Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Pharmacol Exp Ther ; 389(3): 301-309, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38621994

ABSTRACT

δ opioid receptors (DORs) hold potential as a target for neurologic and psychiatric disorders, yet no DOR agonist has proven efficacious in critical phase II clinical trials. The exact reasons for the failure to produce quality drug candidates for the DOR are unclear. However, it is known that certain DOR agonists can induce seizures and exhibit tachyphylaxis. Several studies have suggested that those adverse effects are more prevalent in delta agonists that share the (+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide (SNC80)/4-[(αR*)-α-((2S*,5R*)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-hydroxybenzyl]-N,N-diethylbenzamide chemotype. There is a need to find novel lead candidates for drug development that have improved pharmacological properties to differentiate them from the current failed delta agonists. Our objective in this study was to identify novel DOR agonists. We used a ß-arrestin assay to screen a small G-protein coupled receptors (GPCR)-focused chemical library. We identified a novel chemotype of DOR agonists that appears to bind to the orthosteric site based of docking and molecular dynamic simulation. The most potent agonist hit compound is selective for the DOR over a panel of 167 other GPCRs, is slightly biased toward G-protein signaling and has anti-allodynic efficacy in a complete Freund's adjuvant model of inflammatory pain in C57BL/6 male and female mice. The newly discovered chemotype contrasts with molecules like SNC80 that are highly efficacious ß-arrestin recruiters and may suggest this novel class of DOR agonists could be expanded on to develop a clinical candidate drug. SIGNIFICANCE STATEMENT: δ opioid receptors are a clinical target for various neurological disorders, including migraine and chronic pain. Many of the clinically tested delta opioid agonists share a single chemotype, which carries risks during drug development. Through a small-scale high-throughput screening assay, this study identified a novel δ opioid receptor agonist chemotype, which may serve as alternative for the current analgesic clinical candidates.


Subject(s)
Receptors, Opioid, delta , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Animals , Mice , Male , Humans , Spiro Compounds/pharmacology , Spiro Compounds/chemistry , Piperazines/pharmacology , Piperazines/chemistry , Mice, Inbred C57BL , Molecular Docking Simulation , Benzamides/pharmacology , Benzamides/chemistry , Cricetulus , beta-Arrestins/metabolism , HEK293 Cells , CHO Cells
2.
Headache ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38899347

ABSTRACT

OBJECTIVE: Our aim was to survey astrocyte and microglial activation across four brain regions in a mouse model of chronic migraine. BACKGROUND: Chronic migraine is a leading cause of disability, with higher rates in females. The role of central nervous system neurons and glia in migraine pathophysiology is not fully elucidated. Preclinical studies have shown abnormal glial activation in the trigeminal nucleus caudalis of male rodents. No current reports have investigated glial activation in both sexes in other important brain regions involved with the nociceptive and emotional processing of pain. METHODS: The mouse nitroglycerin model of migraine was used, and nitroglycerin (10 mg/kg) or vehicle was administered every other day for 9 days. Prior to injections on days 1, 5, and 9, cephalic allodynia was determined by periorbital von Frey hair testing. Immunofluorescent staining of astrocyte marker, glial fibrillary protein (GFAP), and microglial marker, ionized calcium binding adaptor molecule 1 (Iba1), in male and female trigeminal nucleus caudalis, periaqueductal gray, somatosensory cortex, and nucleus accumbens was completed. RESULTS: Behavioral testing demonstrated increased cephalic allodynia in nitroglycerin- versus vehicle-treated mice. An increase in the percent area covered by GFAP+ cells in the trigeminal nucleus caudalis and nucleus accumbens, but not the periaqueductal gray or somatosensory cortex, was observed in response to nitroglycerin. No significant differences were observed for Iba1 staining across brain regions. We did not detect significant sex differences in GFAP or Iba1 quantification. CONCLUSIONS: Immunohistochemical analysis suggests that, at the time point tested, immunoreactivity of GFAP+ astrocytes, but not Iba1+ microglia, changes in response to chronic migraine-associated pain. Additionally, there do not appear to be significant differences between males and females in GFAP+ or Iba1+ cells across the four brain regions analyzed.

3.
Headache ; 64(2): 195-210, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38288634

ABSTRACT

OBJECTIVE: To characterize the circadian features of the trigeminal ganglion in a mouse model of headache. BACKGROUND: Several headache disorders, such as migraine and cluster headache, are known to exhibit distinct circadian rhythms of attacks. The circadian basis for these rhythmic pain responses, however, remains poorly understood. METHODS: We examined trigeminal ganglion ex vivo and single-cell cultures from Per2::LucSV reporter mice and performed immunohistochemistry. Circadian behavior and transcriptomics were investigated using a novel combination of trigeminovascular and circadian models: a nitroglycerin mouse headache model with mechanical thresholds measured every 6 h, and trigeminal ganglion RNA sequencing measured every 4 h for 24 h. Finally, we performed pharmacogenomic analysis of gene targets for migraine, cluster headache, and trigeminal neuralgia treatments as well as trigeminal ganglion neuropeptides; this information was cross-referenced with our cycling genes from RNA sequencing data to identify potential targets for chronotherapy. RESULTS: The trigeminal ganglion demonstrates strong circadian rhythms in both ex vivo and single-cell cultures, with core circadian proteins found in both neuronal and non-neuronal cells. Using our novel behavioral model, we showed that nitroglycerin-treated mice display circadian rhythms of pain sensitivity which were abolished in arrhythmic Per1/2 double knockout mice. Furthermore, RNA-sequencing analysis of the trigeminal ganglion revealed 466 genes that displayed circadian oscillations in the control group, including core clock genes and clock-regulated pain neurotransmitters. In the nitroglycerin group, we observed a profound circadian reprogramming of gene expression, as 331 of circadian genes in the control group lost rhythm and another 584 genes gained rhythm. Finally, pharmacogenetics analysis identified 10 genes in our trigeminal ganglion circadian transcriptome that encode target proteins of current medications used to treat migraine, cluster headache, or trigeminal neuralgia. CONCLUSION: Our study unveiled robust circadian rhythms in the trigeminal ganglion at the behavioral, transcriptomic, and pharmacogenetic levels. These results support a fundamental role of the clock in pain pathophysiology. PLAIN LANGUAGE SUMMARY: Several headache diseases, such as migraine and cluster headache, have headaches that occur at the same time each day. We learned that the trigeminal ganglion, an important pain structure in several headache diseases, has a 24-hour cycle that might be related to this daily cycle of headaches. Our genetic analysis suggests that some medications may be more effective in treating migraine and cluster headache when taken at specific times of the day.


Subject(s)
Cluster Headache , Migraine Disorders , Trigeminal Neuralgia , Mice , Animals , Trigeminal Ganglion , Transcriptome , Trigeminal Neuralgia/genetics , Nitroglycerin , Headache , Gene Expression Profiling , Pain , Circadian Rhythm/genetics , Mice, Knockout
4.
Headache ; 62(7): 780-791, 2022 07.
Article in English | MEDLINE | ID: mdl-35676889

ABSTRACT

OBJECTIVE: Our laboratory has recently shown that there is a decrease in neuronal complexity in head pain processing regions in mouse models of chronic migraine-associated pain and aura. Importantly, restoration of this neuronal complexity corresponds with anti-migraine effects of known and experimental pharmacotherapies. The objective of the current study was to expand this work and examine other brain regions involved with pain or emotional processing. We also investigated the generalizability of our findings by analyzing neuronal cytoarchitectural changes in a model of complex regional pain syndrome (CRPS), a peripheral pain disorder. METHODS: We used the nitroglycerin (NTG) model of chronic migraine-associated pain in which mice receive 10 mg/kg NTG every other day for 9 days. Cortical spreading depression (CSD), a physiological corelate of migraine aura, was evoked in anesthetized mice using KCl. CRPS was induced by tibial fracture followed by casting. Neuronal cytoarchitecture was visualized with Golgi stain and analyzed with Simple Neurite Tracer. RESULTS: In the NTG model, we previously showed decreased neuronal complexity in the trigeminal nucleus caudalis (TNC) and periaqueductal gray (PAG). In contrast, we found increased neuronal complexity in the thalamus and no change in the amygdala or caudate putamen in this study. Following CSD, we observed decreased neuronal complexity in the PAG, in line with decreases in the somatosensory cortex and TNC reported with this model previously. In the CRPS model there was decreased neuronal complexity in the hippocampus, as reported by others; increased complexity in the PAG; and no change within the somatosensory cortex. CONCLUSIONS: Collectively these results demonstrate that alterations in neuronal complexity are a feature of both chronic migraine and chronic CRPS. However, each type of pain presents a unique cytoarchitectural signature, which may provide insight on how these pain states differentially transition from acute to chronic conditions.


