Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.127
Filter
Add more filters

Publication year range
1.
Cell ; 186(2): 413-427.e17, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36638794

ABSTRACT

Opioids are effective analgesics, but their use is beset by serious side effects, including addiction and respiratory depression, which contribute to the ongoing opioid crisis. The human opioid system contains four opioid receptors (µOR, δOR, κOR, and NOPR) and a set of related endogenous opioid peptides (EOPs), which show distinct selectivity toward their respective opioid receptors (ORs). Despite being key to the development of safer analgesics, the mechanisms of molecular recognition and selectivity of EOPs to ORs remain unclear. Here, we systematically characterize the binding of EOPs to ORs and present five structures of EOP-OR-Gi complexes, including ß-endorphin- and endomorphin-bound µOR, deltorphin-bound δOR, dynorphin-bound κOR, and nociceptin-bound NOPR. These structures, supported by biochemical results, uncover the specific recognition and selectivity of opioid peptides and the conserved mechanism of opioid receptor activation. These results provide a structural framework to facilitate rational design of safer opioid drugs for pain relief.


Subject(s)
Receptors, Opioid , Humans , Analgesics, Opioid/pharmacology , Opioid Peptides , Receptors, Opioid, mu/metabolism , Receptors, Opioid/chemistry
2.
Cell ; 186(3): 591-606.e23, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36669483

ABSTRACT

Dysregulation of the immune system is a cardinal feature of opioid addiction. Here, we characterize the landscape of peripheral immune cells from patients with opioid use disorder and from healthy controls. Opioid-associated blood exhibited an abnormal distribution of immune cells characterized by a significant expansion of fragile-like regulatory T cells (Tregs), which was positively correlated with the withdrawal score. Analogously, opioid-treated mice also showed enhanced Treg-derived interferon-γ (IFN-γ) expression. IFN-γ signaling reshaped synaptic morphology in nucleus accumbens (NAc) neurons, modulating subsequent withdrawal symptoms. We demonstrate that opioids increase the expression of neuron-derived C-C motif chemokine ligand 2 (Ccl2) and disrupted blood-brain barrier (BBB) integrity through the downregulation of astrocyte-derived fatty-acid-binding protein 7 (Fabp7), which both triggered peripheral Treg infiltration into NAc. Our study demonstrates that opioids drive the expansion of fragile-like Tregs and favor peripheral Treg diapedesis across the BBB, which leads to IFN-γ-mediated synaptic instability and subsequent withdrawal symptoms.


Subject(s)
Interferon-gamma , Opioid-Related Disorders , Substance Withdrawal Syndrome , T-Lymphocytes, Regulatory , Animals , Mice , Analgesics, Opioid/administration & dosage , Interferon-gamma/metabolism , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/pathology
3.
Cell ; 185(23): 4361-4375.e19, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36368306

ABSTRACT

Morphine and fentanyl are among the most used opioid drugs that confer analgesia and unwanted side effects through both G protein and arrestin signaling pathways of µ-opioid receptor (µOR). Here, we report structures of the human µOR-G protein complexes bound to morphine and fentanyl, which uncover key differences in how they bind the receptor. We also report structures of µOR bound to TRV130, PZM21, and SR17018, which reveal preferential interactions of these agonists with TM3 side of the ligand-binding pocket rather than TM6/7 side. In contrast, morphine and fentanyl form dual interactions with both TM3 and TM6/7 regions. Mutations at the TM6/7 interface abolish arrestin recruitment of µOR promoted by morphine and fentanyl. Ligands designed to reduce TM6/7 interactions display preferential G protein signaling. Our results provide crucial insights into fentanyl recognition and signaling of µOR, which may facilitate rational design of next-generation analgesics.


Subject(s)
Fentanyl , Morphine , Humans , Analgesics, Opioid/pharmacology , Arrestin/metabolism , Fentanyl/pharmacology , GTP-Binding Proteins/metabolism , Morphine/pharmacology , Receptors, Opioid, mu
4.
Annu Rev Biochem ; 90: 739-761, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33756098

ABSTRACT

Opioids such as morphine and oxycodone are analgesics frequently prescribed for the treatment of moderate or severe pain. Unfortunately, these medications are associated with exceptionally high abuse potentials and often cause fatal side effects, mainly through the µ-opioid receptor (MOR). Efforts to discover novel, safer, and more efficacious analgesics targeting MOR have encountered challenges. In this review, we summarize alternative strategies and targets that could be used to develop safer nonopioid analgesics. A molecular understanding of G protein-coupled receptor activation and signaling has illuminated not only the complexities of receptor pharmacology but also the potential for pathway-selective agonists and allosteric modulators as safer medications. The availability of structures of pain-related receptors, in combination with high-throughput computational tools, has accelerated the discovery of multitarget ligands with promising pharmacological profiles. Emerging clinical evidence also supports the notion that drugs targeting peripheral opioid receptors have potential as improved analgesic agents.


Subject(s)
Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Receptors, Opioid/chemistry , Receptors, Opioid/metabolism , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , Drug Discovery , High-Throughput Screening Assays/methods , Humans , Ligands , Receptors, G-Protein-Coupled/metabolism
5.
Cell ; 178(3): 653-671.e19, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348890

ABSTRACT

Nociceptin and its receptor are widely distributed throughout the brain in regions associated with reward behavior, yet how and when they act is unknown. Here, we dissected the role of a nociceptin peptide circuit in reward seeking. We generated a prepronociceptin (Pnoc)-Cre mouse line that revealed a unique subpopulation of paranigral ventral tegmental area (pnVTA) neurons enriched in prepronociceptin. Fiber photometry recordings during progressive ratio operant behavior revealed pnVTAPnoc neurons become most active when mice stop seeking natural rewards. Selective pnVTAPnoc neuron ablation, inhibition, and conditional VTA nociceptin receptor (NOPR) deletion increased operant responding, revealing that the pnVTAPnoc nucleus and VTA NOPR signaling are necessary for regulating reward motivation. Additionally, optogenetic and chemogenetic activation of this pnVTAPnoc nucleus caused avoidance and decreased motivation for rewards. These findings provide insight into neuromodulatory circuits that regulate motivated behaviors through identification of a previously unknown neuropeptide-containing pnVTA nucleus that limits motivation for rewards.


Subject(s)
Motivation/drug effects , Opioid Peptides/pharmacology , Reward , Ventral Tegmental Area/metabolism , Action Potentials , Animals , Behavior, Animal/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/physiology , Patch-Clamp Techniques , Protein Precursors/genetics , Receptors, Opioid/agonists , Receptors, Opioid/deficiency , Receptors, Opioid/genetics , Nociceptin Receptor , Nociceptin
6.
Cell ; 172(1-2): 55-67.e15, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29307491

ABSTRACT

The κ-opioid receptor (KOP) mediates the actions of opioids with hallucinogenic, dysphoric, and analgesic activities. The design of KOP analgesics devoid of hallucinatory and dysphoric effects has been hindered by an incomplete structural and mechanistic understanding of KOP agonist actions. Here, we provide a crystal structure of human KOP in complex with the potent epoxymorphinan opioid agonist MP1104 and an active-state-stabilizing nanobody. Comparisons between inactive- and active-state opioid receptor structures reveal substantial conformational changes in the binding pocket and intracellular and extracellular regions. Extensive structural analysis and experimental validation illuminate key residues that propagate larger-scale structural rearrangements and transducer binding that, collectively, elucidate the structural determinants of KOP pharmacology, function, and biased signaling. These molecular insights promise to accelerate the structure-guided design of safer and more effective κ-opioid receptor therapeutics.


Subject(s)
Molecular Docking Simulation , Receptors, Opioid, kappa/chemistry , Analgesics/chemistry , Analgesics/pharmacology , Animals , Binding Sites , HEK293 Cells , Humans , Molecular Dynamics Simulation , Morphinans/chemistry , Morphinans/pharmacology , Protein Binding , Protein Stability , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Sf9 Cells , Spodoptera
7.
Cell ; 172(1-2): 41-54.e19, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29249361

ABSTRACT

Natural genetic variation in the human genome is a cause of individual differences in responses to medications and is an underappreciated burden on public health. Although 108 G-protein-coupled receptors (GPCRs) are the targets of 475 (∼34%) Food and Drug Administration (FDA)-approved drugs and account for a global sales volume of over 180 billion US dollars annually, the prevalence of genetic variation among GPCRs targeted by drugs is unknown. By analyzing data from 68,496 individuals, we find that GPCRs targeted by drugs show genetic variation within functional regions such as drug- and effector-binding sites in the human population. We experimentally show that certain variants of µ-opioid and Cholecystokinin-A receptors could lead to altered or adverse drug response. By analyzing UK National Health Service drug prescription and sales data, we suggest that characterizing GPCR variants could increase prescription precision, improving patients' quality of life, and relieve the economic and societal burden due to variable drug responsiveness. VIDEO ABSTRACT.


Subject(s)
Pharmacogenetics/methods , Pharmacogenomic Variants , Receptors, G-Protein-Coupled/genetics , Software , Binding Sites , Drug Prescriptions/standards , HEK293 Cells , Humans , Protein Binding , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
8.
Cell ; 169(2): 350-360.e12, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388416

ABSTRACT

Cells operate through protein interaction networks organized in space and time. Here, we describe an approach to resolve both dimensions simultaneously by using proximity labeling mediated by engineered ascorbic acid peroxidase (APEX). APEX has been used to capture entire organelle proteomes with high temporal resolution, but its breadth of labeling is generally thought to preclude the higher spatial resolution necessary to interrogate specific protein networks. We provide a solution to this problem by combining quantitative proteomics with a system of spatial references. As proof of principle, we apply this approach to interrogate proteins engaged by G-protein-coupled receptors as they dynamically signal and traffic in response to ligand-induced activation. The method resolves known binding partners, as well as previously unidentified network components. Validating its utility as a discovery pipeline, we establish that two of these proteins promote ubiquitin-linked receptor downregulation after prolonged activation.


Subject(s)
Ascorbate Peroxidases/chemistry , Protein Interaction Maps , Staining and Labeling/methods , Animals , Humans , Lysosomes/metabolism , Protein Transport , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid/metabolism , Ubiquitin/metabolism
9.
Cell ; 171(5): 1165-1175.e13, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149605

ABSTRACT

Biased agonism has been proposed as a means to separate desirable and adverse drug responses downstream of G protein-coupled receptor (GPCR) targets. Herein, we describe structural features of a series of mu-opioid-receptor (MOR)-selective agonists that preferentially activate receptors to couple to G proteins or to recruit ßarrestin proteins. By comparing relative bias for MOR-mediated signaling in each pathway, we demonstrate a strong correlation between the respiratory suppression/antinociception therapeutic window in a series of compounds spanning a wide range of signaling bias. We find that ßarrestin-biased compounds, such as fentanyl, are more likely to induce respiratory suppression at weak analgesic doses, while G protein signaling bias broadens the therapeutic window, allowing for antinociception in the absence of respiratory suppression.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Receptors, Opioid, mu/agonists , Animals , Fentanyl/administration & dosage , GTP-Binding Proteins/metabolism , Mice , Morphine/administration & dosage , Receptors, Opioid, mu/chemistry , Respiratory System/drug effects , Signal Transduction , beta-Arrestins/metabolism
10.
CA Cancer J Clin ; 74(3): 286-313, 2024.
Article in English | MEDLINE | ID: mdl-38108561

ABSTRACT

Pain is one of the most burdensome symptoms in people with cancer, and opioid analgesics are considered the mainstay of cancer pain management. For this review, the authors evaluated the efficacy and toxicities of opioid analgesics compared with placebo, other opioids, nonopioid analgesics, and nonpharmacologic treatments for background cancer pain (continuous and relatively constant pain present at rest), and breakthrough cancer pain (transient exacerbation of pain despite stable and adequately controlled background pain). They found a paucity of placebo-controlled trials for background cancer pain, although tapentadol or codeine may be more efficacious than placebo (moderate-certainty to low-certainty evidence). Nonsteroidal anti-inflammatory drugs including aspirin, piroxicam, diclofenac, ketorolac, and the antidepressant medicine imipramine, may be at least as efficacious as opioids for moderate-to-severe background cancer pain. For breakthrough cancer pain, oral transmucosal, buccal, sublingual, or intranasal fentanyl preparations were identified as more efficacious than placebo but were more commonly associated with toxicities, including constipation and nausea. Despite being recommended worldwide for the treatment of cancer pain, morphine was generally not superior to other opioids, nor did it have a more favorable toxicity profile. The interpretation of study results, however, was complicated by the heterogeneity in the study populations evaluated. Given the limited quality and quantity of research, there is a need to reappraise the clinical utility of opioids in people with cancer pain, particularly those who are not at the end of life, and to further explore the effects of opioids on immune system function and quality of life in these individuals.


Subject(s)
Analgesics, Opioid , Cancer Pain , Humans , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/adverse effects , Cancer Pain/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Nociceptive Pain/drug therapy , Neoplasms/complications , Pain Management/methods
11.
Mol Cell ; 81(20): 4165-4175.e6, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34433090

ABSTRACT

GPCR functional selectivity opens new opportunities for the design of safer drugs. Ligands orchestrate GPCR signaling cascades by modulating the receptor conformational landscape. Our study provides insights into the dynamic mechanism enabling opioid ligands to preferentially activate the G protein over the ß-arrestin pathways through the µ-opioid receptor (µOR). We combine functional assays in living cells, solution NMR spectroscopy, and enhanced-sampling molecular dynamic simulations to identify the specific µOR conformations induced by G protein-biased agonists. In particular, we describe the dynamic and allosteric communications between the ligand-binding pocket and the receptor intracellular domains, through conserved motifs in class A GPCRs. Most strikingly, the biased agonists trigger µOR conformational changes in the intracellular loop 1 and helix 8 domains, which may impair ß-arrestin binding or signaling. The findings may apply to other GPCR families and provide key molecular information that could facilitate the design of biased ligands.


Subject(s)
Analgesics, Opioid/pharmacology , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Signal Transduction/drug effects , Analgesics, Opioid/chemistry , Animals , Binding Sites , Computer-Aided Design , Drug Design , Drug Partial Agonism , HEK293 Cells , Humans , Ligands , Mice , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Sf9 Cells , Structure-Activity Relationship , beta-Arrestins/genetics , beta-Arrestins/metabolism
12.
Annu Rev Neurosci ; 43: 355-374, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32109184

ABSTRACT

Opioid addiction and overdose are at record levels in the United States. This is driven, in part, by their widespread prescription for the treatment of pain, which also increased opportunity for diversion by sensation-seeking users. Despite considerable research on the neurobiology of addiction, treatment options for opioid abuse remain limited. Mood disorders, particularly depression, are often comorbid with both pain disorders and opioid abuse. The endogenous opioid system, a complex neuromodulatory system, sits at the neurobiological convergence point of these three comorbid disease states. We review evidence for dysregulation of the endogenous opioid system as a mechanism for the development of opioid addiction and/or mood disorder. Specifically, individual differences in opioid system function may underlie differences in vulnerability to opioid addiction and mood disorders. We also review novel research, which promises to provide more detailed understanding of individual differences in endogenous opioid neurobiology and its contribution to opioid addiction susceptibility.


Subject(s)
Analgesics, Opioid/therapeutic use , Chronic Pain/drug therapy , Depression/drug therapy , Opioid-Related Disorders/drug therapy , Animals , Drug Overdose/drug therapy , Humans , Precision Medicine/methods
13.
Annu Rev Physiol ; 86: 1-25, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38029388

ABSTRACT

The harmful side effects of opioid drugs such as respiratory depression, tolerance, dependence, and abuse potential have limited the therapeutic utility of opioids for their entire clinical history. However, no previous attempt to develop effective pain drugs that substantially ameliorate these effects has succeeded, and the current opioid epidemic affirms that they are a greater hindrance to the field of pain management than ever. Recent attempts at new opioid development have sought to reduce these side effects by minimizing engagement of the regulatory protein arrestin-3 at the mu-opioid receptor, but there is significant controversy around this approach. Here, we discuss the ongoing effort to develop safer opioids and its relevant historical context. We propose a new model that reconciles results previously assumed to be in direct conflict to explain how different signaling profiles at the mu-opioid receptor contribute to opioid tolerance and dependence. Our goal is for this framework to inform the search for a new generation of lower liability opioid analgesics.


Subject(s)
Analgesics, Opioid , Signal Transduction , Humans , Analgesics, Opioid/adverse effects , Drug Tolerance
14.
Proc Natl Acad Sci U S A ; 121(34): e2405465121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39145932

ABSTRACT

Over half of spinal cord injury (SCI) patients develop opioid-resistant chronic neuropathic pain. Safer alternatives to opioids for treatment of neuropathic pain are gabapentinoids (e.g., pregabalin and gabapentin). Clinically, gabapentinoids appear to amplify opioid effects, increasing analgesia and overdose-related adverse outcomes, but in vitro proof of this amplification and its mechanism are lacking. We previously showed that after SCI, sensitivity to opioids is reduced by fourfold to sixfold in rat sensory neurons. Here, we demonstrate that after injury, gabapentinoids restore normal sensitivity of opioid inhibition of cyclic AMP (cAMP) generation, while reducing nociceptor hyperexcitability by inhibiting voltage-gated calcium channels (VGCCs). Increasing intracellular Ca2+ or activation of L-type VGCCs (L-VGCCs) suffices to mimic SCI effects on opioid sensitivity, in a manner dependent on the activity of the Raf1 proto-oncogene, serine/threonine-protein kinase C-Raf, but independent of neuronal depolarization. Together, our results provide a mechanism for potentiation of opioid effects by gabapentinoids after injury, via reduction of calcium influx through L-VGCCs, and suggest that other inhibitors targeting these channels may similarly enhance opioid treatment of neuropathic pain.


Subject(s)
Analgesics, Opioid , Cyclic AMP , Gabapentin , Neuralgia , Signal Transduction , Spinal Cord Injuries , Animals , Neuralgia/drug therapy , Neuralgia/metabolism , Cyclic AMP/metabolism , Rats , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Analgesics, Opioid/pharmacology , Gabapentin/pharmacology , Signal Transduction/drug effects , Rats, Sprague-Dawley , Male , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Pregabalin/pharmacology , Pregabalin/therapeutic use , Drug Synergism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects
15.
Annu Rev Pharmacol Toxicol ; 63: 491-515, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36170657

ABSTRACT

In ligand bias different agonist drugs are thought to produce distinct signaling outputs when activating the same receptor. If these signaling outputs mediate therapeutic versus adverse drug effects, then agonists that selectively activate the therapeutic signaling pathway would be extremely beneficial. It has long been thought that µ-opioid receptor agonists that selectively activate G protein- over ß-arrestin-dependent signaling pathways would produce effective analgesia without the adverse effects such as respiratory depression. However, more recent data indicate that most of the therapeutic and adverse effects of agonist-induced activation of the µ-opioid receptor are actually mediated by the G protein-dependent signaling pathway, and that a number of drugs described as G protein biased in fact may not be biased, but instead may be low-intrinsic-efficacy agonists. In this review we discuss the current state of the field of bias at the µ-opioid receptor and other opioid receptor subtypes.


Subject(s)
Analgesics, Opioid , Signal Transduction , Humans , Analgesics, Opioid/adverse effects , GTP-Binding Proteins/metabolism , Pain/drug therapy , beta-Arrestins/metabolism
16.
Annu Rev Med ; 75: 83-97, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-37827194

ABSTRACT

Opioid use disorder continues to drive overdose deaths in many countries, including the United States. Illicit fentanyl and its analogues have emerged as key contributors to the complications and mortality associated with opioid use disorder. Medications for opioid use disorder treatment, such as methadone and buprenorphine, are safe and substantially reduce opioid use, infectious complications, and mortality risk, but remain underutilized. Polysubstance use and emerging substances such as xylazine and designer benzodiazepines create additional treatment challenges. Recent clinical and policy innovations in treatment delivery, including telemedicine, bridge clinics, and expanded models for accessing methadone have the potential to increase access to life-saving care for people living with opioid use disorder.


Subject(s)
Buprenorphine , Drug Overdose , Opioid-Related Disorders , Humans , United States/epidemiology , Methadone/therapeutic use , Buprenorphine/therapeutic use , Analgesics, Opioid/therapeutic use
17.
Annu Rev Neurosci ; 41: 453-473, 2018 07 08.
Article in English | MEDLINE | ID: mdl-29852083

ABSTRACT

Opioids are the most commonly used and effective analgesic treatments for severe pain, but they have recently come under scrutiny owing to epidemic levels of abuse and overdose. These compounds act on the endogenous opioid system, which comprises four G protein-coupled receptors (mu, delta, kappa, and nociceptin) and four major peptide families (ß-endorphin, enkephalins, dynorphins, and nociceptin/orphanin FQ). In this review, we first describe the functional organization and pharmacology of the endogenous opioid system. We then summarize current knowledge on the signaling mechanisms by which opioids regulate neuronal function and neurotransmission. Finally, we discuss the loci of opioid analgesic action along peripheral and central pain pathways, emphasizing the pain-relieving properties of opioids against the affective dimension of the pain experience.


Subject(s)
Analgesics, Opioid/metabolism , Analgesics, Opioid/therapeutic use , Pain/drug therapy , Pain/metabolism , Animals , Humans , Pain Perception , Receptors, G-Protein-Coupled/metabolism
18.
Trends Biochem Sci ; 46(4): 315-328, 2021 04.
Article in English | MEDLINE | ID: mdl-33127216

ABSTRACT

Opioid receptors (ORs) are undisputed targets for the treatment of pain. Unfortunately, targeting these receptors therapeutically poses significant challenges including addiction, dependence, tolerance, and the appearance of side effects, such as respiratory depression and constipation. Moreover, misuse of prescription and illicit narcotics has resulted in the current opioid crisis. The mu-opioid receptor (MOR) is the cellular mediator of the effects of most commonly used opioids, and is a prototypical G protein-coupled receptor (GPCR) where new pharmacological, signalling and cell biology concepts have been coined. This review summarises the knowledge of the life cycle of this therapeutic target, including its biogenesis, trafficking to and from the plasma membrane, and how the regulation of these processes impacts its function and is related to pathophysiological conditions.


Subject(s)
Analgesics, Opioid , Receptors, Opioid , Analgesics, Opioid/pharmacology , Animals , Drug Tolerance , Life Cycle Stages
19.
J Neurosci ; 44(6)2024 02 07.
Article in English | MEDLINE | ID: mdl-38124021

ABSTRACT

Prolonged exposure to opioids causes an enhanced sensitivity to painful stimuli (opioid-induced hyperalgesia, OIH) and a need for increased opioid doses to maintain analgesia (opioid-induced tolerance, OIT), but the mechanisms underlying both processes remain obscure. We found that pharmacological block or genetic deletion of HCN2 ion channels in primary nociceptive neurons of male mice completely abolished OIH but had no effect on OIT. Conversely, pharmacological inhibition of central HCN channels alleviated OIT but had no effect on OIH. Expression of C-FOS, a marker of neuronal activity, was increased in second-order neurons of the dorsal spinal cord by induction of OIH, and the increase was prevented by peripheral block or genetic deletion of HCN2, but block of OIT by spinal block of HCN channels had no impact on C-FOS expression in dorsal horn neurons. Collectively, these observations show that OIH is driven by HCN2 ion channels in peripheral nociceptors, while OIT is driven by a member of the HCN family located in the CNS. Induction of OIH increased cAMP in nociceptive neurons, and a consequent shift in the activation curve of HCN2 caused an increase in nociceptor firing. The shift in HCN2 was caused by expression of a constitutively active µ-opioid receptor (MOR) and was reversed by MOR antagonists. We identified the opioid-induced MOR as a six-transmembrane splice variant, and we show that it increases cAMP by coupling constitutively to Gs HCN2 ion channels therefore drive OIH, and likely OIT, and may be a novel therapeutic target for the treatment of addiction.


Subject(s)
Analgesics, Opioid , Hyperalgesia , Mice , Male , Animals , Analgesics, Opioid/adverse effects , Hyperalgesia/metabolism , Ion Channels , Nociceptors , Spinal Cord/metabolism , Pain/metabolism
20.
J Neurosci ; 44(3)2024 Jan 17.
Article in English | MEDLINE | ID: mdl-37985179

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.


Subject(s)
Analgesics, Opioid , Morphine , Male , Female , Mice , Animals , Morphine/pharmacology , Analgesics, Opioid/pharmacology , Phosphorylation , Signal Transduction , Receptors, Opioid , Receptors, Opioid, mu/agonists
SELECTION OF CITATIONS
SEARCH DETAIL