Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Subcell Biochem ; 104: 207-244, 2024.
Article in English | MEDLINE | ID: mdl-38963489

ABSTRACT

The transient receptor potential ion channel TRPA1 is a Ca2+-permeable nonselective cation channel widely expressed in sensory neurons, but also in many nonneuronal tissues typically possessing barrier functions, such as the skin, joint synoviocytes, cornea, and the respiratory and intestinal tracts. Here, the primary role of TRPA1 is to detect potential danger stimuli that may threaten the tissue homeostasis and the health of the organism. The ability to directly recognize signals of different modalities, including chemical irritants, extreme temperatures, or osmotic changes resides in the characteristic properties of the ion channel protein complex. Recent advances in cryo-electron microscopy have provided an important framework for understanding the molecular basis of TRPA1 function and have suggested novel directions in the search for its pharmacological regulation. This chapter summarizes the current knowledge of human TRPA1 from a structural and functional perspective and discusses the complex allosteric mechanisms of activation and modulation that play important roles under physiological or pathophysiological conditions. In this context, major challenges for future research on TRPA1 are outlined.


Subject(s)
TRPA1 Cation Channel , Humans , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/physiology , Cryoelectron Microscopy/methods , Animals , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/physiology , Structure-Activity Relationship , Allosteric Regulation
2.
Nat Commun ; 13(1): 7483, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36470868

ABSTRACT

TRPV2 is a ligand-operated temperature sensor with poorly defined pharmacology. Here, we combine calcium imaging and patch-clamp electrophysiology with cryo-electron microscopy (cryo-EM) to explore how TRPV2 activity is modulated by the phytocannabinoid Δ9-tetrahydrocannabiorcol (C16) and by probenecid. C16 and probenecid act in concert to stimulate TRPV2 responses including histamine release from rat and human mast cells. Each ligand causes distinct conformational changes in TRPV2 as revealed by cryo-EM. Although the binding for probenecid remains elusive, C16 associates within the vanilloid pocket. As such, the C16 binding location is distinct from that of cannabidiol, partially overlapping with the binding site of the TRPV2 inhibitor piperlongumine. Taken together, we discover a new cannabinoid binding site in TRPV2 that is under the influence of allosteric control by probenecid. This molecular insight into ligand modulation enhances our understanding of TRPV2 in normal and pathophysiology.


Subject(s)
Cannabidiol , Cannabinoids , Rats , Humans , Animals , Cannabidiol/pharmacology , TRPV Cation Channels/metabolism , Cannabinoids/pharmacology , Probenecid/pharmacology , Ligands , Cryoelectron Microscopy
3.
Nat Commun ; 13(1): 6113, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36253390

ABSTRACT

TRP channels sense temperatures ranging from noxious cold to noxious heat. Whether specialized TRP thermosensor modules exist and how they control channel pore gating is unknown. We studied purified human TRPA1 (hTRPA1) truncated proteins to gain insight into the temperature gating of hTRPA1. In patch-clamp bilayer recordings, ∆1-688 hTRPA1, without the N-terminal ankyrin repeat domain (N-ARD), was more sensitive to cold and heat, whereas ∆1-854 hTRPA1, also lacking the S1-S4 voltage sensing-like domain (VSLD), gained sensitivity to cold but lost its heat sensitivity. In hTRPA1 intrinsic tryptophan fluorescence studies, cold and heat evoked rearrangement of VSLD and the C-terminus domain distal to the transmembrane pore domain S5-S6 (CTD). In whole-cell electrophysiology experiments, replacement of the CTD located cysteines 1021 and 1025 with alanine modulated hTRPA1 cold responses. It is proposed that hTRPA1 CTD harbors cold and heat sensitive domains allosterically coupled to the S5-S6 pore region and the VSLD, respectively.


Subject(s)
Ankyrin Repeat , Hot Temperature , Alanine , Humans , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/metabolism , Thermosensing , Tryptophan
4.
Biomed Pharmacother ; 152: 113262, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35691156

ABSTRACT

Transient receptor potential canonical 5 (TRPC5) is a polymodal, calcium-permeable, nonselective ion channel that is expressed in the brain and 75 % of human sensory neurons. Its pharmacological or genetic inhibition leads to the relief of neuropathic and inflammatory pain. The clinically approved drug duloxetine is superior to other serotonin and norepinephrine reuptake inhibitors at managing painful neuropathies, but it is not known why. Here we ask whether the TRPC5 receptor is modulated by duloxetine and may contribute to its analgesic effect. Electrophysiological measurements of heterologously expressed human TRPC5 in HEK293T cells were performed to evaluate the effect of duloxetine. The interaction site was identified by molecular docking and molecular dynamics simulations in combination with point mutagenesis. We found that duloxetine inhibits TRPC5 in a concentration-dependent manner with a high potency (IC50 = 0.54 ± 0.03 µM). Our data suggest that duloxetine binds into a voltage sensor-like domain. For the interaction, Glu418 exhibited particular importance due to putative hydrogen bond formation. Duloxetine effectively inhibits TRPC5 currents induced by cooling, voltage, direct agonists and by the stimulation of the PLC pathway. The finding that this TRPC5 inhibitor is widely used and well tolerated provides a scaffold for new pain treatment strategies.


Subject(s)
Pain , TRPC Cation Channels , Duloxetine Hydrochloride/pharmacology , HEK293 Cells , Humans , Molecular Docking Simulation , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
5.
J Cell Physiol ; 237(9): 3614-3626, 2022 09.
Article in English | MEDLINE | ID: mdl-35762104

ABSTRACT

The human transient receptor potential canonical 5 (TRPC5) is a calcium-permeable, nonselective cation channel expressed in the central and peripheral nervous system and also in other tissues such as the kidney, synovium, and odontoblasts. TRPC5 has been recently confirmed to play a key role in spontaneous, inflammatory mechanical, and cold pain. Although TRPC5 activation is known to be cold sensitive, it is unclear whether this property is intrinsic to the channel protein and whether or to what extent it may be determined by the cellular environment. In this study, we explored the cold sensitivity of human TRPC5 at the single-channel level using transiently transfected HEK293T cells. Upon decreasing the temperature, the channel demonstrated prolonged mean open dwell times and a robust increase in the open probability (Po ), whereas the amplitude of unitary currents decreased ~1.5-fold per 10°C of temperature difference. In the absence of any agonists, the temperature dependence of Po was sigmoidal, with a steep slope within the temperature range of 16°C-11°C, and exhibited saturation below 8-5°C. Thermodynamic analysis revealed significant changes in enthalpy and entropy, suggesting that substantial conformational changes accompany cold-induced gating. The mutant channel T970A, in which the regulation downstream of G-protein coupled receptor signaling was abrogated, exhibited higher basal activity at room temperature and a less steep temperature response profile, with an apparent threshold below 22°C. An even more pronounced decrease in the activation threshold was observed in a mutant that disrupted the electrostatic interaction of TRPC5 with the endoplasmic reticulum calcium sensor stromal interaction molecule 1. Thus, TRPC5 exhibits features of an intrinsically cold-gated channel; its sensitivity to cold tightly depends on the phosphorylation status of the protein and intracellular calcium homeostasis.


Subject(s)
Calcium , TRPC Cation Channels , Calcium/metabolism , Calcium Channels/metabolism , Cell Membrane/metabolism , HEK293 Cells , Humans , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
6.
Nat Struct Mol Biol ; 28(7): 564-572, 2021 07.
Article in English | MEDLINE | ID: mdl-34239124

ABSTRACT

Numerous physiological functions rely on distinguishing temperature through temperature-sensitive transient receptor potential channels (thermo-TRPs). Although the function of thermo-TRPs has been studied extensively, structural determination of their heat- and cold-activated states has remained a challenge. Here, we present cryo-EM structures of the nanodisc-reconstituted wild-type mouse TRPV3 in three distinct conformations: closed, heat-activated sensitized and open states. The heat-induced transformations of TRPV3 are accompanied by changes in the secondary structure of the S2-S3 linker and the N and C termini and represent a conformational wave that links these parts of the protein to a lipid occupying the vanilloid binding site. State-dependent differences in the behavior of bound lipids suggest their active role in thermo-TRP temperature-dependent gating. Our structural data, supported by physiological recordings and molecular dynamics simulations, provide an insight for understanding the molecular mechanism of temperature sensing.


Subject(s)
TRPV Cation Channels/metabolism , Thermosensing/physiology , Animals , Cell Line , Cold Temperature , Cryoelectron Microscopy , HEK293 Cells , Hot Temperature , Humans , Ion Channel Gating , Lipids/chemistry , Mice , Protein Binding/physiology , Protein Conformation , Thermodynamics
7.
Sci Adv ; 7(13)2021 03.
Article in English | MEDLINE | ID: mdl-33771873

ABSTRACT

Teeth are composed of many tissues, covered by an inflexible and obdurate enamel. Unlike most other tissues, teeth become extremely cold sensitive when inflamed. The mechanisms of this cold sensation are not understood. Here, we clarify the molecular and cellular components of the dental cold sensing system and show that sensory transduction of cold stimuli in teeth requires odontoblasts. TRPC5 is a cold sensor in healthy teeth and, with TRPA1, is sufficient for cold sensing. The odontoblast appears as the direct site of TRPC5 cold transduction and provides a mechanism for prolonged cold sensing via TRPC5's relative sensitivity to intracellular calcium and lack of desensitization. Our data provide concrete functional evidence that equipping odontoblasts with the cold-sensor TRPC5 expands traditional odontoblast functions and renders it a previously unknown integral cellular component of the dental cold sensing system.

8.
Int J Mol Sci ; 21(21)2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33121177

ABSTRACT

The Transient Receptor Potential Ankyrin 1 (TRPA1) channel is an integrative molecular sensor for detecting environmental irritant compounds, endogenous proalgesic and inflammatory agents, pressure, and temperature. Different post-translational modifications participate in the discrimination of the essential functions of TRPA1 in its physiological environment, but the underlying structural bases are poorly understood. Here, we explored the role of the cytosolic N-terminal residue Ser602 located near a functionally important allosteric coupling domain as a potential target of phosphorylation. The phosphomimetic mutation S602D completely abrogated channel activation, whereas the phosphonull mutations S602G and S602N produced a fully functional channel. Using mutagenesis, electrophysiology, and molecular simulations, we investigated the possible structural impact of a modification (mutation or phosphorylation) of Ser602 and found that this residue represents an important regulatory site through which the intracellular signaling cascades may act to reversibly restrict or "dampen" the conformational space of the TRPA1 channel and promote its transitions to the closed state.


Subject(s)
Mutation , Serine/metabolism , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/metabolism , HEK293 Cells , Humans , Models, Molecular , Molecular Dynamics Simulation , Phosphorylation , Protein Conformation , Protein Domains , TRPA1 Cation Channel/genetics
9.
Front Physiol ; 11: 189, 2020.
Article in English | MEDLINE | ID: mdl-32226391

ABSTRACT

Our understanding of the general principles of the polymodal regulation of transient receptor potential (TRP) ion channels has grown impressively in recent years as a result of intense efforts in protein structure determination by cryo-electron microscopy. In particular, the high-resolution structures of various TRP channels captured in different conformations, a number of them determined in a membrane mimetic environment, have yielded valuable insights into their architecture, gating properties and the sites of their interactions with annular and regulatory lipids. The correct repertoire of these channels is, however, organized by supramolecular complexes that involve the localization of signaling proteins to sites of action, ensuring the specificity and speed of signal transduction events. As such, TRP ankyrin 1 (TRPA1), a major player involved in various pain conditions, localizes into cholesterol-rich sensory membrane microdomains, physically interacts with calmodulin, associates with the scaffolding A-kinase anchoring protein (AKAP) and forms functional complexes with the related TRPV1 channel. This perspective will contextualize the recent biochemical and functional studies with emerging structural data with the aim of enabling a more thorough interpretation of the results, which may ultimately help to understand the roles of TRPA1 under various physiological and pathophysiological pain conditions. We demonstrate that an alteration to the putative lipid-binding site containing a residue polymorphism associated with human asthma affects the cold sensitivity of TRPA1. Moreover, we present evidence that TRPA1 can interact with AKAP to prime the channel for opening. The structural bases underlying these interactions remain unclear and are definitely worth the attention of future studies.

10.
Cells ; 9(1)2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31878344

ABSTRACT

Transient receptor potential ankyrin 1 channel (TRPA1) serves as a key sensor for reactive electrophilic compounds across all species. Its sensitivity to temperature, however, differs among species, a variability that has been attributed to an evolutionary divergence. Mouse TRPA1 was implicated in noxious cold detection but was later also identified as one of the prime noxious heat sensors. Moreover, human TRPA1, originally considered to be temperature-insensitive, turned out to act as an intrinsic bidirectional thermosensor that is capable of sensing both cold and heat. Using electrophysiology and modeling, we compare the properties of human and mouse TRPA1, and we demonstrate that both orthologues are activated by heat, and their kinetically distinct components of voltage-dependent gating are differentially modulated by heat and cold. Furthermore, we show that both orthologues can be strongly activated by cold after the concurrent application of voltage and heat. We propose an allosteric mechanism that could account for the variability in TRPA1 temperature responsiveness.


Subject(s)
TRPA1 Cation Channel/metabolism , Amino Acid Sequence , Animals , Cold Temperature , Electrophysiology/methods , HEK293 Cells , Hot Temperature , Humans , Mice , Models, Biological , Species Specificity , Voltage-Dependent Anion Channels/metabolism , Voltage-Dependent Anion Channels/physiology
11.
Int J Mol Sci ; 20(16)2019 Aug 16.
Article in English | MEDLINE | ID: mdl-31426314

ABSTRACT

The vanilloid transient receptor potential channel TRPV3 is a putative molecular thermosensor widely considered to be involved in cutaneous sensation, skin homeostasis, nociception, and pruritus. Repeated stimulation of TRPV3 by high temperatures above 50 °C progressively increases its responses and shifts the activation threshold to physiological temperatures. This use-dependence does not occur in the related heat-sensitive TRPV1 channel in which responses decrease, and the activation threshold is retained above 40 °C during activations. By combining structure-based mutagenesis, electrophysiology, and molecular modeling, we showed that chimeric replacement of the residues from the TRPV3 cytoplasmic inter-subunit interface (N251-E257) with the homologous residues of TRPV1 resulted in channels that, similarly to TRPV1, exhibited a lowered thermal threshold, were sensitized, and failed to close completely after intense stimulation. Crosslinking of this interface by the engineered disulfide bridge between substituted cysteines F259C and V385C (or, to a lesser extent, Y382C) locked the channel in an open state. On the other hand, mutation of a single residue within this region (E736) resulted in heat resistant channels. We propose that alterations in the cytoplasmic inter-subunit interface produce shifts in the channel gating equilibrium and that this domain is critical for the use-dependence of the heat sensitivity of TRPV3.


Subject(s)
Cytoplasm/metabolism , TRPV Cation Channels/metabolism , HEK293 Cells , Hot Temperature , Humans , Molecular Dynamics Simulation , Mutation , Protein Domains , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics
12.
FEBS J ; 286(18): 3664-3683, 2019 09.
Article in English | MEDLINE | ID: mdl-31116904

ABSTRACT

The transient receptor potential ankyrin 1 (TRPA1) channel is a polymodal sensor of environmental irritant compounds, endogenous proalgesic agents, and cold. Upon activation, TRPA1 channels increase cellular calcium levels via direct permeation and trigger signaling pathways that hydrolyze phosphatidylinositol-4,5-bisphosphate (PIP2 ) in the inner membrane leaflet. Our objective was to determine the extent to which a putative PIP2 -interaction site (Y1006-Q1031) is involved in TRPA1 regulation. The interactions of two specific peptides (L992-N1008 and T1003-P1034) with model lipid membranes were characterized by biophysical approaches to obtain information about affinity, peptide secondary structure, and peptide effect in the lipid organization. The results indicate that the two peptides interact with lipid membranes only if PIP2 is present and their affinities depend on the presence of calcium. Using whole-cell electrophysiology, we demonstrate that mutation at F1020 produced channels with faster activation kinetics and with a rightward shifted voltage-dependent activation curve by altering the allosteric constant that couples voltage sensing to pore opening. We assert that the presence of PIP2 is essential for the interaction of the two peptide sequences with the lipid membrane. The putative phosphoinositide-interacting domain comprising the highly conserved F1020 contributes to the stabilization of the TRPA1 channel gate.


Subject(s)
Lipid Metabolism/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipids/chemistry , TRPA1 Cation Channel/chemistry , Biophysical Phenomena , Calcium/chemistry , HEK293 Cells , Humans , Kinetics , Membrane Potentials/genetics , Peptides/chemistry , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phospholipids/metabolism , Protein Structure, Secondary , Signal Transduction/genetics , TRPA1 Cation Channel/genetics
13.
J Gen Physiol ; 150(8): 1125-1144, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29970412

ABSTRACT

Damage-sensing nociceptors in the skin provide an indispensable protective function thanks to their specialized ability to detect and transmit hot temperatures that would block or inflict irreversible damage in other mammalian neurons. Here we show that the exceptional capacity of skin C-fiber nociceptors to encode noxiously hot temperatures depends on two tetrodotoxin (TTX)-resistant sodium channel α-subunits: NaV1.8 and NaV1.9. We demonstrate that NaV1.9, which is commonly considered an amplifier of subthreshold depolarizations at 20°C, undergoes a large gain of function when temperatures rise to the pain threshold. We also show that this gain of function renders NaV1.9 capable of generating action potentials with a clear inflection point and positive overshoot. In the skin, heat-resistant nociceptors appear as two distinct types with unique and possibly specialized features: one is blocked by TTX and relies on NaV1.9, and the second type is insensitive to TTX and composed of both NaV1.8 and NaV1.9. Independent of rapidly gated TTX-sensitive NaV channels that form the action potential at pain threshold, NaV1.8 is required in all heat-resistant nociceptors to encode temperatures higher than ∼46°C, whereas NaV1.9 is crucial for shaping the action potential upstroke and keeping the NaV1.8 voltage threshold within reach.


Subject(s)
Hot Temperature , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.9 Voltage-Gated Sodium Channel/metabolism , Nociceptors/metabolism , Action Potentials , Animals , Cell Line , Evolution, Molecular , In Vitro Techniques , Mice, Inbred C57BL , Pain Threshold , Patch-Clamp Techniques , Skin
14.
Sci Signal ; 11(514)2018 01 23.
Article in English | MEDLINE | ID: mdl-29363587

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is a temperature-sensitive ion channel activated by various pungent and irritant compounds that can produce pain in humans. Its activation involves an allosteric mechanism whereby electrophilic agonists evoke interactions within cytosolic domains and open the channel pore through an integrated nexus formed by intracellular membrane proximal regions that are densely packed beneath the lower segment of the S1-S4 sensor domain. Studies indicate that this part of the channel may contain residues that form a water-accessible cavity that undergoes changes in solvation during channel gating. We identified conserved polar residues facing the putative lower crevice of the sensor domain that were crucial determinants of the electrophilic, voltage, and calcium sensitivity of the TRPA1 channel. This part of the sensor may also comprise a domain capable of binding to membrane phosphoinositides through which gating of the channel is regulated in a state-dependent manner.


Subject(s)
Calcium/metabolism , Ion Channel Gating , Membrane Potentials , TRPA1 Cation Channel/physiology , Allosteric Regulation , Amino Acid Sequence , HEK293 Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Domains , Sequence Homology , TRPA1 Cation Channel/chemistry
15.
J Cell Mol Med ; 22(2): 1355-1362, 2018 02.
Article in English | MEDLINE | ID: mdl-29210178

ABSTRACT

Exposure to repetitive low-frequency electromagnetic field (LF-EMF) shows promise as a non-invasive approach to treat various sensory and neurological disorders. Despite considerable progress in the development of modern stimulation devices, there is a limited understanding of the mechanisms underlying their biological effects and potential targets at the cellular level. A significant impact of electromagnetic field on voltage-gated calcium channels and downstream signalling pathways has been convincingly demonstrated in many distinct cell types. However, evidence for clear effects on primary sensory neurons that particularly may be responsible for the analgesic actions of LF-EMF is still lacking. Here, we used F11 cells derived from dorsal root ganglia neurons as an in vitro model of peripheral sensory neurons and three different protocols of high-induction magnetic stimulation to determine the effects on chemical responsiveness and spontaneous activity. We show that short-term (<180 sec.) exposure of F11 cells to LF-EMF reduces calcium transients in response to bradykinin, a potent pain-producing inflammatory agent formed at sites of injury. Moreover, we characterize an immediate and reversible potentiating effect of LF-EMF on neuronal spontaneous activity. Our results provide new evidence that electromagnetic field may directly modulate the activity of sensory neurons and highlight the potential of sensory neuron-derived cell line as a tool for studying the underlying mechanisms at the cellular and molecular level.


