Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 492
1.
J Neurophysiol ; 127(2): 463-473, 2022 02 01.
Article En | MEDLINE | ID: mdl-35020516

Unmyelinated tactile (C-tactile or CT) afferents are abundant in arm hairy skin and have been suggested to signal features of social affective touch. Here, we recorded from unmyelinated low-threshold mechanosensitive afferents in the peroneal and radial nerves. The most distal receptive fields were located on the proximal phalanx of the third finger for the superficial branch of the radial nerve and near the lateral malleolus for the peroneal nerve. We found that the physiological properties with regard to conduction velocity and mechanical threshold, as well as their tuning to brush velocity, were similar in CT units across the antebrachial (n = 27), radial (n = 8), and peroneal (n = 4) nerves. Moreover, we found that although CT afferents are readily found during microneurography of the arm nerves, they appear to be much more sparse in the lower leg compared with C-nociceptors. We continued to explore CT afferents with regard to their chemical sensitivity and found that they could not be activated by topical application to their receptive field of either the cooling agent menthol or the pruritogen histamine. In light of previous studies showing the combined effects that temperature and mechanical stimuli have on these neurons, these findings add to the growing body of research suggesting that CT afferents constitute a unique class of sensory afferents with highly specialized mechanisms for transducing gentle touch.NEW & NOTEWORHY Unmyelinated tactile (CT) afferents are abundant in arm hairy skin and are thought to signal features of social affective touch. We show that CTs are also present but are relatively sparse in the lower leg compared with C-nociceptors. CTs display similar physiological properties across the arm and leg nerves. Furthermore, CT afferents do not respond to the cooling agent menthol or the pruritogen histamine, and their mechanical response properties are not altered by these chemicals.


Affect , Antipruritics/pharmacology , Histamine Agonists/pharmacology , Mechanoreceptors/physiology , Menthol/pharmacology , Nerve Fibers, Unmyelinated/physiology , Peroneal Nerve/physiology , Touch Perception/physiology , Adult , Afferent Pathways/drug effects , Afferent Pathways/physiology , Antipruritics/administration & dosage , Female , Histamine/pharmacology , Histamine Agonists/administration & dosage , Humans , Leg/innervation , Male , Mechanoreceptors/drug effects , Menthol/administration & dosage , Nerve Fibers, Unmyelinated/drug effects , Nociceptors/drug effects , Nociceptors/physiology , Peroneal Nerve/drug effects , Radial Nerve/drug effects , Radial Nerve/physiology , Touch Perception/drug effects , Young Adult
2.
Front Immunol ; 12: 787565, 2021.
Article En | MEDLINE | ID: mdl-34950149

The incidence of chronic pain is especially high in women, but the underlying mechanisms remain poorly understood. Interleukin-23 (IL-23) is a pro-inflammatory cytokine and contributes to inflammatory diseases (e.g., arthritis and psoriasis) through dendritic/T cell signaling. Here we examined the IL-23 involvement in sexual dimorphism of pain, using an optogenetic approach in transgenic mice expressing channelrhodopsin-2 (ChR2) in TRPV1-positive nociceptive neurons. In situ hybridization revealed that compared to males, females had a significantly larger portion of small-sized (100-200 µm2) Trpv1+ neurons in dorsal root ganglion (DRG). Blue light stimulation of a hindpaw of transgenic mice induced intensity-dependent spontaneous pain. At the highest intensity, females showed more intense spontaneous pain than males. Intraplantar injection of IL-23 (100 ng) induced mechanical allodynia in females only but had no effects on paw edema. Furthermore, intraplantar IL-23 only potentiated blue light-induced pain in females, and intrathecal injection of IL-23 also potentiated low-dose capsaicin (500 ng) induced spontaneous pain in females but not males. IL-23 expresses in DRG macrophages of both sexes. Intrathecal injection of IL-23 induced significantly greater p38 phosphorylation (p-p38), a marker of nociceptor activation, in DRGs of female mice than male mice. In THP-1 human macrophages estrogen and chemotherapy co-application increased IL-23 secretion, and furthermore, estrogen and IL-23 co-application, but not estrogen and IL-23 alone, significantly increased IL-17A release. These findings suggest a novel role of IL-23 in macrophage signaling and female-dominant pain, including C-fiber-mediated spontaneous pain. Our study has also provided new insight into cytokine-mediated macrophage-nociceptor interactions, in a sex-dependent manner.


Ganglia, Spinal/drug effects , Interleukin-23/toxicity , Nerve Fibers, Unmyelinated/drug effects , Nociceptors/drug effects , Pain Threshold/drug effects , Pain/chemically induced , TRPV Cation Channels/metabolism , Animals , Capsaicin , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Disease Models, Animal , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Humans , Interleukin-17/metabolism , Light , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Nerve Fibers, Unmyelinated/metabolism , Nociceptors/metabolism , Optogenetics , Pain/genetics , Pain/metabolism , Pain/physiopathology , Sex Characteristics , THP-1 Cells , TRPV Cation Channels/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G149-G156, 2021 08 01.
Article En | MEDLINE | ID: mdl-34160291

Bile acid reflux in the esophagus plays a role in the pathogenesis of certain esophageal disorders, where it can induce esophageal pain and heartburn. The present study aimed to determine whether bile acid, deoxycholic acid (DCA), directly activates and sensitizes esophageal vagal nociceptive afferent C-fiber subtypes. DCA-elicited effects on vagal nodose and jugular neurons were studied by calcium imaging. Its effects on esophageal-labeled nodose and jugular neurons were then determined by patch-clamp recording. At nodose and jugular C-fiber nerve endings in the esophagus, DCA-evoked action potentials (APs) were compared by extracellular single-unit recordings in ex vivo esophageal-vagal preparations. DCA application induced calcium influxes in nodose and jugular neurons and elicited inward currents in esophageal-labeled nodose and jugular neurons. In the presence of DCA, the current densities elicited by capsaicin were enhanced in those labeled neurons. Consistently, DCA perfusion at nerve terminals in the esophagus evoked APs in about 50% of esophageal nodose and jugular C-fibers. In DCA-sensitive C-fibers, DCA perfusion also sensitized the fibers such that the subsequent response to capsaicin was amplified. Collectively, these results provide new evidence that DCA directly activates and sensitizes nociceptive nodose and jugular C-fibers in the esophagus. Such activation and sensitization effects may contribute to bile acid-induced esophageal nociceptive symptoms that are refractory to proton-pump inhibitor therapy.NEW & NOTEWORTHY Bile acid reflux in the esophagus can induce pain and heartburn in certain esophageal disorders, but the underlying neuronal mechanism is still unclear. The present study demonstrated that bile acid, deoxycholic acid (DCA), directly activates esophageal vagal afferent nodose and jugular nociceptive C-fibers and sensitizes their response to capsaicin. Such effects may contribute to bile acid-induced esophageal nociceptive symptoms that refractory to proton-pump inhibitors (PPIs) therapy.


Action Potentials , Cholagogues and Choleretics/pharmacology , Deoxycholic Acid/pharmacology , Esophagus/physiology , Nociceptors/physiology , Animals , Calcium Signaling , Cells, Cultured , Esophagus/innervation , Guinea Pigs , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Nociceptors/drug effects , Nociceptors/metabolism , Vagus Nerve/drug effects , Vagus Nerve/physiology
4.
PLoS One ; 16(2): e0246375, 2021.
Article En | MEDLINE | ID: mdl-33529249

Prostaglandin E2 (PGE2)-induced coughs in vivo and vagal nerve depolarization in vitro are inhibited by systemic and local administration of prostaglandin EP3 receptor (L-798106) and TRPV1 antagonists (JNJ 17203212). These results indicate a modulating effect of TRPV1 on the EP3 receptor-mediated cough responses to PGE2 likely through the vagal sensory nerve. This study aimed to determine whether 1) inhalation of aerosolized JNJ 17203212 and L-798106 affected cough responses to citric acid (CA, mainly stimulating TRPV1) and PGE2; 2) TRPV1 and EP3 receptor morphologically are co-expressed and electrophysiologically functioned in the individual of vagal pulmonary C-neurons (cell bodies of bronchopulmonary C-fibers in the nodose/jugular ganglia); and 3) there was a cross-effect of TRPV1 and EP3 receptor on these neural excitations. To this end, aerosolized CA or PGE2 was inhaled by unanesthetized guinea pigs pretreated without or with each antagonist given in aerosol form. Immunofluorescence was applied to identify the co-expression of TRPV1 and EP3 receptor in vagal pulmonary C-neurons (retrogradely traced by DiI). Whole-cell voltage patch clamp approach was used to detect capsaicin (CAP)- and PGE2-induced currents in individual vagal pulmonary C-neurons and determine the effects of the TRPV1 and EP3 receptor antagonists on the evoked currents. We found that PGE2-induced cough was attenuated by JNJ 17203212 or L-798106 and CA-evoked cough greatly suppressed only by JNJ 17203212. Approximately 1/4 of vagal pulmonary C-neurons co-expressed EP3 with a cell size < 20 µm. Both CAP- and PGE2-induced currents could be recorded in the individuals of some vagal pulmonary C-neurons. The former was largely inhibited only by JNJ 17203212, while the latter was suppressed by JNJ 17203212 or L-798106. The similarity of the cross-effect of both antagonists on cough and vagal pulmonary C-neural activity suggests that a subgroup of vagal pulmonary C-neurons co-expressing TRPV1 and EP3 receptor is, at least in part, responsible for the cough response to PGE2.


Bronchi/metabolism , Cough/metabolism , Nerve Fibers, Unmyelinated/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , TRPV Cation Channels/metabolism , Aminopyridines/pharmacology , Animals , Capsaicin , Citric Acid/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone , Guinea Pigs , Ion Channel Gating/drug effects , Male , Models, Biological , Nerve Fibers, Unmyelinated/drug effects , Nodose Ganglion/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pulmonary Ventilation/drug effects , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , TRPV Cation Channels/antagonists & inhibitors , Vagus Nerve/drug effects , Vagus Nerve/metabolism
5.
Anesthesiology ; 134(1): 88-102, 2021 01 01.
Article En | MEDLINE | ID: mdl-33166389

BACKGROUND: Although the widely used single L-enantiomers of local anesthetics have less toxic effects on the cardiovascular and central nervous systems, the mechanisms mediating their antinociceptive actions are not well understood. The authors hypothesized that significant differences in the ion channel blocking abilities of the enantiomers of bupivacaine would be identified. METHODS: The authors performed electrophysiologic analysis on rat dorsal root ganglion neurons in vitro and on spinal transmissions in vivo. RESULTS: In the dorsal root ganglion, these anesthetics decreased the amplitudes of action potentials. The half-maximum inhibitory concentrations of D-enantiomer D-bupivacaine were almost equal for Aß (29.5 µM), Aδ (29.7µM), and C (29.8 µM) neurons. However, the half-maximum inhibitory concentrations of L-bupivacaine was lower for Aδ (19.35 µM) and C (19.5 µM) neurons than for A ß (79.4 µM) neurons. Moreover, D-bupivacaine almost equally inhibited tetrodotoxin-resistant (mean ± SD: 15.8 ± 10.9% of the control, n = 14, P < 0.001) and tetrodotoxin-sensitive (15.4 ± 15.6% of the control, n = 11, P = 0.004) sodium currents. In contrast, L-bupivacaine suppressed tetrodotoxin-resistant sodium currents (26.1 ± 19.5% of the control, n = 18, P < 0.001) but not tetrodotoxin-sensitive sodium currents (74.5 ± 18.2% of the control, n = 11, P = 0.477). In the spinal dorsal horn, L-bupivacaine decreased the area of pinch-evoked excitatory postsynaptic currents (39.4 ± 11.3% of the control, n = 7, P < 0.001) but not touch-evoked responses (84.2 ± 14.5% of the control, n = 6, P = 0.826). In contrast, D-bupivacaine equally decreased pinch- and touch-evoked responses (38.8 ± 9.5% of the control, n = 6, P = 0.001, 42.9 ± 11.8% of the control, n = 6, P = 0.013, respectively). CONCLUSIONS: These results suggest that the L-enantiomer of bupivacaine (L-bupivacaine) effectively inhibits noxious transmission to the spinal dorsal horn by blocking action potential conduction through C and Aδ afferent fibers.


Anesthetics, Local/pharmacology , Bupivacaine/pharmacology , Neurons/drug effects , Nociception/drug effects , Peripheral Nerves/drug effects , Posterior Horn Cells/drug effects , Synaptic Transmission/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Male , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Unmyelinated/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sodium Channels/drug effects , Stereoisomerism , Tetrodotoxin/pharmacology
6.
Respir Physiol Neurobiol ; 278: 103446, 2020 07.
Article En | MEDLINE | ID: mdl-32360368

Inflammation can increase the excitability of bronchopulmonary C-fibers leading to excessive sensations and reflexes (e.g. wheeze and cough). We have previously shown modulation of peripheral nerve terminal mitochondria by antimycin A causes hyperexcitability in TRPV1-expressing bronchopulmonary C-fibers through the activation of protein kinase C (PKC). Here, we have investigated the PKC isoform responsible for this signaling. We found PKCß1, PKCδ and PKCε were expressed by many vagal neurons, with PKCα and PKCß2 expressed by subsets of vagal neurons. In dissociated vagal neurons, antimycin A caused translocation of PKCα but not the other isoforms, and only in TRPV1-lineage neurons. In bronchopulmonary C-fiber recordings, antimycin A increased the number of action potentials evoked by α,ß-methylene ATP. Selective inhibition of PKCα, PKCß1 and PKCß2 with 50 nM bisindolylmaleimide I prevented the antimycin-induced bronchopulmonary C-fiber hyperexcitability, whereas selective inhibition of only PKCß1 and PKCß2 with 50 nM LY333531 had no effect. We therefore conclude that PKCα is required for antimycin-induced increases in bronchopulmonary C-fiber excitability.


Antimycin A/pharmacology , Bronchi/innervation , Nerve Fibers, Unmyelinated/drug effects , Neurons/drug effects , Nodose Ganglion/drug effects , Protein Kinase C-alpha/drug effects , Vagus Nerve , Animals , Lung/innervation , Mice , Nerve Fibers, Unmyelinated/metabolism , Neurons/metabolism , Nodose Ganglion/cytology , Nodose Ganglion/metabolism , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Protein Kinase C-alpha/metabolism , TRPV Cation Channels/metabolism
7.
Physiol Res ; 69(Suppl 1): S43-S54, 2020 03 27.
Article En | MEDLINE | ID: mdl-32228011

Itch is the most common chief complaint in patients visiting dermatology clinics and is analogous to cough and also sneeze of the lower and upper respiratory tract, all three of which are host actions trying to clear noxious stimuli. The pathomechanisms of these symptoms are not completely determined. The itch can originate from a variety of etiologies. Itch originates following the activation of peripheral sensory nerve endings following damage or exposure to inflammatory mediators. More than one sensory nerve subtype is thought to subservepruriceptive itch which includes both unmyelinated C-fibers and thinly myelinated Adelta nerve fibers. There are a lot of mediators capable of stimulating these afferent nerves leading to itch. Cough and itch pathways are mediated by small-diameter sensory fibers. These cough and itch sensory fibers release neuropeptides upon activation, which leads to inflammation of the nerves. The inflammation is involved in the development of chronic conditions of itch and cough. The aim of this review is to point out the role of sensory nerves in the pathogenesis of cough and itching. The common aspects of itch and cough could lead to new thoughts and perspectives in both fields.


Cough/physiopathology , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Unmyelinated/physiology , Pruritus/physiopathology , Sensory Receptor Cells/physiology , Animals , Capsaicin/adverse effects , Cough/chemically induced , Histamine/adverse effects , Histamine Agonists/adverse effects , Humans , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Unmyelinated/drug effects , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Peripheral Nerves/drug effects , Peripheral Nerves/physiopathology , Pruritus/chemically induced , Sensory Receptor Cells/drug effects , Sensory System Agents/adverse effects
8.
J Vis Exp ; (150)2019 08 27.
Article En | MEDLINE | ID: mdl-31524869

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.


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
9.
Cephalalgia ; 39(11): 1358-1365, 2019 10.
Article En | MEDLINE | ID: mdl-31475573

BACKGROUND: Botulinum neurotoxin type A, an FDA-approved prophylactic drug for chronic migraine, is thought to achieve its therapeutic effect through blocking activation of unmyelinated meningeal nociceptors and their downstream communications with myelinated nociceptors and potentially the vasculature and immune cells. Prior investigations to determine botulinum neurotoxin type A effects on meningeal nociceptors were carried out in male rats and tested with stimuli that act outside the blood brain barrier. Here, we sought to explore the effects of extracranial injections of botulinum neurotoxin type A on activation of meningeal nociceptors by cortical spreading depression, an event which occurs inside the blood brain barrier, in female rats. MATERIAL AND METHODS: Using single-unit recording, we studied myelinated C- and unmyelinated Aδ-meningeal nociceptors' responses to cortical spreading depression 7-14 days after injection of botulinum neurotoxin type A or saline along calvarial sutures. RESULTS: In female rats, responses to cortical spreading depression were typically more prolonged and, in some cases, began at relatively longer latencies post-cortical spreading depression, than had been observed in previous studies in male rats. Extracranial administration of botulinum neurotoxin type A reduced significantly the prolonged firing of the meningeal nociceptors, in the combined sample of Aδ- and C-fiber, but not their response probability. DISCUSSION: The findings suggest that the mechanism of action by which botulinum neurotoxin type A prevents migraine differ from the one by which calcitonin gene-related peptide monoclonal antibodies prevent migraine and that even when the origin of migraine is central (i.e. in the cortex), a peripherally acting drug can intercept/prevent the headache.


Botulinum Toxins, Type A/pharmacology , Cortical Spreading Depression/drug effects , Meninges/drug effects , Neuromuscular Agents/pharmacology , Nociceptors/drug effects , Animals , Female , Nerve Fibers, Unmyelinated/drug effects , Rats , Rats, Sprague-Dawley
10.
Respir Physiol Neurobiol ; 270: 103267, 2019 12.
Article En | MEDLINE | ID: mdl-31398537

Cough in respiratory diseases is attributed to the activation of airway C-fibers by inflammation. Inflammatory mediators can act on multiple receptors expressed in airway C-fibers, nonetheless, the action potential initiation in C-fibers depends on a limited number of voltage-gated sodium channel (NaV1) subtypes. We have recently demonstrated that NaV1.8 substantially contributes to the action potential initiation in the airway C-fiber subtype implicated in cough. We therefore hypothesized that the NaV1.8 blocker A-803467 inhibits cough. We evaluated the cough evoked by the inhalation of C-fiber activator capsaicin in awake guinea pigs. Compared to vehicle, intraperitoneal or inhaled A-803467 caused 30-50% inhibition of cough at the doses that did not alter respiratory rate. We conclude that the NaV1.8 blocker A-803467 inhibits cough in a manner consistent with its action on the C-fiber nerve terminals in the airways. Targeting voltage-gated sodium channels mediating action potential initiation in airway C-fibers may offer a means of cough inhibition that is independent of the stimulus.


Aniline Compounds/therapeutic use , Antitussive Agents/therapeutic use , Cough/drug therapy , Furans/therapeutic use , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/therapeutic use , Action Potentials/drug effects , Administration, Inhalation , Aniline Compounds/adverse effects , Animals , Antitussive Agents/adverse effects , Bronchi/innervation , Dose-Response Relationship, Drug , Furans/adverse effects , Guinea Pigs , Injections, Intraperitoneal , Lung/innervation , Male , Nerve Fibers, Unmyelinated/drug effects , Presynaptic Terminals/drug effects , Sodium Channel Blockers/adverse effects
11.
Eur J Pain ; 23(10): 1801-1813, 2019 11.
Article En | MEDLINE | ID: mdl-31314951

BACKGROUND: Delayed onset muscle soreness (DOMS) is characterized by mechanical hyperalgesia after lengthening contractions (LC). It is relatively common and causes disturbance for many people who require continuous exercise, yet its molecular and peripheral neural mechanisms are poorly understood. METHODS: We examined whether muscular myelinated Aδ-fibres, in addition to unmyelinated C-fibres, are involved in LC-induced mechanical hypersensitivity, and whether acid-sensing ion channel (ASIC)-3 expressed in thin-fibre afferents contributes to this type of pain using a rat model of DOMS. The peripheral contribution of ASIC3 was investigated using single-fibre electrophysiological recordings in extensor digitorum longus muscle-peroneal nerve preparations in vitro. RESULTS: Behavioural tests demonstrated a significant decrease of the muscular mechanical withdrawal threshold following LC to ankle extensor muscles, and it was improved by intramuscular injection of APETx2 (2.2 µM), a selective blocker of ASIC3. The lower concentration of APETx2 (0.22 µM) and its vehicle had no effect on the threshold. Intramuscular injection of APETx2 (2.2 µM) in naïve rats without LC did not affect the withdrawal threshold. In the ankle extensor muscles that underwent LC one day before the electrophysiological recordings, the mechanical response of Aδ- and C-fibres was significantly facilitated (i.e. decreased response threshold and increased magnitude of the response). The facilitated mechanical response of the Aδ- and C-fibres was significantly suppressed by selective blockade of ASIC3 with APETx2, but not by its vehicle. CONCLUSIONS: These results clearly indicate that ASIC3 contributes to the augmented mechanical response of muscle thin-fibre receptors in delayed onset muscular mechanical hypersensitivity after LC. SIGNIFICANCE: Here, we show that not only C- but also Aδ-fibre nociceptors in the muscle are involved in mechanical hypersensitivity after lengthening contractions, and that acid-sensing ion channel (ASIC)-3 expressed in the thin-fibre nociceptors is responsible for the mechanical hypersensitivity. ASIC3 might be a novel pharmacological target for pain after exercise.


Acid Sensing Ion Channels/metabolism , Hyperalgesia/metabolism , Muscle, Skeletal/innervation , Myalgia/metabolism , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/metabolism , Physical Conditioning, Animal , Acid Sensing Ion Channel Blockers/pharmacology , Animals , Injections, Intramuscular , Male , Muscle Contraction , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Unmyelinated/drug effects , Neural Conduction , Nociceptors , Pain Measurement , Peroneal Nerve/drug effects , Peroneal Nerve/metabolism , Rats , Rats, Sprague-Dawley
12.
Acta Derm Venereol ; 99(11): 1009-1015, 2019 Oct 01.
Article En | MEDLINE | ID: mdl-31289838

Central sensitization induces pain augmentation in chronic pain states. An analogous mechanism is speculated for chronic pruritus. This study compared patients with chronic pruritus (n = 79) of different origins (atopic dermatitis, chronic pruritus on non-lesional skin, chronic prurigo) and healthy controls (HC, n = 54) with regard to itch intensity and qualities of sensory symptoms after selective peripheral nerve fibre activation by electrical stimulation at 5 Hz (surrogate for C-fibre function) and 2,000 Hz (surrogate for Aß-fibre function) using a Neurometer®. Electrically-induced itch was more intense in patients with chronic pruritus than in HC, but patients with chronic pruritus did not report "itch" more often than HC at 5 Hz. Stimulation at 2,000 Hz induced more pricking and tingling, but less throbbing in patients with chronic pruritus compared with HC. Treatment with cooling compound reduced clinical and experimental itch, but did not alter the distribution of sensory symptoms. These data show hyperknesis in chronic pruritus of various origins, arguing for common central sensitization mechanisms.


Central Nervous System Sensitization , Nerve Fibers, Myelinated , Nerve Fibers, Unmyelinated , Pruritus/physiopathology , Sensory Thresholds , Skin/innervation , Administration, Cutaneous , Adult , Aged , Antipruritics/administration & dosage , Case-Control Studies , Chronic Disease , Electric Stimulation , Female , Humans , Male , Middle Aged , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Unmyelinated/drug effects , Pruritus/diagnosis , Pruritus/drug therapy , Pruritus/etiology , Risk Factors , Sensory Thresholds/drug effects , Time Factors , Treatment Outcome
13.
FASEB J ; 33(10): 10731-10741, 2019 10.
Article En | MEDLINE | ID: mdl-31251077

Prenatal nicotinic exposure (PNE) reportedly sensitizes bronchopulmonary C-fibers (PCFs) and prolongs PCF-mediated apnea in rat pups, contributing to the pathogenesis of sudden infant death syndrome. Serotonin, or 5-hydroxytryptamine (5-HT), induces apnea via acting on 5-HT receptor 3 (5-HT3R) in PCFs, and among the 5-HT3R subunits, 5-HT3B is responsible for shortening the decay time of 5-HT3R-mediated currents. We examined whether PNE would promote pulmonary 5-HT secretion and prolong the apnea mediated by 5-HT3Rs in PCFs via affecting the 5-HT3B subunit. To this end, the following variables were compared between the control and PNE rat pups: 1) the 5-HT content in bronchoalveolar lavage fluid, 2) the apneic response to the right atrial bolus injection of phenylbiguanide (a 5-HT3R agonist) before and after PCF inactivation, 3) 5-HT3R currents and the stimulus threshold of the action currents of vagal pulmonary C-neurons, and 4) the immunoreactivity (IR) and mRNA expression of 5-HT3A and 5-HT3B in these neurons. Our results showed that PNE up-regulated the pulmonary 5-HT concentration and strengthened the PCF 5-HT3R-mediated apnea. PNE significantly facilitated neural excitability by shortening the decay time of 5-HT3R currents, lowering the stimulus threshold, and increasing 5-HT3B IR. In summary, PNE prolongs the apnea mediated by 5-HT3Rs in PCFs, likely by increasing 5-HT3B subunits to enhance the excitability of 5-HT3 channels.-Zhao, L., Gao, X., Zhuang, J., Wallen, M., Leng, S., Xu, F. Prolongation of bronchopulmonary C-fiber-mediated apnea by prenatal nicotinic exposure in rat pups: role of 5-HT3 receptors.


Apnea/etiology , Apnea/physiopathology , Lung/drug effects , Lung/innervation , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Nicotine/toxicity , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/physiopathology , Receptors, Serotonin, 5-HT3/physiology , Animals , Animals, Newborn , Apnea/genetics , Biguanides/administration & dosage , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Female , Humans , Infant, Newborn , Lung/physiopathology , Male , Nicotine/administration & dosage , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT3/genetics , Serotonin/metabolism , Serotonin 5-HT3 Receptor Agonists/administration & dosage , Sudden Infant Death/etiology
14.
Neuroscience ; 410: 55-58, 2019 07 01.
Article En | MEDLINE | ID: mdl-31047975

Acute cutaneous exposure to allergen often leads to itch, but seldom pain. The effect of mast cell activation on cutaneous C-fibers was studied using innervated isolated mouse skin preparation that allows for intra-arterial delivery of chemicals to the nerve terminals in the skin. Allergen (ovalbumin) injection into the isolated skin of actively sensitized mice strongly stimulated chloroquine (CQ)-sensitive C-fibers (also referred to as "itch" nerves); on the other hand, CQ-insensitive C-fibers were activated only modestly, if at all. The histamine H1 receptor antagonist pyrilamine abolished itch C-fibers response to histamine, but failed to significantly reduce the response to ovalbumin. Ovalbumin also strongly activated itch C-fibers in skin isolated from Mrgpr-cluster Δ-/- mice. When pyrilamine was studied in the Mrgpr-cluster Δ-/- mice thereby eliminating the influence of both histamine H1 and Mrgpr receptors (MrgprA3 and C11 are selectively expressed by itch nerves), the ovalbumin response was very nearly eliminated. The data indicate that the acute activation of itch C-fibers in mouse skin is largely secondary to the combined effect of activation of histamine H1 and Mrpgr receptors.


Allergens/toxicity , Histamine/metabolism , Nerve Endings/metabolism , Nerve Fibers, Unmyelinated/metabolism , Pruritus/metabolism , Skin/metabolism , Animals , Histamine H1 Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Endings/drug effects , Nerve Fibers, Unmyelinated/drug effects , Organ Culture Techniques , Pruritus/chemically induced , Skin/drug effects , Skin/innervation
15.
Muscle Nerve ; 60(4): 367-375, 2019 10.
Article En | MEDLINE | ID: mdl-31107560

INTRODUCTION: Topical application of lidocaine-and-prilocaine (LP) cream attenuates the functionality of small cutaneous nerve fibers. The aim of this human study was to measure the underlying excitability modulation of small cutaneous nerve fibers using a novel and fast perception threshold tracking (PTT) technique. METHODS: Small sensory fibers were selectively blocked by 120-minute topical application of LP and confirmed by quantitative sensory testing. Excitability changes of small (activated by a specially designed pin electrode) and large (patch electrode) nerve fibers were assessed as the strength-duration relation and threshold electrotonus. RESULTS: The excitability assessed by the strength-duration relation and threshold electrotonus was significantly modulated for the small afferents (P < 0.05, Wilcoxon's test) but not the large afferents. DISCUSSION: This novel PTT technique was able to assess inhibition of membrane properties of small cutaneous fibers, suggesting the usefulness of the technique as a diagnostic method for assessing impairment of small fibers, as seen in many types of polyneuropathies.


Anesthetics, Local/pharmacology , Lidocaine, Prilocaine Drug Combination/pharmacology , Nerve Fibers, Myelinated/drug effects , Sensory Thresholds/drug effects , Small Fiber Neuropathy/diagnosis , Administration, Cutaneous , Adult , Cross-Over Studies , Double-Blind Method , Electric Stimulation , Electrodiagnosis , Female , Healthy Volunteers , Humans , Male , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Sensory Thresholds/physiology , Young Adult
16.
CNS Drugs ; 33(6): 525-537, 2019 06.
Article En | MEDLINE | ID: mdl-30989485

Migraine is a strongly disabling disease characterized by a unilateral throbbing headache lasting for up to 72 h for each individual attack. There have been many theories on the pathophysiology of migraine throughout the years. Currently, the neurovascular theory dominates, suggesting clear involvement of the trigeminovascular system. The most recent data show that a migraine attack most likely originates in the hypothalamus and activates the trigeminal nucleus caudalis (TNC). Although the mechanisms are unknown, activation of the TNC leads to peripheral release of calcitonin gene-related protein (CGRP), most likely from C-fibers. During the past year monoclonal antibodies against CGRP or the CGRP receptor have emerged as the most promising targets for migraine therapy, and at the same time established the strong involvement of CGRP in the pathophysiology of migraine. The viewpoint presented here focuses further on the activation of the CGRP receptor on the sensory Aδ-fiber, leading to the sensation of pain. The CGRP receptor activates adenylate cyclase, which leads to an increase in cyclic adenosine monophosphate (cAMP). We hypothesize that cAMP activates the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, triggering an action potential sensed as pain. The mechanisms behind migraine pain on a molecular level, particularly their importance to cAMP, provide clues to potential new anti-migraine targets. In this article we focus on the development of targets related to the CGRP system, and further include novel targets such as the pituitary adenylate cyclase-activating peptide (PACAP) system, the serotonin 5-HT1F receptor, purinergic receptors, HCN channels, adenosine triphosphate-sensitive potassium channels (KATP), and the glutaminergic system.


Calcitonin Gene-Related Peptide/metabolism , Drug Development/methods , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Cyclic AMP/metabolism , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Molecular Targeted Therapy , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/metabolism , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism , Trigeminal Nuclei/drug effects , Trigeminal Nuclei/metabolism
17.
Neurogastroenterol Motil ; 31(6): e13585, 2019 06.
Article En | MEDLINE | ID: mdl-30947399

BACKGROUND: Ginger has been used as an herbal medicine worldwide to relieve nausea/vomiting and gastrointestinal discomfort, but the cellular and molecular mechanisms of its neuronal action remain unclear. The present study aimed to determine the effects of ginger constituent 6-shogaol on gastroesophageal vagal nodose C-fibers. METHODS: Extracellular single-unit recording and two-photon nodose neuron imaging were performed, respectively, in ex vivo gastroesophageal-vagal preparations from wild type and Pirt-GCaMP6 transgenic mice. The action potential discharge or calcium influx evoked by mechanical distension and chemical perfusions applied to the gastroesophageal vagal afferent nerve endings were recorded, respectively, at their intact neuronal cell soma in vagal nodose ganglia. The effects of 6-shogaol on nodose C-fiber neurons were then compared and determined. KEY RESULTS: Gastroesophageal application of 6-shogaol-elicited intensive calcium influxes in nodose neurons and evoked robust action potential discharges in most studied nodose C-fibers. Such activation effects were followed by a desensitized response to the second application of 6-shogaol. However, action potential discharges evoked by esophageal mechanical distension, after 6-shogaol perfusion, did not significantly change. Pretreatment with TRPA1 selective blocker HC-030031 inhibited 6-shogaol-induced action potential discharges in gastric and esophageal nodose C-fiber neurons, suggesting that TRPA1 played a role in mediating 6-shogaol-induced activation response. CONCLUSION AND INFERENCES: This study provides evidence that ginger constituent 6-shogaol directly activates vagal afferent C-fiber peripheral gastrointestinal endings. This activation leads to desensitization to subsequent application of 6-shogaol but not subsequent esophageal mechanical distension. Further investigation is required to establish a possible contribution in its anti-emetic effects.


Catechols/pharmacology , Nerve Fibers, Unmyelinated/drug effects , Neurons, Afferent/drug effects , Nodose Ganglion/drug effects , Action Potentials/drug effects , Animals , Esophagus/drug effects , Esophagus/innervation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stomach/drug effects , Stomach/innervation
18.
Pulm Pharmacol Ther ; 56: 15-19, 2019 06.
Article En | MEDLINE | ID: mdl-30872160

Activation of vagal C-fibers is likely involved in some types of pathological coughing, especially coughing that is associated with airway inflammation. This is because stimulation of vagal C-fibers leads to strong urge to cough sensations, and because C-fiber terminals can be strongly activated by mediators associated with airway inflammation. The most direct manner in which a given mediator can activate a C-fiber terminal is through interacting with its receptor expressed in the terminal membrane. The agonist-receptor interaction then must lead to the opening (or potentially closing) of ion channels that lead to a membrane depolarization. This depolarization is referred to as a generator potential. If, and only if, the generator potential reaches the voltage necessary to activate voltage-gated sodium channels, action potentials are initiated and conducted to the central terminals within the CNS. Therefore, there are three target areas to block the inflammatory mediator induced activation of C-fiber terminals. First, at the level of the mediator-receptor interaction, secondly at the level of the generator potential, and third at the level of the voltage-gated sodium channels. Here we provide a brief overview of each of these therapeutic strategies.


Antitussive Agents/pharmacology , Cough/drug therapy , Nerve Fibers, Unmyelinated/drug effects , Action Potentials/drug effects , Animals , Cough/physiopathology , Humans , Nerve Fibers, Unmyelinated/metabolism , Vagus Nerve/metabolism , Voltage-Gated Sodium Channels/drug effects , Voltage-Gated Sodium Channels/metabolism
19.
Eur J Pharmacol ; 849: 154-159, 2019 Apr 15.
Article En | MEDLINE | ID: mdl-30716310

Previously, we showed that the synthetic nitroderivative trans-4-methyl-ß-nitrostyrene (T4MeN) induced vasorelaxant effects in rat isolated aortic rings. Here, we investigated the mechanisms underlying the cardiovascular effects of T4MeN in normotensive rats. In pentobarbital-anesthetized rats, intravenous (i.v.) injection of T4MeN (0.03-0.5 mg/kg) induced a rapid (onset time of 1-2 s) and dose-dependent bradycardia and hypotension. These cardiovascular responses to T4MeN were abolished by bilateral cervical vagotomy or selective blockade of neural conduction of vagal C-fiber afferents by perineural treatment of both cervical vagus nerves with capsaicin. Hypotension and bradycardia were also recorded when T4MeN was directly injected in the right, but not into the left ventricle. Furthermore, they were significantly reduced by i.v. pretreatment with capsazepine but remained unaltered by ondansetron or suramin. In conscious rats, the dose-dependent hypotension and bradycardia evoked by T4MeN were abolished by i.v. methylatropine pretreatment. In conclusion, bradycardiac and depressor responses induced by T4MeN has a vago-vagal reflex origin resulting from the vagal pulmonary afferents stimulation. The transduction mechanism seems to involve the activation of vanilloid TRPV1, but not purinergic (P2X) or 5-HT3 receptors located on vagal pulmonary sensory nerves.


Bradycardia/chemically induced , Lung/innervation , Nerve Fibers, Unmyelinated/drug effects , Reflex/drug effects , Styrenes/pharmacology , TRPV Cation Channels/metabolism , Vagus Nerve/drug effects , Animals , Bradycardia/metabolism , Bradycardia/physiopathology , Male , Nerve Fibers, Unmyelinated/metabolism , Nerve Fibers, Unmyelinated/physiology , Rats , Rats, Wistar
20.
Neurogastroenterol Motil ; 31(4): e13543, 2019 04.
Article En | MEDLINE | ID: mdl-30663188

INTRODUCTION: Acid reflux in the esophagus can induce painful sensations such as heartburn and non-cardiac chest pain. These nociceptive symptoms are initiated by activation of TRPV1-positive afferent C fibers in the esophagus. The present study aimed to explore a novel C fiber inhibition approach. We hypothesized that activation of TRPV1 by acid enabled QX-314, a membrane impermeable sodium channel blocker, to inhibit acid-induced activation of esophageal nociceptive C fiber neurons. METHOD: We determined the inhibitory effect of QX-314 in the presence of acid in guinea pig esophageal nociceptive vagal jugular C fiber neurons by both patch clamp recording in neuron soma and by extra-cellular recording at nerve terminals. KEY RESULTS: Our data demonstrated QX-314 alone did not inhibit sodium currents. However, when applied along with capsaicin to activate TRPV1, QX-314 was able to block sodium currents in esophageal-specific jugular C fiber neurons. We then showed that in the presence of acid, QX-314 significantly blocked acid-evoked activation of jugular C fiber neurons. This effect was attenuated by TRPV1 antagonist AMG9810, suggesting acid-mediated inhibitory effect of QX-314 was TRPV1-dependent. Finally, we provided evidence at nerve endings that acid-evoked action potential discharges in esophageal jugular C fibers were inhibited by QX-314 when applied in the presence of acid. CONCLUSION AND INFERENCES: Our data demonstrated that activation of TRPV1 by acid enabled membrane impermeable sodium channel blocker QX-314 to inhibit acid-induced activation in esophageal nociceptive C fibers. This supports a localized application of QX-314 in the esophagus to block esophageal nociception in acid reflux disorders.


Action Potentials/drug effects , Esophagus/innervation , Lidocaine/analogs & derivatives , Nerve Fibers, Unmyelinated/drug effects , Nociception/drug effects , Acrylamides/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Capsaicin/pharmacology , Esophagus/drug effects , Guinea Pigs , Hydrochloric Acid/pharmacology , Lidocaine/pharmacology , Patch-Clamp Techniques , TRPV Cation Channels/antagonists & inhibitors
...