, mechanistic and computational studies on MAGL inhibitors, in addition to the results of in vitro findings concerning selectivity and potency of inhibitors, using the half maximal inhibitory concentration (IC50) as an indicator of their effect on MAGL. Further, for highlighting the potential usefulness of highly selective and effective inhibitors, we examined the preclinical in vivo reports regarding the promising therapeutic applications of MAGL pharmacological inhibition.
Collapse
Affiliation(s)
| | - Anca Ungurianu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (A.Z.); (D.P.M.); (D.R.); (G.M.N.)
| | | | | | | |
Collapse
|
44
|
Yoon S, Myczek K, Penzes P. cAMP Signaling-Mediated Phosphorylation of Diacylglycerol Lipase α Regulates Interaction With Ankyrin-G and Dendritic Spine Morphology. Biol Psychiatry 2021; 90:263-274. [PMID: 34099188 PMCID: PMC8384113 DOI: 10.1016/j.biopsych.2021.03.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Diacylglycerol lipase α (DAGLα), a major biosynthetic enzyme for endogenous cannabinoid signaling, has emerged as a risk gene in multiple psychiatric disorders. However, its role in the regulation of dendritic spine plasticity is unclear. METHODS DAGLα wild-type or point mutants were overexpressed in primary cortical neurons or human embryonic kidney 293T cells. The effects of mutated variants on interaction, dendritic spine morphology, and dynamics were examined by proximity ligation assay or fluorescence recovery after photobleaching. Behavioral tests and immunohistochemistry were performed with ankyrin-G conditional knockout and wild-type male mice. RESULTS DAGLα modulated dendritic spine size and density, but the effects of changes in its protein level versus enzymatic activity were different, implicating either a 2-arachidonoylglycerol (2-AG)-dependent or -independent mechanism. The 2-AG-independent effects were mediated by the interaction of DAGLα with ankyrin-G, a multifunctional scaffold protein implicated in psychiatric disorders. Using superresolution microscopy, we observed that they colocalized in distinct nanodomains, which correlated with spine size. In situ proximity ligation assay combined with structured illumination microscopy revealed that DAGLα phosphorylation upon forskolin treatment enhanced the interaction with ankyrin-G in spines, leading to increased spine size and decreased DAGLα surface diffusion. Ankyrin-G conditional knockout mice showed significantly decreased DAGLα-positive neurons in the forebrain. In mice, ankyrin-G was required for forskolin-dependent reversal of depression-related behavior. CONCLUSIONS Taken together, ANK3 and DAGLA, both neuropsychiatric disorder genes, interact in a complex to regulate spine morphology. These data reveal novel synaptic signaling mechanisms and potential therapeutic avenues.
Collapse
Affiliation(s)
- Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kristoffer Myczek
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Psychiatry and Behavioral Sciences, and Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
45
|
Balezina OP, Tarasova EO, Gaydukov AE. Noncanonical Activity of Endocannabinoids and Their Receptors in Central and Peripheral Synapses. BIOCHEMISTRY (MOSCOW) 2021; 86:818-832. [PMID: 34284706 DOI: 10.1134/s0006297921070038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review focuses on new aspects of endocannabinoid functions and mechanisms of activity in central and peripheral synapses, different from the general viewpoint that endocannabinoids are retrograde signaling molecules, which inhibit neurotransmitter release by activating specific presynaptic endocannabinoid receptors CB1 and CB2. Biased agonism of the endogenous and synthetic cannabinoids as well as ability of the CB-receptors to couple not only with classical Gi-proteins, but also with Gs- and Gq-proteins and, moreover, with β-arrestins (thereby triggering additional signaling pathways in synapses) are described here in detail. Examples of noncanonical tonic activity of endocannabinoids and their receptors and their role in synaptic function are also presented. The role of endocannabinoids in short-term and long-term potentiation of neurotransmitter release in central synapses and their facilitating effect on quantal size and other parameters of acetylcholine release in mammalian neuromuscular junctions are highlighted in this review. In conclusion, it is stated that the endocannabinoid system has a wider range of various multidirectional modulating effects (both potentiating and inhibiting) on neurotransmitter release than initially recognized. Re-evaluation of the functions of endocannabinoid system with consideration of its noncanonical features will lead to better understanding of its role in the normal and pathological functioning of the nervous system and other systems of the body, which has an enormous practical value.
Collapse
Affiliation(s)
- Olga P Balezina
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | | | | |
Collapse
|
46
|
Winters BL, Vaughan CW. Mechanisms of endocannabinoid control of synaptic plasticity. Neuropharmacology 2021; 197:108736. [PMID: 34343612 DOI: 10.1016/j.neuropharm.2021.108736] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/13/2023]
Abstract
The endogenous cannabinoid transmitter system regulates synaptic transmission throughout the nervous system. Unlike conventional transmitters, specific stimuli induce synthesis of endocannabinoids (eCBs) in the postsynaptic neuron, and these travel backwards to modulate presynaptic inputs. In doing so, eCBs can induce short-term changes in synaptic strength and longer-term plasticity. While this eCB regulation is near ubiquitous, it displays major regional and synapse specific variations with different synapse specific forms of short-versus long-term plasticity throughout the brain. These differences are due to the plethora of pre- and postsynaptic mechanisms which have been implicated in eCB signalling, the intricacies of which are only just being realised. In this review, we shall describe the current understanding and highlight new advances in this area, with a focus on the retrograde action of eCBs at CB1 receptors (CB1Rs).
Collapse
Affiliation(s)
- Bryony Laura Winters
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney at Royal North Shore Hospital, NSW, Australia.
| | - Christopher Walter Vaughan
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney at Royal North Shore Hospital, NSW, Australia
| |
Collapse
|
47
|
Heinbockel T, Straiker A. Cannabinoids Regulate Sensory Processing in Early Olfactory and Visual Neural Circuits. Front Neural Circuits 2021; 15:662349. [PMID: 34305536 PMCID: PMC8294086 DOI: 10.3389/fncir.2021.662349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/11/2021] [Indexed: 12/25/2022] Open
Abstract
Our sensory systems such as the olfactory and visual systems are the target of neuromodulatory regulation. This neuromodulation starts at the level of sensory receptors and extends into cortical processing. A relatively new group of neuromodulators includes cannabinoids. These form a group of chemical substances that are found in the cannabis plant. Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) are the main cannabinoids. THC acts in the brain and nervous system like the chemical substances that our body produces, the endogenous cannabinoids or endocannabinoids, also nicknamed the brain's own cannabis. While the function of the endocannabinoid system is understood fairly well in limbic structures such as the hippocampus and the amygdala, this signaling system is less well understood in the olfactory pathway and the visual system. Here, we describe and compare endocannabinoids as signaling molecules in the early processing centers of the olfactory and visual system, the olfactory bulb, and the retina, and the relevance of the endocannabinoid system for synaptic plasticity.
Collapse
Affiliation(s)
- Thomas Heinbockel
- Department of Anatomy, Howard University College of Medicine, Washington, DC, United States
| | - Alex Straiker
- The Gill Center for Biomolecular Science and the Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| |
Collapse
|
48
|
Oubraim S, Wang R, Hausknecht KA, Shen RY, Haj-Dahmane S. Tonic Endocannabinoid Signaling Gates Synaptic Plasticity in Dorsal Raphe Nucleus Serotonin Neurons Through Peroxisome Proliferator-Activated Receptors. Front Pharmacol 2021; 12:691219. [PMID: 34262460 PMCID: PMC8273699 DOI: 10.3389/fphar.2021.691219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/14/2021] [Indexed: 12/27/2022] Open
Abstract
Endocannabinoids (eCBs), which include 2-arachidonoylglycerol (2-AG) and anandamide (AEA) are lipid signaling molecules involved in the regulation of an array of behavioral and physiological functions. Released by postsynaptic neurons, eCBs mediate both phasic and tonic signaling at central synapses. While the roles of phasic eCB signaling in modulating synaptic functions and plasticity are well characterized, very little is known regarding the physiological roles and mechanisms regulating tonic eCB signaling at central synapses. In this study, we show that both 2-AG and AEA are constitutively released in the dorsal raphe nucleus (DRN), where they exert tonic control of glutamatergic synaptic transmission onto serotonin (5-HT) neurons. The magnitude of this tonic eCB signaling is tightly regulated by the overall activity of neuronal network. Thus, short term in vitro neuronal silencing or blockade of excitatory synaptic transmission abolishes tonic eCB signaling in the DRn. Importantly, in addition to controlling basal synaptic transmission, this study reveals that tonic 2-AG, but not AEA signaling, modulates synaptic plasticity. Indeed, short-term increase in tonic 2-AG signaling impairs spike-timing dependent potentiation (tLTP) of glutamate synapses. This tonic 2-AG-mediated homeostatic control of DRN glutamate synapses is not signaled by canonical cannabinoid receptors, but by intracellular peroxisome proliferator-activated receptor gamma (PPARγ). Further examination reveals that 2-AG mediated activation of PPARγ blocks tLTP by inhibiting nitric oxide (NO), soluble guanylate cyclase, and protein kinase G (NO/sGC/PKG) signaling pathway. Collectively, these results unravel novel mechanisms by which tonic 2-AG signaling integrates network activities and controls the synaptic plasticity in the brain.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Ruixiang Wang
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Kathryn A Hausknecht
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States.,Neuroscience Program, University at Buffalo, Buffalo, NY, United States
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States.,Neuroscience Program, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
49
|
O'Brien LD, Smith TL, Donvito G, Cravatt BF, Newton J, Spiegel S, Reeves TM, Phillips LL, Lichtman AH. Diacylglycerol Lipase-β Knockout Mice Display a Sex-Dependent Attenuation of Traumatic Brain Injury-Induced Mortality with No Impact on Memory or Other Functional Consequences. Cannabis Cannabinoid Res 2021; 6:508-521. [PMID: 34142866 DOI: 10.1089/can.2020.0175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Background: The endogenous cannabinoid system modulates inflammatory signaling in a variety of pathological states, including traumatic brain injury (TBI). The selective expression of diacylglycerol lipase-β (DAGL-β), the 2-arachidonylglycerol biosynthetic enzyme, on resident immune cells of the brain (microglia) and the role of this pathway in neuroinflammation, suggest that this enzyme may contribute to TBI-induced neuroinflammation. Accordingly, we tested whether DAGL-β-/- mice would show a protective phenotype from the deleterious consequences of TBI on cognitive and neurological motor functions. Materials and Methods: DAGL-β-/- and -β+/+ mice were subjected to the lateral fluid percussion model of TBI and assessed for learning and memory in the Morris water maze (MWM) Fixed Platform (reference memory) and Reversal (cognitive flexibility) tasks, as well as in a cued MWM task to infer potential sensorimotor/motivational deficits. In addition, subjects were assessed for motor behavior (Rotarod and the Neurological Severity Score assays) and in the light/dark box and the elevated plus maze to infer whether these manipulations affected anxiety-like behavior. Finally, we also examined whether brain injury disrupts the ceramide/sphingolipid lipid signaling system and if DAGL-β deletion offers protection. Results: TBI disrupted all measures of neurological motor function and reduced body weight, but did not affect body temperature or performance in common assays used to infer anxiety. TBI also impaired performance in MWM Fixed Platform and Reversal tasks, but did not affect cued MWM performance. Although no differences were found between DAGL-β-/- and -β+/+ mice in any of these measures, male DAGL-β-/- mice displayed an unexpected survival-protective phenotype, which persisted at increased injury severities. In contrast, TBI did not elicit mortality in female mice regardless of genotype. TBI also produced significant changes in sphingolipid profiles (a family of lipids, members of which have been linked to both apoptotic and antiapoptotic pathways), in which DAGL-β deletion modestly altered levels of select species. Conclusions: These findings indicate that although DAGL-β does not play a necessary role in TBI-induced cognitive and neurological function, it appears to contribute to the increased vulnerability of male mice to TBI-induced mortality, whereas female mice show high survival rates irrespective of DAGL-β expression.
Collapse
Affiliation(s)
- Lesley D O'Brien
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terry L Smith
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Giulia Donvito
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Benjamin F Cravatt
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Jason Newton
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Thomas M Reeves
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
50
|
Potential and Limits of Cannabinoids in Alzheimer's Disease Therapy. BIOLOGY 2021; 10:biology10060542. [PMID: 34204237 PMCID: PMC8234911 DOI: 10.3390/biology10060542] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary This review was aimed at exploring the potentiality of drugging the endocannabinoid system as a therapeutic option for Alzheimer’s disease (AD). Recent discoveries have demonstrated how the modulation of cannabinoid receptor 1 (CB1) and receptor 2 (CB2) can exert neuroprotective effects without the recreational and pharmacological properties of Cannabis sativa. Thus, this review explores the potential of cannabinoids in AD, also highlighting their limitations in perspective to point out the need for further research on cannabinoids in AD therapy. Abstract Alzheimer’s disease (AD) is a detrimental brain disorder characterized by a gradual cognitive decline and neuronal deterioration. To date, the treatments available are effective only in the early stage of the disease. The AD etiology has not been completely revealed, and investigating new pathological mechanisms is essential for developing effective and safe drugs. The recreational and pharmacological properties of marijuana are known for centuries, but only recently the scientific community started to investigate the potential use of cannabinoids in AD therapy—sometimes with contradictory outcomes. Since the endocannabinoid system (ECS) is highly expressed in the hippocampus and cortex, cannabis use/abuse has often been associated with memory and learning dysfunction in vulnerable individuals. However, the latest findings in AD rodent models have shown promising effects of cannabinoids in reducing amyloid plaque deposition and stimulating hippocampal neurogenesis. Beneficial effects on several dementia-related symptoms have also been reported in clinical trials after cannabinoid treatments. Accordingly, future studies should address identifying the correct therapeutic dosage and timing of treatment from the perspective of using cannabinoids in AD therapy. The present paper aims to summarize the potential and limitations of cannabinoids as therapeutics for AD, focusing on recent pre-clinical and clinical evidence.
Collapse
|