Subject(s)
Complex Regional Pain Syndromes , Cortical Spreading Depression , Migraine Disorders , Animals , Cortical Spreading Depression/physiology , Disease Models, Animal , Headache , Mice , Migraine Disorders/drug therapy , Nitroglycerin/adverse effects
5.
Headache ; 61(1): 170-178, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33326598

ABSTRACT

OBJECTIVE: The aim of this study was to determine if the non-convulsant delta-opioid receptor (DOR) agonist, KNT-127, could inhibit migraine-associated endpoints. BACKGROUND: The DOR has been identified as a therapeutic target for migraine. However, the development of delta agonists is limited as some ligands have seizurogenic properties, which may be related to their ability to induce receptor internalization. While both pro- and non-convulsant delta agonists can reduce migraine-associated allodynia, only the proconvulsant agonist, SNC80, has been shown to decrease cortical spreading depression (CSD). It is unclear if the ability of delta agonists to modulate cortical activity is related to the same signaling mechanisms that produce proconvulsant effects. METHODS: The effects of the non-convulsant delta agonist, KNT-127, were examined. Repetitive CSD was induced in female C57BL6/J (n = 6/group) mice by continuous application of KCl and the effect of KNT-127/vehicle (Veh) on both local field potentials and optical intrinsic signals was determined. To assess the effect of KNT-127 on established chronic migraine-associated pain, male and female C57BL6/J mice were treated with nitroglycerin (NTG; 10 mg/kg, ip) every other day for 9 days and tested with KNT-127 (5 mg/kg, sc) or Veh on day 10 (n = 6/group). DOR-enhanced green fluorescent protein mice (n = 4/group) were used to confirm the internalization properties of KNT-127 in the trigeminal ganglia, trigeminal nucleus caudalis, and somatosensory cortex. RESULTS: KNT-127 inhibited CSD events (t(10)  = 3.570, p = 0.0051). In addition, this delta agonist also reversed established cephalic allodynia in the NTG model of chronic migraine (F(1, 20)  = 12.80, p < 0.01). Furthermore, KNT-127 caused limited internalization of DOR in key migraine processing regions. CONCLUSIONS: This study shows that the antimigraine effects of DOR agonists can be separated from their proconvulsant effects. This data provides valuable information for the continued development of delta agonists for the treatment of migraine.


Subject(s)
Analgesics, Opioid/pharmacology , Cortical Spreading Depression/drug effects , Hyperalgesia/drug therapy , Migraine Disorders/drug therapy , Morphinans/pharmacology , Receptors, Opioid, delta/agonists , Analgesics, Opioid/administration & dosage , Animals , Disease Models, Animal , Female , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C57BL , Morphinans/administration & dosage , Nitroglycerin/pharmacology , Vasodilator Agents/pharmacology
6.
Mol Cell Proteomics ; 18(12): 2447-2458, 2019 12.
Article in English | MEDLINE | ID: mdl-31649062

ABSTRACT

Chronic use of opioids can produce opioid-induced hyperalgesia (OIH), and when used to treat migraine, these drugs can result in increased pain and headache chronicity. We hypothesized that overlapping mechanisms between OIH and chronic migraine occur through neuropeptide dysregulation. Using label-free, non-biased liquid chromatography-mass spectrometry to identify and measure changes in more than 1500 neuropeptides under these two conditions, we observed only 16 neuropeptides that were altered between the two conditions. The known pro-migraine molecule, calcitonin-gene related peptide, was among seven peptides associated with chronic migraine, with several pain-processing neuropeptides among the nine other peptides affected in OIH. Further, composite peptide complements Pituitary adenylate cyclase-activating polypeptide (PACAP), Vasoactive intestinal peptide (VIP) and Secretogranin (SCG) showed significant changes in both chronic migraine and OIH. In a follow-up pharmacological study, we confirmed the role of PACAP in models of these two disorders, validating the effectiveness of our peptidomic approach, and identifying PACAP as a mechanistic link between chronic migraine and OIH. Data are available via ProteomeXchange with identifier PXD013362.


Subject(s)
Analgesics, Opioid/adverse effects , Hyperalgesia/chemically induced , Migraine Disorders/drug therapy , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Analgesics, Opioid/therapeutic use , Animals , Behavior, Animal , Chromatography, Liquid , Disease Models, Animal , Headache Disorders/complications , Headache Disorders/drug therapy , Hyperalgesia/complications , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Migraine Disorders/complications , Migraine Disorders/metabolism , Tandem Mass Spectrometry
7.
Mol Pharmacol ; 98(4): 433-444, 2020 10.
Article in English | MEDLINE | ID: mdl-32958571

ABSTRACT

For decades the broad role of opioids in addiction, neuropsychiatric disorders, and pain states has been somewhat well established. However, in recent years, with the rise of technological advances, not only is the existing dogma being challenged, but we are identifying new disease areas in which opioids play a critical role. This review highlights four new areas of exploration in the opioid field. The most recent addition to the opioid family, the nociceptin receptor system, shows promise as the missing link in understanding the neurocircuitry of motivation. It is well known that activation of the kappa opioid receptor system modulates negative affect and dysphoria, but recent studies now implicate the kappa opioid system in the modulation of negative affect associated with pain. Opioids are critical in pain management; however, the often-forgotten delta opioid receptor system has been identified as a novel therapeutic target for headache disorders and migraine. Lastly, changes to the gut microbiome have been shown to directly contribute to many of the symptoms of chronic opioid use and opioid related behaviors. This review summarizes the findings from each of these areas with an emphasis on identifying new therapeutic targets. SIGNIFICANCE STATEMENT: The focus of this minireview is to highlight new disease areas or new aspects of disease in which opioids have been implicated; this includes pain, motivation, migraine, and the microbiome. In some cases, this has resulted in the pursuit of a novel therapeutic target and resultant clinical trial. We believe this is very timely and will be a refreshing take on reading about opioids and disease.


Subject(s)
Analgesics, Opioid/pharmacology , Migraine Disorders/metabolism , Opioid-Related Disorders/microbiology , Pain/metabolism , Receptors, Opioid/metabolism , Analgesics, Opioid/therapeutic use , Animals , Gastrointestinal Microbiome/drug effects , Humans , Migraine Disorders/drug therapy , Motivation , Opioid-Related Disorders/metabolism , Pain/drug therapy , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Signal Transduction/drug effects , Nociceptin Receptor
9.
Int J Neuropsychopharmacol ; 22(8): 523-527, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31179497

ABSTRACT

BACKGROUND: Increased pain sensitivity is observed following alcohol withdrawal, and attempts to alleviate this hyperalgesia can contribute to the cycle of addiction. The aim of this study was to determine if alcohol withdrawal-induced hyperalgesia was observed in a chronic ethanol exposure model and if this pain was affected by histone deacetylase inhibitors, thus revealing an epigenetic mechanism. METHODS: Adult male Sprague Dawley rats received Lieber-DeCarli liquid control or ethanol (9% v/v) diet for 15 days. Mechanical sensitivity was measured with von Frey hair stimulation of the hindpaw during ethanol administration and 24- and 72-hour withdrawal. RESULTS: Ethanol withdrawal produced severe and sustained mechanical hyperalgesia, an effect not observed in the control or ethanol-maintained groups. Furthermore, this hyperalgesia was attenuated by the histone deacetylase inhibitor, suberoylanilide hydroxamic acid treatment. CONCLUSIONS: Heightened pain sensitivity was observed following withdrawal from chronic ethanol exposure, and histone deacetylase inhibitors could be novel treatments for this alcohol withdrawal-induced hyperalgesia.


Subject(s)
Analgesics/pharmacology , Behavior, Animal/drug effects , Ethanol , Histone Deacetylase Inhibitors/pharmacology , Hyperalgesia/drug therapy , Pain Threshold/drug effects , Substance Withdrawal Syndrome/drug therapy , Vorinostat/pharmacology , Animals , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Male , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/psychology
10.
J Neurosci ; 37(14): 3741-3752, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28264976

ABSTRACT

The δ opioid receptor (δR) is a promising alternate target for pain management because δR agonists show decreased abuse potential compared with current opioid analgesics that target the µ opioid receptor. A critical limitation in developing δR as an analgesic target, however, is that δR agonists show relatively low efficacy in vivo, requiring the use of high doses that often cause adverse effects, such as convulsions. Here we tested whether intracellular retention of δR in sensory neurons contributes to this low δR agonist efficacy in vivo by limiting surface δR expression. Using direct visualization of δR trafficking and localization, we define a phosphatase and tensin homolog (PTEN)-regulated checkpoint that retains δR in the Golgi and decreases surface delivery in rat and mice sensory neurons. PTEN inhibition releases δR from this checkpoint and stimulates delivery of exogenous and endogenous δR to the neuronal surface both in vitro and in vivo PTEN inhibition in vivo increases the percentage of TG neurons expressing δR on the surface and allows efficient δR-mediated antihyperalgesia in mice. Together, we define a critical role for PTEN in regulating the surface delivery and bioavailability of the δR, explain the low efficacy of δR agonists in vivo, and provide evidence that active δR relocation is a viable strategy to increase δR antinociception.SIGNIFICANCE STATEMENT Opioid analgesics, such as morphine, which target the µ opioid receptor (µR), have been the mainstay of pain management, but their use is highly limited by adverse effects and their variable efficacy in chronic pain. Identifying alternate analgesic targets is therefore of great significance. Although the δ opioid receptor (δR) is an attractive option, a critical limiting factor in developing δR as a target has been the low efficacy of δR agonists. Why δR agonists show low efficacy is still under debate. This study provides mechanistic and functional data that intracellular localization of δR in neurons is a key factor that contributes to low agonist efficacy, and presents a proof of mechanism that relocating δR improves efficacy.


Subject(s)
Cell Membrane/metabolism , Neurons/metabolism , PTEN Phosphohydrolase/physiology , Receptors, Opioid, delta/metabolism , Animals , Cell Membrane/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , PC12 Cells , PTEN Phosphohydrolase/antagonists & inhibitors , Phenanthrenes/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley
11.
Cephalalgia ; 38(8): 1471-1484, 2018 07.
Article in English | MEDLINE | ID: mdl-29022756

ABSTRACT

Background Nitric oxide (NO) has been heavily implicated in migraine. Nitroglycerin is a prototypic NO-donor, and triggers migraine in humans. However, nitroglycerin also induces oxidative/nitrosative stress and is a source of peroxynitrite - factors previously linked with migraine etiology. Soluble guanylyl cyclase (sGC) is the high affinity NO receptor in the body, and the aim of this study was to identify the precise role of sGC in acute and chronic migraine. Methods We developed a novel brain-bioavailable sGC stimulator (VL-102), and tested its hyperalgesic properties in mice. We also determined the effect of VL-102 on c-fos and calcitonin gene related peptide (CGRP) immunoreactivity within the trigeminovascular complex. In addition, we also tested the known sGC inhibitor, ODQ, within the chronic nitroglycerin migraine model. Results VL-102-evoked acute and chronic mechanical cephalic and hind-paw allodynia in a dose-dependent manner, which was blocked by the migraine medications sumatriptan, propranolol, and topiramate. In addition, VL-102 also increased c-fos and CGRP expressing cells within the trigeminovascular complex. Importantly, ODQ completely inhibited acute and chronic hyperalgesia induced by nitroglycerin. ODQ also blocked hyperalgesia already established by chronic nitroglycerin, implicating this pathway in migraine chronicity. Conclusions These results indicate that nitroglycerin causes migraine-related pain through stimulation of the sGC pathway, and that super-activation of this receptor may be an important component for the maintenance of chronic migraine. This work opens the possibility for negative sGC modulators as novel migraine therapies.


Subject(s)
Hyperalgesia/chemically induced , Hyperalgesia/enzymology , Migraine Disorders/enzymology , Soluble Guanylyl Cyclase/physiology , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/therapeutic use , Allosteric Regulation , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/therapeutic use , Calcitonin Gene-Related Peptide/biosynthesis , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Female , Hyperalgesia/drug therapy , Male , Mice , Mice, Inbred C57BL , Migraine Disorders/chemically induced , Migraine Disorders/drug therapy , Migraine Disorders/etiology , Molecular Targeted Therapy , Nitric Oxide/adverse effects , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitroglycerin/pharmacology , Oxadiazoles/administration & dosage , Oxadiazoles/metabolism , Oxadiazoles/therapeutic use , Propranolol/administration & dosage , Propranolol/therapeutic use , Proto-Oncogene Proteins c-fos/biosynthesis , Quinoxalines/administration & dosage , Quinoxalines/metabolism , Quinoxalines/therapeutic use , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Soluble Guanylyl Cyclase/metabolism , Sumatriptan/administration & dosage , Sumatriptan/therapeutic use , Topiramate/administration & dosage , Topiramate/therapeutic use
12.
Handb Exp Pharmacol ; 247: 73-85, 2018.
Article in English | MEDLINE | ID: mdl-28689302

ABSTRACT

Delta opioid receptors (δORs) regulate a number of physiological functions, and agonists for this receptor are being pursued for the treatment of mood disorders, chronic pain, and migraine. A major challenge to the development of these compounds is that, like many G-protein coupled receptors (GPCRs), agonists at the δOR can induce very different signaling and receptor trafficking events. This concept, known as ligand-directed signaling, functional selectivity, or biased agonism, can result in different agonists producing highly distinct behavioral consequences. In this chapter, we highlight the in vitro and in vivo evidence for ligand-directed signaling and trafficking at the δOR. A number of biological implications of agonist-directed signaling at the δOR have been demonstrated. Importantly, ligand-specific effects can impact both acute behavioral effects of delta agonists, as well as the long-term adaptations induced by chronic drug treatment. A better understanding of the specific signaling cascades that regulate these differential behavioral effects would help to guide rational drug design, ultimately resulting in δOR agonists with fewer adverse effects.


Subject(s)
Receptors, Opioid, delta/drug effects , Signal Transduction/drug effects , Animals , Drug Design , Humans , In Vitro Techniques , Ligands
13.
J Neurosci ; 36(12): 3541-51, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27013682

ABSTRACT

Ligand-specific recruitment of arrestins facilitates functional selectivity of G-protein-coupled receptor signaling. Here, we describe agonist-selective recruitment of different arrestin isoforms to the delta opioid receptor in mice. A high-internalizing delta opioid receptor agonist (SNC80) preferentially recruited arrestin 2 and, in arrestin 2 knock-outs (KOs), we observed a significant increase in the potency of SNC80 to inhibit mechanical hyperalgesia and decreased acute tolerance. In contrast, the low-internalizing delta agonists (ARM390, JNJ20788560) preferentially recruited arrestin 3 with unaltered behavioral effects in arrestin 2 KOs. Surprisingly, arrestin 3 KO revealed an acute tolerance to these low-internalizing agonists, an effect never observed in wild-type animals. Furthermore, we examined delta opioid receptor-Ca(2+)channel coupling in dorsal root ganglia desensitized by ARM390 and the rate of resensitization was correspondingly decreased in arrestin 3 KOs. Live-cell imaging in HEK293 cells revealed that delta opioid receptors are in pre-engaged complexes with arrestin 3 at the cell membrane and that ARM390 strengthens this membrane interaction. The disruption of these complexes in arrestin 3 KOs likely accounts for the altered responses to low-internalizing agonists. Together, our results show agonist-selective recruitment of arrestin isoforms and reveal a novel endogenous role of arrestin 3 as a facilitator of resensitization and an inhibitor of tolerance mechanisms. SIGNIFICANCE STATEMENT: Agonists that bind to the same receptor can produce highly distinct signaling events and arrestins are a major mediator of this ligand bias. Here, we demonstrate that delta opioid receptor agonists differentially recruit arrestin isoforms. We found that the high-internalizing agonist SNC80 preferentially recruits arrestin 2 and knock-out (KO) of this protein results in increased efficacy of SNC80. In contrast, low-internalizing agonists (ARM390 and JNJ20788560) preferentially recruit arrestin 3 and, surprisingly, KO of arrestin 3 produces acute tolerance and impaired receptor resensitization to these agonists. Arrestin 3 is in pre-engaged complexes with the delta opioid receptor at the cell membrane and low-internalizing agonists promote this interaction. This study reveals a novel role for arrestin 3 as a facilitator of receptor resensitization.


Subject(s)
Arrestins/metabolism , Benzamides/administration & dosage , Hyperalgesia/physiopathology , Pain Perception , Piperazines/administration & dosage , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/metabolism , Animals , Drug Tolerance , Female , Male , Mice , Mice, Knockout , Protein Isoforms
14.
J Neurosci Res ; 95(6): 1347-1354, 2017 06.
Article in English | MEDLINE | ID: mdl-28151589

ABSTRACT

Current events within the military and professional sports have resulted in an increased recognition of the long-term and debilitating consequences of traumatic brain injury. Mild traumatic brain injury accounts for the majority of head injuries, and posttraumatic headache is the most common adverse effect. It is estimated that between 30% to 90% of traumatic brain injuries result in posttraumatic headache, and for a significant number of people this headache disorder can continue for up to and over a year post injury. Often, the most severe and chronic posttraumatic headache has a migraine-like phenotype and is difficult to resolve. In this review we discuss the preclinical findings from animal models of posttraumatic headache. We also describe potential mechanisms by which traumatic brain injury leads to chronic posttraumatic headache, including neuroinflammatory mediators and migraine-associated neuropeptides. There are surprisingly few preclinical studies that have investigated overlapping mechanisms between posttraumatic headache and migraine, especially considering the prevalence and debilitating nature of posttraumatic headache. Given this context, posttraumatic headache is a field with many emerging opportunities for growth. The frequency of posttraumatic headache in the general and military population is rising, and further preclinical research is required to understand, ameliorate, and treat this disabling disorder. © 2017 Wiley Periodicals, Inc.


Subject(s)
Brain Injuries, Traumatic/complications , Post-Traumatic Headache/etiology , Translational Research, Biomedical , Animals , Brain Injuries, Traumatic/epidemiology , Brain Injuries, Traumatic/therapy , Humans , Post-Traumatic Headache/epidemiology , Post-Traumatic Headache/therapy
15.
Headache ; 62(3): 221-222, 2022 03.
Article in English | MEDLINE | ID: mdl-35293618
16.
Curr Opin Neurol ; 29(3): 314-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27023737

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to contrast the properties of the δ-opioid receptor with those of the µ-opioid receptor, which is the primary target of most currently available opioid analgesics. We also discuss preclinical evidence that indicates the potential efficacy of δ-opioid receptor agonists as migraine therapy. RECENT FINDINGS: The use of currently available opioid analgesics is highly problematic for patients with migraine. Delta-opioid receptors have key differences from µ receptors; these differences make the δ receptor an attractive therapeutic target for migraine. Delta-opioid receptors are expressed in both the peripheral and central nervous system in anatomical regions and cell types that are believed to play a role in migraine. Delta-receptor agonists have also shown promising effects in multiple migraine models, including nitroglycerin evoked hyperalgesia and conditioned place aversion, and cortical spreading depression. Evidence from animal models indicates that activation of δ receptors is less likely to cause tolerance and dependence, and less likely to cause hyperalgesia. In addition, δ receptors may have antidepressant and anxiolytic properties that are distinct from those of µ receptors. In human studies investigating other conditions, δ-receptor agonists have been generally safe and well tolerated. SUMMARY: Delta-opioid receptor agonists have promising potential as acute and/or preventive migraine therapies, without the problems associated with currently used opioid analgesics.


Subject(s)
Analgesics, Opioid/therapeutic use , Migraine Disorders/drug therapy , Receptors, Opioid, delta/agonists , Animals , Humans , Receptors, Opioid, mu/agonists , Treatment Outcome
17.
Cephalalgia ; 36(11): 1048-1056, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26682574

ABSTRACT

Background The development of novel migraine therapies has been slow, in part because of the small number of clinically relevant animal models. We have recently developed a new mouse model of chronic migraine using chronic intermittent nitroglycerin, a known human migraine trigger. The objective of this study was to validate this model by testing known and potential migraine-preventive treatments. Methods Migraine therapies were administered to male and female mice for 11 days. On day 3, mice were tested with nitroglycerin every second day for nine days. Basal and nitroglycerin-evoked mechanical hypersensitivity was evaluated using von Frey filaments. Results Chronic intermittent nitroglycerin produced acute hyperalgesia with each administration, and progressive and sustained basal hypersensitivity. The established preventive migraine therapy propranolol effectively blocked the development of acute and chronic nitroglycerin-induced hyperalgesia, while valproate had no effect. Potential migraine-preventive therapies were also tested: Amiloride inhibited nitroglycerin-induced acute and chronic hyperalgesia; while memantine was ineffective. We also tested the acute migraine therapy sumatriptan, which did not alter nitroglycerin-induced hyperalgesia, but instead resulted in acute and chronic hyperalgesia similar to that observed following nitroglycerin administration. Conclusions This study establishes the chronic nitroglycerin model as an additional screening tool to test novel migraine-preventive therapies.


Subject(s)
Disease Models, Animal , Migraine Disorders/drug therapy , Migraine Disorders/prevention & control , Nitroglycerin , Propranolol/administration & dosage , Sumatriptan/administration & dosage , Valproic Acid/administration & dosage , Acute Disease , Amiloride/administration & dosage , Animals , Anticonvulsants/administration & dosage , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Migraine Disorders/chemically induced , Pain Measurement/drug effects , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Species Specificity , Treatment Outcome , Vasodilator Agents/administration & dosage
18.
Alcohol Clin Exp Res (Hoboken) ; 48(3): 478-487, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38378262

ABSTRACT

BACKGROUND: Alcohol withdrawal-induced hyperalgesia (AWH) is characterized as an increased pain sensitivity observed after cessation of chronic alcohol use. Alcohol withdrawal-induced hyperalgesia can contribute to the negative affective state associated with abstinence and can increase susceptibility to relapse. We aimed to characterize pain sensitivity in mice during withdrawal from two different models of alcohol exposure: chronic drinking in the dark (DID) and the Lieber-DeCarli liquid diet. We also investigated whether treatment with a histone deacetylase (HDAC) inhibitor, suberoylanilide hydroxamic acid (SAHA), could ameliorate AWH in mice treated with the Lieber-DeCarli diet. METHODS: Male and female C57BL/6J mice were used for these studies. In the DID model, mice received bottles of 20% ethanol or water during the dark cycle for 4 h per day on four consecutive days per week for 6 weeks. Peripheral mechanical sensitivity was measured weekly the morning of Day 5 using von Frey filaments. In the Lieber-DeCarli model, mice received ethanol (5% v/v) or control liquid diet for 10 days, along with a single binge ethanol gavage (5 g/kg) or control gavage, respectively, on Day 10. Peripheral mechanical sensitivity was measured during the liquid diet administration and at 24 and 72 h into ethanol withdrawal. An independent group of mice that received the Lieber-DeCarli diet were administered SAHA (50 mg/kg, i.p.) during withdrawal. RESULTS: Male mice exhibited mechanical hypersensitivity after consuming ethanol for 5 weeks in the DID procedure. In the Lieber-DeCarli model, ethanol withdrawal led to hyperalgesia in both sexes. Suberoylanilide hydroxamic acid treatment during withdrawal from the ethanol liquid diet alleviated AWH. CONCLUSIONS: These results demonstrate AWH in mice after chronic binge drinking in males and after Lieber-DeCarli liquid diet administration in both sexes. Like previous findings in rats, HDAC inhibition reduced AWH in mice, suggesting that epigenetic mechanisms are involved in AWH.

19.
iScience ; 26(2): 105950, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36756376

ABSTRACT

Opioids prescribed for pain and migraine can produce opioid-induced hyperalgesia (OIH) or medication overuse headache (MOH). We previously demonstrated that pituitary adenylate cyclase activating polypeptide (PACAP) is upregulated in OIH and chronic migraine models. Here we determined if PACAP acts as a bridge between opioids and pain chronification. We tested PACAP-PAC1 receptor inhibition in novel models of opioid-exacerbated trigeminovascular pain. The PAC1 antagonist, M65, reversed chronic allodynia in a model which combines morphine with the migraine trigger, nitroglycerin. Chronic opioids also exacerbated cortical spreading depression, a correlate of migraine aura; and M65 inhibited this augmentation. In situ hybridization showed MOR and PACAP co-expression in trigeminal ganglia, and near complete overlap between MOR and PAC1 in the trigeminal nucleus caudalis and periaqueductal gray. PACAPergic mechanisms appear to facilitate the transition to chronic headache following opioid use, and strategies targeting this system may be particularly beneficial for OIH and MOH.

20.
Neuron ; 110(11): 1745-1746, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35654020

ABSTRACT

The trigeminal ganglia (TG) play a crucial role in migraine pathophysiology. In this issue of Neuron, Yang et al. developed a single-cell atlas profiling the transcriptome and epigenome of mouse and human TG, thus providing a roadmap for therapeutic targeting.


Subject(s)
Migraine Disorders , Trigeminal Ganglion , Humans , Migraine Disorders/genetics , Neurons , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL