Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Front Mol Neurosci ; 14: 709228, 2021.
Article in English | MEDLINE | ID: mdl-34385907

ABSTRACT

Apart from the most prominent symptoms in Autism spectrum disorder (ASD), namely deficits in social interaction, communication and repetitive behavior, patients often show abnormal sensory reactivity to environmental stimuli. Especially potentially painful stimuli are reported to be experienced in a different way compared to healthy persons. In our present study, we identified an ASD patient carrying compound heterozygous mutations in the voltage-gated sodium channel (VGSC) Na v 1.8, which is preferentially expressed in sensory neurons. We expressed both mutations, p.I1511M and p.R512∗, in a heterologous expression system and investigated their biophysical properties using patch-clamp recordings. The results of these experiments reveal that the p.R512∗ mutation renders the channel non-functional, while the p.I1511M mutation showed only minor effects on the channel's function. Behavioral experiments in a Na v 1.8 loss-of-function mouse model additionally revealed that Na v 1.8 may play a role in autism-like symptomatology. Our results present Na v 1.8 as a protein potentially involved in ASD pathophysiology and may therefore offer new insights into the genetic basis of this disease.

3.
Channels (Austin) ; 15(1): 208-228, 2021 12.
Article in English | MEDLINE | ID: mdl-33487118

ABSTRACT

Mutations in the voltage-gated sodium channel Nav1.7 are linked to human pain. The Nav1.7/N1245S variant was described before in several patients suffering from primary erythromelalgia and/or olfactory hypersensitivity. We have identified this variant in a pain patient and a patient suffering from severe and life-threatening orthostatic hypotension. In addition, we report a female patient suffering from muscle pain and carrying the Nav1.7/E1139K variant. We tested both Nav1.7 variants by whole-cell voltage-clamp recordings in HEK293 cells, revealing a slightly enhanced current density for the N1245S variant when co-expressed with the ß1 subunit. This effect was counteracted by an enhanced slow inactivation. Both variants showed similar voltage dependence of activation and steady-state fast inactivation, as well as kinetics of fast inactivation, deactivation, and use-dependency compared to WT Nav1.7. Finally, homology modeling revealed that the N1245S substitution results in different intramolecular interaction partners. Taken together, these experiments do not point to a clear pathogenic effect of either the N1245S or E1139K variant and suggest they may not be solely responsible for the patients' pain symptoms. As discussed previously for other variants, investigations in heterologous expression systems may not sufficiently mimic the pathophysiological situation in pain patients, and single nucleotide variants in other genes or modulatory proteins are necessary for these specific variants to show their effect. Our findings stress that biophysical investigations of ion channel mutations need to be evaluated with care and should preferably be supplemented with studies investigating the mutations in their context, ideally in human sensory neurons.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Erythromelalgia , HEK293 Cells , Humans , Membrane Potentials , Patch-Clamp Techniques
4.
J Biol Chem ; 296: 100227, 2021.
Article in English | MEDLINE | ID: mdl-33361158

ABSTRACT

Mutations in voltage-gated sodium channels (Navs) can cause alterations in pain sensation, such as chronic pain diseases like inherited erythromelalgia. The mutation causing inherited erythromelalgia, Nav1.7 p.I848T, is known to induce a hyperpolarized shift in the voltage dependence of activation in Nav1.7. So far, however, the mechanism to explain this increase in voltage sensitivity remains unknown. In the present study, we show that phosphorylation of the newly introduced Thr residue explains the functional change. We expressed wildtype human Nav1.7, the I848T mutant, or other mutations in HEK293T cells and performed whole-cell patch-clamp electrophysiology. As the insertion of a Thr residue potentially creates a novel phosphorylation site for Ser/Thr kinases and because Nav1.7 had been shown in Xenopus oocytes to be affected by protein kinases C and A, we used different nonselective and selective kinase inhibitors and activators to test the effect of phosphorylation on Nav1.7 in a human system. We identify protein kinase C, but not protein kinase A, to be responsible for the phosphorylation of T848 and thereby for the shift in voltage sensitivity. Introducing a negatively charged amino acid instead of the putative phosphorylation site mimics the effect on voltage gating to a lesser extent. 3D modeling using the published cryo-EM structure of human Nav1.7 showed that introduction of this negatively charged site seems to alter the interaction of this residue with the surrounding amino acids and thus to influence channel function. These results could provide new opportunities for the development of novel treatment options for patients with chronic pain.


Subject(s)
Membrane Potentials/physiology , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Protein Kinase C/metabolism , Protein Processing, Post-Translational , Threonine/chemistry , Amino Acid Substitution , Binding Sites , Chronic Pain/genetics , Chronic Pain/metabolism , Chronic Pain/physiopathology , Erythromelalgia/genetics , Erythromelalgia/metabolism , Erythromelalgia/physiopathology , Gene Expression , HEK293 Cells , Humans , Ion Channel Gating/physiology , Isoleucine/chemistry , Isoleucine/metabolism , Models, Molecular , Mutation , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staurosporine/pharmacology , Threonine/metabolism
5.
Neuropharmacology ; 158: 107749, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31461640

ABSTRACT

The homotrimeric P2X3 receptor, one of the seven members of the ATP-gated P2X receptor family, plays a crucial role in sensory neurotransmission. P2X3 receptor antagonists have been identified as promising drugs to treat chronic cough and are suggested to offer pain relief in chronic pain such as neuropathic pain. Here, we analysed whether compounds affect P2X3 receptor activity by high-throughput screening of the Spectrum Collection of 2000 approved drugs, natural products and bioactive substances. We identified aurintricarboxylic acid (ATA) as a nanomolar-potency antagonist of P2X3 receptor-mediated responses. Two-electrode voltage clamp electrophysiology-based concentration-response analysis and selectivity profiling revealed that ATA strongly inhibits the rP2X1 and rP2X3 receptors (with IC50 values of 8.6 nM and 72.9 nM, respectively) and more weakly inhibits P2X2/3, P2X2, P2X4 or P2X7 receptors (IC50 values of 0.76 µM, 22 µM, 763 µM or 118 µM, respectively). Patch-clamp analysis of mouse DRG neurons revealed that ATA inhibited native P2X3 and P2X2/3 receptors to a similar extent than rat P2X3 and P2X2/3 receptors expressed in Xenopus oocytes. In a radioligand binding assay, up to 30 µM ATA did not compete with [3H]-ATP for rP2X3 receptor binding, indicating a non-competitive mechanism of action. Molecular docking studies, site-directed mutagenesis and concentration-response analysis revealed that ATA binds to the negative allosteric site of the hP2X3 receptor. In summary, ATA as a drug-like pharmacological tool compound is a nanomolar-potency, allosteric antagonist with selectivity towards αß-methylene-ATP-sensitive P2X1 and P2X3 receptors.


Subject(s)
Aurintricarboxylic Acid/pharmacology , Neurons/drug effects , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X1/drug effects , Receptors, Purinergic P2X3/drug effects , Allosteric Regulation , Allosteric Site , Animals , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , High-Throughput Screening Assays , Humans , Mice , Molecular Docking Simulation , Neurons/metabolism , Oocytes , Patch-Clamp Techniques , Rats , Receptors, Purinergic P2X1/metabolism , Receptors, Purinergic P2X3/metabolism , Xenopus laevis
6.
Pain ; 160(6): 1327-1341, 2019 06.
Article in English | MEDLINE | ID: mdl-30720580

ABSTRACT

The chronic pain syndrome inherited erythromelalgia (IEM) is attributed to mutations in the voltage-gated sodium channel (NaV) 1.7. Still, recent studies targeting NaV1.7 in clinical trials have provided conflicting results. Here, we differentiated induced pluripotent stem cells from IEM patients with the NaV1.7/I848T mutation into sensory nociceptors. Action potentials in these IEM nociceptors displayed a decreased firing threshold, an enhanced upstroke, and afterhyperpolarization, all of which may explain the increased pain experienced by patients. Subsequently, we investigated the voltage dependence of the tetrodotoxin-sensitive NaV activation in these human sensory neurons using a specific prepulse voltage protocol. The IEM mutation induced a hyperpolarizing shift of NaV activation, which leads to activation of NaV1.7 at more negative potentials. Our results indicate that NaV1.7 is not active during subthreshold depolarizations, but that its activity defines the action potential threshold and contributes significantly to the action potential upstroke. Thus, our model system with induced pluripotent stem cell-derived sensory neurons provides a new rationale for NaV1.7 function and promises to be valuable as a translational tool to profile and develop more efficacious clinical analgesics.


Subject(s)
Erythromelalgia/physiopathology , Induced Pluripotent Stem Cells/cytology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sensory Receptor Cells/metabolism , Action Potentials/drug effects , Electric Stimulation/methods , Erythromelalgia/genetics , Ganglia, Spinal/cytology , Humans , Membrane Potentials/drug effects , NAV1.7 Voltage-Gated Sodium Channel/genetics , Nociceptors/physiology , Pain/diagnosis , Pain/genetics , Patch-Clamp Techniques/methods , Tetrodotoxin/pharmacology
7.
J Neurophysiol ; 121(2): 427-443, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30485151

ABSTRACT

The transient receptor potential ankyrin 1 (TRPA1) ion channel is expressed in pain-sensing neurons and other tissues and has become a major target in the development of novel pharmaceuticals. A remarkable feature of the channel is its long list of activators, many of which we are exposed to in daily life. Many of these agonists induce pain and inflammation, making TRPA1 a major target for anti-inflammatory and analgesic therapies. Studies in human patients and in experimental animals have confirmed an important role for TRPA1 in a number of pain conditions. Over the recent years, much progress has been made in elucidating the molecular structure of TRPA1 and in discovering binding sites and modulatory sites of the channel. Because the list of published mutations and important molecular sites is steadily growing and because it has become difficult to see the forest for the trees, this review aims at summarizing the current knowledge about TRPA1, with a special focus on the molecular structure and the known binding or gating sites of the channel.


Subject(s)
TRPA1 Cation Channel/metabolism , Animals , Humans , Ion Channel Gating , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/genetics
8.
Pflugers Arch ; 470(12): 1787-1801, 2018 12.
Article in English | MEDLINE | ID: mdl-30099632

ABSTRACT

Mutations in voltage-gated sodium channels are associated with altered pain perception in humans. Most of these mutations studied to date present with a direct and intuitive link between the altered electrophysiological function of the channel and the phenotype of the patient. In this study, we characterize a variant of Nav1.8, D1639N, which has been previously identified in a patient suffering from the chronic pain syndrome "small fiber neuropathy". Using a heterologous expression system and patch-clamp analysis, we show that Nav1.8/D1639N reduces current density without altering biophysical gating properties of Nav1.8. Therefore, the D1639N variant causes a loss-of-function of the Nav1.8 sodium channel in a patient suffering from chronic pain. Using immunocytochemistry and biochemical approaches, we show that Nav1.8/D1639N impairs trafficking of the channel to the cell membrane. Neither co-expression of ß1 or ß3 subunit, nor overnight incubation at 27 °C rescued current density of the D1639N variant. On the other hand, overnight incubation with lidocaine fully restored current density of Nav1.8/D1639N most likely by overcoming the trafficking defect, whereas phenytoin failed to do so. Since lidocaine rescues the loss-of-function of Nav1.8/D1639N, it may offer a future therapeutic option for the patient carrying this variant. These results demonstrate that the D1639N variant, identified in a patient suffering from chronic pain, causes loss-of-function of the channel due to impaired cell surface trafficking and that this trafficking defect can be rescued by lidocaine.


Subject(s)
Anesthetics, Local/pharmacology , Chronic Pain/genetics , Lidocaine/pharmacology , Loss of Function Mutation , NAV1.8 Voltage-Gated Sodium Channel/genetics , Action Potentials , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/physiology , Humans , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Protein Transport/drug effects , Xenopus
9.
Br J Pharmacol ; 175(14): 3007-3020, 2018 07.
Article in English | MEDLINE | ID: mdl-29722437

ABSTRACT

BACKGROUND AND PURPOSE: Oxycodone is a potent semi-synthetic opioid that is commonly used for the treatment of severe acute and chronic pain. However, treatment with oxycodone can lead to cardiac electrical changes, such as long QT syndrome, potentially inducing sudden cardiac arrest. Here, we investigate whether the cardiac side effects of oxycodone can be explained by modulation of the cardiac Nav 1.5 sodium channel. EXPERIMENTAL APPROACH: Heterologously expressed human Nav 1.5, Nav 1.7 (HEK293 cells) or Nav 1.8 channels (mouse N1E-115 cells) were used for whole-cell patch-clamp electrophysiology. A variety of voltage-clamp protocols were used to test the effect of oxycodone on different channel gating modalities. Human stem cell-derived cardiomyocytes were used to measure the effect of oxycodone on cardiomyocyte beating. KEY RESULTS: Oxycodone inhibited Nav 1.5 channels, concentration and use-dependently, with an IC50 of 483 µM. In addition, oxycodone slows recovery of Nav 1.5 channels from fast inactivation and increases slow inactivation. At high concentrations, these effects lead to a reduced beat rate in cardiomyocytes and to arrhythmia. In contrast, no such effects could be observed on Nav 1.7 or Nav 1.8 channels. CONCLUSIONS AND IMPLICATIONS: Oxycodone leads to an accumulation of Nav 1.5 channels in inactivated states, with a slow time course. Although the concentrations needed to elicit cardiac arrhythmias in vitro are relatively high, some patients under long-term treatment with oxycodone as well as drug abusers and addicts might suffer from severe cardiac side effects induced by the slowly developing effects of oxycodone on Nav 1.5 channels.


Subject(s)
Analgesics, Opioid/pharmacology , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/physiology , Oxycodone/pharmacology , Sodium Channel Blockers/pharmacology , Animals , Cell Line , Humans , Mice , Myocytes, Cardiac/physiology
10.
PLoS One ; 12(1): e0170097, 2017.
Article in English | MEDLINE | ID: mdl-28076424

ABSTRACT

The TRPA1 ion channel is expressed in nociceptive (pain-sensitive) somatosensory neurons and is activated by a wide variety of chemical irritants, such as acrolein in smoke or isothiocyanates in mustard. Here, we investigate the enhancement of TRPA1 function caused by inflammatory mediators, which is thought to be important in lung conditions such as asthma and COPD. Protein kinase A is an important kinase acting downstream of inflammatory mediators to cause sensitization of TRPA1. By using site-directed mutagenesis, patch-clamp electrophysiology and calcium imaging we identify four amino acid residues, S86, S317, S428, and S972, as the principal targets of PKA-mediated phosphorylation and sensitization of TRPA1.


Subject(s)
Calcium Channels/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Nerve Tissue Proteins/physiology , Transient Receptor Potential Channels/physiology , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/drug effects , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/physiology , Cymenes , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Monoterpenes/pharmacology , Mutagenesis, Site-Directed , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , TRPA1 Cation Channel , Transfection , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
11.
J Physiol ; 594(22): 6643-6660, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27307078

ABSTRACT

KEY POINTS: The transient receptor potential ankyrin 1 (TRPA1) ion channel is expressed in nociceptive neurons and its activation causes ongoing pain and inflammation; TRPA1 is thought to play an important role in inflammation in the airways. TRPA1 is sensitised by repeated stimulation with chemical agonists in a calcium-free environment and this sensitisation is very long lasting following agonist removal. We show that agonist-induced sensitisation is independent of the agonist's binding site and is also independent of ion channel trafficking or of other typical signalling pathways. We find that sensitisation is intrinsic to the TRPA1 protein and is accompanied by a slowly developing shift in the voltage dependence of TRPA1 towards more negative membrane potentials. Agonist-induced sensitisation may provide an explanation for sensitisation following long-term exposure to harmful irritants and pollutants, particularly in the airways. ABSTRACT: The TRPA1 ion channel is expressed in nociceptive (pain-sensitive) neurons and responds to a wide variety of chemical irritants, such as acrolein in smoke or isothiocyanates in mustard. Here we show that in the absence of extracellular calcium the current passing through TRPA1 gradually increases (sensitises) during prolonged application of agonists. Activation by an agonist is essential, because activation of TRPA1 by membrane depolarisation did not cause sensitisation. Sensitisation is independent of the site of action of the agonist, because covalent and non-covalent agonists were equally effective, and is long lasting following agonist removal. Mutating N-terminal cysteines, the target of covalent agonists, did not affect sensitisation by the non-covalent agonist carvacrol, which activates by binding to a different site. Sensitisation is unaffected by agents blocking ion channel trafficking or by block of signalling pathways involving ATP, protein kinase A or the formation of lipid rafts, and does not require ion flux through the channel. Examination of the voltage dependence of TRPA1 activation shows that sensitisation is accompanied by a slowly developing shift in the voltage dependence of TRPA1 towards more negative membrane potentials, and is therefore intrinsic to the TRPA1 channel. Sensitisation may play a role in exacerbating the pain caused by prolonged activation of TRPA1.


Subject(s)
Monoterpenes/pharmacology , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Cymenes , Female , Humans , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Signal Transduction/drug effects
12.
J Headache Pain ; 16: 57, 2015.
Article in English | MEDLINE | ID: mdl-26109436

ABSTRACT

BACKGROUND: The capsaicin and heat responsive ion channel TRPV1 is expressed on trigeminal nociceptive neurons and has been implicated in the pathophysiology of migraine attacks. Here we investigate the efficacy of two TRPV1 channel antagonists in blocking trigeminal activation using two in vivo models of migraine. METHODS: Male Sprague-Dawley rats were used to study the effects of the TRPV1 antagonists JNJ-38893777 and JNJ-17203212 on trigeminal activation. Expression of the immediate early gene c-fos was measured following intracisternal application of inflammatory soup. In a second model, CGRP release into the external jugular vein was determined following injection of capsaicin into the carotid artery. RESULTS: Inflammatory up-regulation of c-fos in the trigeminal brain stem complex was dose-dependently and significantly reduced by both TRPV1 antagonists. Capsaicin-induced CGRP release was attenuated by JNJ-38893777 only in higher dosage. JNJ-17203212 was effective in all doses and fully abolished CGRP release in a time and dose-dependent manner. CONCLUSION: Our results describe two TRPV1 antagonists that are effective in two in vivo models of migraine. These results suggest that TRPV1 may play a role in the pathophysiological mechanisms, which are relevant to migraine.


Subject(s)
Aminopyridines/therapeutic use , Disease Models, Animal , Migraine Disorders/drug therapy , Piperazines/therapeutic use , TRPV Cation Channels/antagonists & inhibitors , Aminopyridines/pharmacology , Animals , Capsaicin/toxicity , Dose-Response Relationship, Drug , Genes, fos/drug effects , Male , Migraine Disorders/chemically induced , Migraine Disorders/metabolism , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Treatment Outcome , Up-Regulation/drug effects
13.
Trends Mol Med ; 16(4): 153-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20347391

ABSTRACT

Migraine is among the most prevalent headache disorders and results from dysfunctions within the trigeminovascular system (TVS). The inflammatory processes that have been suggested to occur in the cascade of events resulting in migraine sensitise trigeminal nociceptors, possibly causing hyperalgesia and allodynia. Trigeminal nociceptors express the heat- and capsaicin-gated channel TRPV1, which seems to play a significant role in the development of peripheral and central sensitisation and of hyperalgesia and allodynia. Here, we review the molecular mechanisms leading to the sensitisation of TRPV1 and attempt to link them to migraine-relevant pathophysiological processes. We argue that antagonising TRPV1 sensitisation is a promising approach and should receive more attention in future research as well as in the development of anti-migraine drugs.


Subject(s)
Migraine Disorders/physiopathology , TRPV Cation Channels/metabolism , Animals , Calcitonin Gene-Related Peptide/physiology , Capsaicin/metabolism , Female , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Male , Mice , Migraine Disorders/drug therapy , Nociceptors/drug effects , Nociceptors/physiology , Rats , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/physiopathology , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...