Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
iScience ; 26(5): 106669, 2023 May 19.
Article in English | MEDLINE | ID: mdl-37182109

ABSTRACT

The ubiquitous volume-regulated anion channels (VRACs) facilitate cell volume control and contribute to many other physiological processes. Treatment with non-specific VRAC blockers or brain-specific deletion of the essential VRAC subunit LRRC8A is highly protective in rodent models of stroke. Here, we tested the widely accepted idea that the harmful effects of VRACs are mediated by release of the excitatory neurotransmitter glutamate. We produced conditional LRRC8A knockout either exclusively in astrocytes or in the majority of brain cells. Genetically modified mice were subjected to an experimental stroke (middle cerebral artery occlusion). The astrocytic LRRC8A knockout yielded no protection. Conversely, the brain-wide LRRC8A deletion strongly reduced cerebral infarction in both heterozygous (Het) and full KO mice. Yet, despite identical protection, Het mice had full swelling-activated glutamate release, whereas KO animals showed its virtual absence. These findings suggest that LRRC8A contributes to ischemic brain injury via a mechanism other than VRAC-mediated glutamate release.

2.
FASEB J ; 35(10): e21869, 2021 10.
Article in English | MEDLINE | ID: mdl-34469026

ABSTRACT

The leucine-rich repeat-containing family 8 member A (LRRC8A) is an essential subunit of the volume-regulated anion channel (VRAC). VRAC is critical for cell volume control, but its broader physiological functions remain under investigation. Recent studies in the field indicate that Lrrc8a disruption in the brain astrocytes reduces neuronal excitability, impairs synaptic plasticity and memory, and protects against cerebral ischemia. In the present work, we generated brain-wide conditional LRRC8A knockout mice (LRRC8A bKO) using NestinCre -driven Lrrc8aflox/flox excision in neurons, astrocytes, and oligodendroglia. LRRC8A bKO animals were born close to the expected Mendelian ratio and developed without overt histological abnormalities, but, surprisingly, all died between 5 and 9 weeks of age with a seizure phenotype, which was confirmed by video and EEG recordings. Brain slice electrophysiology detected changes in the excitability of pyramidal cells and modified GABAergic inputs in the hippocampal CA1 region of LRRC8A bKO. LRRC8A-null hippocampi showed increased immunoreactivity of the astrocytic marker GFAP, indicating reactive astrogliosis. We also found decreased whole-brain protein levels of the GABA transporter GAT-1, the glutamate transporter GLT-1, and the astrocytic enzyme glutamine synthetase. Complementary HPLC assays identified reduction in the tissue levels of the glutamate and GABA precursor glutamine. Together, these findings suggest that VRAC provides vital control of brain excitability in mouse adolescence. VRAC deletion leads to a lethal phenotype involving progressive astrogliosis and dysregulation of astrocytic uptake and supply of amino acid neurotransmitters and their precursors.


Subject(s)
Astrocytes/pathology , Gliosis/mortality , Glutamic Acid/metabolism , Membrane Proteins/physiology , Seizures/mortality , Animals , Astrocytes/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Female , Gliosis/etiology , Gliosis/pathology , Ion Transport , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Seizures/etiology , Seizures/pathology
3.
J Neurosci Methods ; 294: 1-6, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29061345

ABSTRACT

BACKGROUND: Morphometric analyses of biological features have become increasingly common in recent years with such analyses being subject to a large degree of observer bias, variability, and time consumption. While commercial software packages exist to perform these analyses, they are expensive, require extensive user training, and are usually dependent on the observer tracing the morphology. NEW METHOD: To address these issues, we have developed a broadly applicable, no-cost ImageJ plugin we call 'BranchAnalysis2D/3D', to perform morphometric analyses of structures with branching morphologies, such as neuronal dendritic spines, vascular morphology, and primary cilia. RESULTS: Our BranchAnalysis2D/3D algorithm allows for rapid quantification of the length and thickness of branching morphologies, independent of user tracing, in both 2D and 3D data sets. COMPARISON WITH EXISTING METHODS: We validated the performance of BranchAnalysis2D/3D against pre-existing software packages using trained human observers and images from brain and retina. We found that the BranchAnalysis2D/3D algorithm outputs results similar to available software (i.e., Metamorph, AngioTool, Neurolucida), while allowing faster analysis times and unbiased quantification. CONCLUSIONS: BranchAnalysis2D/3D allows inexperienced observers to output results like a trained observer but more efficiently, thereby increasing the consistency, speed, and reliability of morphometric analyses.