Subject(s)
Electromagnetic Fields , Sensory Receptor Cells/metabolism , Bradykinin/pharmacology , Calcium/metabolism , Cell Line , Humans , Sensory Receptor Cells/drug effects , TRPA1 Cation Channel/metabolism
16.
J Biol Chem ; 292(51): 21083-21091, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29084846

ABSTRACT

The transient receptor potential vanilloid 3 (TRPV3) channel is a Ca2+-permeable thermosensitive ion channel widely expressed in keratinocytes, where together with epidermal growth factor receptor (EGFR) forms a signaling complex regulating epidermal homeostasis. Proper signaling through this complex is achieved and maintained via several pathways in which TRPV3 activation is absolutely required. Results of recent studies have suggested that low-level constitutive activity of TRPV3 induces EGFR-dependent signaling that, in turn, amplifies TRPV3 via activation of the mitogen-activated protein kinase ERK in a positive feedback loop. Here, we explored the molecular mechanism that increases TRPV3 activity through EGFR activation. We used mutagenesis and whole-cell patch clamp experiments on TRPV3 channels endogenously expressed in an immortalized human keratinocyte cell line (HaCaT) and in transiently transfected HEK293T cells and found that the sensitizing effect of EGFR on TRPV3 is mediated by ERK. We observed that ERK-mediated phosphorylation of TRPV3 alters its responsiveness to repeated chemical stimuli. Among several putative ERK phosphorylation sites, we identified threonine 264 in the N-terminal ankyrin repeat domain as the most critical site for the ERK-dependent modulation of TRPV3 channel activity. Of note, Thr264 is in close vicinity to a structurally and functionally important TRPV3 region comprising an atypical finger 3 and oxygen-dependent hydroxylation site. In summary, our findings indicate that Thr264 in TRPV3 is a key ERK phosphorylation site mediating EGFR-induced sensitization of the channel to stimulate signaling pathways involved in regulating skin homeostasis.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/agonists , Keratinocytes/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 3/metabolism , TRPV Cation Channels/metabolism , Up-Regulation , Boron Compounds/pharmacology , Cell Line, Transformed , Cymenes , ErbB Receptors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Keratinocytes/drug effects , Keratinocytes/enzymology , MAP Kinase Signaling System/drug effects , Membrane Transport Modulators/pharmacology , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/genetics , Monoterpenes/pharmacology , Mutagenesis, Site-Directed , Mutation , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics , Threonine/metabolism , Up-Regulation/drug effects
17.
Front Mol Neurosci ; 10: 16, 2017.
Article in English | MEDLINE | ID: mdl-28197074

ABSTRACT

Transient receptor potential ankyrin 1 (TRPA1) is an excitatory ion channel involved in pain, inflammation and itching. This channel gates in response to many irritant and proalgesic agents, and can be modulated by calcium and depolarizing voltage. While the closed-state structure of TRPA1 has been recently resolved, also having its open state is essential for understanding how this channel works. Here we use molecular dynamics simulations combined with electrophysiological measurements and systematic mutagenesis to predict and explore the conformational changes coupled to the expansion of the presumptive channel's lower gate. We show that, upon opening, the upper part of the sensor module approaches the pore domain of an adjacent subunit and the conformational dynamics of the first extracellular flexible loop may govern the voltage-dependence of multimodal gating, thereby serving to stabilize the open state of the channel. These results are generally important in understanding the structure and function of TRPA1 and offer new insights into the gating mechanism of TRPA1 and related channels.

18.
Sci Rep ; 6: 28700, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27345869

ABSTRACT

Human transient receptor potential ankyrin channel 1 (TRPA1) is a polymodal sensor implicated in pain, inflammation and itching. An important locus for TRPA1 regulation is the cytoplasmic N-terminal domain, through which various exogenous electrophilic compounds such as allyl-isothiocyanate from mustard oil or cinnamaldehyde from cinnamon activate primary afferent nociceptors. This major region is comprised of a tandem set of 17 ankyrin repeats (AR1-AR17), five of them contain a strictly conserved T/SPLH tetrapeptide motif, a hallmark of an important and evolutionarily conserved contribution to conformational stability. Here, we characterize the functional consequences of putatively stabilizing and destabilizing mutations in these important structural units and identify AR2, AR6, and AR11-13 to be distinctly involved in the allosteric activation of TRPA1 by chemical irritants, cytoplasmic calcium, and membrane voltage. Considering the potential involvement of the T/SP motifs as putative phosphorylation sites, we also show that proline-directed Ser/Thr kinase CDK5 modulates the activity of TRPA1, and that T673 outside the AR-domain is its only possible target. Our data suggest that the most strictly conserved N-terminal ARs define the energetics of the TRPA1 channel gate and contribute to chemical-, calcium- and voltage-dependence.


Subject(s)
Ion Channel Gating/drug effects , Oligopeptides/pharmacology , TRPA1 Cation Channel/metabolism , Allosteric Regulation , Ankyrin Repeat , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , HEK293 Cells , Humans , Ion Channel Gating/genetics , Mutation , Oligopeptides/chemistry , Protein Domains , TRPA1 Cation Channel/genetics
19.
Temperature (Austin) ; 3(1): 77-91, 2016.
Article in English | MEDLINE | ID: mdl-27227099

ABSTRACT

Currently available behavioral assays to quantify normal cold sensitivity, cold hypersensitivity and cold hyperalgesia in mice have betimes created conflicting results in the literature. Some only capture a limited spectrum of thermal experiences, others are prone to experimenter bias or are not sensitive enough to detect the contribution of ion channels to cold sensing because in mice smaller alterations in cold nociception do not manifest as frank behavioral changes. To overcome current limitations we have designed a novel device that is automated, provides a high degree of freedom, i.e. thermal choice, and eliminates experimenter bias. The device represents a thermal gradient assay designed as a circular running track. It allows discerning exploratory behavior from thermal selection behavior and provides increased accuracy by providing measured values in duplicate and by removing edge artifacts. Our custom-designed automated offline analysis by a blob detection algorithm is devoid of movement artifacts, removes light reflection artifacts and provides an internal quality control parameter which we validated. The assay delivers discrete information on a large range of parameters extracted from the occupancy of thermally defined zones such as preference temperature and skew of the distribution. We demonstrate that the assay allows increasingly accurate phenotyping of thermal sensitivity in transgenic mice by disclosing yet unrecognized details on the phenotypes of TRPM8-, TRPA1- and TRPM8/A1-deficient mice.

20.
Neuropharmacology ; 93: 294-307, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25724085

ABSTRACT

The ankyrin transient receptor potential channel TRPA1 is a polymodal sensor for noxious stimuli, and hence a promising target for treating chronic pain. This tetrameric six-transmembrane segment (S1-S6) channel can be activated by various pungent chemicals, such as allyl isothiocyanate or cinnamaldehyde, but also by intracellular Ca(2+) or depolarizing voltages. Within the S4-S5 linker of human TRPA1, a gain-of-function mutation, N855S, was recently found to underlie familial episodic pain syndrome, manifested by bouts of severe upper body pain, triggered by physical stress, fasting, or cold. To clarify the structural basis for this channelopathy, we derive a structural model of TRPA1 by combining homology modeling, molecular dynamics simulations, point mutagenesis and electrophysiology. In the vicinity of N855, the model reveals inter-subunit salt bridges between E854 and K868. Using the heterologous expression of recombinant wild-type and mutant TRPA1 channels in HEK293T cells, we indeed found that the charge-reversal mutants E854R and K868E exhibited dramatically reduced responses to chemical and voltage stimuli, whereas the charge-swapping mutation E854R/K868E substantially rescued their functionalities. Moreover, mutation analysis of highly conserved charged residues within the S4-S5 region revealed a gain-of-function phenotype for R852E with an increased basal channel activity, a loss of Ca(2+)-induced potentiation and an accelerated Ca(2+)-dependent inactivation. Based on the model and on a comparison with the recently revealed atomic-level structure of the related channel TRPV1, we propose that inter-subunit salt bridges between adjacent S4-S5 regions are crucial for stabilizing the conformations associated with chemically and voltage-induced gating of the TRPA1 ion channel.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/genetics , Ion Channel Gating/physiology , Membrane Potentials/genetics , Models, Molecular , Mutation/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/genetics , Amino Acid Sequence , Animals , Asparagine/genetics , Calcium/metabolism , Calcium Channels/metabolism , Electric Stimulation , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Isothiocyanates/pharmacology , Mutagenesis , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary , Serine/genetics , TRPA1 Cation Channel , Transfection , Transient Receptor Potential Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...