1
|
Andersen HK, Vardakas DG, Lamothe JA, Perault TEA, Walsh KB, Laprairie RB. Comparing CB1 receptor GIRK channel responses to receptor internalization using a kinetic imaging assay. Sci Rep 2024; 14:18314. [PMID: 39112591 PMCID: PMC11306342 DOI: 10.1038/s41598-024-68451-2] [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: 01/29/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
The type 1 cannabinoid receptor (CB1R) mediates neurotransmitter release and synaptic plasticity in the central nervous system. Endogenous, plant-derived, synthetic cannabinoids bind to CB1R, initiating the inhibitory G-protein (Gi) and the β-arrestin signaling pathways. Within the Gi signaling pathway, CB1R activates G protein-gated, inwardly-rectifying potassium (GIRK) channels. The β-arrestin pathway reduces CB1R expression on the cell surface through receptor internalization. Because of their association with analgesia and drug tolerance, GIRK channels and receptor internalization are of interest to the development of pharmaceuticals. This research used immortalized mouse pituitary gland cells transduced with a pH-sensitive, fluorescently-tagged human CB1R (AtT20-SEPCB1) to measure GIRK channel activity and CB1R internalization. Cannabinoid-induced GIRK channel activity is measured by using a fluorescent membrane-potential sensitive dye. We developed a kinetic imaging assay that visualizes and measures CB1R internalization. All cannabinoids stimulated a GIRK channel response with a rank order potency of WIN55,212-2 > (±)CP55,940 > Δ9-THC > AEA. Efficacy was expressed relative to (±)CP55,940 with a rank order efficacy of (±)CP55,940 > WIN55, 212-2 > AEA > Δ9-THC. All cannabinoids stimulated CB1R internalization with a rank order potency of (±)CP55,940 > WIN55, 212-2 > AEA > Δ9-THC. Internalization efficacy was normalized to (±)CP55,940 with a rank order efficacy of WIN55,212-2 > AEA > (±)CP55,940 > Δ9-THC. (±)CP55,940 was significantly more potent and efficacious than AEA and Δ9-THC at stimulating a GIRK channel response; no significant differences between potency and efficacy were observed with CB1R internalization. No significant differences were found when comparing a cannabinoid's GIRK channel and CB1R internalization response. In conclusion, AtT20-SEPCB1 cells can be used to assess cannabinoid-induced CB1R internalization. While cannabinoids display differential Gi signaling when compared to each other, this did not extend to CB1R internalization.
Collapse
Affiliation(s)
- Haley K Andersen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Duncan G Vardakas
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Julie A Lamothe
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Tannis E A Perault
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kenneth B Walsh
- Pharmacology, Physiology, and Neuroscience, School of Medicine Columbia, University of South Carolina, Columbia, SC, USA
| | - Robert B Laprairie
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Pharmacology, College of Medicine, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
2
|
Fletcher-Jones A, Spackman E, Craig TJ, Nakamura Y, Wilkinson KA, Henley JM. SGIP1 binding to the α-helical H9 domain of cannabinoid receptor 1 promotes axonal surface expression. J Cell Sci 2024; 137:jcs261551. [PMID: 38864427 PMCID: PMC11213518 DOI: 10.1242/jcs.261551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 04/30/2024] [Indexed: 06/13/2024] Open
Abstract
Endocannabinoid signalling mediated by cannabinoid receptor 1 (CB1R, also known as CNR1) is critical for homeostatic neuromodulation of both excitatory and inhibitory synapses. This requires highly polarised axonal surface expression of CB1R, but how this is achieved remains unclear. We previously reported that the α-helical H9 domain in the intracellular C terminus of CB1R contributes to axonal surface expression by an unknown mechanism. Here, we show in rat primary neuronal cultures that the H9 domain binds to the endocytic adaptor protein SGIP1 to promote CB1R expression in the axonal membrane. Overexpression of SGIP1 increases CB1R axonal surface localisation but has no effect on CB1R lacking the H9 domain (CB1RΔH9). Conversely, SGIP1 knockdown reduces axonal surface expression of CB1R but does not affect CB1RΔH9. Furthermore, SGIP1 knockdown diminishes CB1R-mediated inhibition of presynaptic Ca2+ influx in response to neuronal activity. Taken together, these data advance mechanistic understanding of endocannabinoid signalling by demonstrating that SGIP1 interaction with the H9 domain underpins axonal CB1R surface expression to regulate presynaptic responsiveness.
Collapse
Affiliation(s)
- Alexandra Fletcher-Jones
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| | - Ellen Spackman
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| | - Tim J. Craig
- School of Applied Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Yasuko Nakamura
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| | - Kevin A. Wilkinson
- School of Physiology, Pharmacology and Neuroscience, Centre for Synaptic Plasticity, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| | - Jeremy M. Henley
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| |
Collapse
|
3
|
Rodríguez-Soacha DA, Steinmüller SAM, Işbilir A, Fender J, Deventer MH, Ramírez YA, Tutov A, Sotriffer C, Stove CP, Lorenz K, Lohse MJ, Hislop JN, Decker M. Development of an Indole-Amide-Based Photoswitchable Cannabinoid Receptor Subtype 1 (CB 1R) "Cis-On" Agonist. ACS Chem Neurosci 2022; 13:2410-2435. [PMID: 35881914 DOI: 10.1021/acschemneuro.2c00160] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Activation of the human cannabinoid receptor type 1 (hCB1R) with high spatiotemporal control is useful to study processes involved in different pathologies related to nociception, metabolic alterations, and neurological disorders. To synthesize new agonist ligands for hCB1R, we have designed different classes of photoswitchable molecules based on an indole core. The modifications made to the central core have allowed us to understand the molecular characteristics necessary to design an agonist with optimal pharmacological properties. Compound 27a shows high affinity for CB1R (Ki (cis-form) = 0.18 μM), with a marked difference in affinity with respect to its inactive "trans-off" form (CB1R Ki trans/cis ratio = 5.4). The novel compounds were evaluated by radioligand binding studies, receptor internalization, sensor receptor activation (GRABeCB2.0), Western blots for analysis of ERK1/2 activation, NanoBiT βarr2 recruitment, and calcium mobilization assays, respectively. The data show that the novel agonist 27a is a candidate for studying the optical modulation of cannabinoid receptors (CBRs), serving as a new molecular tool for investigating the involvement of hCB1R in disorders associated with the endocannabinoid system.
Collapse
Affiliation(s)
- Diego A Rodríguez-Soacha
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Sophie A M Steinmüller
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Ali Işbilir
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Versbacher Str. 9, D-97078 Würzburg, Germany.,Receptor Signaling Group, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Julia Fender
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Versbacher Str. 9, D-97078 Würzburg, Germany
| | - Marie H Deventer
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Yesid A Ramírez
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany.,Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Naturales, Universidad Icesi, Valle del Cauca, 760031 Cali, Colombia
| | - Anna Tutov
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Christoph Sotriffer
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Christophe P Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Kristina Lorenz
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Versbacher Str. 9, D-97078 Würzburg, Germany.,Leibniz-Institut für Analytische Wissenschaften─ISAS e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| | - Martin J Lohse
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Versbacher Str. 9, D-97078 Würzburg, Germany.,Receptor Signaling Group, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany.,ISAR Bioscience Institut, 82152 Planegg/Munich, Germany
| | - James N Hislop
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, United Kingdom
| | - Michael Decker
- Pharmazeutische und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
4
|
Degrandmaison J, Rochon-Haché S, Parent JL, Gendron L. Knock-In Mouse Models to Investigate the Functions of Opioid Receptors in vivo. Front Cell Neurosci 2022; 16:807549. [PMID: 35173584 PMCID: PMC8841419 DOI: 10.3389/fncel.2022.807549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/28/2022] Open
Abstract
Due to their low expression levels, complex multi-pass transmembrane structure, and the current lack of highly specific antibodies, the assessment of endogenous G protein-coupled receptors (GPCRs) remains challenging. While most of the research regarding their functions was performed in heterologous systems overexpressing the receptor, recent advances in genetic engineering methods have allowed the generation of several unique mouse models. These animals proved to be useful to investigate numerous aspects underlying the physiological functions of GPCRs, including their endogenous expression, distribution, interactome, and trafficking processes. Given their significant pharmacological importance and central roles in the nervous system, opioid peptide receptors (OPr) are often referred to as prototypical receptors for the study of GPCR regulatory mechanisms. Although only a few GPCR knock-in mouse lines have thus far been generated, OPr are strikingly well represented with over 20 different knock-in models, more than half of which were developed within the last 5 years. In this review, we describe the arsenal of OPr (mu-, delta-, and kappa-opioid), as well as the opioid-related nociceptin/orphanin FQ (NOP) receptor knock-in mouse models that have been generated over the past years. We further highlight the invaluable contribution of such models to our understanding of the in vivo mechanisms underlying the regulation of OPr, which could be conceivably transposed to any other GPCR, as well as the limitations, future perspectives, and possibilities enabled by such tools.
Collapse
Affiliation(s)
- Jade Degrandmaison
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Network of Junior Pain Investigators, Sherbrooke, QC, Canada
| | - Samuel Rochon-Haché
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Network of Junior Pain Investigators, Sherbrooke, QC, Canada
| | - Jean-Luc Parent
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Médecine, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Jean-Luc Parent,
| | - Louis Gendron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Quebec Pain Research Network, Sherbrooke, QC, Canada
- *Correspondence: Louis Gendron,
| |
Collapse
|
5
|
Andersen HK, Walsh KB. Molecular signaling of synthetic cannabinoids: Comparison of CB1 receptor and TRPV1 channel activation. Eur J Pharmacol 2021; 907:174301. [PMID: 34224700 PMCID: PMC8374946 DOI: 10.1016/j.ejphar.2021.174301] [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: 03/08/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 10/21/2022]
Abstract
Recreational use of synthetic cannabinoids (SCs) is associated with desirable euphoric and relaxation effects as well as adverse effects including anxiety, agitation and psychosis. These SC-mediated actions represent a combination of potentiated cannabinoid receptor signaling and "off-target" receptor activity. The goal of this study was to compare the efficacy of various classes of SCs in stimulating CB1 receptors and activating "off-target" transient receptor potential (TRP) channels. Cannabinoid-type 1 (CB1) receptor activity was determined by measuring SC activation of G protein-gated inward rectifier K+ (GIRK) channels using a membrane potential-sensitive fluorescent dye assay. SC opening of vanilloid type-1 (TRPV1) channels was measured by recording intracellular Ca2+ transients. All of the SCs tested activated the GIRK channel with an efficacy of 4-fluoro MDMB-BUTINACA > 5-fluoro MDMB-PICA > MDMB-4en-PINACA ≈ WIN 55,212-2 > AB-FUBINACA > AM1220 ≈ JWH-122 N-(5-chloropentyl) > AM1248 > JWH-018 ≈ XLR-11 ≈ UR-144. The potency of the SCs at the CB1 receptor was 5-fluoro MDMB-PICA ≈ 4-fluoro MDMB-BUTINACA > AB-FUBINACA ≈ MDMB-4en-PINACA > JWH-018 > AM1220 > XLR-11 > JWH-122 N-(5-chloropentyl) > WIN 55,212-2 ≈ UR-144 > AM1248. In contrast, when tested at a SC concentration that produced a maximal effect on the Gi/GIRK channel, only XLR-11, UR-144 and AM1220 caused a significant activation of the TRPV1 channels. The TRPV1 channel/Ca2+ signal measured during application of 10 μM XLR-11 was similar to the signal induced by the endocannabinoid N-arachidonoylethanolamine (AEA). Thus, while various SCs share the ability to stimulate CB1 receptor/Gi signaling, they display limited efficacy in opening TRPV1 channels.
Collapse
Affiliation(s)
- Haley K Andersen
- Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA
| | - Kenneth B Walsh
- Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA.
| |
Collapse
|
6
|
Rodríguez-Rodríguez I, Kalafut J, Czerwonka A, Rivero-Müller A. A novel bioassay for quantification of surface Cannabinoid receptor 1 expression. Sci Rep 2020; 10:18191. [PMID: 33097803 PMCID: PMC7584592 DOI: 10.1038/s41598-020-75331-y] [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: 05/02/2020] [Accepted: 10/14/2020] [Indexed: 12/04/2022] Open
Abstract
The cannabinoid receptor type 1 (CB1) plays critical roles in multiple physiological processes such as pain perception, brain development and body temperature regulation. Mutations on this gene (CNR1), results in altered functionality and/or biosynthesis such as reduced membrane expression, changes in mRNA stability or changes in downstream signaling that act as triggers for diseases such as obesity, Parkinson’s, Huntington’s, among others; thus, it is considered as a potential pharmacological target. To date, multiple quantification methods have been employed to determine how these mutations affect receptor expression and localization; however, they present serious disadvantages that may arise quantifying errors. Here, we describe a sensitive bioassay to quantify receptor surface expression; in this bioassay the Gaussia Luciferase (GLuc) was fused to the extracellular portion of the CB1. The GLuc activity was assessed by coelenterazine addition to the medium followed by immediate readout. Based on GLuc activity assay, we show that the GLuc signals corelate with CB1 localization, besides, we showed the assay’s functionality and reliability by comparing its results with those generated by previously reported mutations on the CNR1 gene and by using flow cytometry to determine the cell surface receptor expression. Detection of membrane-bound CB1, and potentially other GPCRs, is able to quickly screen for receptor levels and help to understand the effect of clinically relevant mutations or polymorphisms.
Collapse
Affiliation(s)
| | - Joanna Kalafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Arkadiusz Czerwonka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland.,Department of Virology and Immunology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
7
|
Fletcher-Jones A, Hildick KL, Evans AJ, Nakamura Y, Henley JM, Wilkinson KA. Protein Interactors and Trafficking Pathways That Regulate the Cannabinoid Type 1 Receptor (CB1R). Front Mol Neurosci 2020; 13:108. [PMID: 32595453 PMCID: PMC7304349 DOI: 10.3389/fnmol.2020.00108] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/20/2020] [Indexed: 12/29/2022] Open
Abstract
The endocannabinoid system (ECS) acts as a negative feedback mechanism to suppress synaptic transmission and plays a major role in a diverse range of brain functions including, for example, the regulation of mood, energy balance, and learning and memory. The function and dysfunction of the ECS are strongly implicated in multiple psychiatric, neurological, and neurodegenerative diseases. Cannabinoid type 1 receptor (CB1R) is the most abundant G protein-coupled receptor (GPCR) expressed in the brain and, as for any synaptic receptor, CB1R needs to be in the right place at the right time to respond appropriately to changing synaptic circumstances. While CB1R is found intracellularly throughout neurons, its surface expression is highly polarized to the axonal membrane, consistent with its functional expression at presynaptic sites. Surprisingly, despite the importance of CB1R, the interacting proteins and molecular mechanisms that regulate the highly polarized distribution and function of CB1R remain relatively poorly understood. Here we set out what is currently known about the trafficking pathways and protein interactions that underpin the surface expression and axonal polarity of CB1R, and highlight key questions that still need to be addressed.
Collapse
Affiliation(s)
- Alexandra Fletcher-Jones
- Centre for Synaptic Plasticity, School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Keri L Hildick
- Centre for Synaptic Plasticity, School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Ashley J Evans
- Centre for Synaptic Plasticity, School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Yasuko Nakamura
- Centre for Synaptic Plasticity, School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Jeremy M Henley
- Centre for Synaptic Plasticity, School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Kevin A Wilkinson
- Centre for Synaptic Plasticity, School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
8
|
Fletcher-Jones A, Hildick KL, Evans AJ, Nakamura Y, Wilkinson KA, Henley JM. The C-terminal helix 9 motif in rat cannabinoid receptor type 1 regulates axonal trafficking and surface expression. eLife 2019; 8:44252. [PMID: 31036155 PMCID: PMC6491034 DOI: 10.7554/elife.44252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
Cannabinoid type one receptor (CB1R) is only stably surface expressed in axons, where it downregulates neurotransmitter release. How this tightly regulated axonal surface polarity is established and maintained is unclear. To address this question, we used time-resolved imaging to determine the trafficking of CB1R from biosynthesis to mature polarised localisation in cultured rat hippocampal neurons. We show that the secretory pathway delivery of CB1R is axonally biased and that surface expressed CB1R is more stable in axons than in dendrites. This dual mechanism is mediated by the CB1R C-terminus and involves the Helix 9 (H9) domain. Removal of the H9 domain increases secretory pathway delivery to dendrites and decreases surface stability. Furthermore, CB1RΔH9 is more sensitive to agonist-induced internalisation and less efficient at downstream signalling than CB1RWT. Together, these results shed new light on how polarity of CB1R is mediated and indicate that the C-terminal H9 domain plays key roles in this process. The brain contains around 100 billion neurons that are in constant communication with one another. Each consists of a cell body, plus two components specialized for exchanging information. These are the axon, which delivers information, and the dendrites, which receive it. This exchange takes place at contact points between neurons called synapses. To send a message, a neuron releases chemicals called neurotransmitters from its axon terminals into the synapse. The neurotransmitters cross the synapse and bind to receptor proteins on the dendrites of another neuron. In doing so, they pass on the message. Cannabinoid type 1 receptors (CB1Rs) help control the flow of information at synapses. They do this by binding neurotransmitters called endocannabinoids, which are unusual among neurotransmitters. Rather than sending messages from axons to dendrites, endocannabinoids send them in the opposite direction. Thus, it is dendrites that release endocannabinoids, which then bind to CB1Rs in axon terminals. This backwards, or 'retrograde', signalling dampens the release of other neurotransmitters. This slows down brain activity, and gives rise to the 'mellow' sensation that recreational cannabis users often describe. Like most other proteins, CB1Rs are built inside the cell body. So, how do these receptors end up in the axon terminals where they are needed? Are they initially sent to both axons and dendrites, with the CB1Rs that travel to dendrites being rerouted back to axons? Or do the receptors travel directly to the axon itself? Fletcher-Jones et al. tracked newly made CB1Rs in rat neurons growing in a dish. The results revealed that the receptors go directly to the axon, before moving on to the axon terminals. A specific region of the CB1R protein is crucial for sending the receptors to the axon, and for ensuring that they do not get diverted to the dendrite surface. This region stabilizes CB1Rs at the axon surface, and helps to make the receptors available to bind endocannabinoids. CB1Rs also respond to medical marijuana, a topic that continues to generate interest as well as controversy. Activating CB1Rs could help treat a wide range of diseases, such as chronic pain, epilepsy and multiple sclerosis. Future studies should build on our understanding of CB1Rs to explore and optimize new therapeutic approaches.
Collapse
Affiliation(s)
- Alexandra Fletcher-Jones
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Keri L Hildick
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Ashley J Evans
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Yasuko Nakamura
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Kevin A Wilkinson
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| | - Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
9
|
Lambert L, Dubayle D, Fafouri A, Herzog E, Csaba Z, Dournaud P, El Mestikawy S, Bernard V. Endocytosis of Activated Muscarinic m2 Receptor (m2R) in Live Mouse Hippocampal Neurons Occurs via a Clathrin-Dependent Pathway. Front Cell Neurosci 2018; 12:450. [PMID: 30555302 PMCID: PMC6283979 DOI: 10.3389/fncel.2018.00450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/07/2018] [Indexed: 02/02/2023] Open
Abstract
Our aim was to examine the dynamics of the muscarinic m2 receptor (m2R), a G-protein coupled receptor (GPCR), after agonist activation in living hippocampal neurons, and especially clathrin dependency endocytosis. We have previously shown that the m2R undergoes agonist-induced internalization in vivo. However, the nature of the endocytotic pathway used by m2R after activation is still unknown in living neurons. Using live cell imaging and quantitative analyses, we have monitored the effect of stimulation on the fate of the membrane-bound m2R and on its redistribution in intraneuronal compartments. Shortly (6 min) after activation, m2R is internalized into clathrin immunopositive structures. Furthermore, after clathrin-dependent endocytosis, m2R associates with early and late endosomes and with subcellular organelles involved in degradation. Together, these results provide, for the first time, a description of m2R trafficking in living neurons and prove that m2R undergoes clathrin-dependent endocytosis before being degraded.
Collapse
Affiliation(s)
- Lisa Lambert
- Sorbonne Université, Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France
| | - David Dubayle
- Sorbonne Université, Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France.,Université Paris Descartes - CNRS UMR 8119, Centre de Neurophysique, Physiologie et Pathologie, Paris, France
| | - Assia Fafouri
- PROTECT, INSERM U1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Etienne Herzog
- Sorbonne Université, Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France.,Interdisciplinary Institute for Neuroscience, University Bordeaux, UMR 5297, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, Bordeaux, France
| | - Zsolt Csaba
- PROTECT, INSERM U1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Pascal Dournaud
- PROTECT, INSERM U1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Salah El Mestikawy
- Sorbonne Université, Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France.,Department of Psychiatry, Douglas Hospital Research Center, McGill University, Montréal, QC, Canada
| | - Véronique Bernard
- Sorbonne Université, Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France
| |
Collapse
|
10
|
Gendron L, Cahill CM, von Zastrow M, Schiller PW, Pineyro G. Molecular Pharmacology of δ-Opioid Receptors. Pharmacol Rev 2017; 68:631-700. [PMID: 27343248 DOI: 10.1124/pr.114.008979] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Opioids are among the most effective analgesics available and are the first choice in the treatment of acute severe pain. However, partial efficacy, a tendency to produce tolerance, and a host of ill-tolerated side effects make clinically available opioids less effective in the management of chronic pain syndromes. Given that most therapeutic opioids produce their actions via µ-opioid receptors (MOPrs), other targets are constantly being explored, among which δ-opioid receptors (DOPrs) are being increasingly considered as promising alternatives. This review addresses DOPrs from the perspective of cellular and molecular determinants of their pharmacological diversity. Thus, DOPr ligands are examined in terms of structural and functional variety, DOPrs' capacity to engage a multiplicity of canonical and noncanonical G protein-dependent responses is surveyed, and evidence supporting ligand-specific signaling and regulation is analyzed. Pharmacological DOPr subtypes are examined in light of the ability of DOPr to organize into multimeric arrays and to adopt multiple active conformations as well as differences in ligand kinetics. Current knowledge on DOPr targeting to the membrane is examined as a means of understanding how these receptors are especially active in chronic pain management. Insight into cellular and molecular mechanisms of pharmacological diversity should guide the rational design of more effective, longer-lasting, and better-tolerated opioid analgesics for chronic pain management.
Collapse
Affiliation(s)
- Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Catherine M Cahill
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Mark von Zastrow
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Peter W Schiller
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| | - Graciela Pineyro
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Centre de Recherche du CHU de Sherbrooke, Centre d'excellence en neurosciences de l'Univeristé de Sherbrooke, and Institut de Pharmacologie de Sherbrooke, Sherbrooke, Quebec, Canada (L.G.); Québec Pain Research Network, Sherbrooke, Quebec, Canada (L.G.); Departments of Anesthesiology and Perioperative Care and Pharmacology, University of California, Irvine, California (C.M.C.); Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.M.C.); Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco, California (M.v.Z.); Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montréal, Montreal, Quebec, Canada (P.W.S.); and Departments of Psychiatry, Pharmacology, and Neurosciences, Faculty of Medicine, University of Montréal and Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada (G.P.)
| |
Collapse
|
11
|
Blume LC, Patten T, Eldeeb K, Leone-Kabler S, Ilyasov AA, Keegan BM, O'Neal JE, Bass CE, Hantgan RR, Lowther WT, Selley DE, Howlett ALC. Cannabinoid Receptor Interacting Protein 1a Competition with β-Arrestin for CB1 Receptor Binding Sites. Mol Pharmacol 2016; 91:75-86. [PMID: 27895162 DOI: 10.1124/mol.116.104638] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 11/23/2016] [Indexed: 01/11/2023] Open
Abstract
Cannabinoid receptor interacting protein 1a (CRIP1a) is a CB1 receptor (CB1R) distal C-terminal-associated protein that alters CB1R interactions with G-proteins. We tested the hypothesis that CRIP1a is capable of also altering CB1R interactions with β-arrestin proteins that interact with the CB1R at the C-terminus. Coimmunoprecipitation studies indicated that CB1R associates in complexes with either CRIP1a or β-arrestin, but CRIP1a and β-arrestin fail to coimmunoprecipitate with each other. This suggests a competition for CRIP1a and β-arrestin binding to the CB1R, which we hypothesized could attenuate the action of β-arrestin to mediate CB1R internalization. We determined that agonist-mediated reduction of the density of cell surface endogenously expressed CB1Rs was clathrin and dynamin dependent and could be modeled as agonist-induced aggregation of transiently expressed GFP-CB1R. CRIP1a overexpression attenuated CP55940-mediated GFP-CB1R as well as endogenous β-arrestin redistribution to punctae, and conversely, CRIP1a knockdown augmented β-arrestin redistribution to punctae. Peptides mimicking the CB1R C-terminus could bind to both CRIP1a in cell extracts as well as purified recombinant CRIP1a. Affinity pull-down studies revealed that phosphorylation at threonine-468 of a CB1R distal C-terminus 14-mer peptide reduced CB1R-CRIP1a association. Coimmunoprecipitation of CB1R protein complexes demonstrated that central or distal C-terminal peptides competed for the CB1R association with CRIP1a, but that a phosphorylated central C-terminal peptide competed for association with β-arrestin 1, and phosphorylated central or distal C-terminal peptides competed for association with β-arrestin 2. Thus, CRIP1a can compete with β-arrestins for interaction with C-terminal CB1R domains that could affect agonist-driven, β-arrestin-mediated internalization of the CB1R.
Collapse
Affiliation(s)
- Lawrence C Blume
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Theresa Patten
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Khalil Eldeeb
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Sandra Leone-Kabler
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Alexander A Ilyasov
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Bradley M Keegan
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Jeremy E O'Neal
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Caroline E Bass
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Roy R Hantgan
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - W Todd Lowther
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - Dana E Selley
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| | - A Llyn C Howlett
- Department of Physiology and Pharmacology (L.C.B., T.P, K.E., S.L.-K., A.A.I., B.M.K., J.E.O., C.E.B., A.C.H.) and Department of Biochemistry and Center for Structural Biology (R.R.H., W.T.L.), Wake Forest University Health Sciences, Winston-Salem, North Carolina; Department of Chemistry (T.P.) and Center for Molecular Signaling (W.T.L., A.C.H.), Wake Forest University, Winston-Salem, North Carolina; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia (D.E.S.); and AL Azhar Faculty of Medicine, New Damietta, Egypt (K.E.)
| |
Collapse
|
12
|
Ceredig RA, Massotte D. Fluorescent knock-in mice to decipher the physiopathological role of G protein-coupled receptors. Front Pharmacol 2015; 5:289. [PMID: 25610398 PMCID: PMC4284998 DOI: 10.3389/fphar.2014.00289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/12/2014] [Indexed: 12/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) modulate most physiological functions but are also critically involved in numerous pathological states. Approximately a third of marketed drugs target GPCRs, which places this family of receptors in the main arena of pharmacological pre-clinical and clinical research. The complexity of GPCR function demands comprehensive appraisal in native environment to collect in-depth knowledge of receptor physiopathological roles and assess the potential of therapeutic molecules. Identifying neurons expressing endogenous GPCRs is therefore essential to locate them within functional circuits whereas GPCR visualization with subcellular resolution is required to get insight into agonist-induced trafficking. Both remain frequently poorly investigated because direct visualization of endogenous receptors is often hampered by the lack of appropriate tools. Also, monitoring intracellular trafficking requires real-time visualization to gather in-depth knowledge. In this context, knock-in mice expressing a fluorescent protein or a fluorescent version of a GPCR under the control of the endogenous promoter not only help to decipher neuroanatomical circuits but also enable real-time monitoring with subcellular resolution thus providing invaluable information on their trafficking in response to a physiological or a pharmacological challenge. This review will present the animal models and discuss their contribution to the understanding of the physiopathological role of GPCRs. We will also address the drawbacks associated with this methodological approach and browse future directions.
Collapse
Affiliation(s)
- Rhian A Ceredig
- CNRS, Institut des Neurosciences Cellulaires et Intégratives, UPR 3212 Strasbourg, France
| | - Dominique Massotte
- CNRS, Institut des Neurosciences Cellulaires et Intégratives, UPR 3212 Strasbourg, France
| |
Collapse
|
13
|
Ligand-specific endocytic dwell times control functional selectivity of the cannabinoid receptor 1. Nat Commun 2014; 5:4589. [PMID: 25081814 PMCID: PMC4227836 DOI: 10.1038/ncomms5589] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 07/03/2014] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the major transducers of external stimuli and
key therapeutic targets in many pathological conditions. When activated by different
ligands, one receptor can elicit multiple signalling cascades that are mediated by G
proteins or β-arrestin, a process defined as functional selectivity or
ligand bias. However, the dynamic mechanisms underlying β-arrestin
signalling remain unknown. Here by studying the cannabinoid receptor 1 (CB1R), we identify ligand-specific endocytic dwell times, that
is, the time during which receptors are clustered into clathrin pits together with
β-arrestins before endocytosis, as the mechanism controlling
β-arrestin signalling. Agonists inducing short endocytic dwell times
produce little or no β-arrestin signalling, whereas those eliciting
prolonged dwell times induce robust signalling. Remarkably, extending CB1R dwell times by preventing endocytosis
substantially increased β-arrestin signalling. These studies reveal how
receptor activation translates into β-arrestin signalling and identify a
mechanism to control this pathway. G-protein coupled receptors can signal through G-proteins or through
β-arrestin, however mechanisms determining pathway selection remain unclear.
Here the authors show that the duration of cannabinoid receptor clustering in clathrin
coated pits prior to endocytosis determines the strength of β-arrestin
signalling.
Collapse
|
14
|
Gendron L, Mittal N, Beaudry H, Walwyn W. Recent advances on the δ opioid receptor: from trafficking to function. Br J Pharmacol 2014; 172:403-19. [PMID: 24665909 DOI: 10.1111/bph.12706] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Within the opioid family of receptors, δ (DOPrs) and μ opioid receptors (MOPrs) are typical GPCRs that activate canonical second-messenger signalling cascades to influence diverse cellular functions in neuronal and non-neuronal cell types. These receptors activate well-known pathways to influence ion channel function and pathways such as the map kinase cascade, AC and PI3K. In addition new information regarding opioid receptor-interacting proteins, downstream signalling pathways and resultant functional effects has recently come to light. In this review, we will examine these novel findings focusing on the DOPr and, in doing so, will contrast and compare DOPrs with MOPrs in terms of differences and similarities in function, signalling pathways, distribution and interactions. We will also discuss and clarify issues that have recently surfaced regarding the expression and function of DOPrs in different cell types and analgesia. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- Louis Gendron
- Département de physiologie et biophysique, Institut de pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
15
|
Bagher AM, Laprairie RB, Kelly MEM, Denovan-Wright EM. Co-expression of the human cannabinoid receptor coding region splice variants (hCB₁) affects the function of hCB₁ receptor complexes. Eur J Pharmacol 2013; 721:341-54. [PMID: 24091169 DOI: 10.1016/j.ejphar.2013.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 08/26/2013] [Accepted: 09/04/2013] [Indexed: 10/26/2022]
Abstract
The human type 1 cannabinoid (hCB1) receptor is expressed at high levels in the central nervous system. mRNA variants of the coding region of this receptor, human cannabinoid hCB1a and hCB1b receptors, have been identified, their biological function remains unclear. The present study demonstrated that the three human cannabinoid hCB1 coding region variants are expressed in the human and monkey (Macaca fascicularis) brain. Western blot analyses of homogenates from different regions of the monkey brain demonstrated that proteins with the expected molecular weights of the cannabinoid CB1, CB1a and CB1b receptors were co-expressed throughout the brain. Given the co-localization of these receptors, we hypothesized that physical interactions between the three splice variants may affect cannabinoid pharmacology. The human cannabinoid hCB1, hCB1a, and hCB1b receptors formed homodimers and heterodimers, as determined by BRET in transiently transfected HEK 293A cells. We found that the co-expression of the human cannabinoid hCB1 and each of the splice variants increased cell surface expression of the human cannabinoid hCB1 receptor and increased Gi/o-dependent ERK phosphorylation in response to cannabinoid agonists. Therefore, the human cannabinoid hCB1 coding region splice variants play an important physiological role in the activity of the endocannabinoid system.
Collapse
Affiliation(s)
- Amina M Bagher
- Department of Pharmacology, Dalhousie University, 6E Sir Charles Tupper Medical Bldg., 5850 College St., Halifax, NS, Canada B3H 4R2
| | | | | | | |
Collapse
|
16
|
Gyombolai P, Boros E, Hunyady L, Turu G. Differential β-arrestin2 requirements for constitutive and agonist-induced internalization of the CB1 cannabinoid receptor. Mol Cell Endocrinol 2013; 372:116-27. [PMID: 23541635 DOI: 10.1016/j.mce.2013.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/06/2013] [Accepted: 03/15/2013] [Indexed: 01/13/2023]
Abstract
CB1 cannabinoid receptor (CB1R) undergoes both constitutive and agonist-induced internalization, but the underlying mechanisms of these processes and the role of β-arrestins in the regulation of CB1R function are not completely understood. In this study, we followed CB1R internalization using confocal microscopy and bioluminescence resonance energy transfer measurements in HeLa and Neuro-2a cells. We found that upon activation CB1R binds β-arrestin2 (β-arr2), but not β-arrestin1. Furthermore, both the expression of dominant-negative β-arr2 (β-arr2-V54D) and siRNA-mediated knock-down of β-arr2 impaired the agonist-induced internalization of CB1R. In contrast, neither β-arr2-V54D nor β-arr2-specific siRNA had a significant effect on the constitutive internalization of CB1R. However, both constitutive and agonist-induced internalization of CB1R were impaired by siRNA-mediated depletion of clathrin heavy chain. We conclude that although clathrin is required for both constitutive and agonist-stimulated internalization of CB1R, β-arr2 binding is only required for agonist-induced internalization of the receptor suggesting that the molecular mechanisms underlying constitutive and agonist-induced internalization of CB1R are different.
Collapse
Affiliation(s)
- Pál Gyombolai
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| | | | | | | |
Collapse
|
17
|
Sturt BL, Bamber BA. Automated quantification of synaptic fluorescence in C. elegans. J Vis Exp 2012:4090. [PMID: 22907390 DOI: 10.3791/4090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Synapse strength refers to the amplitude of postsynaptic responses to presynaptic neurotransmitter release events, and has a major impact on overall neural circuit function. Synapse strength critically depends on the abundance of neurotransmitter receptors clustered at synaptic sites on the postsynaptic membrane. Receptor levels are established developmentally, and can be altered by receptor trafficking between surface-localized, subsynaptic, and intracellular pools, representing important mechanisms of synaptic plasticity and neuromodulation. Rigorous methods to quantify synaptically-localized neurotransmitter receptor abundance are essential to study synaptic development and plasticity. Fluorescence microscopy is an optimal approach because it preserves spatial information, distinguishing synaptic from non-synaptic pools, and discriminating among receptor populations localized to different types of synapses. The genetic model organism Caenorhabditis elegans is particularly well suited for these studies due to the small size and relative simplicity of its nervous system, its transparency, and the availability of powerful genetic techniques, allowing examination of native synapses in intact animals. Here we present a method for quantifying fluorescently-labeled synaptic neurotransmitter receptors in C. elegans. Its key feature is the automated identification and analysis of individual synapses in three dimensions in multi-plane confocal microscope output files, tabulating position, volume, fluorescence intensity, and total fluorescence for each synapse. This approach has two principal advantages over manual analysis of z-plane projections of confocal data. First, because every plane of the confocal data set is included, no data are lost through z-plane projection, typically based on pixel intensity averages or maxima. Second, identification of synapses is automated, but can be inspected by the experimenter as the data analysis proceeds, allowing fast and accurate extraction of data from large numbers of synapses. Hundreds to thousands of synapses per sample can easily be obtained, producing large data sets to maximize statistical power. Considerations for preparing C. elegans for analysis, and performing confocal imaging to minimize variability between animals within treatment groups are also discussed. Although developed to analyze C. elegans postsynaptic receptors, this method is generally useful for any type of synaptically-localized protein, or indeed, any fluorescence signal that is localized to discrete clusters, puncta, or organelles. The procedure is performed in three steps: 1) preparation of samples, 2) confocal imaging, and 3) image analysis. Steps 1 and 2 are specific to C. elegans, while step 3 is generally applicable to any punctate fluorescence signal in confocal micrographs.
Collapse
Affiliation(s)
- Brianne L Sturt
- Department of Biological Sciences, University of Toledo, USA
| | | |
Collapse
|
18
|
Wang C, Wang Y, Wang M, Chen J, Yu N, Song S, Kaminski NE, Zhang W. Signal peptide and denaturing temperature are critical factors for efficient mammalian expression and immunoblotting of cannabinoid receptors. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2012; 32:299-302. [PMID: 22528237 DOI: 10.1007/s11596-012-0052-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Indexed: 12/01/2022]
Abstract
Many researchers employed mammalian expression system to artificially express cannabinoid receptors, but immunoblot data that directly prove efficient protein expression can hardly be seen in related research reports. In present study, we demonstrated cannabinoid receptor protein was not able to be properly expressed with routine mammalian expression system. This inefficient expression was rescued by endowing an exogenous signal peptide ahead of cannabinoid receptor peptide. In addition, the artificially synthesized cannabinoid receptor was found to aggregate under routine sample denaturing temperatures (i.e., ≥95°C), forming a large molecular weight band when analyzed by immuno-blotting. Only denaturing temperatures ≤75°C yielded a clear band at the predicted molecular weight. Collectively, we showed that efficient mammalian expression of cannabinoid receptors need a signal peptide sequence, and described the requirement for a low sample denaturing temperature in immuno-blot analysis. These findings provide very useful information for efficient mammalian expression and immuno-blotting of membrane receptors.
Collapse
Affiliation(s)
- Chenyun Wang
- Department of Pharmaceutical Administration, PLA General Hospital, Beijing, 100850, China
| | - Yingying Wang
- Outpatient Department of Xiyuan, PLA General Hospital, Beijing, 100850, China
| | - Miao Wang
- Department of Clinical Laboratory, No. 307 Hospital, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Jiankui Chen
- Department of Clinical Laboratory, No. 307 Hospital, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Nong Yu
- Department of Clinical Laboratory, No. 307 Hospital, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Shiping Song
- Department of Clinical Laboratory, No. 307 Hospital, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Norbert E Kaminski
- Department of Pharmacology and Toxicology and the Center for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Wei Zhang
- Department of Clinical Laboratory, No. 307 Hospital, Academy of Military Medical Sciences, Beijing, 100071, China.
| |
Collapse
|
19
|
Pineda Rodó A, Váchová L, Palková Z. In vivo determination of organellar pH using a universal wavelength-based confocal microscopy approach. PLoS One 2012; 7:e33229. [PMID: 22470445 PMCID: PMC3310042 DOI: 10.1371/journal.pone.0033229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 02/12/2012] [Indexed: 11/19/2022] Open
Abstract
Many essential cellular processes are affected by transmembrane H(+) gradients and intracellular pH (pHi). The research of such metabolic events calls for a non-invasive method to monitor pHi within individual subcellular compartments. We present a novel confocal microscopy approach for the determination of organellar pHi in living cells expressing pH-dependent ratiometric fluorescent proteins. Unlike conventional intensity-based fluorometry, our method relies on emission wavelength scans at single-organelle resolution to produce wavelength-based pH estimates both accurate and robust to low-signal artifacts. Analyses of Ato1p-pHluorin and Ato1p-mCherry yeast cells revealed previously unreported wavelength shifts in pHluorin emission which, together with ratiometric mCherry, allowed for high-precision quantification of actual physiological pH values and evidenced dynamic pHi changes throughout the different stages of yeast colony development. Additionally, comparative pH quantification of Ato1p-pHluorin and Met17p-pHluorin cells implied the existence of a significant pHi gradient between peripheral and internal cytoplasm of cells from colonies occurring in the ammonia-producing alkali developmental phase. Results represent a step forward in the study of pHi regulation and subcellular metabolic functions beyond the scope of this study.
Collapse
Affiliation(s)
- Albert Pineda Rodó
- Department of Genetics and Microbiology, Charles University, Prague, Czech Republic
| | - Libuše Váchová
- Department of Genetics and Microbiology, Charles University, Prague, Czech Republic
- Institute of Microbiology, Academy of Sciences, Prague, Czech Republic
| | - Zdena Palková
- Department of Genetics and Microbiology, Charles University, Prague, Czech Republic
| |
Collapse
|
20
|
Bouschet T, Martin S, Henley JM. Regulation of calcium sensing receptor trafficking by RAMPs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 744:39-48. [PMID: 22434106 DOI: 10.1007/978-1-4614-2364-5_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
As mentioned earlier in this book, RAMPs were identified as proteins escorting the Calcitonin Receptor-Like Receptor (CRLR) to the plasma membrane (PM) to generate either CGRP (when associated with RAMP1), or adrenomedullin receptors (when associated with RAMP2 or RAMP3). Some years after this initial discovery, it was established that RAMPs can accompany four additional class B G Protein-Coupled Receptors-GPCRs- (PTH1, PTH2, glucagon receptor and VPAC1) to the PM.(1) By demonstrating that the sorting traffic of the Calcium Sensing Receptor (CaSR), a class C GPCR, is positively modulated by RAMP1 and RAMP3,(2) our data extended the concept of RAMPs as escorting molecules to another class of GPCRs.
Collapse
Affiliation(s)
- Tristan Bouschet
- Institut de Génomique Fonctionnelle, INSERM U661-CNRS UMR5203, Montpellier, France
| | | | | |
Collapse
|
21
|
Activation of spinal mu- and delta-opioid receptors potently inhibits substance P release induced by peripheral noxious stimuli. J Neurosci 2011; 31:13068-77. [PMID: 21917790 DOI: 10.1523/jneurosci.1817-11.2011] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Over the past few years, δ-opioid receptors (DOPRs) and μ-opioid receptors (MOPRs) have been shown to interact with each other. We have previously seen that expression of MOPR is essential for morphine and inflammation to potentiate the analgesic properties of selective DOPR agonists. In vivo, it is not clear whether MOPRs and DOPRs are expressed in the same neurons. Indeed, it was recently proposed that these receptors are segregated in different populations of nociceptors, with MOPRs and DOPRs expressed by peptidergic and nonpeptidergic fibers, respectively. In the present study, the role and the effects of DOPR- and MOPR-selective agonists in two different pain models were compared. Using preprotachykinin A knock-out mice, we first confirmed that substance P partly mediates intraplantar formalin- and capsaicin-induced pain behaviors. These mice had a significant reduction in pain behavior compared with wild-type mice. We then measured the effects of intrathecal deltorphin II (DOPR agonist) and DAMGO (MOPR agonist) on pain-like behavior, neuronal activation, and substance P release following formalin and capsaicin injection. We found that both agonists were able to decrease formalin- and capsaicin-induced pain, an effect that was correlated with a reduction in the number of c-fos-positive neurons in the superficial laminae of the lumbar spinal cord. Finally, visualization of NK(1) (neurokinin 1) receptor internalization revealed that DOPR and MOPR activation strongly reduced formalin- and capsaicin-induced substance P release via direct action on primary afferent fibers. Together, our results indicate that functional MOPRs and DOPRs are both expressed by peptidergic nociceptors.
Collapse
|
22
|
Baines AE, Corrêa SAL, Irving AJ, Frenguelli BG. Differential trafficking of adenosine receptors in hippocampal neurons monitored using GFP- and super-ecliptic pHluorin-tagged receptors. Neuropharmacology 2011; 61:1-11. [PMID: 21315741 DOI: 10.1016/j.neuropharm.2011.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 01/28/2011] [Accepted: 02/02/2011] [Indexed: 01/18/2023]
Abstract
Adenosine receptors (ARs) modulate many cellular and systems-level processes in the mammalian CNS. However, little is known about the trafficking of ARs in neurons, despite their importance in controlling seizure activity and in neuroprotection in cerebral ischaemia. To address this we examined the agonist-dependent internalisation of C-terminal GFP-tagged A(1)Rs, A(2A)Rs and A(3)Rs in primary hippocampal neurons. Furthermore, we developed a novel super-ecliptic pHluorin (SEP)-tagged A(1)R which, via the N-terminal SEP tag, reports the cell-surface expression and trafficking of A(1)Rs in real-time. We demonstrate the differential trafficking of ARs in neurons: A(3)Rs internalise more rapidly than A1Rs, with little evidence of appreciable A(2A)R trafficking over the time-course of the experiments. Furthermore, the novel SEP-A(1)R construct revealed the time-course of internalisation and recovery of cell-surface expression to occur within minutes of agonist exposure and removal, respectively. These observations highlight the labile nature of A(1)R and A(3)Rs when expressed at the neuronal plasma membrane. Given the high levels of adenosine in the brain during ischaemia and seizures, internalisation of the inhibitory A(1)R may result in hyperexcitability, increased brain damage and the development of chronic epileptic states.
Collapse
Affiliation(s)
- A E Baines
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | | | | | | |
Collapse
|
23
|
Abstract
The majority of G-protein-coupled receptors (GPCRs) function at the cell surface, where they are activated by their ligands present in the extracellular milieu. Interestingly, type I cannabinoid receptor (CB(1) R), one of the most abundant GPCRs in the central nervous system, is predominantly intracellular. The important physiological roles of CB(1) R have sparked interest in the elucidation of the molecular mechanisms underlying the trafficking of this receptor and the role of intracellular CB(1) Rs. Thus far, results from different groups have been, at least in part, contradictory and the basis of CB(1) R intracellular localization remains controversial. In this commentary, by comparing the studies examining CB(1) R trafficking and localization, we identify technical or experimental ground responsible for the conflicting results. Finally, we propose a possible mechanism of CB(1) R trafficking that may reconcile the different models.
Collapse
Affiliation(s)
- Raphael Rozenfeld
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
24
|
Dubois D, Gendron L. Delta opioid receptor-mediated analgesia is not altered in preprotachykinin A knockout mice. Eur J Neurosci 2010; 32:1921-9. [DOI: 10.1111/j.1460-9568.2010.07466.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
25
|
Nagakura I, Dunn TW, Farah CA, Heppner A, Li FF, Sossin WS. Regulation of protein kinase C Apl II by serotonin receptors in Aplysia. J Neurochem 2010; 115:994-1006. [PMID: 20964689 DOI: 10.1111/j.1471-4159.2010.06986.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Serotonin (5-hydroxytryptamine, 5HT) is the neurotransmitter that mediates dishabituation in Aplysia. Serotonin mediates this behavioral change through the reversal of synaptic depression in sensory neurons (SNs). However, the 5HT receptors present in SNs and in particular, the receptor important for activation of protein kinase C (PKC) have not been fully identified. Using a recent genome assembly of Aplysia, we identified new receptors from the 5HT(2) , 5HT(4) , and 5HT(7) families. Using RT-PCR from isolated SNs, we found that three 5HT receptors, 5HT(1Apl(a)) , 5HT(2Apl) , and 5HT(7Apl) were expressed in SNs. These receptors were cloned and expressed in a heterologous system. In this system, 5HT(2Apl) could significantly translocate PKC Apl II in response to 5HT and this was blocked by pirenperone, a 5HT(2) receptor antagonist. Surprisingly, pirenperone did not block 5HT-mediated translocation of PKC Apl II in SNs, nor 5HT-mediated reversal of depression. Expression of 5HT(1Apl(a)) in SNs or genistein, an inhibitor of tyrosine kinases inhibited both PKC translocation and reversal of depression. These results suggest a non-canonical mechanism for the translocation of PKC Apl II in SNs.
Collapse
Affiliation(s)
- Ikue Nagakura
- Department of Psychology, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
Astrocytes possess GPCRs (G-protein-coupled receptors) for neuroactive substances and can respond via these receptors to signals originating from neurons as well as astrocytes. Like many transmembrane proteins, GPCRs exist in a dynamic equilibrium between receptors expressed at the plasma membrane and those present within intracellular trafficking compartments. The characteristics of GPCR trafficking within astrocytes have not been investigated. We therefore monitored the trafficking of recombinant fluorescent protein chimeras of the CB1R (cannabinoid receptor 1) that is thought to be expressed natively in astrocytes. CB1R chimeras displayed a marked punctate intracellular localization when expressed in cultured rat visual cortex astrocytes, an expression pattern reminiscent of native CB1R expression in these cells. Based upon trafficking characteristics, we found the existence of two populations of vesicular CB1R puncta: (i) relatively immobile puncta with movement characteristic of diffusion and (ii) mobile puncta with movement characteristic of active transport along cytoskeletal elements. The predominant direction of active transport is oriented radially to/from the nuclear region, which can be abolished by disruption of the microtubule cytoskeleton. CB1R puncta are localized within intracellular acidic organelles, mainly co-localizing with endocytic compartments. Constitutive trafficking of CB1R to and from the plasma membrane is an energetically costly endeavour whose function is at present unclear in astrocytes. However, given that intracellular CB1Rs can engage cell signalling pathways, it is likely that this process plays an important regulatory role.
Collapse
|
27
|
Bouschet T, Martin S, Henley JM. Regulation of calcium-sensing-receptor trafficking and cell-surface expression by GPCRs and RAMPs. Trends Pharmacol Sci 2008; 29:633-9. [PMID: 18930324 DOI: 10.1016/j.tips.2008.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 09/15/2008] [Accepted: 09/16/2008] [Indexed: 12/14/2022]
Abstract
The calcium-sensing (CaS) receptor is a G-protein-coupled receptor (GPCR) that is of fundamental importance for extracellular calcium signalling and calcium homeostasis. The CaS receptor detects changes in free, ionized extracellular calcium concentration and initiates pathways that constantly re-adjust levels of circulating calcium. In addition, the CaS receptor is involved in processes such as stem-cell homing and regulation of neuronal-process outgrowth. To perform these functions, the CaS receptor must be appropriately targeted to the plasma membrane so that its large N-terminal calcium-sensing domain is positioned in the extracellular environment to detect dynamic changes in ionic calcium concentration. Here, we provide an overview of the molecular determinants controlling CaS receptor forward traffic and highlight the roles of CaS receptor interactors such as receptor-activity-modifying proteins and subunits of other class C GPCRs in this process.
Collapse
Affiliation(s)
- Tristan Bouschet
- Department of Anatomy, Medical Research Council Centre for Synaptic Plasticity, School of Medical Sciences, University of Bristol, University Walk, Bristol, UK
| | | | | |
Collapse
|
28
|
Harkany T, Keimpema E, Barabás K, Mulder J. Endocannabinoid functions controlling neuronal specification during brain development. Mol Cell Endocrinol 2008; 286:S84-90. [PMID: 18394789 DOI: 10.1016/j.mce.2008.02.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 02/19/2008] [Accepted: 02/19/2008] [Indexed: 01/30/2023]
Abstract
Endocannabinoids (eCBs) regulate a broad range of physiological functions in the postnatal brain and are implicated in the neuropathogenesis of psychiatric and metabolic diseases. Accumulating evidence indicates that eCB signaling also serves key functions during neurodevelopment; and is inherently involved in the control of neurogenesis, neural progenitor proliferation, lineage segregation, and the migration and phenotypic specification of immature neurons. Recent advances in developmental biology define fundamental eCB-driven cellular mechanisms that also contribute to our understanding of the molecular substrates of prenatal drug, in particular cannabis, actions. Here, we summarize known organizing principles of eCB-signaling systems in the developing telencephalon, and outline the sequence of decision points and underlying signaling pathways upon CB1 cannabinoid receptor activation that contribute to neuronal diversification in the developing brain. Finally, we discuss how these novel principles affect the formation of complex neuronal networks.
Collapse
Affiliation(s)
- Tibor Harkany
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, United Kingdom.
| | | | | | | |
Collapse
|
29
|
Hayakawa K, Mishima K, Hazekawa M, Sano K, Irie K, Orito K, Egawa T, Kitamura Y, Uchida N, Nishimura R, Egashira N, Iwasaki K, Fujiwara M. Cannabidiol potentiates pharmacological effects of Delta(9)-tetrahydrocannabinol via CB(1) receptor-dependent mechanism. Brain Res 2007; 1188:157-64. [PMID: 18021759 DOI: 10.1016/j.brainres.2007.09.090] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 09/16/2007] [Accepted: 09/28/2007] [Indexed: 10/22/2022]
Abstract
Cannabidiol, a non-psychoactive component of cannabis, has been reported to have interactions with Delta(9)-tetrahydrocannabinol (Delta(9)-THC). However, such interactions have not sufficiently been clear and may have important implications for understanding the pharmacological effects of marijuana. In the present study, we investigated whether cannabidiol modulates the pharmacological effects of Delta(9)-THC on locomotor activity, catalepsy-like immobilisation, rectal temperature and spatial memory in the eight-arm radial maze task in mice. In addition, we measured expression level of cannabinoid CB(1) receptor at striatum, cortex, hippocampus and hypothalamus. Delta(9)-THC (1, 3, 6 and 10 mg/kg) induced hypoactivity, catalepsy-like immobilisation and hypothermia in a dose-dependent manner. In addition, Delta(9)-THC (1, 3 and 6 mg/kg) dose-dependently impaired spatial memory in eight-arm radial maze. On the other hand, cannabidiol (1, 3, 10, 25 and 50 mg/kg) did not affect locomotor activity, catalepsy-like immobilisation, rectal temperature and spatial memory on its own. However, higher dose of cannabidiol (10 or 50 mg/kg) exacerbated pharmacological effects of lower dose of Delta(9)-THC, such as hypoactivity, hypothermia and impairment of spatial memory. Moreover, cannabidiol (50 mg/kg) with Delta(9)-THC (1 mg/kg) enhanced the expression level of CB(1) receptor expression in hippocampus and hypothalamus. Cannabidiol potentiated pharmacological effects of Delta(9)-THC via CB(1) receptor-dependent mechanism. These findings may contribute in setting the basis for interaction of cannabinoids and to find a cannabinoid mechanism in central nervous system.
Collapse
Affiliation(s)
- Kazuhide Hayakawa
- Department of Neuropharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka City, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|