Subject(s)
Brain/cytology , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Neurons/cytology , Software , Algorithms , Animals , Mice , Observer Variation , Reproducibility of Results , Retina/anatomy & histology
4.
J Neurosci ; 36(28): 7485-96, 2016 07 13.
Article in English | MEDLINE | ID: mdl-27413158

ABSTRACT

UNLABELLED: The occurrence of recurrent, unprovoked seizures is the hallmark of human epilepsy. Currently, only two-thirds of this patient population has adequate seizure control. New epilepsy models provide the potential for not only understanding the development of spontaneous seizures, but also for testing new strategies to treat this disorder. Here, we characterize a primary generalized seizure model of epilepsy following repeated exposure to the GABAA receptor antagonist, flurothyl, in which mice develop spontaneous seizures that remit within 1 month. In this model, we expose C57BL/6J mice to flurothyl until they experience a generalized seizure. Each of these generalized seizures typically lasts <30 s. We induce one seizure per day for 8 d followed by 24 h video-electroencephalographic recordings. Within 1 d following the last of eight flurothyl-induced seizures, ∼50% of mice have spontaneous seizures. Ninety-five percent of mice tested have seizures within the first week of the recording period. Of the spontaneous seizures recorded, the majority are generalized clonic seizures, with the remaining 7-12% comprising generalized clonic seizures that transition into brainstem seizures. Over the course of an 8 week recording period, spontaneous seizure episodes remit after ∼4 weeks. Overall, the repeated flurothyl paradigm is a model of epileptogenesis with spontaneous seizures that remit. This model provides an additional tool in our armamentarium for understanding the mechanisms underlying epileptogenesis and may provide insights into why spontaneous seizures remit without anticonvulsant treatment. Elucidating these processes could lead to the development of new epilepsy therapeutics. SIGNIFICANCE STATEMENT: Epilepsy is a chronic disorder characterized by the occurrence of recurrent, unprovoked seizures in which the individual seizure-ictal events are self-limiting. Remission of recurrent, unprovoked seizures can be achieved in two-thirds of cases by treatment with anticonvulsant medication, surgical resection, and/or nerve/brain electrode stimulation. However, there are examples in humans of epilepsy with recurrent, unprovoked seizures remitting without any intervention. While elucidating how recurrent, unprovoked seizures develop is critical for understanding epileptogenesis, an understanding of how and why recurrent, unprovoked seizures remit may further our understanding and treatment of epilepsy. Here, we describe a new model of recurrent, unprovoked spontaneous seizures in which the occurrence of spontaneous seizures naturally remits over time without any therapeutic intervention.


Subject(s)
Convulsants/toxicity , Flurothyl/toxicity , Seizures/chemically induced , Analysis of Variance , Animals , Anticonvulsants/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Electroencephalography , Fluoresceins/metabolism , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Seizures/drug therapy , Seizures/pathology , Time Factors , Video Recording
5.
Drug Metab Dispos ; 43(9): 1326-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26109562

ABSTRACT

Cytochrome P450 monooxygenases (P450s), which are well-known drug-metabolizing enzymes, are thought to play a signal transduction role in µ opioid analgesia and may serve as high-affinity (3)H-cimetidine ((3)HCIM) binding sites in the brain. (3)HCIM binding sites may also be related to opioid or nonopioid analgesia. However, of the more than 100 murine P450 enzymes, the specific isoform(s) responsible for either function have not been identified. Presently, three lines of constitutive P450 gene cluster knockout (KO) mice with full-length deletions of 14 Cyp2c, 9 Cyp2d, and 7 Cyp3a genes were studied for deficiencies in (3)HCIM binding and for opioid analgesia. Liver and brain homogenates from all three genotypes showed normal (3)HCIM binding values, indicating that gene products of Cyp2d, Cyp3a, and Cyp2c are not (3)HCIM-binding proteins. Cyp2d KO and Cyp3a KO mice showed normal antinociceptive responses to a moderate systemic dose of morphine (20 mg/kg, s.c.), thereby excluding 16 P450 isoforms as mediators of opioid analgesia. In contrast, Cyp2c KO mice showed a 41% reduction in analgesic responses following systemically (s.c.) administered morphine. However, the significance of brain Cyp2c gene products in opioid analgesia is uncertain because little or no analgesic deficits were noted in Cyp2c KO mice following intracerebroventricular or intrathecalmorphine administration, respectively. These results show that the gene products of Cyp2d and Cyp3a do not contribute to µ opioid analgesia in the central nervous system. A possible role for Cyp2c gene products in opioid analgesia requires further consideration.


