Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
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.

2.
Neurobiol Pain ; 12: 100099, 2022.
Article in English | MEDLINE | ID: mdl-35859654

ABSTRACT

Migraine is one of the most common pain disorders and causes disability in millions of people every year. Delta opioid receptors (DOR) have been identified as a novel therapeutic target for migraine and other headache disorders. DORs are present in both peripheral and central regions and it is unclear which receptor populations regulate migraine-associated effects. The aim of this study was to determine if DOR expressed in peripheral nociceptors regulates headache associated endpoints and the effect of delta agonists within these mouse models. We used a conditional knockout, in which DOR was selectively deleted from Nav1.8 expressing cells. Nav1.8-DOR mice and loxP control littermates were tested in models of chronic migraine-associated allodynia, opioid-induced hyperalgesia, migraine-associated negative affect, and aura. Nav1.8-DOR and loxP mice had comparable effect sizes in all of these models. The anti-allodynic effect of the DOR agonist, SNC80, was slightly diminished in the nitroglycerin model of migraine. Intriguingly, in the OIH model the peripheral effects of SNC80 were completely lost in Nav1.8-DOR mice while the cephalic effects remained intact. Regardless of genotype, SNC80 continued to inhibit conditioned place aversion associated with nitroglycerin and decreased cortical spreading depression events associated with migraine aura. These results suggest that DOR in Nav1.8-expressing nociceptors do not critically regulate the anti-migraine effects of delta agonist; and that brain-penetrant delta agonists would be a more effective drug development strategy.

3.
Elife ; 102021 04 15.
Article in English | MEDLINE | ID: mdl-33856345

ABSTRACT

Migraine is the sixth most prevalent disease worldwide but the mechanisms that underlie migraine chronicity are poorly understood. Cytoskeletal flexibility is fundamental to neuronal-plasticity and is dependent on dynamic microtubules. Histone-deacetylase-6 (HDAC6) decreases microtubule dynamics by deacetylating its primary substrate, α-tubulin. We use validated mouse models of migraine to show that HDAC6-inhibition is a promising migraine treatment and reveal an undiscovered cytoarchitectural basis for migraine chronicity. The human migraine trigger, nitroglycerin, produced chronic migraine-associated pain and decreased neurite growth in headache-processing regions, which were reversed by HDAC6 inhibition. Cortical spreading depression (CSD), a physiological correlate of migraine aura, also decreased cortical neurite growth, while HDAC6-inhibitor restored neuronal complexity and decreased CSD. Importantly, a calcitonin gene-related peptide receptor antagonist also restored blunted neuronal complexity induced by nitroglycerin. Our results demonstrate that disruptions in neuronal cytoarchitecture are a feature of chronic migraine, and effective migraine therapies might include agents that restore microtubule/neuronal plasticity.


Migraines are a common brain disorder that affects 14% of the world's population. For many people the main symptom of a migraine is a painful headache, often on one side of the head. Other symptoms include increased sensitivity to light or sound, disturbed vision, and feeling sick. These sensory disturbances are called aura and they often occur before the headache begins. One particularly debilitating subset of migraines are chronic migraines, in which patients experience more than 15 headache days per month. Migraine therapies are often only partially effective or poorly tolerated, making it important to develop new drugs for this condition, but unfortunately, little is known about the molecular causes of migraines. To bridge this gap, Bertels et al. used two different approaches to cause migraine-like symptoms in mice. One approach consisted on giving mice nitroglycerin, which dilates blood vessels, produces hypersensitivity to touch, and causes photophobia in both humans and mice. In the second approach, mice underwent surgery and potassium chloride was applied onto the dura, a thick membrane that surrounds the brain. This produces cortical spreading depression, an event that is linked to migraine auras and involves a wave of electric changes in brain cells that slowly propagates across the brain, silencing brain electrical activity for several minutes. Using these approaches, Bertels et al. studied whether causing chronic migraine-like symptoms in mice is associated with changes in the structures of neurons, focusing on the effects of migraines on microtubules. Microtubules are cylindrical protein structures formed by the assembly of smaller protein units. In most cells, microtubules assemble and disassemble depending on what the cell needs. Neurons need stable microtubules to establish connections with other neurons. The experiments showed that provoking chronic migraines in mice led to a reduction in the numbers of connections between different neurons. Additionally, Bertels et al. found that inhibiting HDAC6 (a protein that destabilizes microtubules) reverses the structural changes in neurons caused by migraines and decreases migraine symptoms. The same effects are seen when a known migraine treatment strategy, known as CGRP receptor blockade, is applied. These results suggest that chronic migraines may involve decreased neural complexity, and that the restoration of this complexity by HDAC6 inhibitors could be a potential therapeutic strategy for migraine.


Subject(s)
Brain/drug effects , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Microtubules/drug effects , Migraine Disorders/drug therapy , Neuronal Plasticity/drug effects , Neurons/drug effects , Tubulin/metabolism , Acetylation , Animals , Behavior, Animal/drug effects , Brain/enzymology , Brain/physiopathology , Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Cortical Spreading Depression/drug effects , Disease Models, Animal , Female , Histone Deacetylase 6/metabolism , Male , Mice, Inbred C57BL , Microtubules/enzymology , Microtubules/pathology , Migraine Disorders/chemically induced , Migraine Disorders/enzymology , Migraine Disorders/physiopathology , Neuronal Outgrowth/drug effects , Neurons/enzymology , Neurons/pathology , Nitroglycerin , Pain Perception/drug effects , Pain Threshold/drug effects , Protein Processing, Post-Translational , Receptors, Calcitonin Gene-Related Peptide/drug effects , Receptors, Calcitonin Gene-Related Peptide/metabolism
4.
Sci Rep ; 10(1): 17629, 2020 10 19.
Article in English | MEDLINE | ID: mdl-33077757

ABSTRACT

Delta opioid receptor (DOR) agonists have been identified as a promising novel therapy for headache disorders. DORs are broadly expressed in several peripheral and central regions important for pain processing and mood regulation; and it is unclear which receptors regulate headache associated symptoms. In a model of chronic migraine-associated pain using the human migraine trigger, nitroglycerin, we observed increased expression of DOR in cortex, hippocampus, and striatum; suggesting a role for these forebrain regions in the regulation of migraine. To test this hypothesis, we used conditional knockout mice with DORs deleted from forebrain GABAergic neurons (Dlx-DOR), and investigated the outcome of this knockout on the effectiveness of the DOR agonist SNC80 in multiple headache models. In DOR loxP controls SNC80 blocked the development of acute and chronic cephalic allodynia in the chronic nitroglycerin model, an effect that was lost in Dlx-DOR mice. In addition, the anti-allodynic effects of SNC80 were lost in a model of opioid induced hyperalgesia/medication overuse headache in Dlx-DOR conditional knockouts. In a model reflecting negative affect associated with migraine, SNC80 was only effective in loxP controls and not Dlx-DOR mice. Similarly, SNC80 was ineffective in the cortical spreading depression model of migraine aura in conditional knockout mice. Taken together, these data indicate that forebrain DORs are necessary for the action of DOR agonists in relieving headache-related symptoms and suggest that forebrain regions may play an important role in migraine modulation.


Subject(s)
Analgesics, Opioid/therapeutic use , Benzamides/therapeutic use , Hyperalgesia/metabolism , Migraine Disorders/metabolism , Piperazines/therapeutic use , Prosencephalon/metabolism , Receptors, Opioid, delta/metabolism , Analgesics, Opioid/pharmacology , Animals , Benzamides/pharmacology , Cortical Spreading Depression/drug effects , Cortical Spreading Depression/physiology , Disease Models, Animal , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Mice , Mice, Knockout , Migraine Disorders/chemically induced , Migraine Disorders/drug therapy , Nitroglycerin , Piperazines/pharmacology , Prosencephalon/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics
5.
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
6.
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
7.
Cephalalgia ; 39(1): 77-90, 2019 01.
Article in English | MEDLINE | ID: mdl-29771142

ABSTRACT

BACKGROUND: Post-traumatic headache is the most common and long-lasting impairment observed following mild traumatic brain injury, and frequently has migraine-like characteristics. The mechanisms underlying progression from mild traumatic brain injury to post-traumatic headache are not fully understood. The aim of this study was to develop a mouse model of post-traumatic headache and identify mechanisms and novel targets associated with this disorder. METHODS: We combined the closed head weight-drop method and the nitroglycerin chronic migraine model. To induce mild traumatic brain injury, a weight was dropped onto intact crania of mildly anesthetized mice, and mechanical responses to chronic-intermittent administration of nitroglycerin, a human migraine trigger, were determined at multiple time points post-injury. RESULTS: Low dose nitroglycerin (0.1 mg/kg) evoked acute periorbital and hind paw allodynia in both mild traumatic brain injury and sham animals. However, only mild traumatic brain injury mice developed chronic hypersensitivity to low dose nitroglycerin. Migraine medications, sumatriptan and topiramate, inhibited post-traumatic headache-associated allodynia. In addition, the delta opioid receptor agonist, SNC80, also blocked post-traumatic headache-associated allodynia. Finally, we examined the expression of calcitonin gene-related peptide within this model and found that it was increased in trigeminal ganglia two weeks post-mild traumatic brain injury. CONCLUSIONS: Overall, we have established a mouse model of post-traumatic headache and identified the delta opioid receptor as a novel therapeutic target for this disorder.


Subject(s)
Brain Concussion/complications , Disease Models, Animal , Post-Traumatic Headache/etiology , Post-Traumatic Headache/metabolism , Receptors, Opioid, delta/metabolism , Animals , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Migraine Disorders/etiology , Migraine Disorders/metabolism , Trigeminal Ganglion/metabolism
8.
Neuropharmacology ; 148: 77-86, 2019 04.
Article in English | MEDLINE | ID: mdl-30553828

ABSTRACT

Headaches are highly disabling and are among the most common neurological disorders worldwide. Despite the high prevalence of headache, therapeutic options are limited. We recently identified the delta opioid receptor (DOR) as an emerging therapeutic target for migraine. In this study, we examined the effectiveness of a hallmark DOR agonist, SNC80, in disease models reflecting diverse headache disorders including: chronic migraine, post-traumatic headache (PTH), medication overuse headache by triptans (MOH), and opioid-induced hyperalgesia (OIH). To model chronic migraine C57BL/6J mice received chronic intermittent treatment with the known human migraine trigger, nitroglycerin. PTH was modeled by combining the closed head weight drop model with the nitroglycerin model of chronic migraine. For MOH and OIH, mice were chronically treated with sumatriptan or morphine, respectively. The development of periorbital and peripheral allodynia was observed in all four models; and SNC80 significantly inhibited allodynia in all cases. In addition, we also determined if chronic daily treatment with SNC80 would induce MOH/OIH, and we observed limited hyperalgesia relative to sumatriptan or morphine. Together, our results indicate that DOR agonists could be effective in multiple headache disorders, despite their distinct etiology, thus presenting a novel therapeutic target for headache.


