Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Am J Physiol Cell Physiol ; 325(1): C42-C51, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37212545

ABSTRACT

Transient receptor potential channels canonical 1 and 4 (TRPC1 and TRPC4) are proteins belonging to the same TRPC channel family, and the two are known to form a heterotetrameric channel. TRPC4 can form a homotetrameric, nonselective cation channel by itself, but the involvement of the TRPC1 subunit changes several major characteristics of the channel. In this study, we focused on the pore region (selectivity filter, pore helix, and S6 helix) of TRPC1 and TRPC4 as a determinant of the identity and characteristics of a heteromeric TRPC1/4 channel: decreased calcium permeability of the channel and outward-rectifying current-voltage (I-V) curve. Mutants and chimeras of the pore residues were created, and their currents were recorded using whole cell patch clamp. The lower gate mutants of TRPC4 exhibited diminished calcium permeability as measured by GCaMP6 fluorescence. Also, chimeric channels substituting the pore region of TRPC1 to TRPC4 were made to locate the pore region that is critical in the production of an outward-rectifying I-V curve characteristic of TRPC1/4 heteromeric channels.NEW & NOTEWORTHY Heteromer research has been a challenging field due to lack of structural studies. Using chimeras and single mutants, we present evidence that the pore region of TRPC1/4 heteromer contributes to determining the channel's characteristics such as calcium permeability, I-V curve, and conductance.


Subject(s)
Protein Multimerization , Humans , HEK293 Cells , Models, Molecular , Protein Structure, Tertiary , Calcium/metabolism , TRPC Cation Channels/chemistry , Protein Structure, Quaternary , Ion Channel Gating , Cell Membrane/chemistry
2.
Cell Calcium ; 109: 102684, 2023 01.
Article in English | MEDLINE | ID: mdl-36495796

ABSTRACT

Recent structural studies have shown that the carboxyl-terminus of many TRP channels, including TRPC3, are folded into a horizontal rib helix that is connected to the vertical pole helix, which play roles in inter-structural interactions and multimerization. In a previous work we identified I807 located in the pole helix with a role in regulation of TRPC3 by STIM1 (Lee et al., 2014, Liu et al., 2022). To further determine the role of the pole helix in TRPC3 function, here we identified key hydrophobic residues in the pole helix that form tight tunnel-like structure and used mutations to probe their role in TRPC3 regulation by Ca2+ and Calmodulin. Our findings suggest that the hydrophobic starch formed by the I807-L818 residues has several roles, it modulates gating of TRPC3 by Ca2+, affects channel selectivity and the channel Ca2+ permeability. Mutations of I807, I811, L814 and L818 all attenuated the Ca2+-dependent inactivation (CDI) of TRPC3, with I807 having the most prominent effect. The extent of modulation of the CDI depended on the degree of hydrophobicity of I807. Moreover, the TRPC3(I807S) mutant showed altered channel monovalent ion selectivity and increased Ca2+ permeability, without affecting the channel permeability to Mg2+ and Ba2+ and without changing the pore diameter. The CDI of TRPC3 was reduced by an inactive calmodulin mutant and by a pharmacological inhibitor of calmodulin, which was eliminated by the I807S mutation. Notably, deletion of STIM1 caused similar alteration of TRPC3 properties. Taken together, these findings reveal a role of the pole helix in CDI, in addition to its potential role in channel multimerization that required gating of TRPC3 by STIM1. Since all TRPC and most TRP channels have pole helix structures, our findings raise the possibility that the pole helix may have similar roles in all the TRP family.


Subject(s)
Calcium Channels , Calcium , Calmodulin , TRPC Cation Channels , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Calmodulin/chemistry , Hydrophobic and Hydrophilic Interactions , Mutation , TRPC Cation Channels/genetics , TRPC Cation Channels/chemistry , Humans
3.
Biomolecules ; 12(7)2022 06 25.
Article in English | MEDLINE | ID: mdl-35883446

ABSTRACT

Transient receptor potential canonical 3 (TRPC3) channel belongs to the superfamily of transient receptor potential (TRP) channels which mediate Ca2+ influx into the cell. These channels constitute essential elements of cellular signalling and have been implicated in a wide range of diseases. TRPC3 is primarily gated by lipids and its surface expression has been shown to be dependent on cholesterol, yet a comprehensive exploration of its interaction with this lipid has thus far not emerged. Here, through 80 µs of coarse-grained molecular dynamics simulations, we show that cholesterol interacts with multiple elements of the transmembrane machinery of TRPC3. Through our approach, we identify an annular binding site for cholesterol on the pre-S1 helix and a non-annular site at the interface between the voltage-sensor-like domain and pore domains. Here, cholesterol interacts with exposed polar residues and possibly acts to stabilise the domain interface.


