Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Proc Natl Acad Sci U S A ; 115(45): E10566-E10575, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30355767

ABSTRACT

Extracellular calcium flow through neuronal voltage-gated CaV2.2 calcium channels converts action potential-encoded information to the release of pronociceptive neurotransmitters in the dorsal horn of the spinal cord, culminating in excitation of the postsynaptic central nociceptive neurons. The CaV2.2 channel is composed of a pore-forming α1 subunit (CaVα1) that is engaged in protein-protein interactions with auxiliary α2/δ and ß subunits. The high-affinity CaV2.2α1⋅CaVß3 protein-protein interaction is essential for proper trafficking of CaV2.2 channels to the plasma membrane. Here, structure-based computational screening led to small molecules that disrupt the CaV2.2α1⋅CaVß3 protein-protein interaction. The binding mode of these compounds reveals that three substituents closely mimic the side chains of hot-spot residues located on the α-helix of CaV2.2α1 Site-directed mutagenesis confirmed the critical nature of a salt-bridge interaction between the compounds and CaVß3 Arg-307. In cells, compounds decreased trafficking of CaV2.2 channels to the plasma membrane and modulated the functions of the channel. In a rodent neuropathic pain model, the compounds suppressed pain responses. Small-molecule α-helical mimetics targeting ion channel protein-protein interactions may represent a strategy for developing nonopioid analgesia and for treatment of other neurological disorders associated with calcium-channel trafficking.


Subject(s)
Calcium Channel Blockers/pharmacology , Ion Channel Gating/drug effects , Small Molecule Libraries/pharmacology , Animals , Calcium Channel Blockers/pharmacokinetics , Female , HEK293 Cells , Humans , Ion Transport , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mice , Neuralgia/prevention & control , Nociception/drug effects , Protein Binding , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/pharmacokinetics
2.
Sci Rep ; 7(1): 12743, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28986567

ABSTRACT

Central sensitization and network hyperexcitability of the nociceptive system is a basic mechanism of neuropathic pain. We hypothesize that development of cortical hyperexcitability underlying neuropathic pain may involve homeostatic plasticity in response to lesion-induced somatosensory deprivation and activity loss, and can be controlled by enhancing cortical activity. In a mouse model of neuropathic pain, in vivo two-photon imaging and patch clamp recording showed initial loss and subsequent recovery and enhancement of spontaneous firings of somatosensory cortical pyramidal neurons. Unilateral optogenetic stimulation of cortical pyramidal neurons both prevented and reduced pain-like behavior as detected by bilateral mechanical hypersensitivity of hindlimbs, but corpus callosotomy eliminated the analgesic effect that was ipsilateral, but not contralateral, to optogenetic stimulation, suggesting involvement of inter-hemispheric excitatory drive in this effect. Enhancing activity by focally blocking cortical GABAergic inhibition had a similar relieving effect on the pain-like behavior. Patch clamp recordings from layer V pyramidal neurons showed that optogenetic stimulation normalized cortical hyperexcitability through changing neuronal membrane properties and reducing frequency of excitatory postsynaptic events. We conclude that development of neuropathic pain involves abnormal homeostatic activity regulation of somatosensory cortex, and that enhancing cortical excitatory activity may be a novel strategy for preventing and controlling neuropathic pain.


Subject(s)
Homeostasis , Neuralgia/physiopathology , Neuronal Plasticity/physiology , Somatosensory Cortex/physiopathology , Action Potentials , Animals , Behavior, Animal , Channelrhodopsins/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials , Hyperalgesia/complications , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Inhibitory Postsynaptic Potentials , Ischemia/complications , Ischemia/pathology , Ischemia/physiopathology , Mice, Inbred C57BL , Neuralgia/complications , Neuralgia/pathology , Optogenetics , Pyramidal Cells/metabolism , Somatosensory Cortex/pathology , Spinal Cord/pathology , Spinal Cord/physiopathology , Synaptic Transmission , Tibial Nerve/injuries , Tibial Nerve/pathology , Tibial Nerve/physiopathology
3.
Sci Rep ; 7(1): 3741, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623271

ABSTRACT

Accumulating evidence indicates that Toll-like receptor (TLR) signaling adapter protein interactions with Toll/Interleukin-1 Receptor (TIR) domains present in sensory neurons may modulate neuropathic pain states. Following ligand interaction with TLRs, TIR serves to both initiate intracellular signaling and facilitate recruitment of signaling adapter proteins to the intracytoplasmic domain. Although TLR TIR is central to a number of TLR signaling cascades, its role in sensory neurons is poorly understood. In this study we investigated the degree to which TLR TIR decoy peptide modified to include a TAT sequence (Trans-Activator of Transcription gene in HIV; TAT-4BB) affected LPS-induced intracellular calcium flux and excitation in sensory neurons, and behavioral changes due to TLR4 active metabolite, morphine-3-glucuronide (M3G) exposure in vivo. TAT-4BB inhibited LPS-induced calcium changes in a majority of sensory neurons and decreased LPS-dependent neuronal excitability in small diameter neurons. Acute systemic administration of the TAT-4BB reversed M3G-induced tactile allodynia in a dose-dependent manner but did not affect motor activity, anxiety or responses to noxious thermal stimulus. These data suggest that targeting TLR TIR domains may provide novel pharmacological targets to reduce or reverse TLR4-dependent pain behavior in the rodent.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Lipopolysaccharides/toxicity , Morphine Derivatives/pharmacology , Neuralgia , Peptides , Sensory Receptor Cells/metabolism , Toll-Like Receptor 4/metabolism , Animals , Calcium Signaling/drug effects , Female , Mice , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/metabolism , Neuralgia/pathology , Peptides/chemistry , Peptides/pharmacology , Protein Domains , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology
4.
Cancer Res ; 77(6): 1283-1295, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28254863

ABSTRACT

Multiple myeloma patients experience severe bone pain (MMBP) that is undertreated and poorly understood. In this study, we studied MMBP in an intratibial mouse xenograft model that employs JJN3 human multiple myeloma cells. In this model, mice develop MMBP associated in bone with increased sprouting of calcitonin gene-related peptide-positive (CGRP+) sensory nerves and in dorsal root ganglia (DRG) with upregulation of phosphorylated ERK1/2 (pERK1/2) and pCREB, two molecular indicators of neuron excitation. We found that JJN3 cells expressed a vacuolar proton pump (V-ATPase) that induced an acidic bone microenvironment. Inhibition of JJN3-colonized bone acidification by a single injection of the selective V-ATPase inhibitor, bafilomycin A1, decreased MMBP, CGRP+ sensory neuron sprouting, and pERK1/2 and pCREB expression in DRG. CGRP+ sensory nerves also expressed increased levels of the acid-sensing nociceptor ASIC3. Notably, a single injection of the selective ASIC3 antagonist APETx2 dramatically reduced MMBP in the model. Mechanistic investigations in primary DRG neurons cocultured with JJN3 cells showed increased neurite outgrowth and excitation inhibited by bafilomycin A1 or APETx2. Furthermore, combining APETx2 with bafilomycin A1 reduced MMBP to a greater extent than either agent alone. Finally, combining bafilomycin A1 with the osteoclast inhibitor zoledronic acid was sufficient to ameliorate MMBP, which was refractory to zoledronic acid. Overall, our results show that osteoclasts and multiple myeloma cooperate to induce an acidic bone microenvironment that evokes MMBP as a result of the excitation of ASIC3-activated sensory neurons. Furthermore, they present a mechanistic rationale for targeting ASIC3 on neurons along with the multiple myeloma-induced acidic bone microenvironment as a strategy to relieve MMBP in patients. Cancer Res; 77(6); 1283-95. ©2017 AACR.


Subject(s)
Acid Sensing Ion Channels/chemistry , Bone Diseases/prevention & control , Bone Resorption/prevention & control , Multiple Myeloma/complications , Pain/prevention & control , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Animals , Bone Density Conservation Agents/pharmacology , Bone Diseases/etiology , Bone Diseases/metabolism , Bone Resorption/etiology , Bone Resorption/metabolism , Cells, Cultured , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation/drug effects , Humans , Imidazoles/pharmacology , Macrolides/pharmacology , Mice , Mice, SCID , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Pain/etiology , Pain/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Zoledronic Acid
5.
Stem Cells ; 35(5): 1303-1315, 2017 05.
Article in English | MEDLINE | ID: mdl-28299842

ABSTRACT

Electroacupuncture (EA) performed in rats and humans using limb acupuncture sites, LI-4 and LI-11, and GV-14 and GV-20 (humans) and Bai-hui (rats) increased functional connectivity between the anterior hypothalamus and the amygdala and mobilized mesenchymal stem cells (MSCs) into the systemic circulation. In human subjects, the source of the MSC was found to be primarily adipose tissue, whereas in rodents the tissue sources were considered more heterogeneous. Pharmacological disinhibition of rat hypothalamus enhanced sympathetic nervous system (SNS) activation and similarly resulted in a release of MSC into the circulation. EA-mediated SNS activation was further supported by browning of white adipose tissue in rats. EA treatment of rats undergoing partial rupture of the Achilles tendon resulted in reduced mechanical hyperalgesia, increased serum interleukin-10 levels and tendon remodeling, effects blocked in propranolol-treated rodents. To distinguish the afferent role of the peripheral nervous system, phosphoinositide-interacting regulator of transient receptor potential channels (Pirt)-GCaMP3 (genetically encoded calcium sensor) mice were treated with EA acupuncture points, ST-36 and LIV-3, and GV-14 and Bai-hui and resulted in a rapid activation of primary sensory neurons. EA activated sensory ganglia and SNS centers to mediate the release of MSC that can enhance tissue repair, increase anti-inflammatory cytokine production and provide pronounced analgesic relief. Stem Cells 2017;35:1303-1315.


Subject(s)
Central Nervous System/cytology , Electroacupuncture , Mesenchymal Stem Cells/cytology , Achilles Tendon/pathology , Acupuncture Points , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Antigens, CD/metabolism , Forelimb/physiology , Hindlimb/physiology , Humans , Hyperalgesia/therapy , Hypothalamus/cytology , Interleukin-10/blood , Macrophages/cytology , Mice , Nerve Net/physiology , Rats , Rupture , Sensory Receptor Cells/metabolism , Uncoupling Protein 1/metabolism
6.
Pain ; 157(9): 2124-2140, 2016 09.
Article in English | MEDLINE | ID: mdl-27537210

ABSTRACT

Uncoupling the protein-protein interaction between collapsin response mediator protein 2 (CRMP2) and N-type voltage-gated calcium channel (CaV2.2) with an allosteric CRMP2-derived peptide (CBD3) is antinociceptive in rodent models of inflammatory and neuropathic pain. We investigated the efficacy, duration of action, abuse potential, and neurobehavioral toxicity of an improved mutant CRMP2 peptide. A homopolyarginine (R9)-conjugated CBD3-A6K (R9-CBD3-A6K) peptide inhibited the CaV2.2-CRMP2 interaction in a concentration-dependent fashion and diminished surface expression of CaV2.2 and depolarization-evoked Ca influx in rat dorsal root ganglia neurons. In vitro studies demonstrated suppression of excitability of small-to-medium diameter dorsal root ganglion and inhibition of subtypes of voltage-gated Ca channels. Sprague-Dawley rats with tibial nerve injury had profound and long-lasting tactile allodynia and ongoing pain. Immediate administration of R9-CBD3-A6K produced enhanced dopamine release from the nucleus accumbens shell selectively in injured animals, consistent with relief of ongoing pain. R9-CBD3-A6K, when administered repeatedly into the central nervous system ventricles of naive rats, did not result in a positive conditioned place preference demonstrating a lack of abusive liability. Continuous subcutaneous infusion of R9-CBD3-A6K over a 24- to 72-hour period reversed tactile allodynia and ongoing pain, demonstrating a lack of tolerance over this time course. Importantly, continuous infusion of R9-CBD3-A6K did not affect motor activity, anxiety, depression, or memory and learning. Collectively, these results validate the potential therapeutic significance of targeting the CaV-CRMP2 axis for treatment of neuropathic pain.


Subject(s)
Aptamers, Peptide/therapeutic use , Intercellular Signaling Peptides and Proteins/chemistry , Nerve Tissue Proteins/chemistry , Neuralgia/drug therapy , Action Potentials/drug effects , Animals , Anxiety/drug therapy , Anxiety/etiology , Aptamers, Peptide/pharmacology , Disease Models, Animal , Dopamine/metabolism , Electric Stimulation , Exploratory Behavior/drug effects , Female , Ganglia, Spinal/cytology , Hindlimb Suspension , Hyperalgesia/drug therapy , Maze Learning/drug effects , Mice, Inbred C57BL , Neuralgia/pathology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects
7.
Pain Res Treat ; 2016: 8364762, 2016.
Article in English | MEDLINE | ID: mdl-27069684

ABSTRACT

Aim. Nonsteroidal anti-inflammatory drugs or opioids are commonly used to control surgical pain following veterinary and clinical procedures. This study evaluated the efficacy of postoperative ketorolac or buprenorphine following abdominal surgery. Main Methods. Mean arterial pressure (MAP), heart rate, animal activity, corticosterone levels, and a nociceptive sensitivity assay were used to evaluate 18 adult male Sprague-Dawley rats which underwent aortic artery occlusion for implantation of a radiotelemetry device. The animals were treated postoperatively with intraperitoneal injections of vehicle, ketorolac (10 mg/kg), or buprenorphine (0.06 mg/kg) every 8 hours for 3 days. Key Findings. There were no consistent significant changes in any of the telemetry parameters after treatment with ketorolac compared with no saline treatment with the exception of increased MAP in the buprenorphine group during the first 48 hours when compared with other treatment groups. There was a sustained increase in fecal corticosterone levels from baseline on days 2-7 with buprenorphine compared with vehicle- or ketorolac-treated animals. All treatment conditions displayed reduced paw withdrawal thresholds (PWTs) from day 1 to day 21 following surgery. Compared with the vehicle treatment group, buprenorphine-treated animals exhibited significantly lower PWT levels from day 4 to 14 days. Significance. Given the prolonged increase in fecal corticosterone levels and pronounced changes in tactile hyperalgesia behavior in rodents subjected to buprenorphine treatment, these data suggest that ketorolac may be superior to buprenorphine for the treatment of postprocedure pain behavior in rodents.

8.
ACS Chem Neurosci ; 6(2): 316-30, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25418676

ABSTRACT

The functionalized amino acid, lacosamide ((R)-2), and the α-aminoamide, safinamide ((S)-3), are neurological agents that have been extensively investigated and have displayed potent anticonvulsant activities in seizure models. Both compounds have been reported to modulate voltage-gated sodium channel activity. We have prepared a series of chimeric compounds, (R)-7-(R)-10, by merging key structural units in these two clinical agents, and then compared their activities with (R)-2 and (S)-3. Compounds were assessed for their ability to alter sodium channel kinetics for inactivation, frequency (use)-dependence, and steady-state activation and fast inactivation. We report that chimeric compounds (R)-7-(R)-10 in catecholamine A-differentiated (CAD) cells and embryonic rat cortical neurons robustly enhanced sodium channel inactivation at concentrations far lower than those required for (R)-2 and (S)-3, and that (R)-9 and (R)-10, unlike (R)-2 and (S)-3, produce sodium channel frequency (use)-dependence at low micromolar concentrations. We further show that (R)-7-(R)-10 displayed excellent anticonvulsant activities and pain-attenuating properties in the animal formalin model. Of these compounds, only (R)-7 reversed mechanical hypersensitivity in the tibial-nerve injury model for neuropathic pain in rats.


Subject(s)
Acetamides/pharmacology , Alanine/analogs & derivatives , Analgesics/pharmacology , Anticonvulsants/pharmacology , Benzylamines/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Acetamides/chemistry , Alanine/chemistry , Alanine/pharmacology , Analgesics/chemistry , Animals , Anticonvulsants/chemistry , Benzylamines/chemistry , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Disease Models, Animal , Female , Formaldehyde , Lacosamide , Male , Membrane Potentials/drug effects , Mice , Neuralgia/drug therapy , Neuralgia/etiology , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Rats, Sprague-Dawley , Seizures/drug therapy , Tibial Nerve/injuries , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channels/metabolism
9.
PLoS One ; 9(9): e107399, 2014.
Article in English | MEDLINE | ID: mdl-25221944

ABSTRACT

Approximately 60% of morphine is glucuronidated to morphine-3-glucuronide (M3G) which may aggravate preexisting pain conditions. Accumulating evidence indicates that M3G signaling through neuronal Toll-like receptor 4 (TLR4) may be central to this proalgesic signaling event. These events are known to include elevated neuronal excitability, increased voltage-gated sodium (NaV) current, tactile allodynia and decreased opioid analgesic efficacy. Using an in vitro ratiometric-based calcium influx analysis of acutely dissociated small and medium-diameter neurons derived from lumbar dorsal root ganglion (DRG), we observed that M3G-sensitive neurons responded to lipopolysaccharide (LPS) and over 35% of these M3G/LPS-responsive cells exhibited sensitivity to capsaicin. In addition, M3G-exposed sensory neurons significantly increased excitatory activity and potentiated NaV current as measured by current and voltage clamp, when compared to baseline level measurements. The M3G-dependent excitability and potentiation of NaV current in these sensory neurons could be reversed by the addition of carbamazepine (CBZ), a known inhibitor of several NaV currents. We then compared the efficacy between CBZ and morphine as independent agents, to the combined treatment of both drugs simultaneously, in the tibial nerve injury (TNI) model of neuropathic pain. The potent anti-nociceptive effects of morphine (5 mg/kg, i.p.) were observed in TNI rodents at post-injury day (PID) 7-14 and absent at PID21-28, while administration of CBZ (10 mg/kg, i.p.) alone failed to produce anti-nociceptive effects at any time following TNI (PID 7-28). In contrast to either drug alone at PID28, the combination of morphine and CBZ completely attenuated tactile hyperalgesia in the rodent TNI model. The basis for the potentiation of morphine in combination with CBZ may be due to the effects of a latent upregulation of NaV1.7 in the DRG following TNI. Taken together, our observations demonstrate a potential therapeutic use of morphine and CBZ as a combinational treatment for neuropathic pain.


Subject(s)
Analgesics, Opioid/therapeutic use , Carbamazepine/therapeutic use , Morphine/therapeutic use , Neuralgia/drug therapy , Action Potentials/drug effects , Animals , Female , Ganglia, Spinal/drug effects , Male , Morphine Derivatives/therapeutic use , Neuralgia/metabolism , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Toll-Like Receptor 4/metabolism
10.
Sci Transl Med ; 6(252): 252ra124, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25186179

ABSTRACT

Traumatic brain injury (TBI) results in systemic inflammatory responses that affect the lung. This is especially critical in the setting of lung transplantation, where more than half of donor allografts are obtained postmortem from individuals with TBI. The mechanism by which TBI causes pulmonary dysfunction remains unclear but may involve the interaction of high-mobility group box-1 (HMGB1) protein with the receptor for advanced glycation end products (RAGE). To investigate the role of HMGB1 and RAGE in TBI-induced lung dysfunction, RAGE-sufficient (wild-type) or RAGE-deficient (RAGE(-/-)) C57BL/6 mice were subjected to TBI through controlled cortical impact and studied for cardiopulmonary injury. Compared to control animals, TBI induced systemic hypoxia, acute lung injury, pulmonary neutrophilia, and decreased compliance (a measure of the lungs' ability to expand), all of which were attenuated in RAGE(-/-) mice. Neutralizing systemic HMGB1 induced by TBI reversed hypoxia and improved lung compliance. Compared to wild-type donors, lungs from RAGE(-/-) TBI donors did not develop acute lung injury after transplantation. In a study of clinical transplantation, elevated systemic HMGB1 in donors correlated with impaired systemic oxygenation of the donor lung before transplantation and predicted impaired oxygenation after transplantation. These data suggest that the HMGB1-RAGE axis plays a role in the mechanism by which TBI induces lung dysfunction and that targeting this pathway before transplant may improve recipient outcomes after lung transplantation.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/physiopathology , HMGB1 Protein/metabolism , Lung Transplantation , Lung/physiopathology , Receptors, Immunologic/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/physiopathology , Adult , Animals , Antibodies, Neutralizing/pharmacology , Brain Injuries/complications , Cardiac Output/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Interleukin-10/metabolism , Lung/drug effects , Lung/pathology , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Peptides/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Tissue Donors , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/metabolism
11.
Brain Behav Immun ; 42: 169-77, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25014009

ABSTRACT

Recent studies indicate that the release of high mobility group box 1 (HMGB1) following nerve injury may play a central role in the pathogenesis of neuropathic pain. HMGB1 is known to influence cellular responses within the nervous system via two distinct receptor families; the Receptor for Advanced Glycation End-products (RAGE) and Toll-like receptors (TLRs). The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. Though it is known that dorsal root ganglia (DRG) sensory neurons exposed to HMGB1 and TLR4 agonists can influence excitation, the degree to which at-HMGB1 signaling through neuronal RAGE contributes to neuropathic pain is unknown. Here we demonstrate that at-HMGB1 activation of nociceptive neurons is dependent on RAGE and not TLR4. To distinguish the possible role of RAGE on neuropathic pain, we characterized the changes in RAGE mRNA expression up to one month after tibial nerve injury (TNI). RAGE mRNA expression in lumbar dorsal root ganglion (DRG) is substantially increased by post-injury day (PID) 28 when compared with sham injured rodents. Protein expression at PID28 confirms this injury-induced event in the DRG. Moreover, a single exposure to monoclonal antibody to RAGE (RAGE Ab) failed to abrogate pain behavior at PID 7, 14 and 21. However, RAGE Ab administration produced reversal of mechanical hyperalgesia on PID28. Thus, at-HMGB1 activation through RAGE may be responsible for sensory neuron sensitization and mechanical hyperalgesia associated with chronic neuropathic pain states.


Subject(s)
HMGB1 Protein/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Neurons/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Disease Models, Animal , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Male , Neuralgia/etiology , Neuralgia/physiopathology , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Rats , Rats, Sprague-Dawley
12.
J Endourol ; 27(3): 338-42, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22974473

ABSTRACT

PURPOSE: The cardiac and renal protective effects of phosphodiesterase-5 (PDE-5) inhibitors against ischemia-reperfusion injury have recently been demonstrated in animal studies. We evaluated the effect of pretreatment with the PDE-5 inhibitor zaprinast on warm renal ischemia in a rat model. METHODS: Female Sprague-Dawley rats underwent concomitant right nephrectomy and left renal hilar occlusion for 30 minutes. Twelve animals were equally divided into three groups: Group 1 received no pharmacologic pretreatment, group 2 was pretreated with zaprinast 10 mg/kg, and group 3 was pretreated with zaprinast 20 mg/kg. Zaprinast was dissolved in 25% dimethyl sulfoxide and given as a single intraperitoneal injection 30 minutes before surgery. Serum blood urea nitrogen (BUN) and creatinine levels, histopathology, and TUNEL staining for apoptosis were assessed 24 hours postoperatively. RESULTS: The mean creatinine level for groups 1, 2, and 3 was 0.73 mg/dL, 0.55 mg/dL, and 0.38 mg/dL, respectively. These values were not statistically different (P=0.099). The mean BUN levels of 35.8 mg/dL for group 1, 27.3 mg/dL for group 2, and 23.3 mg/dL for group 3 were also statistically similar (P=0.278). There were no objective differences in histopathologic evaluation or TUNEL staining between the groups. CONCLUSION: This study did not demonstrate a beneficial effect of zaprinast pretreatment on renal parameters after warm ischemic injury.


Subject(s)
Phosphodiesterase 5 Inhibitors/therapeutic use , Purinones/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology , Animals , Blood Urea Nitrogen , Creatinine/blood , Female , In Situ Nick-End Labeling , Phosphodiesterase 5 Inhibitors/pharmacology , Purinones/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/blood , Reperfusion Injury/pathology , Staining and Labeling
13.
J Neurochem ; 124(6): 869-79, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23106100

ABSTRACT

The N-type voltage-gated calcium channel (CaV2.2) is a clinically endorsed target in chronic pain treatments. As directly targeting the channel can lead to multiple adverse side effects, targeting modulators of CaV2.2 may prove better. We previously identified ST1-104, a short peptide from the collapsin response mediator protein 2 (CRMP2), which disrupted the CaV2.2-CRMP2 interaction and suppressed a model of HIV-related neuropathy induced by anti-retroviral therapy but not traumatic neuropathy. Here, we report ST2-104 -a peptide wherein the cell-penetrating TAT motif has been supplanted with a homopolyarginine motif, which dose-dependently inhibits the CaV2.2-CRMP2 interaction and inhibits depolarization-evoked Ca(2+) influx in sensory neurons. Ca(2+) influx via activation of vanilloid receptors is not affected by either peptide. Systemic administration of ST2-104 does not affect thermal or tactile nociceptive behavioral changes. Importantly, ST2-104 transiently reduces persistent mechanical hypersensitivity induced by systemic administration of the anti-retroviral drug 2',3'-dideoxycytidine (ddC) and following tibial nerve injury (TNI). Possible mechanistic explanations for the broader efficacy of ST2-104 are discussed.


Subject(s)
Disease Models, Animal , Nerve Tissue Proteins/therapeutic use , Pain/drug therapy , Peptides/therapeutic use , Peripheral Nervous System Diseases/drug therapy , Amino Acid Sequence , Animals , Cells, Cultured , Female , Intercellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Pain/pathology , Pain/psychology , Pain Management/methods , Peptides/genetics , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/psychology , Rats , Rats, Sprague-Dawley
14.
J Neuroinflammation ; 9: 200, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22898544

ABSTRACT

BACKGROUND: Multiple adverse events are associated with the use of morphine for the treatment of chronic non-cancer pain, including opioid-induced hyperalgesia (OIH). Mechanisms of OIH are independent of opioid tolerance and may involve the morphine metabolite morphine-3-glucuronide (M3G). M3G exhibits limited affinity for opioid receptors and no analgesic effect. Previous reports suggest that M3G can act via the Toll-like receptor 4 (TLR4)/myeloid differentiation protein-2 (MD-2) heterodimer in the central nervous system to elicit pain. METHODS: Immunoblot and immunocytochemistry methods were used to characterize the protein expression of TLR4 present in lumbar dorsal root ganglion (DRG). Using in vitro intracellular calcium and current clamp techniques, we determined whether TLR4 activation as elicited by the prototypical agonists of TLR4, lipopolysaccharide (LPS) and M3G, contributed to changes in intracellular calcium and increased excitation. Rodents were also injected with M3G to determine the degree to which M3G-induced tactile hyperalgesia could be diminished using either a small molecule inhibitor of the MD-2/TLR4 complex in rats or TLR4 knockout mice. Whole cell voltage-clamp recordings were made from small- and medium-diameter DRG neurons (25 µm < DRG diameter <45 µm) for both control and M3G-treated neurons to determine the potential influence on voltage-gated sodium channels (NaVs). RESULTS: We observed that TLR4 immunoreactivity was present in peptidergic and non-peptidergic sensory neurons in the DRG. Non-neuronal cells in the DRG lacked evidence of TLR4 expression. Approximately 15% of assayed small- and medium-diameter sensory neurons exhibited a change in intracellular calcium following LPS administration. Both nociceptive and non-nociceptive neurons were observed to respond, and approximately 40% of these cells were capsaicin-insensitive. Increased excitability observed in sensory neurons following LPS or M3G could be eliminated using Compound 15, a small molecule inhibitor of the TLR4/MD-2 complex. Likewise, systemic injection of M3G induced rapid tactile, but not thermal, nociceptive behavioral changes in the rat, which were prevented by pre-treating animals with Compound 15. Unlike TLR4 wild-type mice, TLR4 knockout mice did not exhibit M3G-induced hyperalgesia. As abnormal pain sensitivity is often associated with NaVs, we predicted that M3G acting via the MD-2/TLR4 complex may affect the density and gating of NaVs in sensory neurons. We show that M3G increases tetrodotoxin-sensitive and tetrodotoxin-resistant (NaV1.9) current densities. CONCLUSIONS: These outcomes provide evidence that M3G may play a role in OIH via the TLR4/MD-2 heterodimer complex and biophysical properties of tetrodotoxin-sensitive and tetrodotoxin-resistant NaV currents.


Subject(s)
Central Nervous System Stimulants/pharmacology , Morphine Derivatives/pharmacology , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Action Potentials/drug effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hyperalgesia/physiopathology , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Lectins/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Pain Measurement , Pain Threshold/drug effects , Phosphopyruvate Hydratase/metabolism , Physical Stimulation , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Signal Transduction/genetics , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Toll-Like Receptor 4/genetics , Touch/drug effects
15.
J Biol Chem ; 287(42): 35065-35077, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22891239

ABSTRACT

N-type Ca(2+) channels (CaV2.2) are a nidus for neurotransmitter release and nociceptive transmission. However, the use of CaV2.2 blockers in pain therapeutics is limited by side effects resulting from inhibition of the physiological functions of CaV2.2 within the CNS. We identified an anti-nociceptive peptide (Brittain, J. M., Duarte, D. B., Wilson, S. M., Zhu, W., Ballard, C., Johnson, P. L., Liu, N., Xiong, W., Ripsch, M. S., Wang, Y., Fehrenbacher, J. C., Fitz, S. D., Khanna, M., Park, C. K., Schmutzler, B. S., Cheon, B. M., Due, M. R., Brustovetsky, T., Ashpole, N. M., Hudmon, A., Meroueh, S. O., Hingtgen, C. M., Brustovetsky, N., Ji, R. R., Hurley, J. H., Jin, X., Shekhar, A., Xu, X. M., Oxford, G. S., Vasko, M. R., White, F. A., and Khanna, R. (2011) Suppression of inflammatory and neuropathic pain by uncoupling CRMP2 from the presynaptic Ca(2+) channel complex. Nat. Med. 17, 822-829) derived from the axonal collapsin response mediator protein 2 (CRMP2), a protein known to bind and enhance CaV2.2 activity. Using a peptide tiling array, we identified novel peptides within the first intracellular loop (CaV2.2(388-402), "L1") and the distal C terminus (CaV1.2(2014-2028) "Ct-dis") that bound CRMP2. Microscale thermophoresis demonstrated micromolar and nanomolar binding affinities between recombinant CRMP2 and synthetic L1 and Ct-dis peptides, respectively. Co-immunoprecipitation experiments showed that CRMP2 association with CaV2.2 was inhibited by L1 and Ct-dis peptides. L1 and Ct-dis, rendered cell-penetrant by fusion with the protein transduction domain of the human immunodeficiency virus TAT protein, were tested in in vitro and in vivo experiments. Depolarization-induced calcium influx in dorsal root ganglion (DRG) neurons was inhibited by both peptides. Ct-dis, but not L1, peptide inhibited depolarization-stimulated release of the neuropeptide transmitter calcitonin gene-related peptide in mouse DRG neurons. Similar results were obtained in DRGs from mice with a heterozygous mutation of Nf1 linked to neurofibromatosis type 1. Ct-dis peptide, administered intraperitoneally, exhibited antinociception in a zalcitabine (2'-3'-dideoxycytidine) model of AIDS therapy-induced and tibial nerve injury-related peripheral neuropathy. This study suggests that CaV peptides, by perturbing interactions with the neuromodulator CRMP2, contribute to suppression of neuronal hypersensitivity and nociception.


Subject(s)
AIDS-Associated Nephropathy/drug therapy , Calcium Channels, N-Type/pharmacology , Ganglia, Spinal/metabolism , Neurotransmitter Agents/metabolism , Peptides/pharmacology , Tibial Neuropathy/drug therapy , AIDS-Associated Nephropathy/genetics , AIDS-Associated Nephropathy/metabolism , AIDS-Associated Nephropathy/pathology , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Ganglia, Spinal/pathology , Humans , Mice , Mice, Knockout , Mice, Mutant Strains , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Neurons/metabolism , Neurons/pathology , Neurotransmitter Agents/genetics , Nociception/drug effects , Peptides/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Tibial Neuropathy/genetics , Tibial Neuropathy/metabolism , Tibial Neuropathy/pathology , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/pharmacology
16.
J Neuroinflammation ; 9: 180, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22824385

ABSTRACT

BACKGROUND: High-mobility group box-1 protein (HMGB1) is a nuclear protein that regulates gene expression throughout the body. It can also become cytoplasmic and function as a neuromodulatory cytokine after tissue damage or injury. The manner in which HMGB1 influences the peripheral nervous system following nerve injury is unclear. The present study investigated the degree to which HMGB1 signaling contributes to the maintenance of neuropathic pain behavior in the rodent. RESULTS: Redistribution of HMGB1 from the nucleus to the cytoplasm occurred in both sensory neurons derived from a tibial nerve injured (TNI) rat and in a sensory neuron-like cell line following exposure to a depolarizing stimulus. We also observe that exogenous administration of HMGB1 to acutely dissociated sensory neurons derived from naïve or TNI rodents elicit increased excitability. Furthermore systemic injection of glycyrrhizin (50 mg/kg; i.p.), a known inhibitor of HMGB1, reversed TNI-induced mechanical hyperalgesia at fourteen days and three months following nerve injury. CONCLUSIONS: We have identified that a persistent endogenous release of HMGB1 by sensory neurons may be a potent, physiologically relevant modulator of neuronal excitability. More importantly, the use of the anti-inflammatory compound and known inhibitor of HMGB1, glycyrrhizin, has the ability to diminish persistent pain behavior in a model of peripheral neuropathy, presumably through its ability to neutralize the cyotkine. The identification of HMGB1 as a potential therapeutic target may contribute to a better understanding of mechanisms associated with chronic pain syndromes.


Subject(s)
HMGB1 Protein/metabolism , Hyperalgesia/etiology , Hyperalgesia/metabolism , Neuralgia/complications , Sensory Receptor Cells/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Activating Transcription Factor 3/metabolism , Animals , Calcium/metabolism , Cell Count , Cell Line, Tumor , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Mice , Neuroblastoma/pathology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Tibial Nerve/pathology
17.
Mol Pain ; 8: 54, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22828369

ABSTRACT

BACKGROUND: The ubiquity of protein-protein interactions in biological signaling offers ample opportunities for therapeutic intervention. We previously identified a peptide, designated CBD3, that suppressed inflammatory and neuropathic behavioral hypersensitivity in rodents by inhibiting the ability of collapsin response mediator protein 2 (CRMP-2) to bind to N-type voltage-activated calcium channels (CaV2.2) [Brittain et al. Nature Medicine 17:822-829 (2011)]. RESULTS AND DISCUSSION: Here, we utilized SPOTScan analysis to identify an optimized variation of the CBD3 peptide (CBD3A6K) that bound with greater affinity to Ca²âº channels. Molecular dynamics simulations demonstrated that the CBD3A6K peptide was more stable and less prone to the unfolding observed with the parent CBD3 peptide. This mutant peptide, conjugated to the cell penetrating motif of the HIV transduction domain protein TAT, exhibited greater anti-nociception in a rodent model of AIDS therapy-induced peripheral neuropathy when compared to the parent TAT-CBD3 peptide. Remarkably, intraperitoneal administration of TAT-CBD3A6K produced none of the minor side effects (i.e. tail kinking, body contortion) observed with the parent peptide. Interestingly, excitability of dissociated small diameter sensory neurons isolated from rats was also reduced by TAT-CBD3A6K peptide suggesting that suppression of excitability may be due to inhibition of T- and R-type Ca²âº channels. TAT-CBD3A6K had no effect on depolarization-evoked calcitonin gene related peptide (CGRP) release compared to vehicle control. CONCLUSIONS: Collectively, these results establish TAT-CBD3A6K as a peptide therapeutic with greater efficacy in an AIDS therapy-induced model of peripheral neuropathy than its parent peptide, TAT-CBD3. Structural modifications of the CBD3 scaffold peptide may result in peptides with selectivity against a particular subset of voltage-gated calcium channels resulting in a multipharmacology of action on the target.


Subject(s)
Acquired Immunodeficiency Syndrome/therapy , Calcium Channels, N-Type/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Nerve Tissue Proteins/chemistry , Nociception , Nociceptors/metabolism , Peptides/therapeutic use , Peripheral Nervous System Diseases/drug therapy , Acquired Immunodeficiency Syndrome/complications , Amino Acid Sequence , Animals , Cell Separation , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Ion Channel Gating/drug effects , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis/genetics , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuralgia/drug therapy , Neuralgia/etiology , Neurotransmitter Agents/metabolism , Nociception/drug effects , Nociceptors/drug effects , Nociceptors/pathology , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Peripheral Nervous System Diseases/etiology , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , tat Gene Products, Human Immunodeficiency Virus/metabolism
18.
Transl Neurosci ; 3(1): 1-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22662308

ABSTRACT

Biological, genetic, and clinical data provide compelling proof for N-type voltage-gated calcium channels (CaV2.2) as therapeutic targets for chronic pain. While decreasing channel function is ultimately anti-nociceptive, directly targeting the channel can lead to multiple adverse effects. Targeting regulators of channel activity may facilitate improved analgesic properties associated with channel block and afford a broader therapeutic window. Towards this end, we recently identified a short peptide, designated CBD3, derived from collapsin response mediator protein 2 (CRMP-2) that suppressed inflammatory and neuropathic hypersensitivity by inhibiting CRMP-2 binding to CaV2.2 [Brittain et al., Nature Medicine 17:822-829 (2011)]. Rodents administered CBD3 intraperitoneally, fused to the HIV TAT protein cell penetrating domain, exhibited antinociception lasting ~4 hours highlighting potential instability, limited oral bioavailability, and/or rapid elimination of peptide. This report focuses on improving upon the parental CBD3 peptide. Using SPOTScan analysis of synthetic versions of the parental CBD3 peptide, we identified peptides harboring single amino acid mutations that bound with greater affinity to CaV2.2. One such peptide, harboring a phenylalanine instead of glycine (G14F), was tested in rodent models of migraine and neuropathic pain. In vivo laser Doppler blood flowmetry measure of capsaicin-induced meningeal vascular responses related to headache pain was almost completely suppressed by dural application of the G14F peptide. The G14F mutant peptide, administered intraperitoneally, also exhibited greater antinociception in Stavudine (2'-3'-didehydro-2'-3'-dideoxythymidine (d4T)/Zerit®) model of AIDS therapy-induced peripheral neuropathy compared to the parent CBD3 peptide. These results demonstrate the patent translational value of small biologic drugs targeting CaV2.2 for management of clinical pain.

19.
Channels (Austin) ; 5(5): 449-56, 2011.
Article in English | MEDLINE | ID: mdl-21829088

ABSTRACT

The N-type voltage-gated calcium channel (Cav 2.2) has gained immense prominence in the treatment of chronic pain. While decreased channel function is ultimately anti-nociceptive, directly targeting the channel can lead to multiple adverse side effects. Targeting modulators of channel activity may facilitate improved analgesic properties associated with channel block and a broader therapeutic window. A novel interaction between Cav 2.2 and collapsin response mediator protein 2 (CRMP-2) positively regulates channel function by increasing surface trafficking. We recently identified a CRMP-2 peptide (TAT-CBD3), which effectively blocks this interaction, reduces or completely reverses pain behavior in a number of inflammatory and neuropathic models. Importantly, TAT-CBD3 did not produce many of the typical side effects often observed with Cav 2.2 inhibitors. Notably chronic pain mechanisms offer unique challenges as they often encompass a mix of both neuropathic and inflammatory elements, whereby inflammation likely causes damage to the neuron leading to neuropathic pain, and neuronal injury may produce inflammatory reactions. To this end, we sought to further disseminate the ability of TAT-CBD3 to alter behavioral outcomes in two additional rodent pain models. While we observed that TAT-CBD3 reversed mechanical hypersensitivity associated with a model of chronic inflammatory pain due to lysophosphotidylcholine-induced sciatic nerve focal demyelination (LPC), injury to the tibial nerve (TNI) failed to respond to drug treatment. Moreover, a single amino acid mutation within the CBD3 sequence demonstrated amplified Cav 2.2 binding and dramatically increased efficacy in an animal model of migraine. Taken together, TAT-CBD3 potentially represents a novel class of therapeutics targeting channel regulation as opposed to the channel itself.


Subject(s)
Calcium Channels, N-Type/metabolism , Chronic Pain/drug therapy , Nerve Tissue Proteins/metabolism , Peptides/pharmacology , Signal Transduction/drug effects , Animals , Calcium Channels, N-Type/genetics , Chronic Pain/genetics , Chronic Pain/metabolism , Chronic Pain/pathology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Female , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intercellular Signaling Peptides and Proteins , Migraine Disorders/drug therapy , Migraine Disorders/genetics , Migraine Disorders/metabolism , Migraine Disorders/pathology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Peptides/genetics , Point Mutation , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/chemically induced , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Signal Transduction/genetics , Tibial Nerve/injuries , Tibial Neuropathy/drug therapy , Tibial Neuropathy/genetics , Tibial Neuropathy/metabolism , Tibial Neuropathy/pathology
20.
ACS Chem Neurosci ; 2(6): 317-322, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21765969

ABSTRACT

We recently reported that merging key structural pharmacophores of the anticonvulsant drugs lacosamide (a functionalized amino acid) with safinamide (an α-aminoamide) resulted in novel compounds with anticonvulsant activities superior to that of either drug alone. Here, we examined the effects of six such chimeric compounds on Na(+)-channel function in central nervous system catecholaminergic (CAD) cells. Using whole-cell patch clamp electrophysiology, we demonstrated that these compounds affected Na(+) channel fast and slow inactivation processes. Detailed electrophysiological characterization of two of these chimeric compounds that contained either an oxymethylene ((R)-7) or a chemical bond ((R)-11) between the two aromatic rings showed comparable effects on slow inactivation, use-dependence of block, development of slow inactivation, and recovery of Na(+) channels from inactivation. Both compounds were equally effective at inducing slow inactivation; (R)-7 shifted the fast inactivation curve in the hyperpolarizing direction greater than (R)-11, suggesting that in the presence of (R)-7, a larger fraction of the channels are in an inactivated state. None of the chimeric compounds affected veratridine- or KCl-induced glutamate release in neonatal cortical neurons. There was modest inhibition of KCl-induced calcium influx in cortical neurons. Finally, a single intraperitoneal administration of (R)-7, but not (R)-11, completely reversed mechanical hypersensitivity in a tibial-nerve injury model of neuropathic pain. The strong effects of (R)-7 on slow and fast inactivation of Na(+) channels may contribute to its efficacy and provide a promising novel therapy for neuropathic pain, in addition to its antiepileptic potential.

SELECTION OF CITATIONS
SEARCH DETAIL
...