Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Calcium ; 123: 102927, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38959762

ABSTRACT

In order to understand protein function, the field of structural biology makes extensive use of cryogenic electron microscopy (cryo-EM), a technique that enables structure determination at atomic resolution following embedding of protein particles in vitreous ice. Considering the profound effects of temperature on macromolecule function, an important-but often neglected-question is how the frozen particles relate to the actual protein conformations at physiological temperatures. In a recent study, Hu et al. compare structures of the cation channel TRPM4 "frozen" at 4 °C versus 37 °C, revealing how temperature critically affects the binding of activating Ca2+ ions and other channel modulators.

2.
Sci Adv ; 9(31): eadg8842, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37531421

ABSTRACT

Host-derived succinate accumulates in the airways during bacterial infection. Here, we show that luminal succinate activates murine tracheal brush (tuft) cells through a signaling cascade involving the succinate receptor 1 (SUCNR1), phospholipase Cß2, and the cation channel transient receptor potential channel subfamily M member 5 (TRPM5). Stimulated brush cells then trigger a long-range Ca2+ wave spreading radially over the tracheal epithelium through a sequential signaling process. First, brush cells release acetylcholine, which excites nearby cells via muscarinic acetylcholine receptors. From there, the Ca2+ wave propagates through gap junction signaling, reaching also distant ciliated and secretory cells. These effector cells translate activation into enhanced ciliary activity and Cl- secretion, which are synergistic in boosting mucociliary clearance, the major innate defense mechanism of the airways. Our data establish tracheal brush cells as a central hub in triggering a global epithelial defense program in response to a danger-associated metabolite.


Subject(s)
Acetylcholine , Trachea , Mice , Animals , Trachea/metabolism , Signal Transduction , Succinates/metabolism , Epithelium/metabolism
3.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681657

ABSTRACT

BACKGROUND: The transient receptor potential ankyrin 1 (TRPA1) cation channels function as broadly-tuned sensors of noxious chemicals in many species. Recent studies identified four functional TRPA1 isoforms in Drosophila melanogaster (dTRPA1(A) to (D)), but their responses to non-electrophilic chemicals are yet to be fully characterized. METHODS: We determined the behavioral responses of adult flies to the mammalian TRPA1 non-electrophilic activators citronellal and menthol, and characterized the effects of these compounds on all four dTRPA1 channel isoforms using intracellular Ca2+ imaging and whole-cell patch-clamp recordings. RESULTS: Wild type flies avoided citronellal and menthol in an olfactory test and this behavior was reduced in dTrpA1 mutant flies. Both compounds activate all dTRPA1 isoforms in the heterologous expression system HEK293T, with the following sensitivity series: dTRPA1(C) = dTRPA1(D) > dTRPA1(A) ≫ dTRPA1(B) for citronellal and dTRPA1(A) > dTRPA1(D) > dTRPA1(C) > dTRPA1(B) for menthol. CONCLUSIONS: dTrpA1 was required for the normal avoidance of Drosophila melanogaster towards citronellal and menthol. All dTRPA1 isoforms are activated by both compounds, but the dTRPA1(B) is consistently the least sensitive. We discuss how these findings may guide further studies on the physiological roles and the structural bases of chemical sensitivity of TRPA1 channels.


Subject(s)
Acyclic Monoterpenes/pharmacology , Aldehydes/pharmacology , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Menthol/pharmacology , TRPA1 Cation Channel/metabolism , Animals , Animals, Genetically Modified/metabolism , Calcium/metabolism , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Female , HEK293 Cells , Humans , Insect Repellents/pharmacology , Male , Patch-Clamp Techniques , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , TRPA1 Cation Channel/deficiency , TRPA1 Cation Channel/genetics
4.
J Physiol ; 598(19): 4321-4338, 2020 10.
Article in English | MEDLINE | ID: mdl-32721035

ABSTRACT

KEY POINTS: 25-Hydroxyvitamin D (25OHD) is a partial agonist of TRPV1 whereby 25OHD can weakly activate TRPV1 yet antagonize the stimulatory effects of the full TRPV1 agonists capsaicin and oleoyl dopamine. 25OHD binds to TRPV1 within the same vanilloid binding pocket as capsaicin. 25OHD inhibits the potentiating effects of PKC-mediated TRPV1 activity. 25OHD reduces T-cell activation and trigeminal neuron calcium signalling mediated by TRPV1 activity. These results provide evidence that TRPV1 is a novel receptor for the biological actions of vitamin D in addition to the well-documented effects of vitamin D upon the nuclear vitamin D receptor. The results may have important implications for our current understanding of certain diseases where TRPV1 and vitamin D deficiency have been implicated, such as chronic pain and autoimmune diseases, such as type 1 diabetes. ABSTRACT: The capsaicin receptor TRPV1 plays an important role in nociception, inflammation and immunity and its activity is regulated by exogenous and endogenous lipophilic ligands. As vitamin D is lipophilic and involved in similar biological processes as TRPV1, we hypothesized that it directly regulates TRPV1 activity and function. Our calcium imaging and electrophysiological data demonstrate that vitamin D (25-hydroxyvitamin D (25OHD) and 1,25-hydroxyvitamin D (1,25OHD)) can weakly activate TRPV1 at physiologically relevant concentrations (100 nM). Furthermore, both 25OHD and 1,25OHD can inhibit capsaicin-induced TRPV1 activity (IC50  = 34.3 ± 0.2 and 11.5 ± 0.9 nM, respectively), but not pH-induced TRPV1 activity, suggesting that vitamin D interacts with TRPV1 in the same region as the TRPV1 agonist capsaicin. This hypothesis is supported by our in silico TRPV1 structural modelling studies, which place 25OHD in the same binding region as capsaicin. 25OHD also attenuates PKC-dependent TRPV1 potentiation via interactions with a known PKC phospho-acceptor residue in TRPV1. To provide evidence for a physiological role for the interaction of vitamin D with TRPV1, we employed two different cellular models known to express TRPV1: mouse CD4+ T-cells and trigeminal neurons. Our results indicate that 25OHD reduces TRPV1-induced cytokine release from T-cells and capsaicin-induced calcium activity in trigeminal neurons. In summary, we provide evidence that vitamin D is a novel endogenous regulator of TRPV1 channel activity that may play an important physiological role in addition to its known effects through the canonical nuclear vitamin D receptor pathway.


Subject(s)
Transient Receptor Potential Channels , Animals , Capsaicin/pharmacology , Mice , Neurons , Rats, Sprague-Dawley , TRPV Cation Channels , Vitamin D/pharmacology
5.
Immunity ; 52(4): 683-699.e11, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294408

ABSTRACT

Mucociliary clearance through coordinated ciliary beating is a major innate defense removing pathogens from the lower airways, but the pathogen sensing and downstream signaling mechanisms remain unclear. We identified virulence-associated formylated bacterial peptides that potently stimulated ciliary-driven transport in the mouse trachea. This innate response was independent of formyl peptide and taste receptors but depended on key taste transduction genes. Tracheal cholinergic chemosensory cells expressed these genes, and genetic ablation of these cells abrogated peptide-driven stimulation of mucociliary clearance. Trpm5-deficient mice were more susceptible to infection with a natural pathogen, and formylated bacterial peptides were detected in patients with chronic obstructive pulmonary disease. Optogenetics and peptide stimulation revealed that ciliary beating was driven by paracrine cholinergic signaling from chemosensory to ciliated cells operating through muscarinic M3 receptors independently of nerves. We provide a cellular and molecular framework that defines how tracheal chemosensory cells integrate chemosensation with innate defense.


Subject(s)
Acetylcholine/immunology , Bacterial Proteins/pharmacology , Cilia/immunology , Mucociliary Clearance/immunology , Pulmonary Disease, Chronic Obstructive/immunology , TRPM Cation Channels/immunology , Trachea/immunology , Acetylcholine/metabolism , Animals , Bacterial Proteins/immunology , Biological Transport , Cilia/drug effects , Cilia/metabolism , Female , Formates/metabolism , Gene Expression , Humans , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Optogenetics/methods , Paracrine Communication/immunology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , Taste Buds/immunology , Taste Buds/metabolism , Trachea/drug effects , Trachea/pathology , Virulence
6.
Physiol Rev ; 100(2): 725-803, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31670612

ABSTRACT

The transient receptor potential ankyrin (TRPA) channels are Ca2+-permeable nonselective cation channels remarkably conserved through the animal kingdom. Mammals have only one member, TRPA1, which is widely expressed in sensory neurons and in non-neuronal cells (such as epithelial cells and hair cells). TRPA1 owes its name to the presence of 14 ankyrin repeats located in the NH2 terminus of the channel, an unusual structural feature that may be relevant to its interactions with intracellular components. TRPA1 is primarily involved in the detection of an extremely wide variety of exogenous stimuli that may produce cellular damage. This includes a plethora of electrophilic compounds that interact with nucleophilic amino acid residues in the channel and many other chemically unrelated compounds whose only common feature seems to be their ability to partition in the plasma membrane. TRPA1 has been reported to be activated by cold, heat, and mechanical stimuli, and its function is modulated by multiple factors, including Ca2+, trace metals, pH, and reactive oxygen, nitrogen, and carbonyl species. TRPA1 is involved in acute and chronic pain as well as inflammation, plays key roles in the pathophysiology of nearly all organ systems, and is an attractive target for the treatment of related diseases. Here we review the current knowledge about the mammalian TRPA1 channel, linking its unique structure, widely tuned sensory properties, and complex regulation to its roles in multiple pathophysiological conditions.


Subject(s)
Calcium Signaling , Mechanotransduction, Cellular , Nociception , Sensory Receptor Cells/metabolism , TRPA1 Cation Channel/metabolism , Thermosensing , Animals , Channelopathies/metabolism , Channelopathies/physiopathology , Chemoreceptor Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/physiopathology , Mechanoreceptors/metabolism , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Thermoreceptors/metabolism
7.
Elife ; 82019 06 11.
Article in English | MEDLINE | ID: mdl-31184584

ABSTRACT

The cation channel TRPA1 transduces a myriad of noxious chemical stimuli into nociceptor electrical excitation and neuropeptide release, leading to pain and neurogenic inflammation. Despite emergent evidence that TRPA1 is regulated by the membrane environment, it remains unknown whether this channel localizes in membrane microdomains or whether it interacts with cholesterol. Using total internal reflection fluorescence microscopy and density gradient centrifugation we found that mouse TRPA1 localizes preferably into cholesterol-rich domains and functional experiments revealed that cholesterol depletion decreases channel sensitivity to chemical agonists. Moreover, we identified two structural motifs in transmembrane segments 2 and 4 involved in mTRPA1-cholesterol interactions that are necessary for normal agonist sensitivity and plasma membrane localization. We discuss the impact of such interactions on TRPA1 gating mechanisms, regulation by the lipid environment, and role of this channel in sensory membrane microdomains, all of which helps to understand the puzzling pharmacology and pathophysiology of this channel.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , TRPA1 Cation Channel/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cholesterol/chemistry , Cricetinae , Cricetulus , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Microdomains/metabolism , Mice , Microscopy, Fluorescence/methods , Models, Molecular , Protein Binding , Protein Domains , Sequence Homology, Amino Acid , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/genetics , Red Fluorescent Protein
8.
Int J Mol Sci ; 20(2)2019 01 16.
Article in English | MEDLINE | ID: mdl-30654572

ABSTRACT

Transient Receptor Potential ion channels (TRPs) have been described as polymodal sensors, being responsible for transducing a wide variety of stimuli, and being involved in sensory functions such as chemosensation, thermosensation, mechanosensation, and photosensation. Mechanical and chemical stresses exerted on the membrane can be transduced by specialized proteins into meaningful intracellular biochemical signaling, resulting in physiological changes. Of particular interest are compounds that can change the local physical properties of the membrane, thereby affecting nearby proteins, such as TRP channels, which are highly sensitive to the membrane environment. In this review, we provide an overview of the current knowledge of TRP channel activation as a result of changes in the membrane properties induced by amphipathic structural lipidic components such as cholesterol and diacylglycerol, and by exogenous amphipathic bacterial endotoxins.


Subject(s)
Cell Membrane/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Biomechanical Phenomena , Humans , Lipopolysaccharides/metabolism , Mechanotransduction, Cellular , Membrane Lipids/metabolism
9.
Toxins (Basel) ; 10(8)2018 08 11.
Article in English | MEDLINE | ID: mdl-30103489

ABSTRACT

The cellular and systemic effects induced by bacterial lipopolysaccharides (LPS) have been solely attributed to the activation of the Toll-like receptor 4 (TLR4) signalling cascade. However, recent studies have shown that LPS activates several members of the Transient Receptor Potential (TRP) family of cation channels. Indeed, LPS induces activation of the broadly-tuned chemosensor TRPA1 in sensory neurons in a TLR4-independent manner, and genetic ablation of this channel reduced mouse pain and inflammatory responses triggered by LPS and the gustatory-mediated avoidance to LPS in fruit flies. LPS was also shown to activate TRPV4 channels in airway epithelial cells, an effect leading to an immediate production of bactericidal nitric oxide and to an increase in ciliary beat frequency. In this review, we discuss the role of TRP channels as sensors of bacterial endotoxins, and therefore, as crucial players in the timely detection of invading gram-negative bacteria.


Subject(s)
Endotoxins/pharmacology , Transient Receptor Potential Channels/metabolism , Animals , Humans , Transient Receptor Potential Channels/chemistry
10.
Cell Calcium ; 73: 72-81, 2018 07.
Article in English | MEDLINE | ID: mdl-29689522

ABSTRACT

Acute neurogenic inflammation and pain associated to bacterial infection have been traditionally ascribed to sensitization and activation of sensory nerve afferents secondary to immune cell stimulation. However, we recently showed that lipopolysaccharides (LPS) directly activate the Transient Receptor Potential channels TRPA1 in sensory neurons and TRPV4 in airway epithelial cells. Here we investigated whether LPS activates other sensory TRP channels expressed in sensory neurons. Using intracellular Ca2+ imaging and patch-clamp we determined the effects of LPS on recombinant TRPV1, TRPV2, TRPM3 and TRPM8, heterologously expressed in HEK293T cells. We found that LPS activates TRPV1, although with lower potency than for TRPA1. Activation of TRPV1 by LPS was not affected by mutations of residues required for activation by electrophilic agents or by diacylglycerol and capsaicin. On the other hand, LPS weakly activated TRPM3, activated TRPM8 at 25 °C, but not at 35 °C, and was ineffective on TRPV2. Experiments performed in mouse dorsal root ganglion (DRG) neurons revealed that genetic ablation of Trpa1 did not abolish the responses to LPS, but remain detected in 30% of capsaicin-sensitive cells. The population of neurons responding to LPS was dramatically lower in double Trpa1/Trpv1 KO neurons. Our results show that, in addition to TRPA1, other TRP channels in sensory neurons can be targets of LPS, suggesting that they may contribute to trigger and regulate innate defenses against gram-negative bacterial infections.


Subject(s)
Ganglia, Spinal/metabolism , Lipopolysaccharides/pharmacology , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , TRPA1 Cation Channel/agonists , TRPV Cation Channels/agonists
11.
Nat Commun ; 8(1): 1059, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057902

ABSTRACT

Lipopolysaccharides (LPS), the major components of the wall of gram-negative bacteria, trigger powerful defensive responses in the airways via mechanisms thought to rely solely on the Toll-like receptor 4 (TLR4) immune pathway. Here we show that airway epithelial cells display an increase in intracellular Ca2+ concentration within seconds of LPS application. This response occurs in a TLR4-independent manner, via activation of the transient receptor potential vanilloid 4 cation channel (TRPV4). We found that TRPV4 mediates immediate LPS-induced increases in ciliary beat frequency and the production of bactericidal nitric oxide. Upon LPS challenge TRPV4-deficient mice display exacerbated ventilatory changes and recruitment of polymorphonuclear leukocytes into the airways. We conclude that LPS-induced activation of TRPV4 triggers signaling mechanisms that operate faster and independently from the canonical TLR4 immune pathway, leading to immediate protective responses such as direct antimicrobial action, increase in airway clearance, and the regulation of the inflammatory innate immune reaction.


Subject(s)
Calcium Signaling , Epithelial Cells/immunology , Lipopolysaccharides/immunology , Respiratory Mucosa/immunology , TRPV Cation Channels/metabolism , Animals , Cilia/physiology , Escherichia coli , HEK293 Cells , Humans , Immunity, Innate , Mice, Knockout , Nitric Oxide/metabolism , Patch-Clamp Techniques , Primary Cell Culture
12.
Elife ; 52016 06 14.
Article in English | MEDLINE | ID: mdl-27296646

ABSTRACT

Detecting pathogens and mounting immune responses upon infection is crucial for animal health. However, these responses come at a high metabolic price (McKean and Lazzaro, 2011, Kominsky et al., 2010), and avoiding pathogens before infection may be advantageous. The bacterial endotoxins lipopolysaccharides (LPS) are important immune system infection cues (Abbas et al., 2014), but it remains unknown whether animals possess sensory mechanisms to detect them prior to infection. Here we show that Drosophila melanogaster display strong aversive responses to LPS and that gustatory neurons expressing Gr66a bitter receptors mediate avoidance of LPS in feeding and egg laying assays. We found the expression of the chemosensory cation channel dTRPA1 in these cells to be necessary and sufficient for LPS avoidance. Furthermore, LPS stimulates Drosophila neurons in a TRPA1-dependent manner and activates exogenous dTRPA1 channels in human cells. Our findings demonstrate that flies detect bacterial endotoxins via a gustatory pathway through TRPA1 activation as conserved molecular mechanism.


Subject(s)
Drosophila Proteins/analysis , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Feeding Behavior , Lipopolysaccharides/metabolism , Neurons/physiology , Receptors, Cell Surface/analysis , TRPA1 Cation Channel/metabolism , Animals , Ion Channels , Neurons/chemistry , Neurons/drug effects
13.
Eur Respir J ; 48(2): 380-92, 2016 08.
Article in English | MEDLINE | ID: mdl-27126687

ABSTRACT

Asthma may be induced by chemical sensitisers, via mechanisms that are still poorly understood. This type of asthma is characterised by airway hyperreactivity (AHR) and little airway inflammation. Since potent chemical sensitisers, such as toluene-2,4-diisocyanate (TDI), are also sensory irritants, it is suggested that chemical-induced asthma relies on neuro-immune mechanisms.We investigated the involvement of transient receptor potential channels (TRP) A1 and V1, major chemosensors in the airways, and mast cells, known for their ability to communicate with sensory nerves, in chemical-induced AHR.In vitro intracellular calcium imaging and patch-clamp recordings in TRPA1- and TRPV1-expressing Chinese hamster ovarian cells showed that TDI activates murine TRPA1, but not TRPV1. Using an in vivo model, in which an airway challenge with TDI induces AHR in TDI-sensitised C57Bl/6 mice, we demonstrated that AHR does not develop, despite successful sensitisation, in Trpa1 and Trpv1 knockout mice, and wild-type mice pretreated with a TRPA1 blocker or a substance P receptor antagonist. TDI-induced AHR was also abolished in mast cell deficient Kit(Wsh) (/Wsh) mice, and in wild-type mice pretreated with the mast cell stabiliser ketotifen, without changes in immunological parameters.These data demonstrate that TRPA1, TRPV1 and mast cells play an indispensable role in the development of TDI-elicited AHR.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , Mast Cells/metabolism , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism , Animals , CHO Cells , Calcium/metabolism , Cricetulus , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Patch-Clamp Techniques , Sensory Receptor Cells/immunology , Toluene 2,4-Diisocyanate/chemistry
14.
Pflugers Arch ; 466(3): 507-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23955021

ABSTRACT

The powerful plant-derived irritant allyl isothiocyanate (AITC, aka mustard oil) induces hyperalgesia to heat in rodents and humans through mechanisms that are not yet fully understood. It is generally believed that AITC activates the broadly tuned chemosensory cation channel transient receptor potential cation channel subfamily A member 1 (TRPA1), triggering an inflammatory response that sensitizes the heat sensor transient receptor potential cation channel subfamily V member 1 (TRPV1). In the view of recent data demonstrating that AITC can directly activate TRPV1, we here explored the possibility that this compound sensitizes TRPV1 to heat stimulation in a TRPA1-independent manner. Patch-clamp recordings and intracellular Ca(2+) imaging experiments in HEK293T cells over-expressing mouse TRPV1 revealed that the increase in channel activation induced by heating is larger in the presence of AITC than in control conditions. The analysis of the effects of AITC and heat on the current-voltage relationship of TRPV1 indicates that the mechanism of sensitization is based on additive shifts of the voltage dependence of activation towards negative voltages. Finally, intracellular Ca(2+) imaging experiments in mouse sensory neurons isolated from Trpa1 KO mice yielded that AITC enhances the response to heat, specifically in the subpopulation expressing TRPV1. Furthermore, this effect was strongly reduced by the TRPV1 inhibitor capsazepine and virtually absent in neurons isolated from double Trpa1/Trpv1 KO mice. Taken together, these findings demonstrate that TRPV1 is a locus for cross sensitization between AITC and heat in sensory neurons and may help explaining, at least in part, the role of this channel in AITC-induced hyperalgesia to heat.


Subject(s)
Hot Temperature , Isothiocyanates/pharmacology , TRPV Cation Channels/metabolism , Thermosensing , Transient Receptor Potential Channels/metabolism , Action Potentials , Animals , Calcium/metabolism , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cells, Cultured , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , TRPA1 Cation Channel , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Transient Receptor Potential Channels/genetics
15.
Mol Pharmacol ; 84(3): 325-34, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23757176

ABSTRACT

Allyl isothiocyanate (AITC; aka, mustard oil) is a powerful irritant produced by Brassica plants as a defensive trait against herbivores and confers pungency to mustard and wasabi. AITC is widely used experimentally as an inducer of acute pain and neurogenic inflammation, which are largely mediated by the activation of nociceptive cation channels transient receptor potential ankyrin 1 and transient receptor potential vanilloid 1 (TRPV1). Although it is generally accepted that electrophilic agents activate these channels through covalent modification of cytosolic cysteine residues, the mechanism underlying TRPV1 activation by AITC remains unknown. Here we show that, surprisingly, AITC-induced activation of TRPV1 does not require interaction with cysteine residues, but is largely dependent on S513, a residue that is involved in capsaicin binding. Furthermore, AITC acts in a membrane-delimited manner and induces a shift of the voltage dependence of activation toward negative voltages, which is reminiscent of capsaicin effects. These data indicate that AITC acts through reversible interactions with the capsaicin binding site. In addition, we show that TRPV1 is a locus for cross-sensitization between AITC and acidosis in nociceptive neurons. Furthermore, we show that residue F660, which is known to determine the stimulation by low pH in human TRPV1, is also essential for the cross-sensitization of the effects of AITC and low pH. Taken together, these findings demonstrate that not all reactive electrophiles stimulate TRPV1 via cysteine modification and help understanding the molecular bases underlying the surprisingly large role of this channel as mediator of the algesic properties of AITC.


Subject(s)
Isothiocyanates/pharmacology , TRPV Cation Channels/metabolism , Animals , Binding Sites , Capsaicin/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Ganglia, Spinal/cytology , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Mutation , Patch-Clamp Techniques , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...