Subject(s)
Molecular Dynamics Simulation , TRPC Cation Channels , Binding Sites , Cholesterol , Protein Domains , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism
4.
Biochem Biophys Res Commun ; 594: 69-73, 2022 02 26.
Article in English | MEDLINE | ID: mdl-35074588

ABSTRACT

BacMam system utilizes baculovirus to deliver exogenous genes into mammalian cells and is extensively used for recombinant production of eukaryotic proteins. Here, we described the development of a BacMam vector (pBMCL1), which allows convenient tracing of virus production, provides higher infection efficiency towards mammalian cells, minimizes unwanted transcription of toxic genes in insect cells, and provides the capability for co-expression of multiple proteins via a single virus. We demonstrate the successful application of the pBMCL1 vector for the expression of homo-tetrameric human TRPC3 channel and hetero-octameric KATP channel.


Subject(s)
Baculoviridae/metabolism , Gene Expression Regulation , Animals , DNA, Complementary/metabolism , Electrophysiology , Gene Expression , Genes, Reporter , Genetic Vectors , HEK293 Cells , Humans , Insecta , Mesocricetus , Mice , Recombinant Proteins/chemistry , Sf9 Cells , TRPC Cation Channels/chemistry , Transduction, Genetic
5.
Structure ; 30(1): 139-155.e5, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34453887

ABSTRACT

Transient receptor potential (TRP) channels emerged in fungi as mechanosensitive osmoregulators. The Saccharomyces cerevisiae vacuolar TRP yeast 1 (TRPY1) is the most studied TRP channel from fungi, but the structure and details of channel modulation remain elusive. Here, we describe the full-length cryoelectron microscopy structure of TRPY1 at 3.1 Å resolution in a closed state. The structure, despite containing an evolutionarily conserved and archetypical transmembrane domain, reveals distinctive structural folds for the cytosolic N and C termini, compared with other eukaryotic TRP channels. We identify an inhibitory phosphatidylinositol 3-phosphate (PI(3)P) lipid-binding site, along with two Ca2+-binding sites: a cytosolic site, implicated in channel activation and a vacuolar lumen site, implicated in inhibition. These findings, together with data from microsecond-long molecular dynamics simulations and a model of a TRPY1 open state, provide insights into the basis of TRPY1 channel modulation by lipids and Ca2+, and the molecular evolution of TRP channels.


Subject(s)
Calcium/metabolism , Phosphatidylinositol Phosphates/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Binding Sites , Cryoelectron Microscopy , Cytosol/metabolism , Models, Molecular , Molecular Dynamics Simulation , Phosphatidylinositol Phosphates/chemistry , Protein Conformation , Protein Domains , Protein Multimerization , Saccharomyces cerevisiae/chemistry
6.
J Biol Chem ; 297(4): 101126, 2021 10.
Article in English | MEDLINE | ID: mdl-34461097

ABSTRACT

Transient receptor potential (TRP) cation channels, which are conserved across mammals, flies, fish, sea squirts, worms, and fungi, essentially contribute to cellular Ca2+ signaling. The activity of the unique TRP channel in yeast, TRP yeast channel 1 (TRPY1), relies on the vacuolar and cytoplasmic Ca2+ concentration. However, the mechanism(s) of Ca2+-dependent regulation of TRPY1 and possible contribution(s) of Ca2+-binding proteins are yet not well understood. Our results demonstrate a Ca2+-dependent binding of yeast calmodulin (CaM) to TRPY1. TRPY1 activity was increased in the cmd1-6 yeast strain, carrying a non-Ca2+-binding CaM mutant, compared with the parent strain expressing wt CaM (Cmd1). Expression of Cmd1 in cmd1-6 yeast rescued the wt phenotype. In addition, in human embryonic kidney 293 cells, hypertonic shock-induced TRPY1-dependent Ca2+ influx and Ca2+ release were increased by the CaM antagonist ophiobolin A. We found that coexpression of mammalian CaM impeded the activity of TRPY1 by reinforcing effects of endogenous CaM. Finally, inhibition of TRPY1 by Ca2+-CaM required the cytoplasmic amino acid stretch E33-Y92. In summary, our results show that TRPY1 is under inhibitory control of Ca2+-CaM and that mammalian CaM can replace yeast CaM for this inhibition. These findings add TRPY1 to the innumerable cellular proteins, which include a variety of ion channels, that use CaM as a constitutive or dissociable Ca2+-sensing subunit, and contribute to a better understanding of the modulatory mechanisms of Ca2+-CaM.


Subject(s)
Calcium Signaling , Calcium/metabolism , Calmodulin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , TRPC Cation Channels/metabolism , Vacuoles/metabolism , Calcium/chemistry , Calmodulin/antagonists & inhibitors , Calmodulin/chemistry , Calmodulin/genetics , HEK293 Cells , Humans , Protein Domains , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sesterterpenes/pharmacology , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics , Vacuoles/chemistry , Vacuoles/genetics
7.
Elife ; 102021 03 08.
Article in English | MEDLINE | ID: mdl-33683200

ABSTRACT

TRPC5 channel is a nonselective cation channel that participates in diverse physiological processes. TRPC5 inhibitors show promise in the treatment of anxiety disorder, depression, and kidney disease. However, the binding sites and inhibitory mechanism of TRPC5 inhibitors remain elusive. Here, we present the cryo-EM structures of human TRPC5 in complex with two distinct inhibitors, namely clemizole and HC-070, to the resolution of 2.7 Å. The structures reveal that clemizole binds inside the voltage sensor-like domain of each subunit. In contrast, HC-070 is wedged between adjacent subunits and replaces the glycerol group of a putative diacylglycerol molecule near the extracellular side. Moreover, we found mutations in the inhibitor binding pockets altered the potency of inhibitors. These structures suggest that both clemizole and HC-070 exert the inhibitory functions by stabilizing the ion channel in a nonconductive closed state. These results pave the way for further design and optimization of inhibitors targeting human TRPC5.


Subject(s)
Benzimidazoles/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/chemistry , Benzimidazoles/metabolism , Binding Sites , Heterocyclic Compounds, 4 or More Rings/metabolism , Humans , Models, Molecular , TRPC Cation Channels/metabolism
8.
Cell Calcium ; 95: 102361, 2021 05.
Article in English | MEDLINE | ID: mdl-33578200

ABSTRACT

TRPC4 ion channel was reported to be regulated by small molecular inhibitors and calmodulin. We discuss these findings in the context of other members of TRPC subfamily modulated by different stimulants.


Subject(s)
Calmodulin/metabolism , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , Calmodulin/pharmacology , Humans , Protein Structure, Secondary , TRPC Cation Channels/chemistry
9.
Structure ; 29(4): 330-344.e4, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33326749

ABSTRACT

Drosophila TRP is a calcium-permeable cation channel essential for fly visual signal transduction. During phototransduction, Ca2+ mediates both positive and negative feedback regulation on TRP channel activity, possibly via binding to calmodulin (CaM). However, the molecular mechanism underlying Ca2+ modulated CaM/TRP interaction is poorly understood. Here, we discover an unexpected, Ca2+-dependent binding mode between CaM and TRP. The TRP tail contains two CaM binding sites (CBS1 and CBS2) separated by an ∼70-residue linker. CBS1 binds to the CaM N-lobe and CBS2 recognizes the CaM C-lobe. Structural studies reveal the lobe-specific binding of CaM to CBS1&2. Mutations introduced in both CBS1 and CBS2 eliminated CaM binding in full-length TRP, but surprisingly had no effect on the response to light under physiological conditions, suggesting alternative mechanisms governing Ca2+-mediated feedback on the channel activity. Finally, we discover that TRPC4, the closest mammalian paralog of Drosophila TRP, adopts a similar CaM binding mode.


Subject(s)
Calmodulin/chemistry , Drosophila Proteins/chemistry , Transient Receptor Potential Channels/chemistry , Animals , Binding Sites , Calcium/metabolism , Calmodulin/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , HEK293 Cells , Humans , Mice , Mutation , Protein Binding , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
10.
Elife ; 92020 11 25.
Article in English | MEDLINE | ID: mdl-33236980

ABSTRACT

Canonical transient receptor potential channels (TRPC) are involved in receptor-operated and/or store-operated Ca2+ signaling. Inhibition of TRPCs by small molecules was shown to be promising in treating renal diseases. In cells, the channels are regulated by calmodulin (CaM). Molecular details of both CaM and drug binding have remained elusive so far. Here, we report structures of TRPC4 in complex with three pyridazinone-based inhibitors and CaM. The structures reveal that all the inhibitors bind to the same cavity of the voltage-sensing-like domain and allow us to describe how structural changes from the ligand-binding site can be transmitted to the central ion-conducting pore of TRPC4. CaM binds to the rib helix of TRPC4, which results in the ordering of a previously disordered region, fixing the channel in its closed conformation. This represents a novel CaM-induced regulatory mechanism of canonical TRP channels.


Subject(s)
Calmodulin/metabolism , Membrane Transport Modulators/pharmacology , Pyridazines/pharmacology , TRPC Cation Channels/drug effects , Zebrafish Proteins/drug effects , Animals , Binding Sites , Calmodulin/chemistry , Calmodulin/genetics , HEK293 Cells , Humans , Ligands , Membrane Potentials , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Pyridazines/chemistry , Pyridazines/metabolism , Sf9 Cells , Structure-Activity Relationship , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Xenopus , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
Commun Biol ; 3(1): 704, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230284

ABSTRACT

TRPC1/4/5 channels are non-specific cation channels implicated in a wide variety of diseases, and TRPC1/4/5 inhibitors have recently entered clinical trials. However, fundamental and translational studies require a better understanding of TRPC1/4/5 channel regulation by endogenous and exogenous factors. Although several potent and selective TRPC1/4/5 modulators have been reported, the paucity of mechanistic insights into their modes-of-action remains a barrier to the development of new chemical probes and drug candidates. Xanthine-based modulators include the most potent and selective TRPC1/4/5 inhibitors described to date, as well as TRPC5 activators. Our previous studies suggest that xanthines interact with a, so far, elusive pocket of TRPC1/4/5 channels that is essential to channel gating. Here we report the structure of a small-molecule-bound TRPC1/4/5 channel-human TRPC5 in complex with the xanthine Pico145-to 3.0 Å. We found that Pico145 binds to a conserved lipid binding site of TRPC5, where it displaces a bound phospholipid. Our findings explain the mode-of-action of xanthine-based TRPC1/4/5 modulators, and suggest a structural basis for TRPC1/4/5 modulation by endogenous factors such as (phospho)lipids and Zn2+ ions. These studies lay the foundations for the structure-based design of new generations of TRPC1/4/5 modulators.


Subject(s)
TRPC Cation Channels , Xanthines , Humans , Lipids/chemistry , Molecular Docking Simulation , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Xanthines/chemistry , Xanthines/metabolism
12.
ChemMedChem ; 15(19): 1854-1860, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32717096

ABSTRACT

The nonselective Ca2+ -permeable transient receptor potential channel subfamily member 5 (TRPC5) belongs to the transient receptor potential canonical (TRPC) superfamily and is widely expressed in the brain. Compelling evidence reveals that TRPC5 plays crucial roles in depression and other psychiatric disorders. To develop a TRPC5 radioligand, following up on our previous effort, we synthesized the iodine compound TZ66127 and its iodine-125-labeled counterpart [125 I]TZ66127. The synthesis of TZ66127 was achieved by replacing chloride with iodide in the structure of HC608, and the [125 I]TZ66127 was radiosynthesized using its corresponding tributylstannylated precursor. We established a stable human TRPC5-overexpressed HEK293-hTRPC5 cell line and performed Ca2+ imaging and a cell-binding assay study of TZ66127; these indicated that TZ66127 had good inhibition activity for TRPC5, and the inhibitory efficiency of TZ66127 toward TRPC5 presented in a dose-dependent manner. An in vitro autoradiography and immunohistochemistry study of rat brain sections suggested that [125 I]TZ66127 had binding specificity toward TRPC5. Altogether, [125 I]TZ66127 has high potential to serve as a radioligand for screening the binding activity of other new compounds toward TRPC5. The availability of [125 I]TZ66127 might facilitate the development of therapeutic drugs and PET imaging agents that target TRPC5.


Subject(s)
Radioligand Assay , Radiopharmaceuticals/pharmacology , TRPC Cation Channels/antagonists & inhibitors , Animals , Brain/metabolism , Calcium/analysis , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Iodine Radioisotopes , Molecular Structure , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Rats , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism
13.
Bioorg Med Chem ; 28(8): 115430, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32197812

ABSTRACT

The plausible nitric oxide (NO)-sensing module of TRPC5 was incorporated in a enhanced green fluorescent protein (EGFP) to evaluate its conformational change as an optical response upon the reaction with NO. Two cysteine residues located in the NO-sensing module have been proposed to form a disulfide bond through S-nitrosylation of the thiol group by NO. Modification of the cysteine residues by NO resulted a ratiometric change of EGFP emission through transducing the conformational change of NO-sensing module to the EGFP chromophore. The oxidized form of NO-sensing module fused EGFP changed the intensity of emission spectra upon reduction of the disulfide bond at the NO-reactive module. The NO-sensing module fused EGFP in its reduced form avidly reacted with NO and realized the ratiometric fluorescence intensity changes depending on the formation of disulfide bond. These results support the notion that NO induces a conformational change at the putative NO-sensing segment of TRPC5, and provide a prototype for the genetically encoded cellular NO sensors.


Subject(s)
Gene Expression Regulation/drug effects , Nitric Oxide/pharmacology , TRPC Cation Channels/metabolism , Escherichia coli , Green Fluorescent Proteins , Humans , Hydrogen Peroxide , Optical Imaging , Structure-Activity Relationship , TRPC Cation Channels/chemistry
14.
Pharmacol Ther ; 209: 107497, 2020 05.
Article in English | MEDLINE | ID: mdl-32004513

ABSTRACT

Transient receptor potential canonical (TRPC) channels constitute a group of receptor-operated calcium-permeable nonselective cation channels of the TRP superfamily. The seven mammalian TRPC members, which can be further divided into four subgroups (TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7) based on their amino acid sequences and functional similarities, contribute to a broad spectrum of cellular functions and physiological roles. Studies have revealed complexity of their regulation involving several components of the phospholipase C pathway, Gi and Go proteins, and internal Ca2+ stores. Recent advances in cryogenic electron microscopy have provided several high-resolution structures of TRPC channels. Growing evidence demonstrates the involvement of TRPC channels in diseases, particularly the link between genetic mutations of TRPC6 and familial focal segmental glomerulosclerosis. Because TRPCs were discovered by the molecular identity first, their pharmacology had lagged behind. This is rapidly changing in recent years owning to great efforts from both academia and industry. A number of potent tool compounds from both synthetic and natural products that selective target different subtypes of TRPC channels have been discovered, including some preclinical drug candidates. This review will cover recent advancements in the understanding of TRPC channel regulation, structure, and discovery of novel TRPC small molecular probes over the past few years, with the goal of facilitating drug discovery for the study of TRPCs and therapeutic development.


Subject(s)
Drug Discovery/trends , Molecular Probes/chemistry , Molecular Probes/physiology , TRPC Cation Channels/chemistry , TRPC Cation Channels/physiology , Animals , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Phosphoinositide Phospholipase C/chemistry , Phosphoinositide Phospholipase C/physiology , Protein Structure, Secondary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/physiology
15.
Cells ; 9(1)2020 01 07.
Article in English | MEDLINE | ID: mdl-31936014

ABSTRACT

: Transient Receptor Potential Canonical (TRPC) channels are homologues of Drosophila TRP channel first cloned in mammalian cells. TRPC family consists of seven members which are nonselective cation channels with a high Ca2+ permeability and are activated by a wide spectrum of stimuli. These channels are ubiquitously expressed in different tissues and organs in mammals and exert a variety of physiological functions. Post-translational modifications (PTMs) including phosphorylation, N-glycosylation, disulfide bond formation, ubiquitination, S-nitrosylation, S-glutathionylation, and acetylation play important roles in the modulation of channel gating, subcellular trafficking, protein-protein interaction, recycling, and protein architecture. PTMs also contribute to the polymodal activation of TRPCs and their subtle regulation in diverse physiological contexts and in pathological situations. Owing to their roles in the motor coordination and regulation of kidney podocyte structure, mutations of TRPCs have been implicated in diseases like cerebellar ataxia (moonwalker mice) and focal and segmental glomerulosclerosis (FSGS). The aim of this review is to comprehensively integrate all reported PTMs of TRPCs, to discuss their physiological/pathophysiological roles if available, and to summarize diseases linked to the natural mutations of TRPCs.


Subject(s)
Mutation/genetics , Protein Processing, Post-Translational , TRPC Cation Channels/genetics , Animals , Disease/genetics , Humans , Models, Biological , TRPC Cation Channels/chemistry
16.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118620, 2020 02.
Article in English | MEDLINE | ID: mdl-31812495

ABSTRACT

Protein S-palmitoylation, the covalent lipid modification of the side chain of Cys residues with the 16­carbon fatty acid palmitate, is the most common acylation, and it enhances the membrane stability of ion channels. This post-translational modification (PTM) determines a functional mechanism of ion channel life cycle from maturation and membrane trafficking to localization. Especially, neurodevelopment is regulated by balancing the level of synaptic protein palmitoylation/depalmitoylation. Recently, we revealed the pathological role of the transient receptor potential canonical type 5 (TRPC5) channel in striatal neuronal loss during Huntington's disease (HD), which is abnormally activated by oxidative stress. Here, we report a mechanism of TRPC5 palmitoylation at a conserved cysteine residue, that is critical for intrinsic channel activity. Furthermore, we identified the therapeutic effect of TRPC5 depalmitoylation by enhancing the TRPC5 membrane instability on HD striatal cells in order to lower TRPC5 toxicity. Collectively, these findings suggest that controlling S-palmitoylation of the TRPC5 channel as a potential risk factor can modulate TRPC5 channel expression and activity, providing new insights into a therapeutic strategy for neurodegenerative diseases.


Subject(s)
Neurons/metabolism , Oxidative Stress , TRPC Cation Channels/metabolism , Amino Acid Motifs , Animals , Antineoplastic Agents, Alkylating/toxicity , Apoptosis/drug effects , Carmustine/toxicity , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Lipoylation/drug effects , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Oxidative Stress/drug effects , Palmitates/pharmacology , Protein Stability , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics
17.
Channels (Austin) ; 13(1): 410-423, 2019 12.
Article in English | MEDLINE | ID: mdl-31603369

ABSTRACT

We have previously provided pharmacological evidence that stimulation of calcium-sensing receptors (CaSR) induces endothelium-dependent relaxations of rabbit mesenteric arteries through activation of heteromeric TRPV4/TRPC1 channels and nitric oxide (NO) production. The present study further investigates the role of heteromeric TRPV4/TRPC1 channels in these CaSR-induced vascular responses by comparing responses in mesenteric arteries from wild-type (WT) and TRPC1-/- mice. In WT mice, stimulation of CaSR induced endothelium-dependent relaxations of pre-contracted tone and NO generation in endothelial cells (ECs), which were inhibited by the TRPV4 channel blocker RN1734 and the TRPC1 blocking antibody T1E3. In addition, TRPV4 and TRPC1 proteins were colocalised at, or close to, the plasma membrane of endothelial cells (ECs) from WT mice. In contrast, in TRPC1-/- mice, CaSR-mediated vasorelaxations and NO generation were greatly reduced, unaffected by T1E3, but blocked by RN1734. In addition, the TRPV4 agonist GSK1016790A (GSK) induced endothelium-dependent vasorelaxations which were blocked by RN1734 and T1E3 in WT mice, but only by RN1734 in TRPC1-/- mice. Moreover, GSK activated cation channel activity with a 6pS conductance in WT ECs but with a 52 pS conductance in TRPC1-/- ECs. These results indicate that stimulation of CaSR activates heteromeric TRPV4/TRPC1 channels and NO production in ECs, which are responsible for endothelium-dependent vasorelaxations. This study also suggests that heteromeric TRPV4-TRPC1 channels may form the predominant TRPV4-containing channels in mouse mesenteric artery ECs. Together, our data further implicates CaSR-induced pathways and heteromeric TRPV4/TRPC1 channels in the regulation of vascular tone.


Subject(s)
Mesenteric Arteries/metabolism , Nitric Oxide/metabolism , Receptors, Calcium-Sensing/metabolism , TRPC Cation Channels/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Dimerization , Endothelial Cells/metabolism , Female , Male , Mice , Mice, Knockout , Rabbits , Receptors, Calcium-Sensing/genetics , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics , Vasodilation
18.
Curr Opin Struct Biol ; 58: 314-323, 2019 10.
Article in English | MEDLINE | ID: mdl-31378426

ABSTRACT

Transient Receptor Potential (TRP) channels are a large superfamily of polymodal ion channels, which perform important roles in numerous physiological processes. The architecture of their transmembrane (TM) domains closely resembles that of voltage-gated potassium channels (KV). However, recent cryoEM and crystallographic studies of TRP channels have identified π-helices in functionally important regions, and it is increasingly recognized that they utilize a distinct mechanism of gating that relies on α-to-π secondary structure transitions. Here we review our current understanding of the role of π-helices in TRP channel function and their broader impact on different classes of ion channels.


Subject(s)
Ion Channel Gating , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Animals , Conserved Sequence , Humans , Protein Conformation, alpha-Helical
19.
Sci Adv ; 5(7): eaaw7935, 2019 07.
Article in English | MEDLINE | ID: mdl-31355338

ABSTRACT

The transient receptor potential canonical subfamily member 5 (TRPC5), one of seven mammalian TRPC members, is a nonselective calcium-permeant cation channel. TRPC5 is of considerable interest as a drug target in the treatment of progressive kidney disease, depression, and anxiety. Here, we present the 2.8-Å resolution cryo-electron microscopy (cryo-EM) structure of the mouse TRPC5 (mTRPC5) homotetramer. Comparison of the TRPC5 structure to previously determined structures of other TRPC and TRP channels reveals differences in the extracellular pore domain and in the length of the S3 helix. The disulfide bond at the extracellular side of the pore and a preceding small loop are essential elements for its proper function. This high-resolution structure of mTRPC5, combined with electrophysiology and mutagenesis, provides insight into the lipid modulation and gating mechanisms of the TRPC family of ion channels.


Subject(s)
Conserved Sequence , Cryoelectron Microscopy , TRPC Cation Channels/metabolism , TRPC Cation Channels/ultrastructure , Animals , Binding Sites , Cations , Gadolinium/pharmacology , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Kinetics , Lanthanum/pharmacology , Lipids/chemistry , Mice , Mutation/genetics , Structure-Activity Relationship , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics
20.
Pflugers Arch ; 471(8): 1045-1053, 2019 08.
Article in English | MEDLINE | ID: mdl-31222490

ABSTRACT

Transient receptor potential canonical (TRPC) channels are calcium permeable, non-selective cation channels with wide tissue-specific distribution. Among 7 TRPC channels, TRPC 1/4/5 and TRPC3/6/7 are subdivided based on amino acid sequence homology. TRPC4 and TRPC5 channels exhibit cationic current with homotetrameric form, but they also form heterotetrameric channel such as TRPC1/4 or TRPC1/5 once TRPC1 is incorporated. The expression of TRPC1 is ubiquitous whereas the expressions of TRPC4 and TRPC5 are rather focused in nervous system. With the help of conditional knock-out of TPRC1, 4 and/or 5 genes, TRPC channels made of these constituents are reported to be involved in various pathophysiological functions such as seizure, anxiety-like behaviour, fear, Huntington's disease, Parkinson's disease and many others. In heterologous expression system, many issues such as activation mechanism, stoichiometry and relative cation permeabilites of homomeric or heteromeric channels have been addressed. In this review, we discussed the role of TRPC1 channel per se in plasma membrane, role of TRPC1 in heterotetrameric conformation (TRPC1/4 or TRPC1/5) and relationship between TRPC1/4/5 channels, calcium influx and voltage-gated calcium channels.


Subject(s)
Neurons/metabolism , TRPC Cation Channels/metabolism , Animals , Brain/cytology , Brain/metabolism , Humans , Membrane Potentials , Neurons/physiology , Protein Multimerization , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...