Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Br J Pharmacol ; 181(17): 3192-3214, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38741464

ABSTRACT

BACKGROUND AND PURPOSE: The mechanistic target of rapamycin (mTOR) signalling pathway is a key regulator of cell growth and metabolism. Its deregulation is implicated in several diseases. The macrolide rapamycin, a specific inhibitor of mTOR, has immunosuppressive, anti-inflammatory and antiproliferative properties. Recently, we identified tacrolimus, another macrolide immunosuppressant, as a novel activator of TRPM8 ion channels, involved in cold temperature sensing, thermoregulation, tearing and cold pain. We hypothesized that rapamycin may also have agonist activity on TRPM8 channels. EXPERIMENTAL APPROACH: Using calcium imaging and electrophysiology in transfected HEK293 cells and wildtype or Trpm8 KO mouse DRG neurons, we characterized rapamycin's effects on TRPM8 channels. We also examined the effects of rapamycin on tearing in mice. KEY RESULTS: Micromolar concentrations of rapamycin activated rat and mouse TRPM8 channels directly and potentiated cold-evoked responses, effects also observed in human TRPM8 channels. In cultured mouse DRG neurons, rapamycin increased intracellular calcium levels almost exclusively in cold-sensitive neurons. Responses were markedly decreased in Trpm8 KO mice or by TRPM8 channel antagonists. Cutaneous cold thermoreceptor endings were also activated by rapamycin. Topical application of rapamycin to the eye surface evokes tearing in mice by a TRPM8-dependent mechanism. CONCLUSION AND IMPLICATIONS: These results identify TRPM8 cationic channels in sensory neurons as novel molecular targets of the immunosuppressant rapamycin. These findings may help explain some of its therapeutic effects after topical application to the skin and the eye surface. Moreover, rapamycin could be used as an experimental tool in the clinic to explore cold thermoreceptors.


Subject(s)
Immunosuppressive Agents , Mice, Knockout , Sensory Receptor Cells , Sirolimus , TRPM Cation Channels , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/metabolism , Animals , Humans , HEK293 Cells , Sirolimus/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Immunosuppressive Agents/pharmacology , Rats , Mice , Male , Mice, Inbred C57BL , Cells, Cultured , Calcium/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Cold Temperature
2.
Brain ; 146(2): 475-491, 2023 02 13.
Article in English | MEDLINE | ID: mdl-35871491

ABSTRACT

Chemotherapy-induced peripheral neuropathy is a frequent, disabling side effect of anticancer drugs. Oxaliplatin, a platinum compound used in the treatment of advanced colorectal cancer, often leads to a form of chemotherapy-induced peripheral neuropathy characterized by mechanical and cold hypersensitivity. Current therapies for chemotherapy-induced peripheral neuropathy are ineffective, often leading to the cessation of treatment. Transient receptor potential ankyrin 1 (TRPA1) is a polymodal, non-selective cation-permeable channel expressed in nociceptors, activated by physical stimuli and cellular stress products. TRPA1 has been linked to the establishment of chemotherapy-induced peripheral neuropathy and other painful neuropathic conditions. Sigma-1 receptor is an endoplasmic reticulum chaperone known to modulate the function of many ion channels and receptors. Sigma-1 receptor antagonist, a highly selective antagonist of Sigma-1 receptor, has shown effectiveness in a phase II clinical trial for oxaliplatin chemotherapy-induced peripheral neuropathy. However, the mechanisms involved in the beneficial effects of Sigma-1 receptor antagonist are little understood. We combined biochemical and biophysical (i.e. intermolecular Förster resonance energy transfer) techniques to demonstrate the interaction between Sigma-1 receptor and human TRPA1. Pharmacological antagonism of Sigma-1R impaired the formation of this molecular complex and the trafficking of functional TRPA1 to the plasma membrane. Using patch-clamp electrophysiological recordings we found that antagonists of Sigma-1 receptor, including Sigma-1 receptor antagonist, exert a marked inhibition on plasma membrane expression and function of human TRPA1 channels. In TRPA1-expressing mouse sensory neurons, Sigma-1 receptor antagonists reduced inward currents and the firing of actions potentials in response to TRPA1 agonists. Finally, in a mouse experimental model of oxaliplatin neuropathy, systemic treatment with a Sigma-1 receptor antagonists prevented the development of painful symptoms by a mechanism involving TRPA1. In summary, the modulation of TRPA1 channels by Sigma-1 receptor antagonists suggests a new strategy for the prevention and treatment of chemotherapy-induced peripheral neuropathy and could inform the development of novel therapeutics for neuropathic pain.


Subject(s)
Antineoplastic Agents , Neuralgia , Transient Receptor Potential Channels , Mice , Humans , Animals , Oxaliplatin/toxicity , TRPA1 Cation Channel , Antineoplastic Agents/toxicity , Neuralgia/chemically induced , Neuralgia/prevention & control , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Sigma-1 Receptor
3.
Int J Mol Sci ; 23(24)2022 Dec 18.
Article in English | MEDLINE | ID: mdl-36555804

ABSTRACT

TRPM8 is a non-selective cation channel expressed in primary sensory neurons and other tissues, including the prostate and urothelium. Its participation in different physiological and pathological processes such as thermoregulation, pain, itch, inflammation and cancer has been widely described, making it a promising target for therapeutic approaches. The detection and quantification of TRPM8 seems crucial for advancing the knowledge of the mechanisms underlying its role in these pathophysiological conditions. Antibody-based techniques are commonly used for protein detection and quantification, although their performance with many ion channels, including TRPM8, is suboptimal. Thus, the search for reliable antibodies is of utmost importance. In this study, we characterized the performance of six TRPM8 commercial antibodies in three immunodetection techniques: Western blot, immunocytochemistry and immunohistochemistry. Different outcomes were obtained for the tested antibodies; two of them proved to be successful in detecting TRPM8 in the three approaches while, in the conditions tested, the other four were acceptable only for specific techniques. Considering our results, we offer some insight into the usefulness of these antibodies for the detection of TRPM8 depending on the methodology of choice.


Subject(s)
TRPM Cation Channels , Transient Receptor Potential Channels , Humans , Antibodies/metabolism , Membrane Proteins/metabolism , Pain , Transient Receptor Potential Channels/metabolism , TRPM Cation Channels/metabolism
4.
Methods Mol Biol ; 1987: 83-97, 2019.
Article in English | MEDLINE | ID: mdl-31028675

ABSTRACT

The family of transient receptor potential (TRPs) channels contains 28 mammalian members, each a unique cellular sensor that responds to a wide variety of external and internal signals. TRP channels are expressed by most mammalian cells, where they are involved in many different physiological functions. Canonical TRP channels (TRPCs) form a family of nonselective cationic channels, although with greater selectivity for Ca2+. This family is made up of seven members (TRPC1-7), all of which contain a TRP box in the carboxyl terminal and 3-4 ankyrin repeats in the amino terminal. While these channels share some similar properties, they display diverse gating mechanisms and are involved in different signaling pathways (Gees M et al., Compr Physiol, 2012). The activation or inhibition of these channels has been studied using different approaches and techniques. Here, we characterize the activation of the TRPC5 channel expressed in a heterologous system, using calcium imaging and the patch-clamp technique in whole-cell configuration.


Subject(s)
Calcium/analysis , Patch-Clamp Techniques/methods , TRPC Cation Channels/metabolism , Fura-2/chemistry , HEK293 Cells , Humans , Microscopy, Fluorescence , Osmolar Concentration
5.
Pain ; 159(4): 739-748, 2018 04.
Article in English | MEDLINE | ID: mdl-29319609

ABSTRACT

The mechanisms whereby deposition of monosodium urate (MSU) crystals in gout activates nociceptors to induce joint pain are incompletely understood. We tried to reproduce the signs of painful gouty arthritis, injecting into the knee joint of rats suspensions containing amorphous or triclinic, needle MSU crystals. The magnitude of MSU-induced inflammation and pain behavior signs were correlated with the changes in firing frequency of spontaneous and movement-evoked nerve impulse activity recorded in single knee joint nociceptor saphenous nerve fibers. Joint swelling, mechanical and cold allodynia, and hyperalgesia appeared 3 hours after joint injection of MSU crystals. In parallel, spontaneous and movement-evoked joint nociceptor impulse activity raised significantly. Solutions containing amorphous or needle-shaped MSU crystals had similar inflammatory and electrophysiological effects. Intra-articular injection of hyaluronan (HA, Synvisc), a high-MW glycosaminoglycan present in the synovial fluid with analgesic effects in osteoarthritis, significantly reduced MSU-induced behavioral signs of pain and decreased the enhanced joint nociceptor activity. Our results support the interpretation that pain and nociceptor activation are not triggered by direct mechanical stimulation of nociceptors by MSU crystals, but are primarily caused by the release of excitatory mediators by inflammatory cells activated by MSU crystals. Intra-articular HA decreased behavioral and electrophysiological signs of pain, possibly through its viscoelastic filtering effect on the mechanical forces acting over sensitized joint sensory endings and probably also by a direct interaction of HA molecules with the transducing channels expressed in joint nociceptor terminals.


Subject(s)
Acute Pain/etiology , Adjuvants, Immunologic/therapeutic use , Gout/complications , Gout/drug therapy , Hyaluronic Acid/therapeutic use , Acute Pain/physiopathology , Animals , Antioxidants/toxicity , Disease Models, Animal , Flow Cytometry , Gout/pathology , Inflammation/drug therapy , Inflammation/etiology , Injections, Intra-Articular , Knee Joint/innervation , Knee Joint/pathology , Male , Nerve Fibers/physiology , Pain Threshold/drug effects , Physical Stimulation/adverse effects , Rats , Rats, Wistar , Uric Acid/toxicity , Weight-Bearing/physiology
7.
Nat Commun ; 6: 8095, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26311398

ABSTRACT

Hyaluronan (HA) is present in the extracellular matrix of all body tissues, including synovial fluid in joints, in which it behaves as a filter that buffers transmission of mechanical forces to nociceptor nerve endings thereby reducing pain. Using recombinant systems, mouse-cultured dorsal root ganglia (DRG) neurons and in vivo experiments, we found that HA also modulates polymodal transient receptor potential vanilloid subtype 1 (TRPV1) channels. HA diminishes heat, pH and capsaicin (CAP) responses, thus reducing the opening probability of the channel by stabilizing its closed state. Accordingly, in DRG neurons, HA decreases TRPV1-mediated impulse firing and channel sensitization by bradykinin. Moreover, subcutaneous HA injection in mice reduces heat and capsaicin nocifensive responses, whereas the intra-articular injection of HA in rats decreases capsaicin joint nociceptor fibres discharge. Collectively, these results indicate that extracellular HA reduces the excitability of the ubiquitous TRPV1 channel, thereby lowering impulse activity in the peripheral nociceptor endings underlying pain.


Subject(s)
Adjuvants, Immunologic/pharmacology , Hyaluronic Acid/pharmacology , Neurons/drug effects , Nociceptive Pain , Nociceptors/drug effects , Stifle/drug effects , TRPV Cation Channels/drug effects , Animals , Behavior, Animal/drug effects , Bradykinin/pharmacology , CHO Cells , Calcium/metabolism , Capsaicin/pharmacology , Cell Line, Tumor , Cricetulus , Ganglia, Spinal/cytology , HEK293 Cells , Hot Temperature , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Models, Molecular , Mutagenesis, Site-Directed , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar , Sensory System Agents/pharmacology , Stifle/innervation , TRPA1 Cation Channel , TRPM Cation Channels/drug effects , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism , Vasodilator Agents/pharmacology
8.
Handb Exp Pharmacol ; 222: 547-79, 2014.
Article in English | MEDLINE | ID: mdl-24756721

ABSTRACT

Transient receptor potential melastatin 8 (TRPM8) was originally cloned from prostate tissue. Shortly thereafter, the protein was identified as a cold- and menthol-activated ion channel in peripheral sensory neurons, where it plays a critical role in cold temperature detection. In this chapter, we review our current understanding of the molecular and biophysical properties, the pharmacology, and the modulation by signaling molecules of this TRP channel. Finally, we examine the physiological role of TRPM8 and its emerging link to various human diseases, including pain, prostate cancer, dry eye disease, and metabolic disorders.


Subject(s)
TRPM Cation Channels/metabolism , Animals , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Membrane Potentials , Membrane Transport Modulators/pharmacology , Mice , Mice, Knockout , Phenotype , Protein Conformation , Signal Transduction , Structure-Activity Relationship , TRPM Cation Channels/chemistry , TRPM Cation Channels/deficiency , TRPM Cation Channels/drug effects , TRPM Cation Channels/genetics
9.
PLoS One ; 7(12): e52475, 2012.
Article in English | MEDLINE | ID: mdl-23300680

ABSTRACT

The hippocampus plays an important role in short term memory, learning and spatial navigation. A characteristic feature of the hippocampal region is its expression of different electrical population rhythms and activities during different brain states. Physiological fluctuations in brain temperature affect the activity patterns in hippocampus, but the underlying cellular mechanisms are poorly understood. In this work, we investigated the thermal modulation of hippocampal activity at the cellular network level. Primary cell cultures of mouse E17 hippocampus displayed robust network activation upon light cooling of the extracellular solution from baseline physiological temperatures. The activity generated was dependent on action potential firing and excitatory glutamatergic synaptic transmission. Involvement of thermosensitive channels from the transient receptor potential (TRP) family in network activation by temperature changes was ruled out, whereas pharmacological and immunochemical experiments strongly pointed towards the involvement of temperature-sensitive two-pore-domain potassium channels (K(2P)), TREK/TRAAK family. In hippocampal slices we could show an increase in evoked and spontaneous synaptic activity produced by mild cooling in the physiological range that was prevented by chloroform, a K(2P) channel opener. We propose that cold-induced closure of background TREK/TRAAK family channels increases the excitability of some hippocampal neurons, acting as a temperature-sensitive gate of network activation. Our findings in the hippocampus open the possibility that small temperature variations in the brain in vivo, associated with metabolism or blood flow oscillations, act as a switch mechanism of neuronal activity and determination of firing patterns through regulation of thermosensitive background potassium channel activity.


Subject(s)
Cold Temperature , Hippocampus/cytology , Nerve Net/cytology , Neurons/cytology , Animals , Cells, Cultured , Evoked Potentials/drug effects , Glutamic Acid/metabolism , Hippocampus/physiology , Membrane Potentials/drug effects , Mice , Nerve Net/drug effects , Nerve Net/physiology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Potassium Channels/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Synapses/drug effects , Synapses/metabolism , Tetrodotoxin/pharmacology , Time Factors
10.
J Neurosci ; 29(10): 3120-31, 2009 Mar 11.
Article in English | MEDLINE | ID: mdl-19279249

ABSTRACT

Molecular determinants of threshold differences among cold thermoreceptors are unknown. Here we show that such differences correlate with the relative expression of I(KD), a current dependent on Shaker-like Kv1 channels that acts as an excitability brake, and I(TRPM8), a cold-activated excitatory current. Neurons responding to small temperature changes have high functional expression of TRPM8 (transient receptor potential cation channel, subfamily M, member 8) and low expression of I(KD). In contrast, neurons activated by lower temperatures have a lower expression of TRPM8 and a prominent I(KD). Otherwise, both subpopulations have nearly identical membrane and firing properties, suggesting that they belong to the same neuronal pool. Blockade of I(KD) shifts the threshold of cold-sensitive neurons to higher temperatures and augments cold-evoked nocifensive responses in mice. Similar behavioral effects of I(KD) blockade were observed in TRPA1(-/-) mice. Moreover, only a small percentage of trigeminal cold-sensitive neurons were activated by TRPA1 agonists, suggesting that TRPA1 does not play a major role in the detection of low temperatures by uninjured somatic cold-specific thermosensory neurons under physiological conditions. Collectively, these findings suggest that innocuous cooling sensations and cold discomfort are signaled by specific low- and high-threshold cold thermoreceptor neurons, differing primarily in their relative expression of two ion channels having antagonistic effects on neuronal excitability. Thus, although TRPM8 appears to function as a critical cold sensor in the majority of peripheral sensory neurons, the expression of Kv1 channels in the same terminals seem to play an important role in the peripheral gating of cold-evoked discomfort and pain.


Subject(s)
Cold Temperature , Sensory Thresholds/physiology , Shaker Superfamily of Potassium Channels/physiology , TRPM Cation Channels/physiology , Thermoreceptors/physiology , Trigeminal Ganglion/physiology , Animals , Cells, Cultured , Male , Mice , Mice, Knockout , Neurons/physiology , TRPA1 Cation Channel , Thermosensing/physiology , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/physiology
11.
J Physiol ; 587(Pt 9): 1961-76, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19273581

ABSTRACT

Hyperpolarization-activated currents (I(h)) are mediated by the expression of combinations of hyperpolarization-activated, cyclic nucleotide-gated (HCN) channel subunits (HCN1-4). These cation currents are key regulators of cellular excitability in the heart and many neurons in the nervous system. Subunit composition determines the gating properties and cAMP sensitivity of native I(h) currents. We investigated the functional properties of I(h) in adult mouse cold thermoreceptor neurons from the trigeminal ganglion, identified by their high sensitivity to moderate cooling and responsiveness to menthol. All cultured cold-sensitive (CS) neurons expressed a fast activating I(h), which was fully blocked by extracellular Cs(+) or ZD7288 and had biophysical properties consistent with those of heteromeric HCN1-HCN2 channels. In CS neurons from HCN1(-/-) animals, I(h) was greatly reduced but not abolished. We find that I(h) activity is not essential for the transduction of cold stimuli in CS neurons. Nevertheless, I(h) has the potential to shape the excitability of CS neurons. First, I(h) blockade caused a membrane hyperpolarization in CS neurons of about 5 mV. Furthermore, impedance power analysis showed that all CS neurons had a prominent subthreshold membrane resonance in the 5-7 Hz range, completely abolished upon blockade of I(h) and absent in HCN1 null mice. This frequency range matches the spontaneous firing frequency of cold thermoreceptor terminals in vivo. Behavioural responses to cooling were reduced in HCN1 null mice and after peripheral pharmacological blockade of I(h) with ZD7288, suggesting that I(h) plays an important role in peripheral sensitivity to cold.


Subject(s)
Action Potentials/physiology , Biological Clocks/physiology , Cold Temperature , Membrane Potentials/physiology , Thermoreceptors/physiology , Thermosensing/physiology , Animals , Mice
12.
J Physiol ; 581(Pt 1): 155-74, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17317754

ABSTRACT

TRPM8, a member of the melastatin subfamily of transient receptor potential (TRP) cation channels, is activated by voltage, low temperatures and cooling compounds. These properties and its restricted expression to small sensory neurons have made it the ion channel with the most advocated role in cold transduction. Recent work suggests that activation of TRPM8 by cold and menthol takes place through shifts in its voltage-activation curve, which cause the channel to open at physiological membrane potentials. By contrast, little is known about the actions of inhibitors on the function of TRPM8. We investigated the chemical and thermal modulation of TRPM8 in transfected HEK293 cells and in cold-sensitive primary sensory neurons. We show that cold-evoked TRPM8 responses are effectively suppressed by inhibitor compounds SKF96365, 4-(3-chloro-pyridin-2-yl)-piperazine-1-carboxylic acid (4-tert-butyl-phenyl)-amide (BCTC) and 1,10-phenanthroline. These antagonists exert their effect by shifting the voltage dependence of TRPM8 activation towards more positive potentials. An opposite shift towards more negative potentials is achieved by the agonist menthol. Functionally, the bidirectional shift in channel gating translates into a change in the apparent temperature threshold of TRPM8-expressing cells. Accordingly, in the presence of the antagonist compounds, the apparent response-threshold temperature of TRPM8 is displaced towards colder temperatures, whereas menthol sensitizes the response, shifting the threshold in the opposite direction. Co-application of agonists and antagonists produces predictable cancellation of these effects, suggesting the convergence on a common molecular process. The potential for half maximal activation of TRPM8 activation by cold was approximately 140 mV more negative in native channels compared to recombinant channels, with a much higher open probability at negative membrane potentials in the former. In functional terms, this difference translates into a shift in the apparent temperature threshold for activation towards higher temperatures for native currents. This difference in voltage-dependence readily explains the high threshold temperatures characteristic of many cold thermoreceptors. The modulation of TRPM8 activity by different chemical agents unveils an important flexibility in the temperature-response curve of TRPM8 channels and cold thermoreceptors.


Subject(s)
Cold Temperature , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , TRPM Cation Channels/drug effects , TRPM Cation Channels/physiology , Thermoreceptors/physiology , Animals , Calcium Channel Blockers/pharmacology , Cell Line , Evoked Potentials/drug effects , Evoked Potentials/physiology , Humans , Imidazoles/pharmacology , Membrane Potentials/physiology , Methanol/pharmacology , Mice , Phenanthrolines/pharmacology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Pyridines/pharmacology , Rats , Thermoreceptors/drug effects , Transfection
13.
J Physiol ; 567(Pt 2): 415-26, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15961432

ABSTRACT

Different classes of ion channels have been implicated in sensing cold temperatures at mammalian thermoreceptor nerve endings. A major candidate is TRPM8, a non-selective cation channel of the transient receptor potential family, activated by menthol and low temperatures. We investigated the role of TRPM8 in cold sensing during transient expression in mouse cultured hippocampal neurones, a tissue that lacks endogenous expression of thermosensitive TRPs. In the absence of synaptic input, control hippocampal neurones were not excited by cooling. In contrast, all TRPM8-transfected hippocampal neurones were excited by cooling and menthol. However, in comparison to cold-sensitive trigeminal sensory neurones, hippocampal neurones exhibited much lower threshold temperatures, requiring temperatures below 27 degrees C to fire action potentials. These results directly demonstrate that expression of TRPM8 in mammalian neurones induces cold sensing, albeit at lower temperatures than native TRPM8-expressing neurones, suggesting the presence of additional modulatory mechanisms in the cold response of sensory neurones.


Subject(s)
Cold Temperature , Hippocampus/physiology , Ion Channels/metabolism , Neoplasm Proteins/metabolism , Neurons, Afferent/physiology , Thermosensing/physiology , Animals , Cells, Cultured , Humans , Ion Channels/genetics , Mice , Neoplasm Proteins/genetics , TRPM Cation Channels , Transfection
14.
Proc Natl Acad Sci U S A ; 99(4): 2374-9, 2002 Feb 19.
Article in English | MEDLINE | ID: mdl-11854530

ABSTRACT

Vanilloid receptor subunit 1 (VR1) appears to play a critical role in the transduction of noxious chemical and thermal stimuli by sensory nerve endings in peripheral tissues. Thus, VR1 antagonists are useful compounds to unravel the contribution of this receptor to pain perception, as well as to induce analgesia. We have used a combinatorial approach to identify new, nonpeptidic channel blockers of VR1. Screening of a library of trimers of N-alkylglycines resulted in the identification of two molecules referred to as DD161515 [N-[2-(2-(N-methylpyrrolidinyl)ethyl]glycyl]-[N-[2,4-dichlorophenethyl]glycyl]-N-(2,4-dichlorophenethyl)glycinamide] and DD191515 [[N-[3-(N,N-diethylamino)propyl]glycyl]-[N-[2,4-dichlorophenethyl]glycyl]-N-(2,4-dichlorophenethyl)glycinamide] that selectively block VR1 channel activity with micromolar efficacy, rivaling that characteristic of vanilloid-related inhibitors. These compounds appear to be noncompetitive VR1 antagonists that recognize a receptor site distinct from that of capsaicin. Intraperitoneal administration of both trialkylglycines into mice significantly attenuated thermal nociception as measured in the hot plate test. It is noteworthy that these compounds eliminated pain and neurogenic inflammation evoked by intradermal injection of capsaicin into the animal hindpaw, as well as the thermal hyperalgesia induced by tissue irritation with nitrogen mustard. In contrast, responses to mechanical stimuli were not modified by either compound. Modulation of sensory nerve fibers excitability appears to underlie the peptoid analgesic activity. Collectively, these results indicate that blockade of VR1 activity attenuates chemical and thermal nociception and hyperalgesia, supporting the tenet that this ionotropic receptor contributes to chemical and thermal sensitivity and pain perception in vivo. These trialkylglycine-based, noncompetitive VR1 antagonists may likely be developed into analgesics to treat inflammatory pain.


Subject(s)
Hot Temperature , Hyperalgesia , Pain/drug therapy , Receptors, Drug/antagonists & inhibitors , Animals , Calcium/metabolism , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Glycine/pharmacology , Inflammation/drug therapy , Knee/physiology , Male , Mice , Mice, Inbred ICR , Mustard Plant , Neurons/metabolism , Pain Threshold , Peptoids , Plant Extracts/pharmacology , Plant Oils , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Stress, Mechanical , Time Factors , Xenopus
15.
Nat Neurosci ; 5(3): 254-60, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11836533

ABSTRACT

Sensations of cold are mediated by specific thermoreceptor nerve endings excited by low temperature and menthol. Here we identify a population of cold-sensitive cultured mouse trigeminal ganglion neurons with a unique set of biophysical properties. Their impulse activity during cooling and menthol application was similar to that of cold thermoreceptor fibers in vivo. We show that cooling closes a background K+ channel, causing depolarization and firing that is limited by the slower reduction of a cationic inward current (Ih). In cold-insensitive neurons, firing is prevented by a slow, transient, 4-AP-sensitive K+ current (IKD) that acts as an excitability brake. In addition, pharmacological blockade of IKD induced thermosensitivity in cold-insensitive neurons, a finding that may explain cold allodynia in neuropathic pain. These results suggest that cold sensitivity is not associated to a specific transduction molecule but instead results from a favorable blend of ionic channels expressed in a small subset of sensory neurons.


Subject(s)
Cold Temperature , Potassium Channels/metabolism , Thermoreceptors/metabolism , Thermosensing/physiology , 4-Aminopyridine/pharmacology , Action Potentials/physiology , Animals , Calcium/metabolism , Menthol/pharmacology , Mice , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Signal Transduction/physiology , Thermoreceptors/cytology , Thermoreceptors/drug effects , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL