Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Life Sci ; 264: 118682, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33127519

ABSTRACT

AIMS: Menthacarin is a herbal combination that is clinically used for the treatment of functional gastrointestinal disorders (FGIDs). In several clinical studies, Menthacarin reduced visceral hypersensitivity-related symptoms. Pathogenesis of visceral hypersensitivity is multifactorial. This involves several cell types and different transient receptor potential ion channels (TRPs); these ion channels are highly conductive for calcium ions. Since transient changes in cytosolic calcium levels are crucial for many functions of living cells, we investigated if Menthacarin can induce calcium influx in sensory, largely nociceptive, neurons from dorsal root ganglia (DRG), peritoneal macrophages (PMs) and colonic organoids. MAIN METHODS: We employed the calcium imaging technique on sensory neurons from DRG, PMs and colonic organoids isolated from mice. All cells were superfused by Menthacarin at several concentrations (600, 1200, 1800 µg/ml) during the experiments, followed by calcium ionophor ionomycin (Iono., 1 µM) as a positive control. KEY FINDINGS: Menthacarin induced concentration-dependent calcium ion influx in all investigated cell types. Furthermore, repeated applications of Menthacarin induced tachyphylaxis (desensitisation) of calcium responses in sensory neurons and colonic organoids. SIGNIFICANCE: Menthacarin-induced calcium influx into sensory neurons, macrophages and colonic organoids is probably related to its clinical desensitising effects in patients with FGIDs.


Subject(s)
Calcium Channels/metabolism , Colon/metabolism , Macrophages/metabolism , Organoids/metabolism , Plant Preparations/pharmacology , Sensory Receptor Cells/metabolism , Animals , Colon/cytology , Colon/drug effects , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Oils, Volatile/chemistry , Oils, Volatile/pharmacology , Organ Culture Techniques , Organoids/drug effects , Plant Preparations/chemistry , Sensory Receptor Cells/drug effects
2.
Life Sci ; 257: 118112, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32682914

ABSTRACT

AIMS: STW 5 is an herbal drug combination used for the treatment of functional gastrointestinal disorders (FGIDs) with visceral hypersensitivity as the therapy-resistant hallmark. STW 5 has been clinically proven to alleviate visceral hypersensitivity-related symptoms, including abdominal pain, bloating, nausea, and early satiety. However, the molecular and cellular mechanisms underlying the antinociceptive action of STW 5 remain unknown. Here, we investigate the role of STW 5 in the calcium mobilisation of dorsal root ganglion (DRG) sensory neurons. MAIN METHODS: Calcium imaging experiments were performed with freshly dissociated cultured murine DRG neurons isolated from mice by microfluorometry. TRPA1-deficient DRGs, TRPV1-deficient DRGs, TRPA1/V1 double-deficient DRGs, and wild-type DRGs have been used to investigate the role of TRPs ion channels in mediating STW 5 action. KEY FINDINGS: STW 5 (1.74 and 5.8 mg/ml) induced calcium ion influx into DRG neurons in a concentration-dependent manner. Calcium transients were desensitised during repeated exposure to STW 5, an effect that was facilitated in TRPA1-deficient DRGs and less pronounced in TRPV1-deficient DRGs compared to wild-type (WT) DRGs. SIGNIFICANCE: Repeated exposure to STW 5 induced desensitisation of sensory neurons and may ultimately contribute to its proven clinical efficacy against sensory-related symptoms in patients with FGID, including abdominal pain, bloating, nausea, and early satiety. This effect is modulated by the two prominent irritant sensors in nociceptors, TRPA1 and TRPV1.


Subject(s)
Ganglia, Spinal/drug effects , Plant Extracts/pharmacology , Transient Receptor Potential Channels/drug effects , Animals , Calcium/metabolism , Mice , Mice, Inbred C57BL , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
3.
Sci Rep ; 7(1): 543, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28373680

ABSTRACT

The voltage-gated sodium channel subtype NaV1.8 is expressed in the peripheral nervous system in primary afferent nociceptive C-fibers and is essential for noxious cold signaling. We utilized functional magnetic resonance imaging on NaV1.8-deficient (NaV1.8-/-) compared with wildtype (WT) mice to identify brain structures decoding noxious cold and/or heat signals. In NaV1.8-/- mice functional activity patterns, activated volumes and BOLD signal amplitudes are significantly reduced upon noxious cold stimulation whereas differences of noxious heat processing are less pronounced. Graph-theoretical analysis of the functional connectivity also shows dramatic alterations in noxious cold sensation in NaV1.8-/- mice and clearly reduced interactions between certain brain structures. In contrast, upon heat stimulation qualitatively quite the same functional connectivity pattern and consequently less prominent connectivity differences were observed between NaV1.8-/- and WT mice. Thus, the fact that NaV1.8-/- mice do not perceive nociceptive aspects of strong cooling in contrast to their WT littermates seems not only to be a pure peripheral phenomenon with diminished peripheral transmission, but also consists of upstream effects leading to altered subsequent nociceptive processing in the central nervous system and consequently altered connectivity between pain-relevant brain structures.


Subject(s)
Cold Temperature , Ion Channel Gating , Magnetic Resonance Imaging , Molecular Imaging , NAV1.8 Voltage-Gated Sodium Channel/deficiency , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Signal Transduction , Analysis of Variance , Animals , Brain/physiopathology , Computational Biology , Image Processing, Computer-Assisted , Male , Mice , Mice, Knockout , Pain/physiopathology , Physical Stimulation
4.
Mucosal Immunol ; 9(6): 1500-1513, 2016 11.
Article in English | MEDLINE | ID: mdl-26982596

ABSTRACT

The transient receptor potential (TRP) ion channel family is well characterized in sensory neurons; however, little is known about its role in the immune system. Here we show that the cold-sensing TRPM8 has an unexpected role in innate immunity. TRPM8 expression and function in macrophages were demonstrated in vitro using molecular techniques and calcium imaging. In addition, adoptive macrophage transfer and systemic interleukin (IL)-10 overexpression were performed in experimental colitis. TRPM8 activation induced calcium-transients in murine peritoneal macrophages (PM) and bone marrow-derived macrophages of wild-type (WT) but not TRPM8-deficient mice. TRPM8-deficient PM exhibited defective phagocytosis and increased motility compared with those in WT, whereas the opposite effects of TRPM8 activation were induced in WT PM. TRPM8 activation or blockage/genetic deletion induced a anti- or pro-inflammatory macrophage cytokine profile, respectively. WT mice treated with repeated menthol (TRPM8 agonist) enemas were consistently protected from experimental colitis, whereas TRPM8-deficient mice showed increased colitis susceptibility. Adoptive transfer of TRPM8-deficient macrophages aggravated colitis, whereas systemic IL-10 overexpression rescued this phenotype. TRPM8 activation in peptidergic sensory neurons did not affect neuropeptide release from the inflamed colon. TRPM8 in macrophages determines pro- or anti-inflammatory actions by regulating tumor necrosis factor-α and interleukin-10 production. These findings suggest novel TRPM8-based options for immunomodulatory intervention.


Subject(s)
Colitis/metabolism , Interleukin-10/biosynthesis , Macrophages/metabolism , TRPM Cation Channels/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Movement , Colitis/genetics , Colitis/immunology , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Gene Expression , Macrophages/immunology , Mice , Mice, Knockout , Phagocytes/immunology , Phagocytes/metabolism , TRPM Cation Channels/genetics
5.
Eur J Pain ; 20(8): 1326-34, 2016 09.
Article in English | MEDLINE | ID: mdl-26914965

ABSTRACT

BACKGROUND: Injection of the noxious peptide Bv8 has previously been shown to induce a biphasic thermal hyperalgesia in rodents, the first peak presumably due to peripheral sensitization. This hypothesis has never been directly confirmed. We have assessed whether Bv8 can indeed sensitize peripheral nerve fibres in the mouse to heat. METHODS: We used recordings from single cutaneous fibres, cutaneous calcitonin gene-related peptide (CGRP) release and immunostaining in nerves and plantar skin to evaluate the Bv8 effects on cutaneous nerves. RESULTS: Application of Bv8 at nanomolar concentrations (30-310 nmol/L) to skin preparations significantly increased the heat-induced discharge, the heat-induced afterdischarge and reduced threshold temperature of single unmyelinated polymodal fibres. Furthermore, application of Bv8 to hind-paw skin flaps or trigeminal ganglia significantly elevated their heat-induced CGRP release. Capsaicin-induced and to a lesser extent also KCl-induced CGRP releases were also augmented after Bv8 treatment. Immunohistochemistry revealed co-localization of prokineticin 2 (Bv8 ortholog in rodents) and CGRP in both plantar skin and nerve tissues. These results confirm that Bv8 sensitizes cutaneous nerve endings to heat, partly, although not exclusively through TRPV1 activation. CONCLUSION: Our results thus support the hypothesis that the first hyperalgesic phase to follow Bv8 injection to hind paws of intact animals is due to peripheral sensitization of nociceptors. WHAT DOES THIS STUDY ADD?: Our data provide mechanistic insights into the effect Bv8 application exerts on afferent nerve endings and into the concomitant development of thermal hyperalgesia.


Subject(s)
Gastrointestinal Hormones , Hyperalgesia/etiology , Nerve Fibers/drug effects , Neuropeptides , Peripheral Nerves/drug effects , Skin/innervation , Animals , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Disease Models, Animal , Female , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Nociceptors/physiology
6.
Neurosci Lett ; 592: 12-6, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25731910

ABSTRACT

We assess in mice, the electrophysiological criteria developed in humans and rats in vivo for unmyelinated (C) fibre differentiation into sub-classes, derived from the activity-induced latency increase ("slowing") in response to electrical stimulation during 6 min at 0.25 Hz followed by 3 min at 2 Hz. Fibres are considered nociceptors if they show more than 10% slowing at 2 Hz; nociceptors are further divided into mechanosensitive ("polymodal") and mechanoinsensitive ("silent") ones according to a latency shift of less and more than 1% during the first minute at 0.25 Hz, respectively. Sympathetic postganglionics are recognised by 2-10% slowing at 2 Hz; units slowing less than 2% at 2 Hz remain uncategorised. For assessment of these criteria, we also developed a novel in vivo technique for recording of peripheral single-fibres in the mouse. We compared the theoretical slowing-rate discriminator criteria with experimental data obtained from mice in vivo/in vitro and rats in vitro. Out of 69 cutaneous mouse C-fibres in vitro and 19 in vivo, only 38 (67%) and 9 (47%) met the above 1% criterion, respectively; sympathetics were not identified. In contrast, out of 20 rats nerve fibres in vitro, 19 (95%) met this criterion. We conclude that (A) our novel electrophysiological technique is a practical method for examining mouse cutaneous single-fibres in vivo and (B) the published criterion for identifying silent nociceptors in rats and humans is not applicable in mice.


Subject(s)
Femoral Nerve/physiology , Neural Conduction , Animals , Electric Stimulation , Humans , Male , Mice, Inbred C57BL , Nerve Fibers, Unmyelinated/classification , Nerve Fibers, Unmyelinated/physiology , Rats, Wistar , Species Specificity
7.
Exp Clin Endocrinol Diabetes ; 122(4): 201-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24623503

ABSTRACT

The clinical symptoms of diabetic neuropathy (DN) manifest in a time dependent manner as a positive symptoms (i. e. pain, hypersensitivity, tingling, cramps, cold feet etc.) during its early stages and by a loss of function (i. e. loss of sensory perception, delayed wound healing etc.) predominating in the later stages. Elevated blood glucose alone cannot explain the development and progression of DN and the lowering of blood glucose is insufficient in preventing and/or reversing neuropathy in patients with type 2 diabetes. Recently it has been shown that the endogenous reactive metabolite methylglyoxal (MG) can contribute to the gain of function via post-translational modification in DN of neuronal ion channels involved in chemosensing and action potential generation in nociceptive nerve endings. Dicarbonyls, such as MG, that are elevated in diabetic patients, modify DNA as well as extra- and intracellular proteins, leading to the formation of advanced glycation endproducts (AGEs). Increased formation of AGEs leads to increased cellular stress, dysfunction and ultimately cell death. The interaction of AGE-modified proteins through cell surface receptors, such as RAGE, can lead to increased cellular activation and sustained inflammatory responses, which are the molecular hallmarks of the later, degenerative, stages of DN. The direct and indirect effects of dicarbonyls on nerves or neuronal microvascular network provides a unifying mechanism for the development and progression of DN. Targeting the accumulation of MG and/or prevention of RAGE interactions may therefore provide new, more effective, therapeutic approaches for the treatment of DN.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/therapy , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/therapy , Diabetic Neuropathies/blood , Glycation End Products, Advanced/blood , Glycation End Products, Advanced/metabolism , Glyoxal/blood , Glyoxal/metabolism , Humans , Receptor for Advanced Glycation End Products , Receptors, Immunologic/blood , Receptors, Immunologic/metabolism
8.
Br J Pharmacol ; 171(2): 438-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117225

ABSTRACT

BACKGROUND AND PURPOSE: Selective nociceptor fibre block is achieved by introducing the cell membrane impermeant sodium channel blocker lidocaine N-ethyl bromide (QX-314) through transient receptor potential V1 (TRPV1) channels into nociceptors. We screened local anaesthetics for their capacity to activate TRP channels, and characterized the nerve block obtained by combination with QX-314. EXPERIMENTAL APPROACH: We investigated TRP channel activation in dorsal root ganglion (DRG) neurons by calcium imaging and patch-clamp recordings, and cellular QX-314 uptake by MS. To characterize nerve block, compound action potential (CAP) recordings from isolated nerves and behavioural responses were analysed. KEY RESULTS: Of the 12 compounds tested, bupivacaine was the most potent activator of ruthenium red-sensitive calcium entry in DRG neurons and activated heterologously expressed TRPA1 channels. QX-314 permeated through TRPA1 channels and accumulated intracellularly after activation of these channels. Upon sciatic injections, QX-314 markedly prolonged bupivacaine's nociceptive block and also extended (to a lesser degree) its motor block. Bupivacaine's blockade of C-, but not A-fibre, CAPs in sciatic nerves was extended by co-application of QX-314. Surprisingly, however, this action was the same in wild-type, TRPA1-knockout and TRPV1/TRPA1-double knockout mice, suggesting a TRP-channel independent entry pathway. Consistent with this, high doses of bupivacaine promoted a non-selective, cellular uptake of QX-314. CONCLUSIONS AND IMPLICATIONS: Bupivacaine, combined with QX-314, produced a long-lasting sensory nerve block. This did not require QX-314 permeation through TRPA1, although bupivacaine activated these channels. Regardless of entry pathway, the greatly extended duration of block produced by QX-314 and bupivacaine may be clinically useful.


Subject(s)
Anesthetics, Local/pharmacology , Bupivacaine/pharmacology , Lidocaine/analogs & derivatives , Nerve Block , Sodium Channel Blockers/metabolism , Anesthetics, Local/administration & dosage , Animals , Behavior, Animal/drug effects , Bupivacaine/administration & dosage , Calcium/metabolism , Cell Line , Foot Injuries , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Injections , Lidocaine/metabolism , Male , Mice, Knockout , Patch-Clamp Techniques , Peripheral Nerves/drug effects , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , TRPA1 Cation Channel , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
9.
Eur J Pain ; 17(10): 1472-82, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23720338

ABSTRACT

BACKGROUND: Two transient receptor potential (TRP) channels, TRPV1 and TRPA1, have been physiologically studied with regard to noxious heat transduction. Evidence argues against these channels as sole transducers of noxious heat or cold, respectively. Moreover, in submammalian species the TRPA1 orthologue shows heat sensitivity. METHODS: In vitro, single-fibre and compound action potential recordings from C-fibres as well as measurements of stimulated cutaneous CGRP release are combined with behavioural experiments to assess heat responsiveness in wild type mice, TRPA1 and TRPV1 as well as double-null mutants. RESULTS: Heat thresholds of cutaneous C-mechano-heat sensitive fibres were significantly higher in TRPA1-/- (43 °C) than +/+ (40 °C) mice, and averaged heat responses were clearly weaker, whereas TRPV1-/- showed normal heat thresholds and responses (up to 46 °C). Compound action potential recordings revealed much less activity-dependent slowing of conduction velocity upon noxious heat stimulation in TRPA1-/- and a delayed deficit in TRPV1-/- in comparison to controls. Heat-induced calcitonin gene-related peptide release was reduced in TRPV1-/- but not TRPA1-/- animals. Paw withdrawal latencies to radiant heat were significantly elevated in TRPA1-/-, more so in TRPV1-/- animals. In general, double-null mutants were similar to TRPV1-/- except for the single-fibre heat responses which appeared as weak as in TRPA1-/-. CONCLUSIONS: Our results indicate that in addition to TRPV1, TRPA1 plays a role in heat nociception, in particular in definition of the heat threshold, and might therefore serve as a therapeutic target in acute inflammatory pain.


Subject(s)
Nociception/physiology , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Female , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Fibers, Unmyelinated/metabolism , Pain/genetics , Pain/metabolism , TRPA1 Cation Channel
10.
Neuropeptides ; 45(6): 391-400, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21868092

ABSTRACT

Vagal sensory afferents innervating airways and abdominal tissues express TRPV1 and TRPA1, two depolarizing calcium permeable ion channels playing a major role in sensing environmental irritants and endogenous metabolites which cause neuropeptide release and neurogenic inflammation. Here we have studied axonal chemosensitivity and control of neuropeptide release from the isolated rat and mouse vagus nerve by using prototypical agonists of these transduction channels - capsaicin, mustard oil and the specific endogenous activators, anandamide (methyl arachidonyl ethanolamide, mAEA), and acrolein, respectively. Capsaicin evoked iCGRP release from the rat vagus nerve with an EC50 of 0.12 µM. Co-application of mAEA had a dual effect: nanomolar concentrations of mAEA (0.01 µM) significantly reduced capsaicin-evoked iCGRP release while concentrations ≥ 1 µM mAEA had sensitizing effects. Only 100 µM mAEA directly augmented iCGRP release by itself. In the mouse, 310 µM mAEA increased release in wildtype and TRPA1-/- mice which could be inhibited by capsazepine (10 µM) and was completely absent in TRPV1-/- mice. CB1-/- and CB1/CB2 double -/- mice equally displayed increased sensitivity to mAEA (100 µM) and a sensitizing effect to capsaicin, in contrast to wildtypes. Acrolein and mustard oil (MO)--at µM concentrations--induced a TRPA1-dependent iCGRP release; however, millimolar concentrations of mustard oil (>1mM) evoked iCGRP release by activating TRPV1, confirming recent evidence for TRPV1 agonism of high mustard oil concentrations. Taken together, we present evidence for functional expression of excitatory TRPV1, TRPA1, and inhibitory CB1 receptors along the sensory fibers of the vagus nerve which lend pathophysiological relevance to the axonal membrane and the control of neuropeptide release that may become important in cases of inflammation or neuropathy. Sensitization and possible ectopic discharge may contribute to the development of autonomic dysregulation in visceral tissues that are innervated by the vagus nerve.


Subject(s)
Axons/metabolism , Neuropeptides/metabolism , Receptor, Cannabinoid, CB1/metabolism , TRPC Cation Channels/metabolism , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Vagus Nerve/metabolism , Animals , Arachidonic Acids/pharmacology , Axons/drug effects , Calcitonin Gene-Related Peptide/metabolism , Cannabinoid Receptor Modulators/pharmacology , Capsaicin/pharmacology , Endocannabinoids , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mustard Plant , Plant Oils/pharmacology , Polyunsaturated Alkamides/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Sensory System Agents/pharmacology , TRPA1 Cation Channel , TRPC Cation Channels/genetics , TRPV Cation Channels/genetics , Transient Receptor Potential Channels/genetics , Vagus Nerve/cytology , Vagus Nerve/drug effects
11.
Pain ; 151(2): 496-505, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20832942

ABSTRACT

Painful neuropathy is a common complication of diabetes. Particularly in the early stage of diabetic neuropathy, patients are characterized by burning feet, hyperalgesia to heat, and mechanical stimuli, as if residual nociceptors were sensitized. Such symptoms are barely explained by common pathophysiological concepts of diabetic neuropathy. Diabetes was induced in Wistar rats by streptozotocin (STZ). After 4 weeks behavioral testing (Plantar test, Randall-Selitto) was conducted. Basal and stimulated release of calcitonin gene-related peptide (CGRP), Substance P (SP) and prostaglandin E(2) (PGE(2)) from isolated skin and sciatic nerve were assessed by enzyme immunoassays. Electrophysiological properties of identified nociceptors under hyperglycemic, hypoxic, and acidotic conditions were investigated using the skin-nerve preparation. The diabetic rats showed hyperalgesia to heat and pressure stimulation. The basal CGRP/SP release was reduced, but chemical stimulation with bradykinin induced greater release of SP, CGRP and PGE(2) than in control animals. In contrast, capsaicin-stimulated CGRP release was reduced in sciatic nerves. Hypoxia per se lowered von Frey thresholds of most C-nociceptors to half. Hyperglycemic hypoxia induced ongoing discharge in all diabetic but not control C-fibers which was further enhanced under acidosis. Sensory and neurosecretory nociceptor functions are sensitized in diabetes. Diabetic C-fibers show exaggerated sensitivity to hyperglycemic hypoxia with and without additional acidosis, conditions that are thought to mimic ischemic episodes in diabetic nerves. Ongoing C-fiber discharge is known to induce spinal sensitization. Together with altered receptor and ion channel expressions this may contribute to painful episodes in diabetic neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Hyperalgesia/etiology , Nociceptors/physiology , Pain Threshold/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Behavior, Animal , Bradykinin/pharmacology , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Dinoprostone/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Nerve Fibers/physiology , Pain Measurement/methods , Pain Threshold/drug effects , Rats , Reaction Time/drug effects , Reaction Time/physiology , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Skin/drug effects , Skin/metabolism , Streptozocin/adverse effects , Substance P/metabolism
12.
Neurogastroenterol Motil ; 22(6): 680-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20100279

ABSTRACT

BACKGROUND: Acid-sensing and regulating reactions are vitally important in the upper gastrointestinal tract and disturbances are common. Sensory neurons in the mucosa detect the intrusion of hydrogen ions and, by their release of vasoactive neuropeptides, seem to play a predominantly protective role in these tissues. METHODS: The model to investigate sensory transduction of proton stimuli in the isolated everted mouse stomach was to measure the induced calcitonin gene-related peptide (CGRP) release as an index of neuronal activation. KEY RESULTS: Proton concentrations in the range of pH 2.5-0.5 stimulated the release of CGRP and substance P and profoundly decreased the prostaglandin E2 formation in outbred CD mice. A similar linearly pH-dependent CGRP release was observed in inbred C57BL/6 mice, fully dependent on extracellular calcium at pH 2, partially at pH 1. Both transient receptor potential vanilloid type 1 (TRPV1) and acid-sensing ion channel type 3 (ASIC3) are expressed in the sensory neurons innervating the stomach walls and are responsible for the transduction of acidic stimuli in other visceral organs. However, the proton-induced gastric CGRP release in mice lacking the TRPV1 or the ASIC3 receptor-channels was the same as in corresponding wild-type mice. Nonetheless, the pharmacological blockers N-(4-tertiarybutylphenyl)-4-(3-chlorophyridin-2-yl)tetrahydropyrazine-1(2H)carboxamide and amiloride, respectively, inhibited the acid-stimulated CGRP release, although to the same extend in wild types as TRPV1 and ASIC3 knockout mice. CONCLUSIONS & INFERENCES: Adequate proton concentrations inhibit prostaglandin and stimulate CGRP release from the stomach wall, however, the transduction mechanism in the gastric sensory neurons remains unclear.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Gastric Mucosa/metabolism , Sodium Channels/physiology , TRPV Cation Channels/physiology , Acid Sensing Ion Channels , Acids , Amiloride/pharmacology , Animals , Data Interpretation, Statistical , Dinoprostone/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/innervation , Hydrogen-Ion Concentration , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Sodium Channels/drug effects , Sodium Channels/genetics , Stomach/drug effects , Substance P/metabolism , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics
13.
Klin Padiatr ; 222(1): 45-50, 2010.
Article in English | MEDLINE | ID: mdl-20084592

ABSTRACT

BACKGROUND: With simple prevention measures SIDS (Sudden Infant Death Syndrome) incidence rates can be reduced. A cross-sectional survey in Bavaria in 2005 showed that maternity units fall short in acting as role models and informing parents regarding SIDS prevention and that parents did not sufficiently apply prevention measures. An information campaign in Bavaria was initialized and evaluated after 18 months. METHODS: Flyers and posters for SIDS prevention using consistent information were developed for dissemination by the relevant occupational groups. The campaign impact was assessed in repeating the cross-sectional survey. RESULTS: A response rate of 99% for the maternity units and 60% for the parent questionnaires could be achieved. Significant improvements (p<0.05) in the maternity units regarded a higher proportion of parents informed about SIDS prevention measures (97 vs. 92%) and an increased use of sleeping bags (37 vs. 12%), whereas the proportion of children placed in supine position to sleep was not increased. More parents reported to have obtained advice for SIDS prevention (83 vs. 73%) and to have used sleeping bags (66 vs. 56%). A subgroup analysis revealed poorer information and implementation of the recommendations in families with three or more children and less educated families. CONCLUSIONS: After initiation of the information campaign reported knowledge about SIDS prevention measures increased. Continuing shortfalls exist regarding maternity units acting as role models and implementation of the recommendations by the parents. Future interventions should focus in particular on the role model function of maternity units and target groups.


Subject(s)
Sudden Infant Death/prevention & control , Austria , Cross-Sectional Studies , Health Promotion , Humans , Incidence , Infant , Infant Care/methods , Sudden Infant Death/epidemiology
14.
Eur J Pain ; 13(1): 35-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18993095

ABSTRACT

Distinct sensory properties of unmyelinated axons in the isolated rat sciatic nerve have previously been revealed by measuring stimulated neuropeptide (CGRP) release in response to noxious stimuli. Axonal sensitization to heat by inflammatory mediators has been demonstrated and shown to depend on the heat- and proton-activated ion channel TRPV1. Recently, we have demonstrated in vitro that heat stimulation of nociceptive axons generates ectopic action potential discharge which resembles the heat response of the corresponding cutaneous nerve endings. It remained however, to be established whether adequate axonal stimulation could also generate projected sensations in a conscious human subject. In a singular human trial, the superficial radial nerve (SR) was exposed and stimulated mechanically as well as with noxious cold (3 degrees C). These stimuli were unable to induce any conscious local or projected sensations. However, controlled radiant heat applied to the nerve resulted in intense slowly adapting burning pain sensations projected into the center of the SR innervation area. No local sensation was reported. Thus, presumably activated nervi nevorum in the sheath of a healthy nerve do not cause conscious sensations, while axons of passage in mid-nerve exhibit a sensory transduction capacity for noxious heat though not for mechanical and cold stimulation. Axonal heat transduction may therefore become a source of ectopic discharge and neuropathic pain when heat threshold drops to body temperature as is the case with peripheral nerve endings in inflamed skin.


Subject(s)
Hot Temperature , Pain/physiopathology , Peripheral Nerves/physiopathology , Animals , Axons/physiology , Cold Temperature , Humans , Male , Middle Aged , Myelin Sheath/physiology , Neurosurgical Procedures , Pain Measurement , Physical Stimulation , Radial Nerve/pathology , Radial Nerve/physiopathology , Rats
16.
Gesundheitswesen ; 70 Suppl 1: S29-33, 2008 Mar.
Article in German | MEDLINE | ID: mdl-18368653

ABSTRACT

INTRODUCTION: The pathomechanisms of SIDS are not yet completely understood. However some risk factors are known which can be influenced by simple prevention measures. In order to spread this knowledge and inform young parents, an information campaign was launched in Bavaria in the year 2005. Different occupational groups were involved. METHODS: Firstly, an analysis of interviews and questionnaires of maternal units and parents concerning knowledge and implementation of recommendations for SIDS prevention was done. Subsequently information material was developed and distributed and a publicity was started. A flyer containing simple and evidence-based recommendations was also translated in foreign languages to reach disadvantaged people. RESULTS: Analysis of the questionnaires showed shortfalls concerning information and implementation of recommendations for SIDS prevention. There was high request for the information material. The demand increased impressively after public relations work. Up to the end of October 2007, 233 046 flyers had been dispatched, 42 959 in foreign languages. The greatest part was sent to material units and local health departments. DISCUSSION: The information campaign had a good response. Further intensification and inclusion of different professional workers and extension is necessary as well as extension of publicity. All efforts have to be done to reduce the risk of SIDS.


Subject(s)
Health Promotion/methods , Health Promotion/organization & administration , Infant Mortality , Outcome Assessment, Health Care , Risk Assessment/methods , Sudden Infant Death/epidemiology , Sudden Infant Death/prevention & control , Germany/epidemiology , Humans , Infant, Newborn , Risk Factors
17.
Neuroscience ; 151(3): 836-42, 2008 Feb 06.
Article in English | MEDLINE | ID: mdl-18178321

ABSTRACT

We have previously shown that capsaicin, noxious heat, protons and potassium ions (K(+)) induce a graded, calcium- and receptor-dependent increase of immunoreactive calcitonin gene-related peptide (iCGRP) release from isolated rat sciatic axons. Morphological evidence for axonal vesicular exocytosis has also been presented. Here we determine the differential contribution of voltage-gated calcium and sodium channels to high extracellular potassium and capsaicin-induced iCGRP secretion. Blockade of L-type calcium channels significantly decreased the K(+)-induced axonal response (nimodipine (10 microM) by 66% and methoxyverapamil, D600 (50 microM), by 77%). Interestingly, however, D600 was unable to reduce the capsaicin-induced iCGRP release. Omega-Conotoxin GVIA (1 microM), a N-type blocker, and omega-agatoxin TK (0.1 microM), a P/Q-type blocker, had no significant effect. Also the anticonvulsant gabapentin (50 microM and 100 microM), reported to impede calcium channels, was ineffective. Inhibition of low threshold T-type calcium channels by mibefradil (10 microM) significantly reduced potassium (by 47%) but not capsaicin-stimulated iCGRP release. Reduction of total sodium channel conductance by tetrodotoxin (1 microM), lidocaine (10 microM, 50 microM or 500 microM) or by replacement of extracellular sodium with choline-chloride did not result in a reduction of either potassium- or capsaicin-induced axonal iCGRP release. These results suggest that slow depolarization by high extracellular potassium activates axonal low threshold (T-type) as well as high threshold-activated (L-type) voltage-gated calcium channels to mediate iCGRP release, and that capsaicin-induced release is largely dependent on calcium influx through TRPV1. Action potential generation and propagation are not required for axonal release mechanisms.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Calcium Channels, L-Type/physiology , Calcium Channels, T-Type/physiology , Capsaicin/pharmacology , Extracellular Fluid/drug effects , Potassium/pharmacology , TRPV Cation Channels/physiology , Amines/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Excitatory Amino Acid Antagonists/pharmacology , Gabapentin , Immunoenzyme Techniques/methods , In Vitro Techniques , Male , Rats , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Statistics, Nonparametric , gamma-Aminobutyric Acid/pharmacology
18.
Neuroscience ; 151(1): 242-54, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18065157

ABSTRACT

A recently described family of "orphan" receptors, called Mas-related G-protein-coupled receptors (Mrg), is preferentially expressed in small nociceptive neurons of the rodent and human dorsal root ganglia (DRG). Mrg are activated by high affinity peptide fragments derived from the proenkephalin A gene, e.g. BAM22 (bovine adrenal medullary). To study the histological distribution and functional properties of these receptors, we combined confocal immunohistochemistry in rat DRG and dermis whole mounts, using new antibodies against the rat Mas-related G-protein-coupled receptor C (MrgC), with single-fiber recordings and neurochemical experiments using isolated hind-paw skin and sciatic nerve. In lumbar DRG we found cytoplasmic MrgC labeling mainly in small- and medium-sized neurons; coexpression with isolectin B4 (46%) and transient receptor potential vanilloid receptor 1 channel protein (TRPV1) (52%) occurred frequently, whereas calcitonin gene-related peptide (CGRP) was rarely colocalized with MrgC in DRG (11%) and dermal nerve fibers (6%). One of the MrgC agonists, BAM22, more than doubled the heat-induced cutaneous CGRP release from rat and mouse skin. The effect of BAM22, also known to activate opioid receptors, was further enhanced by combination with naloxone that had no effect on its own. This sensitizing effect proved to be independent of secondary prostaglandin formation, mast cell degranulation, protein kinase C (PKC) activation and independent of TRPV1. Nonetheless, the capsaicin-induced CGRP release was also sensitized. Receptive fields of 26 mechano-heat sensitive C-fibers were treated with MrgC agonists. Only one unit was strongly and repeatedly excited and showed a profound sensitization to heat upon BAM22+naloxone. Two other established MrgC agonists (gamma2-melanocyte stimulating hormone and BAM8-22) were ineffective. Thus, BAM22 sensitizes the capsaicin- and heat-induced CGRP release in an apparently MrgC-unrelated way. The sensitization to heat appears unusually resistant against pharmacological interventions and does not involve TRPV1.


Subject(s)
Hydrogen-Ion Concentration , Receptors, G-Protein-Coupled/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Electrophysiology , Female , Fluorescent Antibody Technique , Ganglia, Spinal/drug effects , Ganglia, Spinal/ultrastructure , Histamine Release/drug effects , Immunohistochemistry , Male , Molecular Sequence Data , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nerve Fibers/drug effects , Nerve Fibers/physiology , Prostaglandins/biosynthesis , Rats , Rats, Wistar , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism , Skin/drug effects , Skin/innervation , Skin/metabolism , TRPV Cation Channels/biosynthesis , TRPV Cation Channels/genetics , Tissue Fixation
19.
J Physiol ; 582(Pt 1): 229-42, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17478536

ABSTRACT

Emerging evidence indicates a nociceptive role of vagal afferents. A distinct oesophageal innervation in the rat, with muscular and mucosal afferents travelling predominantly in the recurrent (RLN) and superior laryngeal nerve (SLN), respectively, enabled characterization of mucosal afferents with nociceptive properties, using novel isolated oesophagus-nerve preparations. SLN and RLN single-fibre recordings identified 55 and 14 units, respectively, with none conducting faster than 8.7 m s(-1). Mucosal response characteristics in the SLN distinguished mechanosensors (n = 13), mechanosensors with heat sensitivity (18) from those with cold sensitivity (19) and a mechanoinsensitive group (5). The mechanosensitive fibres, all slowly adapting, showed a unimodal distribution of mechanical thresholds (1.4-128 mN, peak approximately 5.7 mN). No difference in response characteristics of C and Adelta fibres was encountered. Mucosal proton stimulation (pH 5.4 for 3 min), mimicking gastro-oesophageal reflux disease (GORD), revealed in 31% of units a desensitizing response that peaked around 20 s and faded within 60 s. Cold stimulation (15 degrees C) was proportionally encoded but the response showed slow adaptation. In contrast, the noxious heat (48 degrees C) response showed no obvious adaptation with discharge rates reflecting the temperature's time course. Polymodal (69%) mucosal units, > 30% proton sensitive, were found in each fibre category and were considered nociceptors; they are tentatively attributed to vagal nerve endings type I, IV and V, previously morphologically described. All receptive fields were mapped and the distribution indicates that the posterior upper oesophagus may serve as a 'cutbank', detecting noxious matters, ingested or regurgitated, and triggering nocifensive reflexes such as bronchoconstriction in GORD.


Subject(s)
Esophagus/innervation , Esophagus/metabolism , Gastroesophageal Reflux/metabolism , Mechanotransduction, Cellular , Neurons, Afferent/metabolism , Nociceptors/metabolism , Pain/etiology , Vagus Nerve/metabolism , Action Potentials , Animals , Cold Temperature , Electrophysiology/methods , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/physiopathology , Hot Temperature , Hydrogen-Ion Concentration , In Vitro Techniques , Kinetics , Laryngeal Nerves/metabolism , Male , Mucous Membrane/innervation , Mucous Membrane/metabolism , Muscle, Smooth/innervation , Muscle, Smooth/metabolism , Neural Conduction , Pain/metabolism , Pain/physiopathology , Rats , Rats, Wistar , Reaction Time , Recurrent Laryngeal Nerve/metabolism , Sensory Thresholds , Skin/innervation , Vagus Nerve/cytology
20.
Neuroscience ; 135(4): 1277-84, 2005.
Article in English | MEDLINE | ID: mdl-16165301

ABSTRACT

TRPV1 gene disruption results in a loss of capsaicin and proton responsiveness, but has minimal effects on heat-induced nocifensive behavior, suggesting that sensory transduction of heat is independent of TRPV1. TRPV3, another heat-activated ion channel but insensitive to capsaicin, was shown to be expressed in keratinocytes as well as in sensory neurons projecting to the skin. Recently, 2-aminoethoxydiphenyl borate was introduced as a TRPV3 agonist, but its selectivity was questioned by showing that it activated recombinant TRPV1 and TRPV2 as well. We used the isolated mouse skin-saphenous nerve preparation and whole-cell patch-clamping of cultured dorsal root ganglia neurons from TRPV1-/- and wildtype mice. We found no phenotypic differences between the heat responses of polymodal C-fibers, whereas cultured dorsal root ganglia neurons of TRPV1-/- hardly showed any heat-activated currents. Only C-fibers of wildtype but not TRPV1-/- mice were clearly sensitized to heat by 2-aminoethoxydiphenyl borate 10 and 100 microM; heat-activated current in wildtype neurons was only facilitated at 100 microM. Noxious heat-induced calcitonin gene-related peptide release showed clear deficits (<50%) in TRPV1 deficient skin, but the stimulated calcitonin gene-related peptide release from the isolated skull dura was unaffected. In both models, 2-aminoethoxydiphenyl borate was able to potentiate the heat response (46 degrees C, 5 min) in a concentration-dependent manner, again, only in wildtype but not TRPV1-/- mice, suggesting that TRPV2/3 are not involved in this sensitization to heat. The results further suggest that TRPV1 is not responsible for the normal heat response of native nociceptors but plays the essential role in thermal sensitization and a prominent one in controlling dermal calcitonin gene-related peptide release, i.e. neurogenic inflammation.


Subject(s)
Boron Compounds/pharmacology , Hot Temperature , Nociceptors/drug effects , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism , Animals , Calcitonin Gene-Related Peptide/drug effects , Calcitonin Gene-Related Peptide/metabolism , Cells, Cultured , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Mice , Mice, Knockout , Nociceptors/metabolism , Organ Culture Techniques , Pain/metabolism , Pain/physiopathology , Patch-Clamp Techniques , Skin/drug effects , Skin/innervation , Skin/metabolism , TRPV Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...