Subject(s)
Benzamides/therapeutic use , Headache Disorders/drug therapy , Piperazines/therapeutic use , Receptors, Opioid, delta/agonists , Animals , Benzamides/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Headache Disorders/chemically induced , Hyperalgesia/drug therapy , Male , Mice , Morphine , Nitroglycerin , Piperazines/adverse effects , Receptors, Opioid, delta/therapeutic use , Sumatriptan
9.
Br J Pharmacol ; 175(14): 3050-3059, 2018 07.
Article in English | MEDLINE | ID: mdl-29722902

ABSTRACT

BACKGROUND AND PURPOSE: Opioid δ receptor agonists are potent antihyperalgesics in chronic pain models, but tolerance develops after prolonged use. Previous evidence indicates that distinct forms of tolerance occur depending on the internalization properties of δ receptor agonists. As arrestins are important in receptor internalization, we investigated the role of arrestin 2 (ß-arrestin 1) in mediating the development of tolerance induced by high- and low-internalizing δ receptor agonists. EXPERIMENTAL APPROACH: We evaluated the effect of two δ receptor agonists with similar analgesic potencies, but either high-(SNC80) or low-(ARM390) internalization properties in wild-type (WT) and arrestin 2 knockout (KO) mice. We compared tolerance to the antihyperalgesic effects of these compounds in a model of inflammatory pain. We also examined tolerance to the convulsant effect of SNC80. Furthermore, effect of chronic treatment with SNC80 on δ agonist-stimulated [35 S]-GTPγS binding was determined in WT and KO mice. KEY RESULTS: Arrestin 2 KO resulted in increased drug potency, duration of action and decreased acute tolerance to the antihyperalgesic effects of SNC80. In contrast, ARM390 produced similar effects in both WT and KO animals. Following chronic treatment, we found a marked decrease in the extent of tolerance to SNC80-induced antihyperalgesia and convulsions in arrestin 2 KO mice. Accordingly, δ receptors remained functionally coupled to G proteins in arrestin 2 KO mice chronically treated with SNC80. CONCLUSIONS AND IMPLICATIONS: Overall, these results suggest that δ receptor agonists interact with arrestins in a ligand-specific manner, and tolerance to high- but not low-internalizing agonists are preferentially regulated by arrestin 2.


Subject(s)
Analgesics/pharmacology , Benzamides/pharmacology , Drug Tolerance/physiology , Piperazines/pharmacology , Receptors, Opioid, delta/agonists , beta-Arrestin 1/metabolism , Analgesics/therapeutic use , Animals , Benzamides/therapeutic use , Brain/drug effects , Brain/metabolism , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Mice, Knockout , Pain/drug therapy , Pain/metabolism , Piperazines/therapeutic use , Receptors, Opioid, delta/metabolism , Seizures/chemically induced , beta-Arrestin 1/genetics
10.
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
11.
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
12.
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
13.
Methods Mol Biol ; 1230: 87-104, 2015.
Article in English | MEDLINE | ID: mdl-25293318

ABSTRACT

G-protein-coupled receptors (GPCRs) regulate a remarkable diversity of biological functions, and are thus often targeted for drug therapies. Receptor internalization is commonly observed following agonist binding and activation. Receptor trafficking events have been well characterized in cell systems, but the in vivo significance of GPCR internalization is still poorly understood. To address this issue, we have developed an innovative knock-in mouse model, where an opioid receptor is directly visible in vivo. These knockin mice express functional fluorescent delta opioid receptors (DOR-eGFP) in place of the endogenous receptor, and these receptors are expressed at physiological levels within their native environment. DOR-eGFP mice have proven to be an extraordinary tool in studying receptor neuroanatomy, real-time receptor trafficking in live neurons, and in vivo receptor internalization. We have used this animal model to determine the relationship between receptor trafficking in neurons and receptor function at a behavioral level. Here, we describe in detail the construction and characterization of this knockin mouse. We also outline how to use these mice to examine the behavioral consequences of agonist-specific trafficking at the delta opioid receptor. These techniques are potentially applicable to any GPCR, and highlight the powerful nature of this imaging tool.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, Opioid, delta/metabolism , Receptors, Opioid/chemistry , Animals , Mice , Neurons/metabolism , Phosphorylation , Protein Transport/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid/metabolism , Receptors, Opioid, delta/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...