Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
PLoS One ; 8(8): e71809, 2013.
Article in English | MEDLINE | ID: mdl-23977150

ABSTRACT

The KCNQ2 gene product, Kv7.2, is a subunit of the M-channel, a low-threshold voltage-gated K(+) channel that regulates mammalian and human neuronal excitability. Spontaneous mutations one of the KCNQ2 genes cause disorders of neural excitability such as Benign Familial Neonatal Seizures. However there appear to be no reports in which both human KCNQ2 genes are mutated. We therefore asked what happens to M-channel function when both KCNQ2 genes are disrupted. We addressed this using sympathetic neurons isolated from mice in which the KCNQ2 gene was truncated at a position corresponding to the second transmembrane domain of the Kv7.2 protein. Since homozygote KCNQ2-/- mice die postnatally, experiments were largely restricted to neurons from late embryos. Quantitative PCR revealed an absence of KCNQ2 mRNA in ganglia from KCNQ2-/- embryos but 100-120% increase of KCNQ3 and KCNQ5 mRNAs; KCNQ2+/- ganglia showed ∼30% less KCNQ2 mRNA than wild-type (+/+) ganglia but 40-50% more KCNQ3 and KCNQ5 mRNA. Neurons from KCNQ2-/- embryos showed a complete absence of M-current, even after applying the Kv7 channel enhancer, retigabine. Neurons from heterozygote KCNQ2+/- embryos had ∼60% reduced M-current. In contrast, M-currents in neurons from adult KCNQ2+/- mice were no smaller than those in neurons from wild-type mice. Measurements of tetraethylammonium block did not indicate an increased expression of Kv7.5-containing subunits, implying a compensatory increase in Kv7.2 expression from the remaining KCNQ2 gene. We conclude that mouse embryonic M-channels have an absolute requirement for Kv7.2 subunits for functionality, that the reduced M-channel activity in heterozygote KCNQ2+/- mouse embryos results primarily from a gene-dosage effect, and that there is a compensatory increase in Kv7.2 expression in adult mice.


Subject(s)
Action Potentials , KCNQ2 Potassium Channel/genetics , Nerve Tissue Proteins/genetics , Neurons/physiology , Animals , Carbamates/pharmacology , Cells, Cultured , Female , Gene Expression , Humans , KCNQ2 Potassium Channel/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Potassium Channel Blockers/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Superior Cervical Ganglion/cytology , Tetraethylammonium/pharmacology
2.
Pflugers Arch ; 465(9): 1371-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23592175

ABSTRACT

Skin keratinocytes fulfil important signalling and protective functions. Immunocytochemical experiments revealed the unexpected presence of immunoreactivity for the M-type potassium channel subunit Kv7.2 in the keratinocyte layer of intact rat paw skin and in keratinocytes isolated from the skin of 1-day-old rats and cultured in vitro for 3-10 days. Application of the M-channel enhancer retigabine (3-10 µM) to isolated cultured rat keratinocytes: (a) increased outward membrane currents recorded under voltage clamp, (b) produced ~3 mV hyperpolarization at rest, (c) enhanced ~3-fold the release of ATP induced by the TRPV3 agonist carvacrol (1 mM) and (d) increased the amplitude of the carvacrol-induced intracellular Ca(2+) transient measured with Fura-2. The effect of retigabine on ATP release was prevented by the M-channel blocking agent XE991. We conclude that rat skin keratinocytes possess M-channels that, when activated, can modify their physiological properties, with potential significance for their sensory and other biological functions.


Subject(s)
KCNQ2 Potassium Channel/metabolism , Keratinocytes/metabolism , Skin/metabolism , Action Potentials , Adenosine Triphosphate/metabolism , Animals , Anthracenes/pharmacology , Calcium/metabolism , Carbamates/pharmacology , Cells, Cultured , Cymenes , KCNQ2 Potassium Channel/antagonists & inhibitors , Keratinocytes/physiology , Monoterpenes/pharmacology , Phenylenediamines/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Skin/cytology , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism
3.
Front Mol Neurosci ; 5: 63, 2012.
Article in English | MEDLINE | ID: mdl-22593734

ABSTRACT

M-channels carry slowly activating potassium currents that regulate excitability in a variety of central and peripheral neurons. Functional M-channels and their Kv7 channel correlates are expressed throughout the somatosensory nervous system where they may play an important role in controlling sensory nerve activity. Here we show that Kv7.2 immunoreactivity is expressed in the peripheral terminals of nociceptive primary afferents. Electrophysiological recordings from single afferents in vitro showed that block of M-channels by 3 µM XE991 sensitized Aδ- but not C-fibers to noxious heat stimulation and induced spontaneous, ongoing activity at 32°C in many Aδ-fibers. These observations were extended in vivo: intraplantar injection of XE991 selectively enhanced the response of deep dorsal horn (DH) neurons to peripheral mid-range mechanical and higher range thermal stimuli, consistent with a selective effect on Aδ-fiber peripheral terminals. These results demonstrate an important physiological role of M-channels in controlling nociceptive Aδ-fiber responses and provide a rationale for the nocifensive behaviors that arise following intraplantar injection of the M-channel blocker XE991.

4.
J Clin Invest ; 120(5): 1380-3, 2010 May.
Article in English | MEDLINE | ID: mdl-20424321

ABSTRACT

Bradykinin is the most potent endogenous inducer of acute pain. However, the way in which it excites nociceptive sensory nerve endings is still unclear. In an article recently published in the JCI, Liu et al. suggest a new mechanism via which bradykinin induces acute spontaneous pain. The authors report that the stimulation of B2 bradykinin receptors by bradykinin triggers the release of intracellular calcium ions from nociceptive sensory neurons of rat dorsal root ganglia. This depolarizes the sensory nerve endings by simultaneously closing M-type potassium channels and opening TMEM16A chloride channels, resulting in the production of nociceptive signals. Here, we discuss the relationship between this effect and a previously described mechanism for pain sensitization and evaluate its potential significance for therapeutic pain control. A separate study by Patwardhan et al. in this issue of the JCI identifies oxidized linoleic acid metabolites as novel mediators of thermally induced pain.


Subject(s)
Bradykinin/metabolism , Pain Management , Pain/metabolism , Animals , Calcium/metabolism , Ganglia, Spinal/metabolism , Humans , Hydrolysis , Ions , Kinetics , Linoleic Acid/metabolism , Models, Biological , Nociceptors/metabolism , Potassium Channels/metabolism , Rats , Receptor, Bradykinin B2/metabolism
5.
Br J Pharmacol ; 156(8): 1185-95, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19298256

ABSTRACT

KCNQ genes encode five Kv7 K(+) channel subunits (Kv7.1-Kv7.5). Four of these (Kv7.2-Kv7.5) are expressed in the nervous system. Kv7.2 and Kv7.3 are the principal molecular components of the slow voltage-gated M-channel, which widely regulates neuronal excitability, although other subunits may contribute to M-like currents in some locations. M-channels are closed by receptors coupled to Gq such as M1 and M3 muscarinic receptors; this increases neuronal excitability and underlies some forms of cholinergic excitation. Muscarinic closure results from activation of phospholipase C and consequent hydrolysis and depletion of membrane phosphatidylinositol-4,5-bisphosphate, which is required for channel opening. Some effects of M-channel closure, determined from transmitter action, selective blocking drugs (linopirdine and XE991) and KCNQ2 gene disruption or manipulation, are as follows: (i) in sympathetic neurons: facilitation of repetitive discharges and conversion from phasic to tonic firing; (ii) in sensory nociceptive systems: facilitation of A-delta peripheral sensory fibre responses to noxious heat; and (iii) in hippocampal pyramidal neurons: facilitation of repetitive discharges, enhanced after-depolarization and burst-firing, and induction of spontaneous firing through a reduction of action potential threshold at the axon initial segment. Several drugs including flupirtine and retigabine enhance neural Kv7/M-channel activity, principally through a hyperpolarizing shift in their voltage gating. In consequence they reduce neural excitability and can inhibit nociceptive stimulation and transmission. Flupirtine is in use as a central analgesic; retigabine is under clinical trial as a broad-spectrum anticonvulsant and is an effective analgesic in animal models of chronic inflammatory and neuropathic pain.


Subject(s)
KCNQ Potassium Channels/metabolism , Neurons/metabolism , Potassium/metabolism , Synaptic Transmission , Acetylcholine/metabolism , Action Potentials , Adrenergic Fibers/metabolism , Analgesics/pharmacology , Animals , Anticonvulsants/pharmacology , CA1 Region, Hippocampal/metabolism , Humans , Ion Channel Gating , KCNQ Potassium Channels/drug effects , KCNQ Potassium Channels/genetics , Muscarinic Agonists/pharmacology , Neurons/drug effects , Potassium Channel Blockers/pharmacology , Pyramidal Cells/metabolism , Receptors, Muscarinic/metabolism , Sensory Receptor Cells/metabolism , Synaptic Transmission/drug effects
6.
PLoS One ; 3(4): e1961, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18398477

ABSTRACT

Recent studies have demonstrated the importance of local protein synthesis for neuronal plasticity. In particular, local mRNA translation through the mammalian target of rapamycin (mTOR) has been shown to play a key role in regulating dendrite excitability and modulating long-term synaptic plasticity associated with learning and memory. There is also increased evidence to suggest that intact adult mammalian axons have a functional requirement for local protein synthesis in vivo. Here we show that the translational machinery is present in some myelinated sensory fibers and that active mTOR-dependent pathways participate in maintaining the sensitivity of a subpopulation of fast-conducting nociceptors in vivo. Phosphorylated mTOR together with other downstream components of the translational machinery were localized to a subset of myelinated sensory fibers in rat cutaneous tissue. We then showed with electromyographic studies that the mTOR inhibitor rapamycin reduced the sensitivity of a population of myelinated nociceptors known to be important for the increased mechanical sensitivity that follows injury. Behavioural studies confirmed that local treatment with rapamycin significantly attenuated persistent pain that follows tissue injury, but not acute pain. Specifically, we found that rapamycin blunted the heightened response to mechanical stimulation that develops around a site of injury and reduced the long-term mechanical hypersensitivity that follows partial peripheral nerve damage--a widely used model of chronic pain. Our results show that the sensitivity of a subset of sensory fibers is maintained by ongoing mTOR-mediated local protein synthesis and uncover a novel target for the control of long-term pain states.


Subject(s)
Electromyography/methods , Neurons, Afferent/physiology , Nociceptors/metabolism , Animals , Electrophysiology/methods , Male , Neuronal Plasticity , Pain Management , Pain Measurement , Phosphorylation , Protein Kinases/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , TOR Serine-Threonine Kinases
7.
J Pharmacol Toxicol Methods ; 51(3): 201-8, 2005.
Article in English | MEDLINE | ID: mdl-15862465

ABSTRACT

Ion channels represent attractive targets in the development of novel analgesics for the treatment of pain. Dorsal root ganglion (DRG) neurones in culture can share characteristics with nociceptors in vivo and are frequently used to investigate the ion channels that underlie the transduction of noxious stimuli into electrical activity during sensory processing. In this article, I describe the methods used to prepare cultures of DRG neurones including the procedures for the dissection, enzymatic dissociation and plating. The criteria used to identify putative nociceptors in vitro are reviewed and using the M-current as an example I highlight how potential analgesic targets can be identified by combining the use of the voltage clamp technique with the use of selective pharmacological agents.


Subject(s)
Analgesics/pharmacology , Ganglia, Spinal/physiology , Neurons/physiology , Animals , Capsaicin/pharmacology , Carbamates/pharmacology , Cell Size , Cells, Cultured , Culture Media , Cytological Techniques , Electrophysiology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ion Channels/drug effects , Neurons/drug effects , Neurons/ultrastructure , Nociceptors/drug effects , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Rats , Rats, Sprague-Dawley
8.
J Neurosci ; 23(18): 7227-36, 2003 Aug 06.
Article in English | MEDLINE | ID: mdl-12904483

ABSTRACT

Neuronal hyperexcitability is a feature of epilepsy and both inflammatory and neuropathic pain. M currents [IK(M)] play a key role in regulating neuronal excitability, and mutations in neuronal KCNQ2/3 subunits, the molecular correlates of IK(M), have previously been linked to benign familial neonatal epilepsy. Here, we demonstrate that KCNQ/M channels are also present in nociceptive sensory systems. IK(M) was identified, on the basis of biophysical and pharmacological properties, in cultured neurons isolated from dorsal root ganglia (DRGs) from 17-d-old rats. Currents were inhibited by the M-channel blockers linopirdine (IC50, 2.1 microm) and XE991 (IC50, 0.26 microm) and enhanced by retigabine (10 microm). The expression of neuronal KCNQ subunits in DRG neurons was confirmed using reverse transcription-PCR and single-cell PCR analysis and by immunofluorescence. Retigabine, applied to the dorsal spinal cord, inhibited C and Adelta fiber-mediated responses of dorsal horn neurons evoked by natural or electrical afferent stimulation and the progressive "windup" discharge with repetitive stimulation in normal rats and in rats subjected to spinal nerve ligation. Retigabine also inhibited responses to intrapaw application of carrageenan in a rat model of chronic pain; this was reversed by XE991. It is suggested that IK(M) plays a key role in controlling the excitability of nociceptors and may represent a novel analgesic target.


Subject(s)
Neurons, Afferent/metabolism , Pain Management , Pain/metabolism , Potassium Channels/metabolism , Animals , Anthracenes/pharmacology , Anura , CHO Cells , Carbamates/pharmacology , Cells, Cultured , Cricetinae , Disease Models, Animal , Ganglia, Spinal/cytology , Hyperalgesia/physiopathology , Indoles/pharmacology , Male , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Oocytes/metabolism , Pain Measurement , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/genetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transfection
9.
J Neurosci ; 23(12): 5012-9, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12832524

ABSTRACT

KCNQ2 and KCNQ3 potassium-channel subunits can form both homomeric and heteromeric channels; the latter are thought to constitute native ganglionic M channels. We have tried to deduce the stoichiometric contributions of KCNQ2 and KCNQ3 subunits to currents generated by the coexpression of KCNQ2 and KCNQ3 cDNA plasmids in Chinese hamster ovary (CHO) cells, and to native M currents in dissociated rat superior cervical ganglion (SCG) neurons, by comparing the block of these currents produced by tetraethylammonium (TEA) with the block of currents generated by a tandem KCNQ3/2 construct. TEA concentration-inhibition curves against coexpressed KCNQ2 plus KCNQ3 currents, and against native M currents in SCG neurons from 6-week-old [postnatal day 45 (P45)] rats, were indistinguishable from those for the expressed tandem construct, and fully accorded with a 1:1 stoichiometry. Inhibition curves in neurons from younger (P17) rats could be better fitted assuming an additional small proportion of current carried by KCNQ2 homomultimers. Single-cell PCR yielded signals for KCNQ2, KCNQ3, and KCNQ5 mRNAs in all SCG neurons tested from both P17 and P45 rats. Quantitative PCR of whole-ganglion mRNA revealed stable levels of KCNQ2 and KCNQ5 mRNA between P7 and P45, but excess and incrementing levels of KCNQ3 mRNA. Increasing levels of KCNQ3 protein between P17 and P45 were confirmed by immunocytochemistry. We conclude that coexpressed KCNQ2 plus KCNQ3 cDNAs generate channels with 1:1 (KCNQ2:KCNQ3) stoichiometry in CHO cells and that native M channels in SCG neurons adopt the same conformation during development, assisted by the increased expression of KCNQ3 mRNA and protein.


Subject(s)
Neurons/metabolism , Potassium Channels/biosynthesis , Superior Cervical Ganglion/metabolism , Tetraethylammonium/pharmacology , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , KCNQ Potassium Channels , KCNQ2 Potassium Channel , KCNQ3 Potassium Channel , Kinetics , Neurons/cytology , Neurons/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/genetics , Potassium Channels, Voltage-Gated , Protein Subunits/biosynthesis , Protein Subunits/drug effects , Protein Subunits/genetics , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL