Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 728, 2022 02 07.
Article in English | MEDLINE | ID: mdl-35132099

ABSTRACT

Postsynaptic NMDARs at spinal synapses are required for postsynaptic long-term potentiation and chronic pain. However, how presynaptic NMDARs (PreNMDARs) in spinal nociceptor terminals control presynaptic plasticity and pain hypersensitivity has remained unclear. Here we report that PreNMDARs in spinal nociceptor terminals modulate synaptic transmission in a nociceptive tone-dependent manner. PreNMDARs depresses presynaptic transmission in basal state, while paradoxically causing presynaptic potentiation upon injury. This state-dependent modulation is dependent on Ca2+ influx via PreNMDARs. Small conductance Ca2+-activated K+ (SK) channels are responsible for PreNMDARs-mediated synaptic depression. Rather, tissue inflammation induces PreNMDARs-PKG-I-dependent BDNF secretion from spinal nociceptor terminals, leading to SK channels downregulation, which in turn converts presynaptic depression to potentiation. Our findings shed light on the state-dependent characteristics of PreNMDARs in spinal nociceptor terminals on modulating nociceptive transmission and revealed a mechanism underlying state-dependent transition. Moreover, we identify PreNMDARs in spinal nociceptor terminals as key constituents of activity-dependent pain sensitization.


Subject(s)
Chronic Pain/physiopathology , Nociceptors/metabolism , Presynaptic Terminals/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Calcium/metabolism , Chronic Pain/genetics , Chronic Pain/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Inflammation , Long-Term Potentiation , Long-Term Synaptic Depression , Mice , Mice, Transgenic , Periaqueductal Gray/cytology , Periaqueductal Gray/physiology , Potassium Channels, Calcium-Activated/genetics , Potassium Channels, Calcium-Activated/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Synaptic Transmission
2.
J Vis Exp ; (150)2019 08 27.
Article in English | MEDLINE | ID: mdl-31524869

ABSTRACT

Single-fiber recording has been a classical and effective electrophysiological technique over the last few decades because of its specific application for nerve fibers in the central and peripheral nervous systems. This method is particularly applicable to dorsal root ganglia (DRG), which are primary sensory neurons that exhibit a pseudo-unipolar structure of nervous processes. The patterns and features of the action potentials passed along axons are recordable in these neurons. The present study uses in vivo single-fiber recordings to observe the conduction failure of sciatic nerves in complete Freund's adjuvant (CFA)-treated rats. As the underlying mechanism cannot be studied using in vivo single-fiber recordings, patch-clamp-recordings of DRG neurons are performed on preparations of intact DRG with the attached sciatic nerve. These recordings reveal a positive correlation between conduction failure and the rising slope of the after-hyperpolarization potential (AHP) of DRG neurons in CFA-treated animals. The protocol for in vivo single fiber-recordings allows the classification of nerve fibers via the measurement of conduction velocity and monitoring of abnormal conditions in nerve fibers in certain diseases. Intact DRG with attached peripheral nerve allows observation of the activity of DRG neurons in most physiological conditions. Conclusively, single-fiber recording combined with electrophysiological recording of intact DRGs is an effective method to examine the role of conduction failure during the analgesic process.


Subject(s)
Ganglia, Spinal/diagnostic imaging , Ganglia, Spinal/physiopathology , Nerve Fibers, Unmyelinated/physiology , Neural Conduction/physiology , Sciatic Nerve/diagnostic imaging , Sciatic Nerve/physiopathology , Animals , Freund's Adjuvant/pharmacology , Ganglia, Spinal/drug effects , Male , Nerve Fibers, Unmyelinated/drug effects , Neural Conduction/drug effects , Rats, Sprague-Dawley , Sciatic Nerve/drug effects
3.
Int J Mol Sci ; 19(1)2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29303989

ABSTRACT

Sensory neuron types have been distinguished by distinct morphological and transcriptional characteristics. Excitability is the most fundamental functional feature of neurons. Mathematical models described by Hodgkin have revealed three types of neuronal excitability based on the relationship between firing frequency and applied current intensity. However, whether natural sensory neurons display different functional characteristics in terms of excitability and whether this excitability type undergoes plastic changes under pathological pain states have remained elusive. Here, by utilizing whole-cell patch clamp recordings, behavioral and pharmacological assays, we demonstrated that large dorsal root ganglion (DRG) neurons can be classified into three classes and four subclasses based on their excitability patterns, which is similar to mathematical models raised by Hodgkin. Analysis of hyperpolarization-activated cation current (Ih) revealed different magnitude of Ih in different excitability types of large DRG neurons, with higher Ih in Class 2-1 than that in Class 1, 2-2 and 3. This indicates a crucial role of Ih in the determination of excitability type of large DRG neurons. More importantly, this pattern of excitability displays plastic changes and transition under pathological pain states caused by peripheral nerve injury. This study sheds new light on the functional characteristics of large DRG neurons and extends functional classification of large DRG neurons by integration of transcriptomic and morphological characteristics.


Subject(s)
Action Potentials , Ganglia, Spinal/cytology , Neuralgia/physiopathology , Neurons, Afferent/physiology , Animals , Cells, Cultured , Ganglia, Spinal/physiopathology , Male , Neuronal Plasticity , Neurons, Afferent/classification , Rats , Rats, Sprague-Dawley
4.
Mol Pain ; 13: 1744806917707127, 2017.
Article in English | MEDLINE | ID: mdl-28587505

ABSTRACT

Cervical radiculopathic pain is a very common symptom that may occur with cervical spondylosis. Mechanical allodynia is often associated with cervical radiculopathic pain and is inadequately treated with current therapies. However, the precise mechanisms underlying cervical radiculopathic pain-associated mechanical allodynia have remained elusive. Compelling evidence from animal models suggests a role of large-diameter dorsal root ganglion neurons and plasticity of spinal circuitry attached with Aß fibers in mediating neuropathic pain. Whether cervical radiculopathic pain condition induces plastic changes of large-diameter dorsal root ganglion neurons and what mechanisms underlie these changes are yet to be known. With combination of patch-clamp recording, immunohistochemical staining, as well as behavioral surveys, we demonstrated that upon chronic compression of C7/8 dorsal root ganglions, large-diameter cervical dorsal root ganglion neurons exhibited frequent spontaneous firing together with hyperexcitability. Quantitative analysis of hyperpolarization-activated cation current ( Ih) revealed that Ih was greatly upregulated in large dorsal root ganglion neurons from cervical radiculopathic pain rats. This increased Ih was supported by the enhanced expression of hyperpolarization-activated, cyclic nucleotide-modulated channels subunit 3 in large dorsal root ganglion neurons. Blockade of Ih with selective antagonist, ZD7288 was able to eliminate the mechanical allodynia associated with cervical radiculopathic pain. This study sheds new light on the functional plasticity of a specific subset of large-diameter dorsal root ganglion neurons and reveals a novel mechanism that could underlie the mechanical allodynia associated with cervical radiculopathy.


Subject(s)
Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Neurons/cytology , Neurons/metabolism , Radiculopathy/etiology , Radiculopathy/metabolism , Animals , Chronic Pain/etiology , Chronic Pain/metabolism , Chronic Pain/pathology , Male , Membrane Potentials/physiology , Neuralgia/pathology , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Radiculopathy/pathology , Rats , Rats, Sprague-Dawley
5.
Sci Rep ; 6: 37251, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27853254

ABSTRACT

Tissue injury is known to produce inflammation and pain. Synaptic potentiation between peripheral nociceptors and spinal lamina I neurons has been proposed to serve as a trigger for chronic inflammatory pain. Gastrodin is a main bioactive constituent of the traditional Chinese herbal medicine Gastrodia elata Blume, which has been widely used as an analgesic since ancient times. However, its underlying cellular mechanisms have remained elusive. The present study demonstrated for the first time that gastrodin exhibits an analgesic effect at the spinal level on spontaneous pain, mechanical and thermal pain hypersensitivity induced by peripheral inflammation, which is not dependent on opioid receptors and without tolerance. This analgesia by gastrodin is at least in part mediated by depressing spinal synaptic potentiation via blockade of acid-sensing ion channels. Further studies with miniature EPSCs and paired-pulse ratio analysis revealed the presynaptic origin of the action of gastrodin, which involves a decrease in transmitter release probability. In contrast, neither basal nociception nor basal synaptic transmission was altered. This study revealed a dramatic analgesic action of gastrodin on inflammatory pain and uncovered a novel spinal mechanism that could underlie the analgesia by gastrodin, pointing the way to a new analgesic for treating chronic inflammatory pain.


Subject(s)
Acid Sensing Ion Channel Blockers/pharmacology , Analgesia/methods , Benzyl Alcohols/pharmacology , Chronic Pain , Glucosides/pharmacology , Spine/metabolism , Synaptic Potentials/drug effects , Acid Sensing Ion Channels/metabolism , Animals , Chronic Pain/drug therapy , Chronic Pain/metabolism , Chronic Pain/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Mice , Spine/pathology
6.
Pain ; 157(10): 2235-2247, 2016 10.
Article in English | MEDLINE | ID: mdl-27583680

ABSTRACT

Although conduction failure has been observed in nociceptive C-fibers, little is known regarding its significance or therapeutic potential. In a previous study, we demonstrated that C-fiber conduction failure, which is regarded as an intrinsic self-inhibition mechanism, was reduced in circumstances of painful diabetic neuropathy. In this study, we extend this finding in the complete Freund's adjuvant model of inflammatory pain and validate that the degree of conduction failure decreased and led to a greater amount of pain signals conveyed to the central nervous system. In complete Freund's adjuvant-injected animals, conduction failure occurred in a C-fiber-selective, activity-dependent manner and was associated with an increase in the rising slope of the C-fiber after-hyperpolarization potential. To target conduction failure in a therapeutic modality, we used ZD7288, an antagonist of hyperpolarization-activated, cyclic nucleotide-modulated channels which are activated by hyperpolarization and play a pivotal role in both inflammatory and neuropathic pain. ZD7288 promoted conduction failure by suppressing Ih as a mechanism to reduce the rising slope of the after-hyperpolarization potential. Moreover, perineuronal injection of ZD7288 inhibited abnormal mechanical allodynia and thermal hyperalgesia without affecting motor function or heart rate. Our data highlight the analgesic potential of local ZD7288 application and identify conduction failure as a novel target for analgesic therapeutic development.


Subject(s)
Nerve Fibers, Unmyelinated/physiology , Neural Conduction/physiology , Neurons/physiology , Pain/pathology , Animals , Biophysics , Calcium/metabolism , Disease Models, Animal , Female , Freund's Adjuvant/toxicity , Ganglia, Spinal/cytology , Hyperalgesia/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Inflammation/chemically induced , Inflammation/complications , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nerve Fibers, Unmyelinated/drug effects , Neural Conduction/drug effects , Neurons/drug effects , Pain/etiology , Pain Threshold/drug effects , Patch-Clamp Techniques , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley
7.
J Neurophysiol ; 115(2): 947-57, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26609114

ABSTRACT

It is known that some patients with diabetic neuropathy are usually accompanied by abnormal painful sensations. Evidence has accumulated that diabetic neuropathic pain is associated with the hyperexcitability of peripheral nociceptors. Previously, we demonstrated that reduced conduction failure of polymodal nociceptive C-fibers and enhanced voltage-dependent sodium currents of small dorsal root ganglion (DRG) neurons contribute to diabetic hyperalgesia. To further investigate whether and how potassium channels are involved in the conduction failure, α-dendrotoxin (α-DTX), a selective blocker of the low-threshold sustained Kv1 channel, was chosen to examine its functional capability in modulating the conduction properties of polymodal nociceptive C-fibers and the excitability of sensory neurons. We found that α-DTX reduced the conduction failure of C-fibers from coccygeal nerve in vivo accompanied by an increased initial conduction velocity but a decreased activity-dependent slowing of conduction velocity. In addition, the number of APs evoked by step currents was significantly enhanced after the treatment with α-DTX in small-diameter sensory neurons. Further study of the mechanism indicates α-DTX-sensitive K(+) current significantly reduced and the activation of this current in peak and steady state shifted to depolarization for diabetic neurons. Expression of Kv channel subunits Kv1.2 and Kv1.6 was downregulated in both small dorsal root ganglion neurons and peripheral C-fibers. Taken together, these results suggest that α-DTX-sensitive Kv1 channels might play an important role in regulating the conduction properties of polymodal nociceptive C-fibers and firing properties of sensory neurons.


Subject(s)
Action Potentials , Diabetic Neuropathies/metabolism , Nerve Fibers, Unmyelinated/metabolism , Nociception , Shaker Superfamily of Potassium Channels/metabolism , Animals , Cells, Cultured , Diabetic Neuropathies/physiopathology , Down-Regulation , Elapid Venoms/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , Male , Nerve Fibers, Unmyelinated/physiology , Neurons/metabolism , Neurons/physiology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Shaker Superfamily of Potassium Channels/genetics
8.
Front Pharmacol ; 6: 251, 2015.
Article in English | MEDLINE | ID: mdl-26578961

ABSTRACT

Postoperative cognitive dysfunction (POCD), mainly characterized by short-term decline of learning and memory, occurs after operations under anesthesia. However, the underlying mechanisms are poorly understood. The µ-opioid receptors (MOR) are highly expressed in interneurons of hippocampus, and is believed to be critical for the dysfunction of synaptic plasticity between hippocampal neurons. Therefore, we investigated the effect of fentanyl, a strong agonist of MOR and often used for anesthesia and analgesia in clinical settings, on hippocampal synaptic plasticity in the Schaffer-collateral CA1 pathway during acute exposure and washout in vitro. Our results revealed that acute fentanyl exposure (0.01, 0.1, 1 µM) dose-dependently increased the field excitatory postsynaptic potentials (fEPSPs), which was prevented by pre-administration of picrotoxin (50 µM) or MOR antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Phe-Thr-NH2 (CTOP, 10 µM). While fentanyl exposure-increased fEPSPs amplitude was prevented by picrotoxin [an inhibitor of γ-aminobutyric acid receptor (GABAR)] treatment or fentanyl washout, pretreatment of picrotoxin failed to prevent the fentanyl-impaired long-term potentiation (LTP) of synaptic strength as well as the fentanyl-enhanced long-term depression (LTD). These results demonstrated that fentanyl acute exposure and washout increases hippocampal excitability in the Schaffer-collateral CA1 pathway, depending on disinhibiting interneurons after MOR activation. In addition, fentanyl acute exposure and washout modulated synaptic plasticity, but the inhibitory activation was not critical. Elucidating the detailed mechanisms for synaptic dysfunction after fentanyl exposure and washout may provide insights into POCD generation after fentanyl anesthesia.

9.
Neural Plast ; 2015: 752782, 2015.
Article in English | MEDLINE | ID: mdl-26609438

ABSTRACT

Evidence has accumulated that reactive oxygen species and inflammation play crucial roles in the development of chronic pain, including radicular low back pain. Nonsteroid anti-inflammatory drugs (NSAIDs), for example, salicylic acid, aspirin, provided analgesic effects in various types of pain. However, long-term use of these drugs causes unwanted side effects, which limits their implication. Stable nitronyl (NIT) nitroxide radicals have been extensively studied as a unique and interesting class of new antioxidants for protection against oxidative damage. The present study synthesized a novel NIT nitroxide radical with salicylic acid framework (SANR) to provide synergistic effect of both antioxidation and antiinflammation. We demonstrated for the first time that both acute and repeated SANR treatment exerted dramatic analgesic effect in radicular low back pain mimicked by chronic compression of dorsal root ganglion in rats. This analgesic potency was more potent than that produced by classical NSAIDs aspirin and traditional nitroxide radical Tempol alone. Furthermore, SANR-induced behavioral analgesia is found to be mediated, at least in partial, by a reduction of ectopic spontaneous discharges in injured DRG neurons. Therefore, the synthesized NIT nitroxide radical coupling with salicylic acid framework may represent a novel potential therapeutic candidate for treatment of chronic pain, including radicular low back pain.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclic N-Oxides/therapeutic use , Hyperalgesia/drug therapy , Imidazoles/therapeutic use , Low Back Pain/drug therapy , Neurons/drug effects , Radiculopathy/drug therapy , Salicylic Acid/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Aspirin/pharmacology , Behavior, Animal/drug effects , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , Drug Synergism , Ganglia, Spinal/pathology , Hyperalgesia/etiology , Imidazoles/chemistry , Low Back Pain/pathology , Low Back Pain/psychology , Male , Radiculopathy/pathology , Radiculopathy/psychology , Rats , Rats, Sprague-Dawley , Spin Labels , Spinal Cord Compression/drug therapy , Spinal Cord Compression/pathology
10.
Sci Rep ; 5: 16713, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26577374

ABSTRACT

Cervical radiculopathy represents aberrant mechanical hypersensitivity. Primary sensory neuron's ability to sense mechanical force forms mechanotransduction. However, whether this property undergoes activity-dependent plastic changes and underlies mechanical hypersensitivity associated with cervical radiculopathic pain (CRP) is not clear. Here we show a new CRP model producing stable mechanical compression of dorsal root ganglion (DRG), which induces dramatic behavioral mechanical hypersensitivity. Amongst nociceptive DRG neurons, a mechanically sensitive neuron, isolectin B4 negative Aδ-type (IB4(-) Aδ) DRG neuron displays spontaneous activity with hyperexcitability after chronic compression of cervical DRGs. Focal mechanical stimulation on somata of IB4(-) Aδ neuron induces abnormal hypersensitivity. Upregulated HCN1 and HCN3 channels and increased Ih current on this subset of primary nociceptors underlies the spontaneous activity together with neuronal mechanical hypersensitivity, which further contributes to the behavioral mechanical hypersensitivity associated with CRP. This study sheds new light on the functional plasticity of a specific subset of nociceptive DRG neurons to mechanical stimulation and reveals a novel mechanism that could underlie the mechanical hypersensitivity associated with cervical radiculopathy.


Subject(s)
Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Mechanotransduction, Cellular , Nociceptors/metabolism , Radiculopathy/genetics , Radiculopathy/physiopathology , Animals , Disease Models, Animal , Gene Expression , Genes, fos , Hyperalgesia/etiology , Membrane Potentials , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Fibers, Myelinated/metabolism , Neuralgia/etiology , Phosphorylation , Radiculopathy/complications , Radiculopathy/etiology , Rats , Up-Regulation
11.
Neural Plast ; 2015: 197392, 2015.
Article in English | MEDLINE | ID: mdl-26101670

ABSTRACT

Recently several lines of evidence demonstrated that methylcobalamin (MeCbl) might have potential analgesic effect in experimental and clinical studies. However, it was reported that MeCbl had no effect on treating lumbar spinal stenosis induced pain. Thus, the effects of short-term and long-term administration of MeCbl were examined in the chronic compression of dorsal root ganglion (CCD) model. We found that mechanical allodynia was significantly inhibited by a continuous application of high dose and a single treatment of a super high dose of MeCbl. Little is known about mechanisms underlying the analgesia of MeCbl. We examined the effect of MeCbl on the spontaneous activity (SA), the excitability, and hyperpolarization-activated nonselective cation ion current in compressed medium-sized dorsal root ganglion (DRG) neurons using extracellular single fiber recording in vivo and whole-cell patch clamp in vitro. We found that MeCbl significantly inhibited the SA of A-type sensory neurons in a dose-dependent manner and inhibited the excitability of medium-sized DRG neurons. In addition, MeCbl also decreased I h current density in injured medium-sized DRG neurons. Our results proved that MeCbl might exert an analgesic effect through the inhibition I h current and then might inhibit the hyperexcitability of primary sensory neurons under neuropathic pain state.


Subject(s)
Analgesics/administration & dosage , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiopathology , Hyperalgesia/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Neurons/drug effects , Vitamin B 12/analogs & derivatives , Action Potentials/drug effects , Analgesics/therapeutic use , Animals , Female , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Neurons/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord Compression/complications , Vitamin B 12/administration & dosage , Vitamin B 12/therapeutic use
12.
Brain Res ; 1596: 1-12, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25449887

ABSTRACT

Intrinsic resonant frequency properties of neurons in the brain provide a basis for different behavioral states and determine the responding modality (i.e. excitability) of different neurons. Previously, three types of excitability behaviors in rat Mesencephalic V neurons were found. To investigate the different resonant frequency and ionic current mechanisms underlying different resonances among three types of excitability, we performed whole-cell patch recordings and applied ZAP current on Mesencephalic V cells (N=70) in rat slices. The results show that: Class 2 neurons (30/30) have two resonances with U-shaped voltage dependence. One is a high frequency resonance (75.4 ± 2.11 Hz) significant at depolarized potentials (about -50 mV) and the other is a low frequency resonance (5.46 ± 0.31 Hz) significant at hyperpolarized potentials (about -70 mV). Voltage clamp experiments reveal two non-inactivating currents operating in the subthreshold voltage range: (1) 4-aminopyridine sensitive K(+) current, which activates at membrane potentials positive than -60 mV and was blocked by 4-AP (50 µM), was underlying the high frequency resonance of Class 2 neuron; (2) h-current, which activates negative than -60mV and was blocked by the hyperpolarization-activated cyclic nucleotide-gated channel blocker ZD7288 (10 µM), was underlying the low frequency resonance. Class 1 neurons do not show voltage-dependence resonant behaviors (10/10). Class 3 neurons (23/30) have two resonances, which are similar with Class 2 in both resonant frequency and currents. Our results provide clear evidence for the existence of multiple kinds of frequency resonances and how the relationship between the resonant frequency and the ionic current is based in Mesencephalic V neurons.


Subject(s)
Biophysical Phenomena/physiology , Ion Channels/physiology , Membrane Potentials/physiology , Mesencephalon/cytology , Neurons/physiology , 4-Aminopyridine/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Biophysical Phenomena/drug effects , Electric Stimulation , Female , In Vitro Techniques , Ion Channels/antagonists & inhibitors , Male , Potassium Channel Blockers/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
13.
Neurosignals ; 22(2): 79-91, 2014.
Article in English | MEDLINE | ID: mdl-25592674

ABSTRACT

Mesencephalic trigeminal nucleus (Mes V) neurons represent an uncommon class of primary sensory neurons. Besides receiving somatosensory information, Mes V neurons are also involved in regulating multisensory information. The present review first describes the passive features as well as three important currents, followed by a distinct excitability classification and a description of the excitability transition of Mes V neurons. Furthermore, their resonance property, the existence of membrane oscillation and electrical coupling which may promote strong synchronization, as well as their function in controlling stretch reflex activity, are discussed.


Subject(s)
Electrophysiological Phenomena/physiology , Mesencephalon/physiology , Sensory Receptor Cells/physiology , Trigeminal Nuclei/physiology , Animals , Humans , Mesencephalon/cytology , Sensory Receptor Cells/cytology , Trigeminal Nuclei/cytology
14.
Neurosignals ; 21(3-4): 213-28, 2013.
Article in English | MEDLINE | ID: mdl-22869293

ABSTRACT

Usually, the main axon is assumed to faithfully conduct action potentials (APs). Recent data have indicated that neural processing can occur along the axonal path. However, the patterns and mechanisms of temporal coding are not clear. In the present study, single fiber recording was used to analyze activity-dependent modulation of AP trains in the main axons of C fibers in the rabbit saphenous nerve. Trains of 5 superthreshold electrical pulses at interstimulus intervals of 20 or 50 ms were applied to the nerve trunk for 200 s. The interspike intervals (ISIs) for these trains were compared to the input interstimulus intervals. Three basic types of C fibers were observed in response to repeated stimuli: first, the ISI between the first and second AP (ISI1-2) of type 1 was longer than the interstimulus interval; second, the ISI1-2 of type 2 showed wavelike fluctuations around the interstimulus interval, and third, the ISI1-2 of type 3 exhibited shorter intervals for a long period. Furthermore, both 4-aminopyridine-sensitive potassium and hyperpolarization-activated cation currents were involved in the modulation of ISI1-2 of train pulses. These data provide new evidence that multiple modes of neural conduction can occur along the main axons of C fibers.


Subject(s)
Action Potentials/physiology , Femoral Nerve/physiology , Nerve Fibers, Unmyelinated/physiology , Animals , Axons/physiology , Electric Stimulation , Electrophysiology , Female , Male , Neural Conduction/physiology , Rabbits
15.
Neural Plast ; 2013: 424651, 2013.
Article in English | MEDLINE | ID: mdl-24455309

ABSTRACT

Methylcobalamin (MeCbl), the activated form of vitamin B12, has been used to treat some nutritional diseases and other diseases in clinic, such as Alzheimer's disease and rheumatoid arthritis. As an auxiliary agent, it exerts neuronal protection by promoting regeneration of injured nerves and antagonizing glutamate-induced neurotoxicity. Recently several lines of evidence demonstrated that MeCbl may have potential analgesic effects in experimental and clinical studies. For example, MeCbl alleviated pain behaviors in diabetic neuropathy, low back pain and neuralgia. MeCbl improved nerve conduction, promoted the regeneration of injured nerves, and inhibited ectopic spontaneous discharges of injured primary sensory neurons. This review aims to summarize the analgesic effect and mechanisms of MeCbl at the present.


Subject(s)
Analgesics, Non-Narcotic , Pain/drug therapy , Vitamin B 12/analogs & derivatives , Vitamins/pharmacology , Vitamins/therapeutic use , Animals , Humans , Nerve Regeneration/drug effects , Neural Conduction/drug effects , Neuralgia/drug therapy , Vitamin B 12/pharmacology , Vitamin B 12/therapeutic use
16.
PLoS One ; 7(11): e50694, 2012.
Article in English | MEDLINE | ID: mdl-23209812

ABSTRACT

Epilepsy is one of the most common neurological disorders, yet its treatment remains unsatisfactory. Saikosaponin a (SSa), a triterpene saponin derived from Bupleurum chinensis DC., has been demonstrated to have significant antiepileptic activity in a variety of epilepsy models in vivo. However, the electrophysiological activities and mechanisms of the antiepileptic properties of SSa remain unclear. In this study, whole-cell current-clamp recordings were used to evaluate the anticonvulsant activities of SSa in the hippocampal neuronal culture (HNC) models of acquired epilepsy (AE) and status epilepticus (SE). Whole-cell voltage-clamp recordings were used to evaluate the modulation effects of SSa on NMDA-evoked current and sodium currents in cultured hippocampal neurons. We found that SSa effectively terminated spontaneous recurrent epileptiform discharges (SREDs) in the HNC model of AE and continuous epileptiform high-frequency bursts (SE) in the HNC model of SE, in a concentration-dependent manner with an IC(50) of 0.42 µM and 0.62 µM, respectively. Furthermore, SSa significantly reduced the peak amplitude of NMDA-evoked current and the peak current amplitude of I(NaP). These results suggest for the first time that the inhibitions of NMDA receptor current and I(NaP) may be the underlying mechanisms of SSa's anticonvulsant properties, including the suppression of SREDs and SE in the HNC models of AE and SE. In addition, effectively abolishing the refractory SE implies that SSa may be a potential anticonvulsant candidate for the clinical treatment of epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Neurons/drug effects , Neurons/metabolism , Oleanolic Acid/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/metabolism , Saponins/pharmacology , Sodium/metabolism , Animals , Cells, Cultured , Electrophysiology , Epilepsy/metabolism , Hippocampus/cytology , Oleanolic Acid/pharmacology , Rats , Rats, Sprague-Dawley , Status Epilepticus/metabolism
17.
Neurosci Bull ; 28(5): 618-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23054639

ABSTRACT

As sciatica and low back pain are among the most common medical complaints, many studies have duplicated these conditions in animals. Chronic compression of the dorsal root ganglion (CCD) is one of these models. The surgery is simple: after exposing the L4/L5 intervertebral foramina, stainless steel rods are implanted unilaterally, one rod for each vertebra, to chronically compress the lumbar dorsal root ganglion (DRG). Then, CCD can be used to simulate the clinical conditions caused by stenosis, such as a laterally herniated disc or foraminal stenosis. As the intraforaminal implantation of a rod results in neuronal somal hyperexcitability and spontaneous action potentials associated with hyperalgesia, spontaneous pain, and mechanical allodynia, CCD provides an animal model that mimics radicular pain in humans. This review concerns the mechanisms of neuronal hyperexcitability, focusing on various patterns of spontaneous discharge including one possible pain signal for mechanical allodynia - evoked bursting. Also, new data regarding its significant property of maintaining peripheral input are also discussed. Investigations using this animal model will enhance our understanding of the neural mechanisms for low back pain and sciatica. Furthermore, the peripheral location of the DRG facilitates its use as a locus for controlling pain with minimal central effects, in the hope of ultimately uncovering analgesics that block neuropathic pain without influencing physiological pain.


Subject(s)
Disease Models, Animal , Ganglia, Spinal/pathology , Low Back Pain/pathology , Nerve Compression Syndromes/pathology , Sciatica/pathology , Animals , Ganglia, Spinal/surgery , Humans , Low Back Pain/surgery , Lumbar Vertebrae/pathology , Lumbar Vertebrae/surgery , Nerve Compression Syndromes/surgery , Sciatica/surgery
18.
PLoS One ; 7(6): e39647, 2012.
Article in English | MEDLINE | ID: mdl-22761855

ABSTRACT

Painful diabetic neuropathy (PDN) is a common complication of diabetes mellitus and adversely affects the patients' quality of life. Evidence has accumulated that PDN is associated with hyperexcitability of peripheral nociceptive primary sensory neurons. However, the precise cellular mechanism underlying PDN remains elusive. This may result in the lacking of effective therapies for the treatment of PDN. The phenolic glucoside, gastrodin, which is a main constituent of the Chinese herbal medicine Gastrodia elata Blume, has been widely used as an anticonvulsant, sedative, and analgesic since ancient times. However, the cellular mechanisms underlying its analgesic actions are not well understood. By utilizing a combination of behavioral surveys and electrophysiological recordings, the present study investigated the role of gastrodin in an experimental rat model of STZ-induced PDN and to further explore the underlying cellular mechanisms. Intraperitoneal administration of gastrodin effectively attenuated both the mechanical allodynia and thermal hyperalgesia induced by STZ injection. Whole-cell patch clamp recordings were obtained from nociceptive, capsaicin-sensitive small diameter neurons of the intact dorsal root ganglion (DRG). Recordings from diabetic rats revealed that the abnormal hyperexcitability of neurons was greatly abolished by application of GAS. To determine which currents were involved in the antinociceptive action of gastrodin, we examined the effects of gastrodin on transient sodium currents (I(NaT)) and potassium currents in diabetic small DRG neurons. Diabetes caused a prominent enhancement of I(NaT) and a decrease of potassium currents, especially slowly inactivating potassium currents (I(AS)); these effects were completely reversed by GAS in a dose-dependent manner. Furthermore, changes in activation and inactivation kinetics of I(NaT) and total potassium current as well as I(AS) currents induced by STZ were normalized by GAS. This study provides a clear cellular basis for the peripheral analgesic action of gastrodin for the treatment of chronic pain, including PDN.


Subject(s)
Benzyl Alcohols/pharmacology , Diabetic Neuropathies/physiopathology , Glucosides/pharmacology , Hyperalgesia/prevention & control , Sensory Receptor Cells/physiology , Animals , Capsaicin/pharmacology , Hyperalgesia/physiopathology , Rats , Sensory Receptor Cells/drug effects , Streptozocin
19.
PLoS Biol ; 10(3): e1001283, 2012.
Article in English | MEDLINE | ID: mdl-22427743

ABSTRACT

Synaptic long-term potentiation (LTP) at spinal neurons directly communicating pain-specific inputs from the periphery to the brain has been proposed to serve as a trigger for pain hypersensitivity in pathological states. Previous studies have functionally implicated the NMDA receptor-NO pathway and the downstream second messenger, cGMP, in these processes. Because cGMP can broadly influence diverse ion-channels, kinases, and phosphodiesterases, pre- as well as post-synaptically, the precise identity of cGMP targets mediating spinal LTP, their mechanisms of action, and their locus in the spinal circuitry are still unclear. Here, we found that Protein Kinase G1 (PKG-I) localized presynaptically in nociceptor terminals plays an essential role in the expression of spinal LTP. Using the Cre-lox P system, we generated nociceptor-specific knockout mice lacking PKG-I specifically in presynaptic terminals of nociceptors in the spinal cord, but not in post-synaptic neurons or elsewhere (SNS-PKG-I(-/-) mice). Patch clamp recordings showed that activity-induced LTP at identified synapses between nociceptors and spinal neurons projecting to the periaqueductal grey (PAG) was completely abolished in SNS-PKG-I(-/-) mice, although basal synaptic transmission was not affected. Analyses of synaptic failure rates and paired-pulse ratios indicated a role for presynaptic PKG-I in regulating the probability of neurotransmitter release. Inositol 1,4,5-triphosphate receptor 1 and myosin light chain kinase were recruited as key phosphorylation targets of presynaptic PKG-I in nociceptive neurons. Finally, behavioural analyses in vivo showed marked defects in SNS-PKG-I(-/-) mice in several models of activity-induced nociceptive hypersensitivity, and pharmacological studies identified a clear contribution of PKG-I expressed in spinal terminals of nociceptors. Our results thus indicate that presynaptic mechanisms involving an increase in release probability from nociceptors are operational in the expression of synaptic LTP on spinal-PAG projection neurons and that PKG-I localized in presynaptic nociceptor terminals plays an essential role in this process to regulate pain sensitivity.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Long-Term Potentiation , Nociceptors/metabolism , Pain/pathology , Aminoquinolines/pharmacology , Animals , Behavior, Animal , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/genetics , Enzyme Activation , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Deletion , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nerve Fibers/metabolism , Nerve Fibers/pathology , Nociceptors/drug effects , Nociceptors/pathology , Pain/metabolism , Patch-Clamp Techniques , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Signal Transduction , Substrate Specificity , Synaptic Transmission
20.
Brain ; 135(Pt 2): 359-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22271663

ABSTRACT

Painful diabetic neuropathy is a common complication of diabetes mellitus and can affect many aspects of life and severely limit patients' daily functions. Signals of painful diabetic neuropathy are believed to originate in the peripheral nervous system. However, its peripheral mechanism of hyperalgesia has remained elusive. Numerous studies have accumulated that polymodal nociceptive C-fibres play a crucial role in the generation and conduction of pain signals and sensitization of which following injury or inflammation leads to marked hyperalgesia. Traditionally, the number of nociceptive primary afferent firings is believed to be determined at the free nerve endings, while the extended main axon of unmyelinated C-fibres only involves the reliable and faithful propagation of firing series to the central terminals. We challenged this classic view by showing that conduction of action potential can fail to occur in response to repetitive activity when they travel down the main axon of polymodal nociceptive C-fibres. Quantitative analysis of conduction failure revealed that the degree of conduction failure displays a frequency-dependent manner. Local administration of low threshold, rapidly activating potassium current blocker, α-dendrotoxin (0.5 nM) and persistent sodium current blocker, low doses of tetrodotoxin (<100 nM) on the main axon of C-fibres can reciprocally regulate the degree of conduction failure, confirming that conduction failure did occur along the main axon of polymodal nociceptive C-fibres. Following streptozotocin-induced diabetes, a subset of polymodal nociceptive C-fibres exhibited high-firing-frequency to suprathreshold mechanical stimulation, which account for about one-third of the whole population of polymodal nociceptive C-fibres tested. These high-firing-frequency polymodal nociceptive C-fibres in rats with diabetes displayed a marked reduction of conduction failure. Delivery of low concentrations of tetrodotoxin and Nav1.8 selective blocker, A-803467 on the main axon of C-fibres was found to markedly enhance the conduction failure in a dose-dependent manner in diabetic rats. Upregulated expression of sodium channel subunits Nav1.7 and Nav1.8 in both small dorsal root ganglion neurons and peripheral C-fibres as well as enhanced transient and persistent sodium current and increased excitability in small dorsal root ganglion neurons from diabetic rats might underlie the reduced conduction failure in the diabetic high-firing-frequency polymodal nociceptive C-fibres. This study shed new light on the functional capability in the pain signals processing for the main axon of polymodal nociceptive C-fibres and revealed a novel mechanism underlying diabetic hyperalgesia.


Subject(s)
Axons/physiology , Diabetic Neuropathies/physiopathology , Hyperalgesia/physiopathology , Neural Conduction/physiology , Nociceptors/physiology , Action Potentials/physiology , Animals , Male , Nerve Fibers/physiology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...