1
|
Jaeckel ER, Arias-Hervert ER, Perez-Medina AL, Schulz S, Birdsong WT. Chronic morphine treatment induces sex- and synapse-specific cellular tolerance on thalamo-cortical mu opioid receptor signaling. J Neurophysiol 2024; 132:968-978. [PMID: 39110512 PMCID: PMC11427077 DOI: 10.1152/jn.00265.2024] [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: 06/24/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
How cellular adaptations give rise to opioid analgesic tolerance to opioids like morphine is not well understood. For one, pain is a complex phenomenon comprising both sensory and affective components, largely mediated through separate circuits. Glutamatergic projections from the medial thalamus (MThal) to the anterior cingulate cortex (ACC) are implicated in processing of affective pain, a relatively understudied component of the pain experience. The goal of this study was to determine the effects of chronic morphine exposure on mu-opioid receptor (MOR) signaling on MThal-ACC synaptic transmission within the excitatory and feedforward inhibitory pathways. Using whole cell patch-clamp electrophysiology and optogenetics to selectively target these projections, we measured morphine-mediated inhibition of optically evoked postsynaptic currents in ACC layer V pyramidal neurons in drug-naïve and chronically morphine-treated mice. We found that morphine perfusion inhibited the excitatory and feedforward inhibitory pathways similarly in females but caused greater inhibition of the inhibitory pathway in males. Chronic morphine treatment robustly attenuated morphine presynaptic inhibition within the inhibitory pathway in males, but not females, and mildly attenuated presynaptic inhibition within the excitatory pathway in both sexes. These effects were not observed in MOR phosphorylation-deficient mice. This study indicates that chronic morphine treatment induces cellular tolerance to morphine within a thalamo-cortical circuit relevant to pain and opioid analgesia. Furthermore, it suggests this tolerance may be driven by MOR phosphorylation. Overall, these findings improve our understanding of how chronic opioid exposure alters cellular signaling in ways that may contribute to opioid analgesic tolerance.NEW & NOTEWORTHY Opioid signaling within the anterior cingulate cortex (ACC) is important for opioid modulation of affective pain. Glutamatergic medial thalamus (MThal) neurons synapse in the ACC and opioids, acting through mu opioid receptors (MORs), acutely inhibit synaptic transmission from MThal synapses. However, the effect of chronic opioid exposure on MThal-ACC synaptic transmission is not known. Here, we demonstrate that chronic morphine treatment induces cellular tolerance at these synapses in a sex-specific and phosphorylation-dependent manner.
Collapse
Affiliation(s)
- Elizabeth R Jaeckel
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States
| | - Erwin R Arias-Hervert
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States
| | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | - William T Birdsong
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
2
|
Gonzalez-Hernandez AJ, Munguba H, Levitz J. Emerging modes of regulation of neuromodulatory G protein-coupled receptors. Trends Neurosci 2024; 47:635-650. [PMID: 38862331 PMCID: PMC11324403 DOI: 10.1016/j.tins.2024.05.008] [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: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024]
Abstract
In the nervous system, G protein-coupled receptors (GPCRs) control neuronal excitability, synaptic transmission, synaptic plasticity, and, ultimately, behavior through spatiotemporally precise initiation of a variety of signaling pathways. However, despite their critical importance, there is incomplete understanding of how these receptors are regulated to tune their signaling to specific neurophysiological contexts. A deeper mechanistic picture of neuromodulatory GPCR function is needed to fully decipher their biological roles and effectively harness them for the treatment of neurological and psychiatric disorders. In this review, we highlight recent progress in identifying novel modes of regulation of neuromodulatory GPCRs, including G protein- and receptor-targeting mechanisms, receptor-receptor crosstalk, and unique features that emerge in the context of chemical synapses. These emerging principles of neuromodulatory GPCR tuning raise critical questions to be tackled at the molecular, cellular, synaptic, and neural circuit levels in the future.
Collapse
Affiliation(s)
| | - Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA; Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
3
|
Jaeckel ER, Herrera YN, Schulz S, Birdsong WT. Chronic Morphine Induces Adaptations in Opioid Receptor Signaling in a Thalamostriatal Circuit That Are Location Dependent, Sex Specific, and Regulated by μ-Opioid Receptor Phosphorylation. J Neurosci 2024; 44:e0293232023. [PMID: 37985179 PMCID: PMC10860620 DOI: 10.1523/jneurosci.0293-23.2023] [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: 02/16/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023] Open
Abstract
Chronic opioid exposure induces tolerance to the pain-relieving effects of opioids but sensitization to some other effects. While the occurrence of these adaptations is well understood, the underlying cellular mechanisms are less clear. This study aimed to determine how chronic treatment with morphine, a prototypical opioid agonist, induced adaptations to subsequent morphine signaling in different subcellular contexts. Opioids acutely inhibit glutamatergic transmission from medial thalamic (MThal) inputs to the dorsomedial striatum (DMS) via activity at μ-opioid receptors (MORs). MORs are present in somatic and presynaptic compartments of MThal neurons terminating in the DMS. We investigated the effects of chronic morphine treatment on subsequent morphine signaling at MThal-DMS synapses and MThal cell bodies in male and female mice. Surprisingly, chronic morphine treatment increased subsequent morphine inhibition of MThal-DMS synaptic transmission (morphine facilitation) in male, but not female, mice. At MThal cell bodies, chronic morphine treatment decreased subsequent morphine activation of potassium conductance (morphine tolerance) in both male and female mice. In knock-in mice expressing phosphorylation-deficient MORs, chronic morphine treatment resulted in tolerance to, rather than facilitation of, subsequent morphine signaling at MThal-DMS terminals, suggesting phosphorylation deficiency unmasks adaptations that counter the facilitation observed at presynaptic terminals in wild-type mice. The results of this study suggest that the effects of chronic morphine exposure are not ubiquitous; rather adaptations in MOR function may be determined by multiple factors such as subcellular receptor distribution, influence of local circuitry, and sex.
Collapse
Affiliation(s)
- Elizabeth R Jaeckel
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109
| | - Yoani N Herrera
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller University, D-07747 Jena, Germany
| | - William T Birdsong
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
4
|
Bouchet CA, McPherson KB, Coutens B, Janowsky A, Ingram SL. Monoacylglycerol Lipase Protects the Presynaptic Cannabinoid 1 Receptor from Desensitization by Endocannabinoids after Persistent Inflammation. J Neurosci 2023; 43:5458-5467. [PMID: 37414560 PMCID: PMC10376933 DOI: 10.1523/jneurosci.0037-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/08/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023] Open
Abstract
Cannabinoid-targeted pain therapies are increasing with the expansion of cannabis legalization, however, their efficacy may be limited by pain-induced adaptations in the cannabinoid system. Cannabinoid receptor subtype 1 (CB1R) inhibition of spontaneous, GABAergic miniature IPSCs (mIPSCs) and evoked IPSCs (eIPSCs) in the ventrolateral periaqueductal gray (vlPAG) were compared in slices from naive and inflamed male and female Sprague Dawley rats. Complete Freund's Adjuvant (CFA) injections into the hindpaw induced persistent inflammation. In naive rats, exogenous cannabinoid agonists robustly reduce both eIPSCs and mIPSCs. After 5-7 d of inflammation, the effects of exogenous cannabinoids are significantly reduced because of CB1R desensitization via GRK2/3, as function is recovered in the presence of the GRK2/3 inhibitor, Compound 101 (Cmp101). Inhibition of GABA release by presynaptic μ-opioid receptors in the vlPAG does not desensitize with persistent inflammation. Unexpectedly, while CB1R desensitization significantly reduces the inhibition produced by exogenous agonists, depolarization-induced suppression of inhibition protocols that promote 2-arachidonoylglycerol (2-AG) synthesis exhibit prolonged CB1R activation after inflammation. 2-AG tone is detected in slices from CFA-treated rats when GRK2/3 is blocked, suggesting an increase in 2-AG synthesis after persistent inflammation. Inhibiting 2-AG degradation with the monoacylglycerol lipase (MAGL) inhibitor JZL184 during inflammation results in the desensitization of CB1Rs by endocannabinoids that is reversed with Cmp101. Collectively, these data indicate that persistent inflammation primes CB1Rs for desensitization, and MAGL degradation of 2-AG protects CB1Rs from desensitization in inflamed rats. These adaptations with inflammation have important implications for the development of cannabinoid-based pain therapeutics targeting MAGL and CB1Rs.SIGNIFICANCE STATEMENT Presynaptic G-protein-coupled receptors are resistant to desensitization. Here we find that persistent inflammation increases endocannabinoid levels, priming presynaptic cannabinoid 1 receptors for desensitization on subsequent addition of exogenous agonists. Despite the reduced efficacy of exogenous agonists, endocannabinoids have prolonged efficacy after persistent inflammation. Endocannabinoids readily induce cannabinoid 1 receptor desensitization if their degradation is blocked, indicating that endocannabinoid concentrations are maintained at subdesensitizing levels and that degradation is critical for maintaining endocannabinoid regulation of presynaptic GABA release in the ventrolateral periaqueductal gray during inflammatory states. These adaptations with inflammation have important implications for the development of cannabinoid-based pain therapies.
Collapse
Affiliation(s)
- Courtney A Bouchet
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
- Neuroscience Graduate Program, Vollum Institute, Portland, Oregon 97239
| | - Kylie B McPherson
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Basile Coutens
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Aaron Janowsky
- Research Service, VA Portland Health Care System, Portland, Oregon 97239
- Departments of Psychiatry, and Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Susan L Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon 97239
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
5
|
Coutens B, Ingram SL. Key differences in regulation of opioid receptors localized to presynaptic terminals compared to somas: Relevance for novel therapeutics. Neuropharmacology 2023; 226:109408. [PMID: 36584882 PMCID: PMC9898207 DOI: 10.1016/j.neuropharm.2022.109408] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/05/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
Opioid receptors are G protein-coupled receptors (GPCRs) that regulate activity within peripheral, subcortical and cortical circuits involved in pain, reward, and aversion processing. Opioid receptors are expressed in both presynaptic terminals where they inhibit neurotransmitter release and postsynaptic locations where they act to hyperpolarize neurons and reduce activity. Agonist activation of postsynaptic receptors at the plasma membrane signal via ion channels or cytoplasmic second messengers. Agonist binding initiates regulatory processes that include phosphorylation by G protein receptor kinases (GRKs) and recruitment of beta-arrestins that desensitize and internalize the receptors. Opioid receptors also couple to effectors from endosomes activating intracellular enzymes and kinases. In contrast to postsynaptic opioid receptors, receptors localized to presynaptic terminals are resistant to desensitization such that there is no loss of signaling in the continuous presence of opioids over the same time scale. Thus, the balance of opioid signaling in circuits expressing pre- and postsynaptic opioid receptors is shifted toward inhibition of presynaptic neurotransmitter release during continuous opioid exposure. The functional implication of this shift is not often acknowledged in behavioral studies. This review covers what is currently understood about regulation of opioid/nociceptin receptors, with an emphasis on opioid receptor signaling in pain and reward circuits. Importantly, the review covers regulation of presynaptic receptors and the critical gaps in understanding this area, as well as the opportunities to further understand opioid signaling in brain circuits. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
Collapse
Affiliation(s)
- Basile Coutens
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Susan L Ingram
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
6
|
Jaeckel ER, Arias-Hervert ER, Perez-Medina AL, Herrera YN, Schulz S, Birdsong WT. Chronic morphine induces adaptations in opioid receptor signaling in a thalamo-cortico-striatal circuit that are projection-dependent, sex-specific and regulated by mu opioid receptor phosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528057. [PMID: 36824766 PMCID: PMC9949156 DOI: 10.1101/2023.02.13.528057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Chronic opioid exposure induces tolerance to the pain-relieving effects of opioids but sensitization to some other effects. While the occurrence of these adaptations is well-understood, the underlying cellular mechanisms are less clear. This study aimed to determine how chronic treatment with morphine, a prototypical opioid agonist, induced adaptations to subsequent morphine signaling in different subcellular contexts. Opioids acutely inhibit glutamatergic transmission from medial thalamic (MThal) inputs to the dorsomedial striatum (DMS) and anterior cingulate cortex (ACC) via activity at μ-opioid receptors (MORs). MORs are present in somatic and presynaptic compartments of MThal neurons terminating in both the DMS and ACC. We investigated the effects of chronic morphine treatment on subsequent morphine signaling at MThal-DMS synapses, MThal-ACC synapses, and MThal cell bodies in male and female mice. Surprisingly, chronic morphine treatment increased subsequent morphine inhibition of MThal-DMS synaptic transmission (morphine facilitation), but decreased subsequent morphine inhibition of transmission at MThal-ACC synapses (morphine tolerance) in a sex-specific manner; these adaptations were present in male but not female mice. Additionally, these adaptations were not observed in knockin mice expressing phosphorylation-deficient MORs, suggesting a role of MOR phosphorylation in mediating both facilitation and tolerance to morphine within this circuit. The results of this study suggest that the effects of chronic morphine exposure are not ubiquitous; rather adaptations in MOR function may be determined by multiple factors such as subcellular receptor distribution, influence of local circuitry and sex.
Collapse
Affiliation(s)
| | | | | | - Yoani N. Herrera
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller University, Jena, Germany
| | | |
Collapse
|
7
|
McPherson KB, Ingram SL. Cellular and circuit diversity determines the impact of endogenous opioids in the descending pain modulatory pathway. Front Syst Neurosci 2022; 16:963812. [PMID: 36045708 PMCID: PMC9421147 DOI: 10.3389/fnsys.2022.963812] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/15/2022] [Indexed: 01/31/2023] Open
Abstract
The descending pain modulatory pathway exerts important bidirectional control of nociceptive inputs to dampen and/or facilitate the perception of pain. The ventrolateral periaqueductal gray (vlPAG) integrates inputs from many regions associated with the processing of nociceptive, cognitive, and affective components of pain perception, and is a key brain area for opioid action. Opioid receptors are expressed on a subset of vlPAG neurons, as well as on both GABAergic and glutamatergic presynaptic terminals that impinge on vlPAG neurons. Microinjection of opioids into the vlPAG produces analgesia and microinjection of the opioid receptor antagonist naloxone blocks stimulation-mediated analgesia, highlighting the role of endogenous opioid release within this region in the modulation of nociception. Endogenous opioid effects within the vlPAG are complex and likely dependent on specific neuronal circuits activated by acute and chronic pain stimuli. This review is focused on the cellular heterogeneity within vlPAG circuits and highlights gaps in our understanding of endogenous opioid regulation of the descending pain modulatory circuits.
Collapse
Affiliation(s)
- Kylie B. McPherson
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy,Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States,*Correspondence: Susan L. Ingram
| |
Collapse
|
8
|
Adhikary S, Williams JT. Cellular Tolerance Induced by Chronic Opioids in the Central Nervous System. Front Syst Neurosci 2022; 16:937126. [PMID: 35837149 PMCID: PMC9273719 DOI: 10.3389/fnsys.2022.937126] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/08/2022] [Indexed: 01/21/2023] Open
Abstract
Opioids are powerful analgesics that elicit acute antinociceptive effects through their action the mu opioid receptor (MOR). However opioids are ineffective for chronic pain management, in part because continuous activation of MORs induces adaptive changes at the receptor level and downstream signaling molecules. These adaptations include a decrease in receptor-effector coupling and changes to second messenger systems that can counteract the persistent activation of MORs by opioid agonists. Homeostatic regulation of MORs and downstream signaling cascades are viewed as precursors to developing tolerance. However, despite numerous studies identifying crucial mechanisms that contribute to opioid tolerance, no single regulatory mechanism that governs tolerance in at the cellular and systems level has been identified. Opioid tolerance is a multifaceted process that involves both individual neurons that contain MORs and neuronal circuits that undergo adaptations following continuous MOR activation. The most proximal event is the agonist/receptor interaction leading to acute cellular actions. This review discusses our understanding of mechanisms that mediate cellular tolerance after chronic opioid treatment that, in part, is mediated by agonist/receptor interaction acutely.
Collapse
|
9
|
Effah F, de Gusmão Taveiros Silva NK, Vijayanathan K, Camarini R, Joly F, Taiwo B, Rabot S, Champeil-Potokar G, Bombail V, Bailey A. SEX-DEPENDENT IMPACT OF MICROBIOTA STATUS ON CEREBRAL μ -OPIOID RECEPTOR DENSITY IN FISCHER RATS. Eur J Neurosci 2022; 55:1917-1933. [PMID: 35393704 PMCID: PMC9324823 DOI: 10.1111/ejn.15666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/08/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022]
Abstract
μ‐opioid receptors (MOPr) play a critical role in social play, reward and pain, in a sex‐ and age‐dependent manner. There is evidence to suggest that sex and age differences in brain MOPr density may be responsible for this variability; however, little is known about the factors driving these differences in cerebral MOPr density. Emerging evidence highlights gut microbiota's critical influence and its bidirectional interaction with the brain on neurodevelopment. Therefore, we aimed to determine the impact of gut microbiota on MOPr density in male and female brains at different developmental stages. Quantitative [3H]DAMGO autoradiographic binding was carried out in the forebrain of male and female conventional (CON) and germ‐free (GF) rats at postnatal days (PND) 8, 22 and 116–150. Significant ‘microbiota status X sex’, ‘age X brain region’ interactions and microbiota status‐ and age‐dependent effects on MOPr binding were uncovered. Microbiota status influenced MOPr levels in males but not females, with higher MOPr levels observed in GF versus CON rats overall regions and age groups. In contrast, no overall sex differences were observed in GF or CON rats. Interestingly, within‐age planned comparison analysis conducted in frontal cortical and brain regions associated with reward revealed that this microbiota effect was restricted only to PND22 rats. Thus, this pilot study uncovers the critical sex‐dependent role of gut microbiota in regulating cerebral MOPr density, which is restricted to the sensitive developmental period of weaning. This may have implications in understanding the importance of microbiota during early development on opioid signalling and associated behaviours.
Collapse
Affiliation(s)
- Felix Effah
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | | | - Katie Vijayanathan
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | - Rosana Camarini
- Pharmacology Department, Universidade de Sao Paulo, São Paulo, Brazil
| | - Fatima Joly
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Benjamin Taiwo
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Vincent Bombail
- UMR PNCA, AgroParisTech, INRAE, Université Paris-Saclay, Paris, France
| | - Alexis Bailey
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| |
Collapse
|
10
|
Metz MJ, Daimon CM, King CM, Rau AR, Hentges ST. Individual arcuate nucleus proopiomelanocortin neurons project to select target sites. Am J Physiol Regul Integr Comp Physiol 2021; 321:R982-R989. [PMID: 34755553 PMCID: PMC8714814 DOI: 10.1152/ajpregu.00169.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/22/2022]
Abstract
Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARH) are a diverse group of neurons that project widely to different brain regions. It is unknown how this small population of neurons organizes its efferent projections. In this study, we hypothesized that individual ARH POMC neurons exclusively innervate select target regions. To investigate this hypothesis, we first verified that only a fraction of ARH POMC neurons innervate the lateral hypothalamus (LH), the paraventricular nucleus of the hypothalamus (PVN), the periaqueductal gray (PAG), or the ventral tegmental area (VTA) using the retrograde tracer cholera toxin B (CTB). Next, two versions of CTB conjugated to distinct fluorophores were injected bilaterally into two of the regions such that PVN and VTA, PAG and VTA, or LH and PVN received tracers simultaneously. These pairs of target sites were chosen based on function and location. Few individual ARH POMC neurons projected to two brain regions at once, suggesting that there are ARH POMC neuron subpopulations organized by their efferent projections. We also investigated whether increasing the activity of POMC neurons could increase the number of ARH POMC neurons labeled with CTB, implying an increase in new synaptic connections to downstream regions. However, chemogenetic enhancement of POMC neuron activity did not increase retrograde tracing of CTB back to ARH POMC neurons from either the LH, PVN, or VTA. Overall, subpopulations of ARH POMC neurons with distinct efferent projections may serve as a way for the POMC population to organize its many functions.
Collapse
Affiliation(s)
- Marissa J Metz
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Caitlin M Daimon
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Connie M King
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Andrew R Rau
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
11
|
He XJ, Patel J, Weiss CE, Ma X, Bloodgood BL, Banghart MR. Convergent, functionally independent signaling by mu and delta opioid receptors in hippocampal parvalbumin interneurons. eLife 2021; 10:69746. [PMID: 34787079 PMCID: PMC8716102 DOI: 10.7554/elife.69746] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Functional interactions between G protein-coupled receptors are poised to enhance neuronal sensitivity to neuromodulators and therapeutic drugs. Mu and Delta opioid receptors (MORs and DORs) can interact when overexpressed in the same cells, but whether co-expression of endogenous MORs and DORs in neurons leads to functional interactions is unclear. Here, in mice, we show that both MORs and DORs inhibit parvalbumin-expressing basket cells (PV-BCs) in hippocampal CA1 through partially occlusive signaling pathways that terminate on somato-dendritic potassium channels and presynaptic calcium channels. Using photoactivatable opioid neuropeptides, we find that DORs dominate the response to enkephalin in terms of both ligand-sensitivity and kinetics, which may be due to relatively low expression levels of MOR. Opioid-activated potassium channels do not show heterologous desensitization, indicating that MORs and DORs signal independently. In a direct test for heteromeric functional interactions, the DOR antagonist TIPP-Psi does not alter the kinetics or potency of either the potassium channel or synaptic responses to photorelease of the MOR agonist DAMGO. Thus, aside from largely redundant and convergent signaling, MORs and DORs do not functionally interact in PV-BCs in a way that impacts somato-dendritic potassium currents or synaptic transmission. These findings imply that crosstalk between MORs and DORs, either in the form of physical interactions or synergistic intracellular signaling, is not a preordained outcome of co-expression in neurons.
Collapse
Affiliation(s)
- Xinyi Jenny He
- Biological Sciences, University of California San Diego, La Jolla, United States
| | - Janki Patel
- University of California San Diego, San Diego, United States
| | - Connor E Weiss
- University of California San Diego, San Diego, United States
| | - Xiang Ma
- University of California San Diego, San Diego, United States
| | - Brenda L Bloodgood
- Biological Sciences, University of California San Diego, La Jolla, United States
| | | |
Collapse
|
12
|
Jullié D, Gondin AB, von Zastrow M, Canals M. Opioid Pharmacology under the Microscope. Mol Pharmacol 2020; 98:425-432. [PMID: 32198210 DOI: 10.1124/mol.119.119321] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/10/2020] [Indexed: 12/18/2022] Open
Abstract
The powerful analgesic effects of opioid drugs have captivated the interest of physicians and scientists for millennia, and the ability of opioid drugs to produce serious undesired effects has been recognized for a similar period of time (Kieffer and Evans, 2009). Many of these develop progressively with prolonged or repeated drug use and then persist, motivating particular interest in understanding how opioid drugs initiate adaptive or maladaptive modifications in neural function or regulation. Exciting advances have been made over the past several years in elucidating drug-induced changes at molecular, cellular, and physiologic scales of analysis. The present review will highlight some recent cellular studies that we believe bridge across scales and will focus on optical imaging approaches that put opioid drug action "under the microscope." SIGNIFICANCE STATEMENT: Opioid receptors are major pharmacological targets, but their signaling at the cellular level results from a complex interplay between pharmacology, regulation, subcellular localization, and membrane trafficking. This minireview discusses recent advances in understanding the cellular biology of opioid receptors, emphasizing particular topics discussed at the 50th anniversary of the International Narcotics Research Conference. Our goal is to highlight distinct signaling and regulatory properties emerging from the cellular biology of opioid receptors and discuss potential relevance to therapeutics.
Collapse
Affiliation(s)
- Damien Jullié
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| | - Arisbel B Gondin
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| | - Mark von Zastrow
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| | - Meritxell Canals
- Departments of Psychiatry and Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California (D.J., M.v.Z.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia (A.B.G.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (M.C.); and Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom (M.C.)
| |
Collapse
|
13
|
Birdsong WT, Williams JT. Recent Progress in Opioid Research from an Electrophysiological Perspective. Mol Pharmacol 2020; 98:401-409. [PMID: 32198208 PMCID: PMC7562972 DOI: 10.1124/mol.119.119040] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Electrophysiological approaches provide powerful tools to further our understanding of how different opioids affect signaling through opioid receptors; how opioid receptors modulate circuitry involved in processes such as pain, respiration, addiction, and feeding; and how receptor signaling and circuits are altered by physiologic challenges, such as injury, stress, and chronic opioid treatment. The use of genetic manipulations to alter or remove μ-opioid receptors (MORs) with anatomic and cell type specificity and the ability to activate or inhibit specific circuits through opto- or chemogenetic approaches are being used in combination with electrophysiological, pharmacological, and systems-level physiology experiments to expand our understanding of the beneficial and maladaptive roles of opioids and opioid receptor signaling. New approaches for studying endogenous opioid peptide signaling and release and the dynamics of these systems in response to chronic opioid use, pain, and stress will add another layer to our understanding of the intricacies of opioid modulation of brain circuits. This understanding may lead to new targets or approaches for drug development or treatment regimens that may affect both acute and long-term effects of manipulating the activity of circuits involved in opioid-mediated physiology and behaviors. This review will discuss recent advancements in our understanding of the role of phosphorylation in regulating MOR signaling, as well as our understanding of circuits and signaling pathways mediating physiologic behaviors such as respiratory control, and discuss how electrophysiological tools combined with new technologies have and will continue to advance the field of opioid research.
Collapse
Affiliation(s)
- William T Birdsong
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (W.T.B.) and Vollum Institute, Oregon Health & Science University, Portland, Oregon (J.T.W.)
| | - John T Williams
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (W.T.B.) and Vollum Institute, Oregon Health & Science University, Portland, Oregon (J.T.W.)
| |
Collapse
|
14
|
Jullié D, Stoeber M, Sibarita JB, Zieger HL, Bartol TM, Arttamangkul S, Sejnowski TJ, Hosy E, von Zastrow M. A Discrete Presynaptic Vesicle Cycle for Neuromodulator Receptors. Neuron 2020; 105:663-677.e8. [PMID: 31837915 PMCID: PMC7035187 DOI: 10.1016/j.neuron.2019.11.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 08/02/2019] [Accepted: 11/11/2019] [Indexed: 01/27/2023]
Abstract
A major function of GPCRs is to inhibit presynaptic neurotransmitter release, requiring ligand-activated receptors to couple locally to effectors at terminals. The current understanding of how this is achieved is through receptor immobilization on the terminal surface. Here, we show that opioid peptide receptors, GPCRs that mediate highly sensitive presynaptic inhibition, are instead dynamic in axons. Opioid receptors diffuse rapidly throughout the axon surface and internalize after ligand-induced activation specifically at presynaptic terminals. We delineate a parallel regulated endocytic cycle for GPCRs operating at the presynapse, separately from the synaptic vesicle cycle, which clears activated receptors from the surface of terminals and locally reinserts them to maintain the diffusible surface pool. We propose an alternate strategy for achieving local control of presynaptic effectors that, opposite to using receptor immobilization and enforced proximity, is based on lateral mobility of receptors and leverages the inherent allostery of GPCR-effector coupling.
Collapse
Affiliation(s)
- Damien Jullié
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California 94158, USA,Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, California 94158, USA
| | - Miriam Stoeber
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California 94158, USA,Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, California 94158, USA,Department of Cell Physiology and Metabolism, University of Geneva, 1211 Geneva, Switzerland
| | - Jean-Baptiste Sibarita
- Interdisciplinary Institute for Neuroscience, UMR 5297, Centre National de la Recherche Scientifique, 33077 Bordeaux, France,Interdisciplinary Institute for Neuroscience, University of Bordeaux, 33077 Bordeaux, France
| | - Hanna L. Zieger
- Interdisciplinary Institute for Neuroscience, UMR 5297, Centre National de la Recherche Scientifique, 33077 Bordeaux, France,Interdisciplinary Institute for Neuroscience, University of Bordeaux, 33077 Bordeaux, France
| | - Thomas M. Bartol
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Seksiri Arttamangkul
- The Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Terrence J. Sejnowski
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Eric Hosy
- Interdisciplinary Institute for Neuroscience, UMR 5297, Centre National de la Recherche Scientifique, 33077 Bordeaux, France,Interdisciplinary Institute for Neuroscience, University of Bordeaux, 33077 Bordeaux, France
| | - Mark von Zastrow
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California 94158, USA,Department of Psychiatry, University of California, San Francisco School of Medicine, San Francisco, California 94158, USA,Lead contact,Correspondence:
| |
Collapse
|
15
|
Lau BK, Winters BL, Vaughan CW. Opioid presynaptic disinhibition of the midbrain periaqueductal grey descending analgesic pathway. Br J Pharmacol 2020; 177:2320-2332. [PMID: 31971607 DOI: 10.1111/bph.14982] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE The midbrain periaqueductal grey (PAG) plays a central role in modulating pain through a descending pathway that projects indirectly to the spinal cord via the rostroventral medial medulla (RVM). While opioids are potent analgesics that target the PAG, their cellular actions on descending projection neurons are unclear. EXPERIMENTAL APPROACH Patch clamp recordings in voltage- and current-clamp mode were made from acutely prepared PAG slices from animals that received retrograde tracer injections into the RVM. KEY RESULTS The μ-agonist DAMGO reduced GABAergic evoked inhibitory postsynaptic currents (IPSCs) in retro-labelled, RVM-projecting neurons to a greater extent than in unlabelled neurons. The κ-opioid agonist U69593 reduced evoked IPSCs to a similar extent in both neuronal groups, while the δ-opioid agonist deltorphin-II was without effect. DAMGO and U69593 both produced a reduction in the rate, but not amplitude of spontaneous miniature IPSCs and asynchronous evoked IPSCs in retro-labelled neurons. DAMGO and U69593 also suppressed glutamatergic EPSCs in retro-labelled and unlabelled neurons. The DAMGO inhibition of evoked EPSCs, however, was less than that for evoked IPSCs in retro-labelled, but not unlabelled neurons. In current clamp, DAMGO produced a depolarizing increase in evoked postsynaptic potentials in retro-labelled neurons, but directly inhibited unlabelled neurons. CONCLUSION AND IMPLICATIONS These findings suggest that μ-opioids activate the descending analgesic pathway from the midbrain PAG by a combination of presynaptic disinhibition of RVM-projecting neurons and postsynaptic inhibition of presumptive interneurons.
Collapse
Affiliation(s)
- Benjamin K Lau
- Pain Management Research Institute and Kolling Institute of Medical Research, The University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| | - Bryony L Winters
- Pain Management Research Institute and Kolling Institute of Medical Research, The University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| | - Christopher W Vaughan
- Pain Management Research Institute and Kolling Institute of Medical Research, The University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| |
Collapse
|
16
|
Velasquez-Martinez MC, Santos-Vera B, Velez-Hernandez ME, Vazquez-Torres R, Jimenez-Rivera CA. Alpha-1 Adrenergic Receptors Modulate Glutamate and GABA Neurotransmission onto Ventral Tegmental Dopamine Neurons during Cocaine Sensitization. Int J Mol Sci 2020; 21:E790. [PMID: 31991781 PMCID: PMC7036981 DOI: 10.3390/ijms21030790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
The ventral tegmental area (VTA) plays an important role in the reward and motivational processes that facilitate the development of drug addiction. Presynaptic α1-AR activation modulates glutamate and Gamma-aminobutyric acid (GABA) release. This work elucidates the role of VTA presynaptic α1-ARs and their modulation on glutamatergic and GABAergic neurotransmission during cocaine sensitization. Excitatory and inhibitory currents (EPSCs and IPSCs) measured by a whole cell voltage clamp show that α1-ARs activation increases EPSCs amplitude after 1 day of cocaine treatment but not after 5 days of cocaine injections. The absence of a pharmacological response to an α1-ARs agonist highlights the desensitization of the receptor after repeated cocaine administration. The desensitization of α1-ARs persists after a 7-day withdrawal period. In contrast, the modulation of α1-ARs on GABA neurotransmission, shown by decreases in IPSCs' amplitude, is not affected by acute or chronic cocaine injections. Taken together, these data suggest that α1-ARs may enhance DA neuronal excitability after repeated cocaine administration through the reduction of GABA inhibition onto VTA dopamine (DA) neurons even in the absence of α1-ARs' function on glutamate release and protein kinase C (PKC) activation. α1-AR modulatory changes in cocaine sensitization increase our knowledge of the role of the noradrenergic system in cocaine addiction and may provide possible avenues for therapeutics.
Collapse
Affiliation(s)
- Maria Carolina Velasquez-Martinez
- Grupo de Neurociencias y Comportamiento, Departamento de Ciencias Básicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga 680006, Colombia;
| | - Bermary Santos-Vera
- Department of Biology, Cayey Campus, University of Puerto Rico, Cayey, PR 00737, USA;
| | - Maria E. Velez-Hernandez
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, Kingsville, TX 78363, USA;
| | - Rafael Vazquez-Torres
- Department of Physiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00925, USA;
| | - Carlos A. Jimenez-Rivera
- Department of Physiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR 00925, USA;
| |
Collapse
|
17
|
DiCello JJ, Saito A, Rajasekhar P, Sebastian BW, McQuade RM, Gondin AB, Veldhuis NA, Canals M, Carbone SE, Poole DP. Agonist-dependent development of delta opioid receptor tolerance in the colon. Cell Mol Life Sci 2019; 76:3033-3050. [PMID: 30904952 PMCID: PMC11105391 DOI: 10.1007/s00018-019-03077-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/26/2019] [Accepted: 03/18/2019] [Indexed: 10/27/2022]
Abstract
The use of opioid analgesics is severely limited due to the development of intractable constipation, mediated through activation of mu opioid receptors (MOR) expressed by enteric neurons. The related delta opioid receptor (DOR) is an emerging therapeutic target for chronic pain, depression and anxiety. Whether DOR agonists also promote sustained inhibition of colonic transit is unknown. This study examined acute and chronic tolerance to SNC80 and ARM390, which were full and partial DOR agonists in neural pathways controlling colonic motility, respectively. Excitatory pathways developed acute and chronic tolerance to SNC80, whereas only chronic tolerance developed in inhibitory pathways. Both pathways remained functional after acute or chronic ARM390 exposure. Propagating colonic motor patterns were significantly reduced after acute or chronic SNC80 treatment, but not by ARM390 pre-treatment. These findings demonstrate that SNC80 has a prolonged inhibitory effect on propagating colonic motility. ARM390 had no effect on motor patterns and thus may have fewer gastrointestinal side-effects.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Benzamides/pharmacology
- Colon/drug effects
- Colon/physiology
- Drug Tolerance
- Electric Stimulation
- Mice
- Mice, Inbred C57BL
- Microscopy, Confocal
- Muscle Contraction/drug effects
- Neurons/metabolism
- Piperazines/pharmacology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Jesse J DiCello
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia.
| | - Ayame Saito
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Pradeep Rajasekhar
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Benjamin W Sebastian
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Rachel M McQuade
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Nicholas A Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Meritxell Canals
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Simona E Carbone
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Parkville, VIC, Australia.
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
18
|
Abstract
Whilst the nociceptin/orphanin FQ (N/OFQ) receptor (NOP) has similar intracellular coupling mechanisms to opioid receptors, it has distinct modulatory effects on physiological functions such as pain. These actions range from agonistic to antagonistic interactions with classical opioids within the spinal cord and brain, respectively. Understanding the electrophysiological actions of N/OFQ has been crucial in ascertaining the mechanisms by which these agonistic and antagonistic interactions occur. These similarities and differences between N/OFQ and opioids are due to the relative location of NOP versus opioid receptors on specific neuronal elements within these CNS regions. These mechanisms result in varied cellular actions including postsynaptic modulation of ion channels and presynaptic regulation of neurotransmitter release.
Collapse
|
19
|
Heblinski M, Bladen C, Connor M. Regulation of heterologously expressed 5-HT 1B receptors coupling to potassium channels in AtT-20 cells. Br J Pharmacol 2018; 176:451-465. [PMID: 30447001 DOI: 10.1111/bph.14547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/08/2018] [Accepted: 10/17/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE 5-HT1B receptors are widely expressed GPCRs and a target of triptans, the most commonly prescribed anti-migraine drugs. There is very limited information about the acute, agonist-induced regulation of 5-HT1B receptor signalling and so we sought to characterize this in a neuron-like system. EXPERIMENTAL APPROACH Epitope-tagged human 5-HT1B receptors were expressed in mouse AtT20 cells. 5-HT1B receptor signalling was assessed using whole-cell patch-clamp recordings of endogenous G protein-gated inwardly rectified potassium (GIRK) channels, and receptor localization measured using immunofluorescence. KEY RESULTS 5-HT (EC50 65 nM) and sumatriptan (EC50 165 nM) activated GIRK channels in AtT20 cells expressing 5-HT1B receptors. Continuous application of both 5-HT (EC50 120 nM) and sumatriptan (EC50 280 nM) produced profound desensitization of 5-HT1B receptor signalling within a few minutes. Complete recovery from desensitization was observed after 10 min. Both 5-HT and sumatriptan induced significant heterologous desensitization of SRIF (somatostatin)-activated GIRK currents, with the 5-HT-induced heterologous desensitization being blocked by the protein kinase inhibitor staurosporine. Both agonists induced modest 5-HT1B receptor internalization, with a time course much slower than receptor desensitization. CONCLUSIONS AND IMPLICATIONS In AtT-20 cells, 5-HT1B receptors undergo rapid and reversible desensitization at concentrations of agonist similar to those required to activate the receptor. Desensitization is incomplete, and the continued signalling of the receptor in the presence of the agonist may lead to cellular adaptations. Finally, 5-HT1B receptor activation causes significant heterologous desensitization, which may lead to a reduced effectiveness of unrelated drugs in vivo.
Collapse
Affiliation(s)
- Marika Heblinski
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Christopher Bladen
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mark Connor
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
20
|
Maiarù M, Leese C, Certo M, Echeverria-Altuna I, Mangione AS, Arsenault J, Davletov B, Hunt SP. Selective neuronal silencing using synthetic botulinum molecules alleviates chronic pain in mice. Sci Transl Med 2018; 10:10/450/eaar7384. [DOI: 10.1126/scitranslmed.aar7384] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/20/2018] [Accepted: 06/28/2018] [Indexed: 12/16/2022]
|
21
|
Orts-Del'Immagine A, Pugh JR. Activity-dependent plasticity of presynaptic GABA B receptors at parallel fiber synapses. Synapse 2018; 72:e22027. [PMID: 29360168 DOI: 10.1002/syn.22027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/05/2018] [Accepted: 01/21/2018] [Indexed: 01/10/2023]
Abstract
Parallel fiber synapses in the cerebellum express a wide range of presynaptic receptors. However, presynaptic receptor expression at individual parallel fiber synapses is quite heterogeneous, suggesting physiological mechanisms regulate presynaptic receptor expression. We investigated changes in presynaptic GABAB receptors at parallel fiber-stellate cell synapses in acute cerebellar slices from juvenile mice. GABAB receptor-mediated inhibition of excitatory postsynaptic currents (EPSCs) is remarkably diverse at these synapses, with transmitter release at some synapses inhibited by >50% and little or no inhibition at others. GABAB receptor-mediated inhibition was significantly reduced following 4 Hz parallel fiber stimulation but not after stimulation at other frequencies. The reduction in GABAB receptor-mediated inhibition was replicated by bath application of forskolin and blocked by application of a PKA inhibitor, suggesting activation of adenylyl cyclase and PKA are required. Immunolabeling for an extracellular domain of the GABAB2 subunit revealed reduced surface expression in the molecular layer after exposure to forskolin. GABAB receptor-mediated inhibition of action potential evoked calcium transients in parallel fiber varicosities was also reduced following bath application of forskolin, confirming presynaptic receptors are responsible for the reduced EPSC inhibition. These data demonstrate that presynaptic GABAB receptor expression can be a plastic property of synapses, which may compliment other forms of synaptic plasticity. This opens the door to novel forms of receptor plasticity previously confined primarily to postsynaptic receptors.
Collapse
Affiliation(s)
- Adeline Orts-Del'Immagine
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| | - Jason R Pugh
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229
| |
Collapse
|
22
|
Levitt ES, Williams JT. Desensitization and Tolerance of Mu Opioid Receptors on Pontine Kölliker-Fuse Neurons. Mol Pharmacol 2018; 93:8-13. [PMID: 29097440 PMCID: PMC5708089 DOI: 10.1124/mol.117.109603] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
Acute desensitization of mu opioid receptors is thought to be an initial step in the development of tolerance to opioids. Given the resistance of the respiratory system to develop tolerance, desensitization of neurons in the Kölliker-Fuse (KF), a key area in the respiratory circuit, was examined. The activation of G protein-coupled inwardly rectifying potassium current was measured using whole-cell voltage-clamp recordings from KF and locus coeruleus (LC) neurons contained in acute rat brain slices. A saturating concentration of the opioid agonist [Met5]-enkephalin (ME) caused significantly less desensitization in KF neurons compared with LC neurons. In contrast to LC, desensitization in KF neurons was not enhanced by activation of protein kinase C or in slices from morphine-treated rats. Cellular tolerance to ME and morphine was also lacking in KF neurons from morphine-treated rats. The lack of cellular tolerance in KF neurons correlates with the relative lack of tolerance to the respiratory depressant effect of opioids.
Collapse
Affiliation(s)
- Erica S Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida (E.S.L.) and Vollum Institute, Oregon Health and Science University, Portland, Oregon (J.T.W.)
| | - John T Williams
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida (E.S.L.) and Vollum Institute, Oregon Health and Science University, Portland, Oregon (J.T.W.)
| |
Collapse
|
23
|
Differential Desensitization Observed at Multiple Effectors of Somatic μ-Opioid Receptors Underlies Sustained Agonist-Mediated Inhibition of Proopiomelanocortin Neuron Activity. J Neurosci 2017; 37:8667-8677. [PMID: 28821664 DOI: 10.1523/jneurosci.1030-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/06/2017] [Accepted: 07/31/2017] [Indexed: 01/17/2023] Open
Abstract
Activation of somatic μ-opioid receptors (MORs) in hypothalamic proopiomelanocortin (POMC) neurons leads to the activation of G-protein-coupled inward rectifier potassium (GIRK) channels and hyperpolarization, but in response to continued signaling MORs undergo acute desensitization resulting in robust reduction in the peak GIRK current after minutes of agonist exposure. We hypothesized that the attenuation of the GIRK current would lead to a recovery of neuronal excitability whereby desensitization of the receptor would lead to a new steady state of POMC neuron activity reflecting the sustained GIRK current observed after the initial decline from peak with continued agonist exposure. However, electrophysiologic recordings and GCaMP6f Ca2+ imaging in POMC neurons in mouse brain slices indicate that maximal inhibition of cellular activity by these measures can be maintained after the GIRK current declines. Blockade of the GIRK current by Ba2+ or Tertiapin-Q did not disrupt the sustained inhibition of Ca2+ transients in the continued presence of agonist, indicating the activation of an effector other than GIRK channels. Use of an irreversible MOR antagonist and Furchgott analysis revealed a low receptor reserve for the activation of GIRK channels but a >90% receptor reserve for the inhibition of Ca2+ events. Altogether, the data show that somatodendritic MORs in POMC neurons inhibit neuronal activity through at least two effectors with distinct levels of receptor reserve and that differentially reflect receptor desensitization. Thus, in POMC cells, the decline in the GIRK current during prolonged MOR agonist exposure does not reflect an increase in cellular activity as expected.SIGNIFICANCE STATEMENT Desensitization of the μ-opioid receptor (MOR) is thought to underlie the development of cellular tolerance to opiate therapy. The present studies focused on MOR desensitization in hypothalamic proopiomelanocortin (POMC) neurons as these neurons produce the endogenous opioid β-endorphin and are heavily regulated by opioids. Prolonged activation of somatic MORs in POMC neurons robustly inhibited action potential firing and Ca2+ activity despite desensitization of the MOR and reduced activation of a potassium current over the same time course. The data show that somatic MORs in POMC neurons couple to multiple effectors that have differential sensitivity to desensitization of the receptor. Thus, in these cells, the cellular consequence of MOR desensitization cannot be defined by the activity of a single effector system.
Collapse
|
24
|
Kallupi M, Scuppa G, de Guglielmo G, Calò G, Weiss F, Statnick MA, Rorick-Kehn LM, Ciccocioppo R. Genetic Deletion of the Nociceptin/Orphanin FQ Receptor in the Rat Confers Resilience to the Development of Drug Addiction. Neuropsychopharmacology 2017; 42:695-706. [PMID: 27562376 PMCID: PMC5240182 DOI: 10.1038/npp.2016.171] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 12/14/2022]
Abstract
The nociceptin (NOP) receptor is a G-protein-coupled receptor whose natural ligand is the NOP/orphanin FQ (N/OFQ) peptide. Evidence from pharmacological studies suggests that the N/OFQ system is implicated in the regulation of several addiction-related phenomena, such as drug intake, withdrawal, and relapse. Here, to further explore the role of NOP system in addiction, we used NOP (-/-) rats to study the motivation for cocaine, heroin, and alcohol self-administration in the absence of N/OFQ function. Conditioned place preference (CPP) and saccharin (0.2% w/v) self-administration were also investigated. Results showed that NOP (-/-) rats self-administer less cocaine (0.25, 0.125, or 0.5 mg/infusion) both under a fixed ratio 1 and a progressive ratio schedule of reinforcement compared with wild-type (Wt) controls. Consistently, cocaine (10 mg/kg, i.p.) was able to induce CPP in Wt but not in NOP (-/-). When NOP (-/-) rats were tested for heroin (20 μg/infusion) and ethanol (10% v/v) self-administration, they showed significantly lower drug intake compared with Wt. Conversely, saccharin self-administration was not affected by NOP deletion, excluding the possibility of nonspecific learning deficits or generalized disruption of reward mechanisms in NOP (-/-) rats. These findings were confirmed with pharmacological experiments using two selective NOP antagonists, SB-612111 and LY2817412. Both drugs attenuated alcohol self-administration in Wt rats but not in NOP (-/-) rats. In conclusion, our results demonstrate that genetic deletion of NOP receptors confers resilience to drug abuse and support a role for NOP receptor antagonism as a potential treatment option for drug addiction.
Collapse
Affiliation(s)
- Marsida Kallupi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Giulia Scuppa
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Girolamo Calò
- Department of Medical Science, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Friedbert Weiss
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA
| | - Michael A Statnick
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN USA
| | | | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy,School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032 Italy, Tel: +39 07 3740 3313, Fax: +39 07 3740 3325, E-mail:
| |
Collapse
|
25
|
Howell RD, Pugh JR. Biphasic modulation of parallel fibre synaptic transmission by co-activation of presynaptic GABAA and GABAB receptors in mice. J Physiol 2016; 594:3651-66. [PMID: 27061582 DOI: 10.1113/jp272124] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/30/2016] [Indexed: 01/23/2023] Open
Abstract
KEY POINTS Many excitatory synapses co-express presynaptic GABAA and GABAB receptors, despite their opposing actions on synaptic transmission. It is still unclear how co-activation of these receptors modulates synapse function. We measured presynaptic GABA receptor function at parallel fibre synapses onto stellate cells in the cerebellum using whole-cell patch-clamp recording and photolytic uncaging of RuBi-GABA. Activation of presynaptic GABA receptors results in a transient (∼100 ms) enhancement of synaptic transmission (mediated by GABAA receptors) followed by a long lasting (>500 ms) inhibition of transmission (mediated by GABAB receptors). When activated just prior to high-frequency trains of stimulation, presynaptic GABAA and GABAB receptors work together to reduce short-term facilitation/enhance depression, altering the filtering properties of synaptic transmission. Inhibition of synaptic transmission by GABAB receptors is more sensitive to GABA than enhancement by GABAA receptors, suggesting GABAB receptors may be activated by ambient GABA or release from greater distances. ABSTRACT GABAA and GABAB receptors are co-expressed at many presynaptic terminals in the central nervous system. Previous studies have shown that GABAA receptors typically enhance vesicle release while GABAB receptors inhibit release. However, it is not clear how the competing actions of these receptors modulate synaptic transmission when co-activated, as is likely in vivo. We investigated this question at parallel fibre synapses in the cerebellum, which co-express presynaptic GABAA and GABAB receptors. In acute slices from C57BL/6 mice, we find that co-activation of presynaptic GABA receptors by photolytic uncaging of RuBi-GABA has a biphasic effect on EPSC amplitudes recorded from stellate cells. Synchronous and asynchronous EPSCs evoked within ∼100 ms of GABA uncaging were increased, while EPSCs evoked ∼300-600 ms after GABA uncaging were reduced compared to interleaved control sweeps. We confirmed these effects are presynaptic by measuring the paired-pulse ratio, variance of EPSC amplitudes, and response probability. During trains of high-frequency stimulation GABAA and GABAB receptors work together (rather than oppose one another) to reduce short-term facilitation when GABA is uncaged just prior to the onset of stimulation. We also find that GABAB receptor-mediated inhibition can be elicited by lower GABA concentrations than GABAA receptor-mediated enhancement of EPSCs, suggesting GABAB receptors may be selectively activated by ambient GABA or release from more distance synapses. These data suggest that GABA, acting through both presynaptic GABAA and GABAB receptors, modulate the amplitude and short-term plasticity of excitatory synapses, a result not possible from activation of either receptor type alone.
Collapse
Affiliation(s)
- Rebecca D Howell
- University of Texas Health Science Center at San Antonio, Department of Physiology, San Antonio, TX 78229, USA
| | - Jason R Pugh
- University of Texas Health Science Center at San Antonio, Department of Physiology, San Antonio, TX 78229, USA
| |
Collapse
|
26
|
Toll L, Bruchas MR, Calo' G, Cox BM, Zaveri NT. Nociceptin/Orphanin FQ Receptor Structure, Signaling, Ligands, Functions, and Interactions with Opioid Systems. Pharmacol Rev 2016; 68:419-57. [PMID: 26956246 PMCID: PMC4813427 DOI: 10.1124/pr.114.009209] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The NOP receptor (nociceptin/orphanin FQ opioid peptide receptor) is the most recently discovered member of the opioid receptor family and, together with its endogenous ligand, N/OFQ, make up the fourth members of the opioid receptor and opioid peptide family. Because of its more recent discovery, an understanding of the cellular and behavioral actions induced by NOP receptor activation are less well developed than for the other members of the opioid receptor family. All of these factors are important because NOP receptor activation has a clear modulatory role on mu opioid receptor-mediated actions and thereby affects opioid analgesia, tolerance development, and reward. In addition to opioid modulatory actions, NOP receptor activation has important effects on motor function and other physiologic processes. This review discusses how NOP pharmacology intersects, contrasts, and interacts with the mu opioid receptor in terms of tertiary structure and mechanism of receptor activation; location of receptors in the central nervous system; mechanisms of desensitization and downregulation; cellular actions; intracellular signal transduction pathways; and behavioral actions with respect to analgesia, tolerance, dependence, and reward. This is followed by a discussion of the agonists and antagonists that have most contributed to our current knowledge. Because NOP receptors are highly expressed in brain and spinal cord and NOP receptor activation sometimes synergizes with mu receptor-mediated actions and sometimes opposes them, an understanding of NOP receptor pharmacology in the context of these interactions with the opioid receptors will be crucial to the development of novel therapeutics that engage the NOP receptor.
Collapse
Affiliation(s)
- Lawrence Toll
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Michael R Bruchas
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Girolamo Calo'
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Brian M Cox
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| | - Nurulain T Zaveri
- Torrey Pines Institute for Molecular Studies, Port St. Lucie, Florida (L.T.); Departments of Anesthesiology, and Neuroscience, Washington University School of Medicine, St. Louis, Missouri (M.R.B.); Section of Pharmacology, Department of Medical Science, and National Institute of Neurosciences, University of Ferrara, Ferrara, Italy (G.C.); Professor of Pharmacology & Neuroscience, Uniformed Services University, Bethesda, Maryland (B.M.C.); and Astraea Therapeutics, LLC, Mountain View, California (N.T.Z.)
| |
Collapse
|
27
|
Pennock RL, Hentges ST. Desensitization-resistant and -sensitive GPCR-mediated inhibition of GABA release occurs by Ca2+-dependent and -independent mechanisms at a hypothalamic synapse. J Neurophysiol 2016; 115:2376-88. [PMID: 26912590 DOI: 10.1152/jn.00535.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 02/19/2016] [Indexed: 11/22/2022] Open
Abstract
Whereas the activation of Gαi/o-coupled receptors commonly results in postsynaptic responses that show acute desensitization, the presynaptic inhibition of transmitter release caused by many Gαi/o-coupled receptors is maintained during agonist exposure. However, an exception has been noted where GABAB receptor (GABABR)-mediated inhibition of inhibitory postsynaptic currents (IPSCs) recorded in mouse proopiomelanocortin (POMC) neurons exhibit acute desensitization in ∼25% of experiments. To determine whether differential effector coupling confers sensitivity to desensitization, voltage-clamp recordings were made from POMC neurons to compare the mechanism by which μ-opioid receptors (MORs) and GABABRs inhibit transmitter release. Neither MOR- nor GABABR-mediated inhibition of release relied on the activation of presynaptic K(+) channels. Both receptors maintained the ability to inhibit release in the absence of external Ca(2+) or in the presence of ionomycin-induced Ca(2+) influx, indicating that inhibition of release can occur through a Ca(2+)-independent mechanism. Replacing Ca(2+) with Sr(2+) to disrupt G-protein-mediated inhibition of release occurring directly at the release machinery did not alter MOR- or GABAB -mediated inhibition of IPSCs, suggesting that reductions in evoked release can occur through the inhibition of Ca(2+) channels. Additionally, both receptors inhibited evoked IPSCs in the presence of selective blockers of N- or P/Q-type Ca(2+) channels. Altogether, the results show that MORs and GABABRs can inhibit transmitter release through the inhibition of calcium influx and by direct actions at the release machinery. Furthermore, since both the desensitizing and nondesensitizing presynaptic receptors are similarly coupled, differential effector coupling is unlikely responsible for differential desensitization of the inhibition of release.
Collapse
Affiliation(s)
- Reagan L Pennock
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
28
|
Next generation effects of female adolescent morphine exposure: sex-specific alterations in response to acute morphine emerge before puberty. Behav Pharmacol 2014; 25:173-81. [PMID: 24561499 DOI: 10.1097/fbp.0000000000000032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Prescription opiate use by adolescent girls has increased significantly in the past decade. Preclinical studies using rats report alterations in morphine sensitivity in the adult offspring of adolescent morphine-exposed females (MOR-F1) when compared with the offspring of adolescent saline-exposed females (SAL-F1). To begin to elucidate the development of these next generation modifications, the present study examined the effects of acute morphine administration on sedation and corticosterone secretion in prepubescent SAL-F1 and MOR-F1 male and female rats. In addition, alterations in proopiomelanocortin (POMC) gene expression in the arcuate nucleus, as well as in tyrosine hydroxylase (TH) and μ-opioid receptor (OPRM1) gene expressions in the ventral tegmental area, were analyzed using quantitative PCR, to determine whether differential regulation of these genes was correlated with the observed behavioral and/or endocrine effects. Increased morphine-induced sedation, coupled with an attenuation of morphine-induced corticosterone secretion, was observed in MOR-F1 males. Significant alterations in both POMC and OPRM1 gene expressions were also observed in MOR-F1 males, with no change in TH mRNA expression. Overall, these data suggest that the transgenerational effects of adolescent morphine exposure can be discerned before pubertal development and are more pronounced in males, and suggest dysregulation of the hypothalamic-pituitary-adrenal axis in the offspring of adolescent morphine-exposed females.
Collapse
|
29
|
Henderson G. The μ-opioid receptor: an electrophysiologist's perspective from the sharp end. Br J Pharmacol 2014; 172:260-7. [PMID: 24640948 DOI: 10.1111/bph.12633] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/02/2013] [Accepted: 12/10/2013] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Morphine, the prototypical opioid analgesic drug, produces its behavioural effects primarily through activation of μ-opioid receptors expressed in neurones of the central and peripheral nervous systems. This perspective provides a historical view of how, over the past 40 years, the use of electrophysiological recording techniques has helped to reveal the molecular mechanisms by which acute and chronic activation of μ-opioid receptors by morphine and other opioid drugs modify neuronal function. 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)
- Graeme Henderson
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| |
Collapse
|
30
|
Connor M, Bagley EE, Chieng BC, Christie MJ. β-Arrestin-2 knockout prevents development of cellular μ-opioid receptor tolerance but does not affect opioid-withdrawal-related adaptations in single PAG neurons. Br J Pharmacol 2014; 172:492-500. [PMID: 24597632 DOI: 10.1111/bph.12673] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/24/2014] [Accepted: 03/01/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Tolerance to the behavioural effects of morphine is blunted in β-arrestin-2 knockout mice, but opioid withdrawal is largely unaffected. The cellular mechanisms of tolerance have been studied in some neurons from β-arrestin-2 knockouts, but tolerance and withdrawal mechanisms have not been examined at the cellular level in periaqueductal grey (PAG) neurons, which are crucial for central tolerance and withdrawal phenomena. EXPERIMENTAL APPROACH μ-Opioid receptor (MOPr) inhibition of voltage-gated calcium channel currents (ICa ) was examined by patch-clamp recordings from acutely dissociated PAG neurons from wild-type and β-arrestin-2 knockout mice treated chronically with morphine (CMT) or vehicle. Opioid withdrawal-induced activation of GABA transporter type 1 (GAT-1) currents was determined using perforated patch recordings from PAG neurons in brain slices. KEY RESULTS MOPr inhibition of ICa in PAG neurons was unaffected by β-arrestin-2 deletion. CMT impaired coupling of MOPrs to ICa in PAG neurons from wild-type mice, but this cellular tolerance was not observed in neurons from CMT β-arrestin-2 knockouts. However, β-arrestin-2 knockouts displayed similar opioid-withdrawal-induced activation of GAT-1 currents as wild-type PAG neurons. CONCLUSIONS AND IMPLICATIONS In β-arrestin-2 knockout mice, the central neurons involved in the anti-nociceptive actions of opioids also fail to develop cellular tolerance to opioids following chronic morphine. The results also provide the first cellular physiological evidence that opioid withdrawal is not disrupted by β-arrestin-2 deletion. However, the unaffected basal sensitivity to opioids in PAG neurons provides further evidence that changes in basal MOPr sensitivity cannot account for the enhanced acute nociceptive response to morphine reported in β-arrestin-2 knockouts. 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)
- M Connor
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
31
|
Lowe JD, Bailey CP. Functional selectivity and time-dependence of μ-opioid receptor desensitization at nerve terminals in the mouse ventral tegmental area. Br J Pharmacol 2014; 172:469-81. [PMID: 24467517 PMCID: PMC4292961 DOI: 10.1111/bph.12605] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 01/08/2014] [Accepted: 01/17/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE The majority of studies examining desensitization of the μ-opioid receptor (MOR) have examined those located at cell bodies. However, MORs are extensively expressed at nerve terminals throughout the mammalian nervous system. This study is designed to investigate agonist-induced MOR desensitization at nerve terminals in the mouse ventral tegmental area (VTA). EXPERIMENTAL APPROACH MOR function was measured in mature mouse brain slices containing the VTA using whole-cell patch-clamp electrophysiology. Presynaptic MOR function was isolated from postsynaptic function and the functional selectivity, time-dependence and mechanisms of agonist-induced MOR desensitization were examined. KEY RESULTS MORs located at GABAergic nerve terminals in the VTA were completely resistant to rapid desensitization induced by the high-efficacy agonists DAMGO and Met-enkephalin. MORs located postsynaptically on GABAergic cell bodies readily underwent rapid desensitization in response to DAMGO. However, after prolonged (>7 h) treatment with Met-enkephalin, profound homologous MOR desensitization was observed. Morphine could induce rapid MOR desensitization at nerve terminals when PKC was activated. CONCLUSIONS AND IMPLICATIONS Agonist-induced MOR desensitization in GABAergic neurons in the VTA is compartment-selective as well as agonist-selective. When MORs are located at cell bodies, higher-efficacy agonists induce greater levels of rapid desensitization than lower-efficacy agonists. However, the converse is true at nerve terminals where agonists that induce MOR desensitization via PKC are capable of rapid agonist-induced desensitization while higher-efficacy agonists are not. MOR desensitization induced by higher-efficacy agonists at nerve terminals only takes place after prolonged receptor activation. 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)
- J D Lowe
- Department of Pharmacy & Pharmacology, University of Bath, Bath, UK; School of Physiology & Pharmacology, University of Bristol, Bristol, UK
| | | |
Collapse
|
32
|
Matsui A, Jarvie BC, Robinson BG, Hentges ST, Williams JT. Separate GABA afferents to dopamine neurons mediate acute action of opioids, development of tolerance, and expression of withdrawal. Neuron 2014; 82:1346-56. [PMID: 24857021 DOI: 10.1016/j.neuron.2014.04.030] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2014] [Indexed: 01/01/2023]
Abstract
GABA release from interneurons in VTA, projections from the nucleus accumbens (NAc), and rostromedial tegmental nucleus (RMTg) was selectively activated in rat brain slices. The inhibition induced by μ-opioid agonists was pathway dependent. Morphine induced a 46% inhibition of IPSCs evoked from the RMTg, 18% from NAc, and IPSCs evoked from VTA interneurons were almost insensitive (11% inhibition). In vivo morphine treatment resulted in tolerance to the inhibition of RMTg, but not local interneurons or NAc, inputs. One common sign of opioid withdrawal is an increase in adenosine-dependent inhibition. IPSCs evoked from the NAc were potently inhibited by activation of presynaptic adenosine receptors, whereas IPSCs evoked from RMTg were not changed. Blockade of adenosine receptors selectively increased IPSCs evoked from the NAc during morphine withdrawal. Thus, the acute action of opioids, the development of tolerance, and the expression of withdrawal are mediated by separate GABA afferents to dopamine neurons.
Collapse
Affiliation(s)
- Aya Matsui
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Brooke C Jarvie
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Brooks G Robinson
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - John T Williams
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
33
|
Abstract
This paper is the thirty-fifth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2012 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
34
|
Wageman CR, Marks MJ, Grady SR. Effectiveness of nicotinic agonists as desensitizers at presynaptic α4β2- and α4α5β2-nicotinic acetylcholine receptors. Nicotine Tob Res 2013; 16:297-305. [PMID: 24052501 DOI: 10.1093/ntr/ntt146] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Nicotine interacts with nicotinic acetylcholine receptors (nAChRs) and modifies neuronal functions. The net result of nicotine exposure is difficult to assess because multiple nAChR subtypes exist and are expressed on multiple classes of neurons. Nicotine, unlike the natural agonist acetylcholine, remains in tissues for hours, and during this extended exposure nAChRs desensitize. Therefore, agonists can block the natural functions of nAChRs. Higher nicotine concentrations are required to desensitize α4β2-nAChRs containing the α5 subunit. The aim of these experiments was to determine if this property holds true for compounds other than nicotine. METHODS [(3)H]-dopamine release from crude mouse striatal synaptosomal preparations was used to measure activation and desensitization of the [(α4β2)2β2] and [(α4β2)2α5] nAChR subtypes. Affinity was measured by competition with [(125)I]-epibatidine. RESULTS Nine compounds of varying affinity and efficacy were tested. All compounds partially desensitized both subtypes; concentration necessary for desensitization correlated with binding site affinity but not efficacy. All compounds showed a similar, significant shift in concentration necessary for a 50% effect when the α5 subunit was included (averaging 8-fold higher). The extent of desensitization produced by a 10-min exposure did not correlate with affinity or efficacy of compound. CONCLUSION Full or partial nicotinic agonists used as medications may effectively desensitize α4β2-nAChRs. However, significantly higher concentrations of all compounds tested were required to elicit desensitization of α4α5β2-nAChRs than α4β2-nAChRs. If desensitization is the important property for a smoking cessation drug, basic screening at both subtypes may provide a mechanistic foundation for effectiveness.
Collapse
Affiliation(s)
- Charles R Wageman
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO
| | | | | |
Collapse
|
35
|
Liu J, Ren Y, Li G, Liu ZL, Liu R, Tong Y, Zhang L, Yang K. GABAB receptors resist acute desensitization in both postsynaptic and presynaptic compartments of periaqueductal gray neurons. Neurosci Lett 2013; 543:146-51. [DOI: 10.1016/j.neulet.2013.03.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 03/17/2013] [Accepted: 03/18/2013] [Indexed: 10/27/2022]
|
36
|
Mercer AJ, Hentges ST, Meshul CK, Low MJ. Unraveling the central proopiomelanocortin neural circuits. Front Neurosci 2013; 7:19. [PMID: 23440036 PMCID: PMC3579188 DOI: 10.3389/fnins.2013.00019] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 02/04/2013] [Indexed: 11/16/2022] Open
Abstract
Central proopiomelanocortin (POMC) neurons form a potent anorexigenic network, but our understanding of the integration of this hypothalamic circuit throughout the central nervous system (CNS) remains incomplete. POMC neurons extend projections along the rostrocaudal axis of the brain, and can signal with both POMC-derived peptides and fast amino acid neurotransmitters. Although recent experimental advances in circuit-level manipulation have been applied to POMC neurons, many pivotal questions still remain: how and where do POMC neurons integrate metabolic information? Under what conditions do POMC neurons release bioactive molecules throughout the CNS? Are GABA and glutamate or neuropeptides released from POMC neurons more crucial for modulating feeding and metabolism? Resolving the exact stoichiometry of signals evoked from POMC neurons under different metabolic conditions therefore remains an ongoing endeavor. In this review, we analyze the anatomical atlas of this network juxtaposed to the physiological signaling of POMC neurons both in vitro and in vivo. We also consider novel genetic tools to further characterize the function of the POMC circuit in vivo. Our goal is to synthesize a global view of the POMC network, and to highlight gaps that require further research to expand our knowledge on how these neurons modulate energy balance.
Collapse
Affiliation(s)
- Aaron J Mercer
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, MI, USA
| | | | | | | |
Collapse
|
37
|
Abstract
Neuropeptides are found in many mammalian CNS neurons where they play key roles in modulating neuronal activity. In contrast to amino acid transmitter release at the synapse, neuropeptide release is not restricted to the synaptic specialization, and after release, a neuropeptide may diffuse some distance to exert its action through a G protein-coupled receptor. Some neuropeptides such as hypocretin/orexin are synthesized only in single regions of the brain, and the neurons releasing these peptides probably have similar functional roles. Other peptides such as neuropeptide Y (NPY) are synthesized throughout the brain, and neurons that synthesize the peptide in one region have no anatomical or functional connection with NPY neurons in other brain regions. Here, I review converging data revealing a complex interaction between slow-acting neuromodulator peptides and fast-acting amino acid transmitters in the control of energy homeostasis, drug addiction, mood and motivation, sleep-wake states, and neuroendocrine regulation.
Collapse
|
38
|
von Zastrow M, Williams JT. Modulating neuromodulation by receptor membrane traffic in the endocytic pathway. Neuron 2012; 76:22-32. [PMID: 23040804 DOI: 10.1016/j.neuron.2012.09.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cellular responsiveness to many neuromodulators is controlled by endocytosis of the transmembrane receptors that transduce their effects. Endocytic membrane trafficking of particular neuromodulator receptors exhibits remarkable diversity and specificity, determined largely by molecular sorting operations that guide receptors at trafficking branchpoints after endocytosis. In this Review, we discuss recent progress in elucidating mechanisms mediating the molecular sorting of neuromodulator receptors in the endocytic pathway. There is emerging evidence that endocytic trafficking of neuromodulator receptors, in addition to influencing longer-term cellular responsiveness under conditions of prolonged or repeated activation, may also affect the acute response. Physiological and pathological consequences of defined receptor trafficking events are only now being elucidated, but it is already apparent that endocytosis of neuromodulator receptors has a significant impact on the actions of therapeutic drugs. The present data also suggest, conversely, that mechanisms of receptor endocytosis and molecular sorting may themselves represent promising targets for therapeutic manipulation.
Collapse
Affiliation(s)
- Mark von Zastrow
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA 94158, USA.
| | | |
Collapse
|
39
|
Seddik R, Jungblut SP, Silander OK, Rajalu M, Fritzius T, Besseyrias V, Jacquier V, Fakler B, Gassmann M, Bettler B. Opposite effects of KCTD subunit domains on GABA(B) receptor-mediated desensitization. J Biol Chem 2012; 287:39869-77. [PMID: 23035119 DOI: 10.1074/jbc.m112.412767] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
GABA(B) receptors assemble from principle and auxiliary subunits. The principle subunits GABA(B1) and GABA(B2) form functional heteromeric GABA(B(1,2)) receptors that associate with homotetramers of auxiliary KCTD8, -12, -12b, or -16 (named after their K(+) channel tetramerization domain) subunits. These auxiliary subunits constitute receptor subtypes with distinct functional properties. KCTD12 and -12b generate desensitizing receptor responses while KCTD8 and -16 generate largely non-desensitizing receptor responses. The structural elements of the KCTDs underlying these differences in desensitization are unknown. KCTDs are modular proteins comprising a T1 tetramerization domain, which binds to GABA(B2), and a H1 homology domain. KCTD8 and -16 contain an additional C-terminal H2 homology domain that is not sequence-related to the H1 domains. No functions are known for the H1 and H2 domains. Here we addressed which domains and sequence motifs in KCTD proteins regulate desensitization of the receptor response. We found that the H1 domains in KCTD12 and -12b mediate desensitization through a particular sequence motif, T/NFLEQ, which is not present in the H1 domains of KCTD8 and -16. In addition, the H2 domains in KCTD8 and -16 inhibit desensitization when expressed C-terminal to the H1 domains but not when expressed as a separate protein in trans. Intriguingly, the inhibitory effect of the H2 domain is sequence-independent, suggesting that the H2 domain sterically hinders desensitization by the H1 domain. Evolutionary analysis supports that KCTD12 and -12b evolved desensitizing properties by liberating their H1 domains from antagonistic H2 domains and acquisition of the T/NFLEQ motif.
Collapse
Affiliation(s)
- Riad Seddik
- Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Levitt ES, Williams JT. Morphine desensitization and cellular tolerance are distinguished in rat locus ceruleus neurons. Mol Pharmacol 2012; 82:983-92. [PMID: 22914548 DOI: 10.1124/mol.112.081547] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
μ-Opioid receptor desensitization is considered an initial step in the development of tolerance. Curiously, the commonly used opioid morphine produces robust tolerance but minimal acute desensitization. This study was designed to test the hypothesis that desensitization is indeed present in morphine-treated animals and is distinguished from cellular tolerance by time course of recovery and mechanism. To induce tolerance, rats were treated with continuously released morphine for 1 week. Morphine-mediated activation of G protein-coupled inwardly rectifying potassium conductance was measured using voltage-clamp recordings from locus ceruleus neurons in brain slices from naive or morphine-treated rats. Cellular tolerance was observed as a decrease in morphine efficacy in slices from morphine-treated rats. This tolerance persisted for at least 6 h. An additional reduction in morphine-mediated current was observed when slices from morphine-treated rats were continuously maintained in morphine at approximately the circulating plasma concentration. This additional reduction recovered within 1 h after removal of morphine from the slice and represents desensitization that developed in the tolerant animal. Recovery from desensitization, but not long-lasting tolerance, was facilitated by protein phosphatase 1 (PP1) activity. Furthermore, desensitization, but not tolerance, was reversed by protein kinase C (PKC) inhibitor but not by an inhibitor of c-Jun N-terminal kinase. Therefore, morphine treatment leads to both long-lasting cellular tolerance and readily reversible desensitization, which are differentially dependent on PP1 and PKC activity and combine to result in a substantial decrease in morphine effectiveness. This PKC-mediated desensitization may contribute to the previously reported PKC-dependent reversal of behavioral tolerance.
Collapse
Affiliation(s)
- Erica S Levitt
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | | |
Collapse
|