Subject(s)
Analgesics, Opioid/administration & dosage , Cytochrome P-450 Enzyme System/metabolism , Isoenzymes/metabolism , Analgesics, Opioid/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , Isoenzymes/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Brain Res ; 1616: 10-8, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-25935691

ABSTRACT

Recent studies suggest a functional role for neuronal cytochrome P450 monooxygenase (P450) activity in opioid analgesia. To characterize the relevant receptors, brain areas, and circuits, detailed in vitro and in vivo studies were performed with the highly selective µ opioid receptor agonist DAMGO in neuronal P450-deficient mutant (Null) and control mice. Homogenates of brain regions and spinal cord showed no differences in DAMGO-induced activation of [(35)S]- GTPγS binding between Null and control mice, indicating no genotype differences in µ opioid receptor signaling, receptor affinities or receptor densities. Intracerebroventricular (icv) DAMGO produced robust, near-maximal, analgesic responses in control mice which were attenuated by 50% in Null mice, confirming a role for µ opioid receptors in activating P450-associated responses. Intra-periaqueductal gray (PAG) and intra-rostral ventromedial medulla (RVM) injections of DAMGO revealed deficits in Null (vs. control) analgesic responses, yet no such genotype differences were observed after intrathecal DAMGO administration. Taken with earlier published findings, the present results suggest that activation of µ opioid receptors in both the PAG and in the RVM relieves pain by mechanisms which include nerve-terminal P450 enzymes within inhibitory PAG-RVM projections. Spinal opioid analgesia, however, does not seem to require such P450 enzyme activity.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , Brain/metabolism , Cytochrome P-450 Enzyme System/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Neurons/metabolism , Nociception/drug effects , Analysis of Variance , Animals , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , Drug Administration Routes , Female , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Male , Mice , Mice, Transgenic , Microinjections , Neurons/drug effects , Periaqueductal Gray/cytology , Protein Binding/drug effects , Protein Binding/genetics , Reaction Time/drug effects , Reaction Time/genetics , Time Factors
7.
Free Radic Biol Med ; 77: 168-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224033

ABSTRACT

The contribution of oxidative stress to ischemic brain damage is well established. Nevertheless, for unknown reasons, several clinically tested antioxidant therapies have failed to show benefits in human stroke. Based on our previous in vitro work, we hypothesized that the neuroprotective potency of antioxidants is related to their ability to limit the release of the excitotoxic amino acids glutamate and aspartate. We explored the effects of two antioxidants, tempol and edaravone, on amino acid release in the brain cortex, in a rat model of transient occlusion of the middle cerebral artery (MCAo). Amino acid levels were quantified using a microdialysis approach, with the probe positioned in the ischemic penumbra as verified by a laser Doppler technique. Two-hour MCAo triggered a dramatic increase in the levels of glutamate, aspartate, taurine, and alanine. Microdialysate delivery of 10mM tempol reduced the amino acid release by 60-80%, whereas matching levels of edaravone had no effect. In line with these data, an intracerebroventricular injection of tempol but not edaravone (500 nmol each, 15 min before MCAo) reduced infarction volumes by ~50% and improved neurobehavioral outcomes. In vitro assays showed that tempol was superior at removing superoxide anion, whereas edaravone was more potent at scavenging hydrogen peroxide, hydroxyl radical, and peroxynitrite. Overall, our data suggest that the neuroprotective properties of tempol are probably related to its ability to reduce tissue levels of the superoxide anion and pathological glutamate release and, in such a way, limit progression of brain infarction within ischemic penumbra. These new findings may be instrumental in developing new antioxidant therapies for treatment of stroke.


Subject(s)
Cyclic N-Oxides/pharmacology , Glutamic Acid/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Alanine/metabolism , Animals , Antipyrine/analogs & derivatives , Antipyrine/chemistry , Antipyrine/pharmacology , Astrocytes/metabolism , Brain/drug effects , Brain/pathology , Cells, Cultured , Cyclic N-Oxides/chemistry , Drug Evaluation, Preclinical , Edaravone , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Molecular Mimicry , Neuroprotective Agents/chemistry , Oxidative Stress , Rats, Sprague-Dawley , Spin Labels , Superoxides/metabolism , Synaptosomes/drug effects , Taurine/metabolism
8.
Eur J Pharmacol ; 740: 255-62, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-25062792

ABSTRACT

Morphine-like analgesics act on µ opioid receptors in the CNS to produce highly effective pain relief, but the same class of receptors also mediates non-therapeutic side effects. The analgesic properties of morphine were recently shown to require the activity of a brain neuronal cytochrome P450 epoxygenase, but the significance of this pathway for opioid side effects is unknown. Here we show that brain P450 activity is not required for three of morphine׳s major side effects (respiratory depression, constipation, and locomotor stimulation). Following systemic or intracerebroventricular administration of morphine, transgenic mice with brain neuron - specific reductions in P450 activity showed highly attenuated analgesic responses as compared with wild-type (control) mice. However, brain P450-deficient mice showed normal morphine-induced side effects (respiratory depression, locomotor stimulation, and inhibition of intestinal motility). Pretreatment of control mice with the P450 inhibitor CC12 similarly reduced the analgesia, but not these side effects of morphine. Because activation of brain µ opioid receptors produces both opioid analgesia and opioid side effects, dissociation of the mechanisms for the therapeutic and therapy-limiting effects of opioids has important consequences for the development of analgesics with reduced side effects and/or limited addiction liability.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/enzymology , Morphine/pharmacology , NADPH-Ferrihemoprotein Reductase/deficiency , Neurons/enzymology , Analgesia , Analgesics, Opioid/adverse effects , Animals , Behavior, Animal/drug effects , Body Temperature/drug effects , Female , Gastrointestinal Motility/drug effects , Male , Mice, Knockout , Morphine/adverse effects , Motor Activity/drug effects , NADPH-Ferrihemoprotein Reductase/genetics , Pain Threshold/drug effects , Respiratory Rate/drug effects
9.
Brain Res ; 1578: 30-7, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25020125

ABSTRACT

Stressful environmental changes can suppress nociceptive transmission, a phenomenon known as "stress-induced analgesia". Depending on the stressor and the subject, opioid or non-opioid mechanisms are activated. Brain µ opioid receptors mediate analgesia evoked either by exogenous agents (e.g. morphine), or by the release of endogenous opioids following stressful procedures. Recent work with morphine and neuronal cytochrome P450 (P450)-deficient mice proposed a signal transduction role for P450 enzymes in µ analgesia. Since µ opioid receptors also mediate some forms of stress-induced analgesia, the present studies assessed the significance of brain P450 activity in opioid-mediated stress-induced analgesia. Two widely-used models of opioid stress-induced analgesia (restraint and warm water swim) were studied in both sexes of wild-type control and P450-deficient (Null) mice. In control mice, both stressors evoked moderate analgesic responses which were blocked by pretreatment with the opioid antagonist naltrexone, confirming the opioid nature of these responses. Consistent with literature, sex differences (control female>control male) were seen in swim-induced, but not restraint-induced, analgesia. Null mice showed differential responses to the two stress paradigms. As compared with control subjects, Null mice showed highly attenuated restraint-induced analgesia, showing a critical role for neuronal P450s in this response. However, warm water swim-induced analgesia was unchanged in Null vs. control mice. Additional control experiments confirmed the absence of morphine analgesia in Null mice. These results are the first to show that some forms of opioid-mediated stress-induced analgesia require brain neuronal P450 activity.


Subject(s)
Analgesics, Opioid/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Morphine/pharmacology , Stress, Psychological/enzymology , Analgesia , Animals , Brain/enzymology , Cytochrome P-450 Enzyme System/genetics , Female , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Neurons/enzymology , Nociception/physiology , Restraint, Physical , Swimming
10.
Eur J Pharmacol ; 714(1-3): 464-71, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23834775

ABSTRACT

Improgan, a non-opioid, antinociceptive drug, activates descending analgesic circuits following brain administration, but the improgan receptor remains unidentified. Since biotinylation of drugs can enhance drug potency or facilitate discovery of new drug targets, a biotinylated congener of improgan (CC44) and several related compounds were synthesized and tested for antinociceptive activity. In rats and mice, intracerebroventricular (i.c.v.) administration of CC44 produced dose-dependent reductions in thermal nociceptive (tail flick and hot plate) responses, with 5-fold greater potency than improgan. CC44 also robustly attenuated mechanical (tail pinch) nociception in normal rats and mechanical allodynia in a spinal nerve ligation model of neuropathic pain. Similar to the effects of improgan, CC44 antinociception was reversed by the GABAA agonist muscimol (consistent with activation of analgesic circuits), and was resistant to the opioid antagonist naltrexone (implying a non-opioid mechanism). Also like improgan, CC44 produced thermal antinociception when microinjected into the rostral ventromedial medulla (RVM). Unlike improgan, CC44 (i.c.v.) produced antinociception which was resistant to antagonism by the cannabinoid CB1 antagonist/inverse agonist rimonabant. CC44 was inactive in mice following systemic administration, indicating that CC44 does not penetrate the brain. Preliminary findings with other CC44 congeners suggest that the heteroaromatic nucleus (imidazole), but not the biotin moiety, is required for CC44's antinociceptive activity. These findings demonstrate that CC44 is a potent analgesic compound with many improgan-like characteristics. Since powerful techniques are available to characterize and identify the binding partners for biotin-containing ligands, CC44 may be useful in searching for new receptors for analgesic drugs.


Subject(s)
Analgesics/chemistry , Analgesics/pharmacology , Biotinylation , Cimetidine/analogs & derivatives , Analgesics/metabolism , Analgesics/therapeutic use , Animals , Avidin/metabolism , Cimetidine/chemistry , Cimetidine/metabolism , Cimetidine/pharmacology , Cimetidine/therapeutic use , Hyperalgesia/drug therapy , Male , Medulla Oblongata/pathology , Mice , Neurons/drug effects , Neurons/pathology , Rats , Streptavidin/metabolism
11.
Brain Res ; 1499: 1-11, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23298831

ABSTRACT

Brain cytochrome P450 epoxygenases were recently shown to play an essential role in mediating the pain-relieving properties of morphine. To identify the CNS sites containing the morphine-relevant P450s, the effects of intracerebral (ic) microinjections of the P450 inhibitor CC12 were determined on morphine antinociception in rats. CC12 inhibited morphine antinociception when both drugs were injected into the rostral ventromedial medulla (RVM), but not following co-injections into the periaqueductal gray (PAG) or into the spinal subarachnoid space. In addition, intra-RVM CC12 pretreatment nearly completely blocked the effects of morphine following intracerebroventricular (icv) administration. Although morphine is thought to act in both the PAG and RVM by pre-synaptic inhibition of inhibitory GABAergic transmission, the present findings show that 1) the mechanism of morphine action differs between these two brainstem areas, and 2) P450 activity within the RVM is important for supraspinal morphine antinociception. Characterization of morphine-P450 interactions within RVM circuits will further enhance the understanding of the biochemistry of pain relief.


Subject(s)
Analgesics, Opioid/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Imidazoles/pharmacology , Medulla Oblongata/drug effects , Morphine/pharmacology , Sulfides/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Male , Pain/physiopathology , Rats , Rats, Sprague-Dawley
12.
J Neurophysiol ; 108(9): 2393-404, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956800

ABSTRACT

Respiratory depression is a therapy-limiting side effect of opioid analgesics, yet our understanding of the brain circuits mediating this potentially lethal outcome remains incomplete. Here we studied the contribution of the rostral ventromedial medulla (RVM), a region long implicated in pain modulation and homeostatic regulation, to opioid-induced respiratory depression. Microinjection of the µ-opioid agonist DAMGO in the RVM of lightly anesthetized rats produced both analgesia and respiratory depression, showing that neurons in this region can modulate breathing. Blocking opioid action in the RVM by microinjecting the opioid antagonist naltrexone reversed the analgesic and respiratory effects of systemically administered morphine, showing that this region plays a role in both the analgesic and respiratory-depressant properties of systemically administered morphine. The distribution of neurons directly inhibited by RVM opioid microinjection was determined with a fluorescent opioid peptide, dermorphin-Alexa 594, and found to be concentrated in and around the RVM. The non-opioid analgesic improgan, like DAMGO, produced antinociception but, unlike DAMGO, stimulated breathing when microinjected into the RVM. Concurrent recording of RVM neurons during improgan microinjection showed that this agent activated RVM ON-cells, OFF-cells, and NEUTRAL-cells. Since opioids are known to activate OFF-cells but suppress ON-cell firing, the differential respiratory response to these two analgesic drugs is best explained by their opposing effects on the activity of RVM ON-cells. These findings show that pain relief can be separated pharmacologically from respiratory depression and identify RVM OFF-cells as important central targets for continued development of potent analgesics with fewer side effects.


Subject(s)
Analgesics, Opioid/toxicity , Medulla Oblongata/drug effects , Neurons/physiology , Nociceptive Pain/physiopathology , Respiratory Insufficiency/chemically induced , Analgesics, Opioid/antagonists & inhibitors , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/antagonists & inhibitors , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Evoked Potentials/drug effects , Evoked Potentials/physiology , Male , Medulla Oblongata/cytology , Medulla Oblongata/physiology , Morphine/antagonists & inhibitors , Morphine/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Nociception/drug effects , Nociception/physiology , Rats , Rats, Sprague-Dawley , Respiratory Insufficiency/physiopathology
13.
Brain Res ; 1424: 32-7, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-22015352

ABSTRACT

Improgan, a non-opioid analgesic, is known to act in the rodent brain stem to produce highly effective antinociception in several acute pain tests. However, improgan has not been studied in any models of chronic pain. To assess the efficacy of improgan in an animal model of neuropathic pain, the effects of this drug were studied on mechanical allodynia following unilateral spinal nerve ligation (SNL) in rats. Intracerebroventricular (icv) improgan (40-80 µg) produced complete, reversible, dose-dependent attenuation of hind paw mechanical allodynia for up to 1h after administration, with no noticeable behavioral or motor side effects. Intracerebral (ic) microinjections of improgan (5-30 µg) into the rostral ventromedial medulla (RVM) also reversed the allodynia, showing this brain area to be an important site for improgan's action. The recently-demonstrated suppression of RVM ON-cell activity by improgan may account for the presently-observed anti-allodynic activity. The present findings suggest that brain-penetrating, improgan-like drugs developed for human use could be effective medications for the treatment of neuropathic pain.


Subject(s)
Analgesics, Non-Narcotic/administration & dosage , Cimetidine/analogs & derivatives , Neuralgia/drug therapy , Animals , Axotomy , Chronic Pain/drug therapy , Cimetidine/administration & dosage , Injections, Intraventricular , Male , Medulla Oblongata/drug effects , Rats , Rats, Sprague-Dawley
14.
Pain ; 152(4): 878-887, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21316152

ABSTRACT

The search for the mechanism of action of improgan (a nonopioid analgesic) led to the recent discovery of CC12, a compound that blocks improgan antinociception. Because CC12 is a cytochrome P450 inhibitor, and brain P450 mechanisms were recently shown to be required in opioid analgesic signaling, pharmacological and transgenic studies were performed in rodents to test the hypothesis that improgan antinociception requires brain P450 epoxygenase activity. Intracerebroventricular (i.c.v.) administration of the P450 inhibitors miconazole and fluconazole, and the arachidonic acid (AA) epoxygenase inhibitor N-methylsulfonyl-6-(2-propargyloxyphenyl)hexanamide (MS-PPOH) potently inhibited improgan antinociception in rats at doses that were inactive alone. MW06-25, a new P450 inhibitor that combines chemical features of CC12 and miconazole, also potently blocked improgan antinociception. Although miconazole and CC12 were weakly active at opioid and histamine H(3) receptors, MW06-25 showed no activity at these sites, yet retained potent P450-inhibiting properties. The P450 hypothesis was also tested in Cpr(low) mice, a viable knock-in model with dramatically reduced brain P450 activity. Improgan (145 nmol, i.c.v.) antinociception was reduced by 37% to 59% in Cpr(low) mice, as compared with control mice. Moreover, CC12 pretreatment (200 nmol, i.c.v.) abolished improgan action (70% to 91%) in control mice, but had no significant effect in Cpr(low) mice. Thus, improgan's activation of bulbospinal nonopioid analgesic circuits requires brain P450 epoxygenase activity. A model is proposed in which (1) improgan activates an unknown receptor to trigger downstream P450 activity, and (2) brainstem epoxygenase activity is a point of convergence for opioid and nonopioid analgesic signaling.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Brain/drug effects , Cimetidine/analogs & derivatives , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Enzymologic/drug effects , 14-alpha Demethylase Inhibitors/pharmacology , Amides/pharmacology , Analgesics, Opioid/pharmacokinetics , Animals , Brain/metabolism , Cell Line, Transformed , Cimetidine/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Injections, Intraventricular/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Miconazole/pharmacology , NADPH-Ferrihemoprotein Reductase/deficiency , Naltrexone/analogs & derivatives , Naltrexone/pharmacokinetics , Narcotic Antagonists/pharmacokinetics , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptors, Histamine H3/metabolism , Sulfides/pharmacology , Time Factors , Tritium/pharmacokinetics
15.
J Neurophysiol ; 104(6): 3222-30, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926616

ABSTRACT

Many analgesic drugs, including µ-opioids, cannabinoids, and the novel nonopioid analgesic improgan, produce antinociception by actions in the rostral ventromedial medulla (RVM). There they activate pain-inhibiting neurons, termed "OFF-cells," defined by a nociceptive reflex-related pause in activity. Based on recent functional evidence that neuronal P450 epoxygenases are important for the central antinociceptive actions of morphine and improgan, we explored the convergence of opioid and nonopioid analgesic drug actions in RVM by studying the effects of the P450 epoxygenase inhibitor CC12 on the analgesic drug-induced activation of these OFF-cells and on behavioral antinociception. In rats lightly anesthetized with isoflurane, we recorded the effects of intraventricular morphine and improgan, with and without CC12 pretreatment, on tail flick latency and activity of identified RVM neurons: OFF-cells, ON-cells (pronociceptive neurons), and neutral cells (unresponsive to analgesic drugs). CC12 pretreatment preserved reflex-related changes in OFF-cell firing and blocked the analgesic actions of both drugs, without interfering with the increase in spontaneous firing induced by improgan or morphine. CC12 blocked suppression of evoked ON-cell firing by improgan, but not morphine. CC12 pretreatment had no effect by itself on RVM neurons or behavior. These data show that the epoxygenase inhibitor CC12 works downstream from receptors for both µ-opioid and improgan, at the inhibitory input mediating the OFF-cell pause. This circuit-level analysis thus provides a cellular basis for the convergence of opioid and nonopioid analgesic actions in the RVM. A presynaptic P450 epoxygenase may therefore be an important target for development of clinically useful nonopioid analgesic drugs.


Subject(s)
Analgesics/antagonists & inhibitors , Cimetidine/analogs & derivatives , Imidazoles/pharmacology , Medulla Oblongata/drug effects , Morphine/antagonists & inhibitors , Pain Perception/drug effects , Receptors, Opioid, mu/drug effects , Sulfides/pharmacology , Action Potentials/drug effects , Animals , Cimetidine/antagonists & inhibitors , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System , Male , Medulla Oblongata/cytology , Medulla Oblongata/physiology , Models, Neurological , Pain Perception/physiology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Receptor, Cannabinoid, CB1/physiology , Receptors, Opioid, mu/physiology , Receptors, Presynaptic/drug effects , Receptors, Presynaptic/physiology , Signal Transduction/drug effects , gamma-Aminobutyric Acid/physiology
16.
Eur J Pharmacol ; 632(1-3): 33-8, 2010 Apr 25.
Article in English | MEDLINE | ID: mdl-20138862

ABSTRACT

[(3)H]cimetidine, a radiolabeled histamine H(2) receptor antagonist, binds with high affinity to an unknown hemoprotein in the brain which is not the histamine H(2) receptor. Improgan, a close chemical congener of cimetidine, is a highly effective pain-relieving drug following CNS administration, yet its mechanism of action remains unknown. To test the hypothesis that the [(3)H]cimetidine-binding site is the improgan antinociceptive target, improgan, cimetidine, and 8 other chemical congeners were studied as potential inhibitors of [(3)H]cimetidine binding in membrane fractions from the rat brain. All compounds produced a concentration-dependent inhibition of [(3)H]cimetidine binding over a 500-fold range of potencies (K(i) values were 14.5 to >8000nM). However, antinociceptive potencies in rats did not significantly correlate with [(3)H]cimetidine-binding affinities (r=0.018, p=0.97, n=10). These results suggest that the [(3)H]cimetidine-binding site is not the analgesic target for improgan-like drugs.


Subject(s)
Analgesics/pharmacology , Brain/metabolism , Cimetidine/analogs & derivatives , Cimetidine/antagonists & inhibitors , Analgesics/chemistry , Animals , Binding Sites , Cimetidine/chemistry , Cimetidine/pharmacology , Dose-Response Relationship, Drug , Histamine/metabolism , Histamine H2 Antagonists/metabolism , Male , Molecular Structure , Pain/metabolism , Rats , Rats, Sprague-Dawley
17.
Nat Neurosci ; 13(3): 284-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20139973

ABSTRACT

To assess the importance of brain cytochrome P450 (P450) activity in mu opioid analgesic action, we generated a mutant mouse with brain neuron-specific reductions in P450 activity; these mice showed highly attenuated morphine antinociception compared with controls. Pharmacological inhibition of brain P450 arachidonate epoxygenases also blocked morphine antinociception in mice and rats. Our findings indicate that a neuronal P450 epoxygenase mediates the pain-relieving properties of morphine.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , Cytochrome P-450 Enzyme System/drug effects , Neurons/drug effects , Pain/drug therapy , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/administration & dosage , Animals , Brain/enzymology , Brain/metabolism , Cytochrome P-450 CYP2J2 , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Male , Mice , Mice, Transgenic , Morphine/administration & dosage , Morphine/pharmacology , Neural Pathways/drug effects , Neural Pathways/enzymology , Neural Pathways/metabolism , Neurons/enzymology , Neurons/metabolism , Pain/enzymology , Pain/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Time Factors
18.
Brain Res ; 1247: 62-70, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-18983834

ABSTRACT

Improgan, a cimetidine derivative which lacks activity at known histamine, opioid or cannabinoid receptors, acts by an unknown mechanism in the periaqueductal gray (PAG) and raphe magnus (RM) to stimulate descending, analgesic circuits. These circuits may utilize cannabinoid mechanisms. To characterize further the nature of these circuits, the effects of intracerebral (i.c.) microinjections of rimonabant (a CB(1) receptor inverse agonist) were studied on antinociceptive responses following i.c. microinjections of improgan and the cannabinoid agonist WIN 55,212 (WIN) in rats. Separate intra-RM injections of improgan (30 microg) and WIN (8 microg) produced near-maximal antinociception on both the hot plate (HP) and tail flick (TF) nociceptive tests. Pretreatment with intra-RM rimonabant (20 microg) antagonized the antinociception produced by both intra-RM improgan and intra-RM WIN, but had no effects when given alone. Similar studies with improgan demonstrated rimonabant-sensitive sites within the dorsal and ventrolateral PAG. However, intra-RM pretreatment with rimonabant had no effect on antinociceptive responses following intra-PAG improgan. These studies show that improgan activates pain-relieving mechanisms in the PAG and the RM, both of which may utilize local cannabinoid mechanisms.


Subject(s)
Brain Stem/drug effects , Cannabinoid Receptor Modulators/metabolism , Cimetidine/analogs & derivatives , Nociceptors/drug effects , Pain/drug therapy , Analgesics/pharmacology , Animals , Benzoxazines/pharmacology , Brain Stem/anatomy & histology , Brain Stem/metabolism , Cimetidine/pharmacology , Male , Microinjections , Morpholines/pharmacology , Naphthalenes/pharmacology , Neural Pathways/anatomy & histology , Neural Pathways/drug effects , Neural Pathways/metabolism , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Pain Measurement/drug effects , Pain Threshold/drug effects , Pain Threshold/physiology , Periaqueductal Gray/anatomy & histology , Periaqueductal Gray/drug effects , Periaqueductal Gray/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Raphe Nuclei/anatomy & histology , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Rhombencephalon/anatomy & histology , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Rimonabant
19.
Drug Metab Dispos ; 36(3): 614-21, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094038

ABSTRACT

[(3)H]Cimetidine (3HCIM) specifically binds to an unidentified site in the rat brain. Because recently described ligands for this site have pharmacological activity, 3HCIM binding was characterized. 3HCIM binding was saturable, heat-labile, and distinct from the histamine H(2) receptor. To test the hypothesis that 3HCIM binds to a cytochrome P450 (P450), the effects of nonselective and isoform-selective P450 inhibitors were studied. The heme inhibitor KCN and the nonselective P450 inhibitor metyrapone both produced complete, concentration-dependent inhibition of 3HCIM binding (K(i) = 1.3 mM and 11.9 muM, respectively). Binding was largely unaffected by inhibitors of CYP1A2, 2B6, 2C8, 2C9, 2D6, 2E1, and 19A1 but was eliminated by inhibitors of CYP2C19 (tranylcypromine) and CYP3A4 (ketoconazole). Synthesis and testing of CC11 [4(5)-(benzylthiomethyl)-1H-imidazole] and CC12 [4(5)-((4-iodobenzyl)-thiomethyl)-1H-imidazole] confirmed both drugs to be high-affinity inhibitors of 3HCIM binding. On recombinant human P450s, CC12 was a potent inhibitor of CYP2B6 (IC(50) = 11.7 nM), CYP2C19 (51.4 nM), and CYP19A1 (140.7 nM) and had a range of activities (100-494 nM) on nine other isoforms. Although the 3HCIM binding site pharmacologically resembles some P450s, eight recombinant human P450s and three recombinant rat P450s did not exhibit 3HCIM binding. Inhibition by KCN and metyrapone suggests that 3HCIM binds to a heme-containing brain protein (possibly a P450). However, results with selective P450 inhibitors, recombinant P450 isoforms, and a P450 antibody did not identify a 3HCIM-binding P450 isoform. Finally, CC12 is a new, potent inhibitor of CYP2B6 and CYP2C19 that may be a valuable tool for P450 research.


Subject(s)
Brain/metabolism , Cimetidine/metabolism , Cytochrome P-450 Enzyme System/metabolism , Animals , Binding Sites , Binding, Competitive , Cimetidine/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Histamine H2 Antagonists/metabolism , Histamine H2 Antagonists/pharmacology , Isoenzymes , Kinetics , Ligands , Protein Binding , Rats , Rats, Sprague-Dawley , Tritium
20.
J Pain ; 8(11): 850-60, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17644043

ABSTRACT

UNLABELLED: Improgan is a congener of the H(2) antagonist cimetidine, which produces potent antinociception. Because a) the mechanism of action of improgan remains unknown and b) this drug may indirectly activate cannabinoid CB(1) receptors, the effects of the CB(1) antagonist/inverse agonist rimonabant (SR141716A) and 3 congeners with varying CB(1) potencies were studied on improgan antinociception after intracerebroventricular (icv) dosing in rats. Consistent with blockade of brain CB(1) receptors, rimonabant (K(d) = 0.23 nM), and O-1691 (K(d) = 0.22 nM) inhibited improgan antinociception by 48% and 70% after icv doses of 43 nmol and 25 nmol, respectively. However, 2 other derivatives with much lower CB(1) affinity (O-1876, K(d) = 139 nM and O-848, K(d) = 352 nM) unexpectedly blocked improgan antinociception by 65% and 50% after icv doses of 300 nmol and 30 nmol, respectively. These derivatives have 600-fold to 1500-fold lower CB(1) potencies than that of rimonabant, yet they retained improgan antagonist activity in vivo. In vitro dose-response curves with (35)S-GTPgammaS on CB(1) receptor-containing membranes confirmed the approximate relative potency of the derivatives at the CB(1) receptor. Although antagonism of improgan antinociception by rimonabant has previously implicated a mechanistic role for the CB(1) receptor, current findings with rimonabant congeners suggest that receptors other than, or in addition to CB(1) may participate in the pain-relieving mechanisms activated by this drug. The use of congeners such as O-848, which lack relevant CB(1)-blocking properties, will help to identify these cannabinoid-like, non-CB(1) mechanisms. PERSPECTIVE: This article describes new pharmacological characteristics of improgan, a pain-relieving drug that acts by an unknown mechanism. Improgan may use a marijuana-like (cannabinoid) pain-relieving mechanism, but it is shown presently that the principal cannabinoid receptor in the brain (CB(1)) is not solely responsible for improgan analgesia.


Subject(s)
Analgesics/administration & dosage , Cimetidine/analogs & derivatives , Pain Threshold/drug effects , Pain/drug therapy , Receptor, Cannabinoid, CB1/physiology , Analysis of Variance , Animals , Cimetidine/administration & dosage , Cimetidine/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Injections, Intraventricular/methods , Male , Pain Measurement/methods , Piperidines/administration & dosage , Piperidines/chemistry , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...