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










Publication year range
1.
Nat Commun ; 14(1): 1269, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36882406

ABSTRACT

PIEZO proteins are unusually large, mechanically-activated trimeric ion channels. The central pore features structural similarities with the pore of other trimeric ion channels, including purinergic P2X receptors, for which optical control of channel gating has been previously achieved with photoswitchable azobenzenes. Extension of these chemical optogenetics methods to mechanically-activated ion channels would provide tools for specific manipulation of pore activity alternative to non-specific mechanical stimulations. Here we report a light-gated mouse PIEZO1 channel, in which an azobenzene-based photoswitch covalently tethered to an engineered cysteine, Y2464C, localized at the extracellular apex of the transmembrane helix 38, rapidly triggers channel gating upon 365-nm-light irradiation. We provide evidence that this light-gated channel recapitulates mechanically-activated PIEZO1 functional properties, and show that light-induced molecular motions are similar to those evoked mechanically. These results push the limits of azobenzene-based methods to unusually large ion channels and provide a simple stimulation means to specifically interrogate PIEZO1 function.


Subject(s)
Azo Compounds , Cysteine , Animals , Mice , Motion , Optogenetics , Ion Channels
2.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207150

ABSTRACT

P2X7 receptors (P2X7) are cationic channels involved in many diseases. Following their activation by extracellular ATP, distinct signaling pathways are triggered, which lead to various physiological responses such as the secretion of pro-inflammatory cytokines or the modulation of cell death. P2X7 also exhibit unique behaviors, such as "macropore" formation, which corresponds to enhanced large molecule cell membrane permeability and current facilitation, which is caused by prolonged activation. These two phenomena have often been confounded but, thus far, no clear mechanisms have been resolved. Here, by combining different approaches including whole-cell and single-channel recordings, pharmacological and biochemical assays, CRISPR/Cas9 technology and cell imaging, we provide evidence that current facilitation and macropore formation involve functional complexes comprised of P2X7 and TMEM16, a family of Ca2+-activated ion channel/scramblases. We found that current facilitation results in an increase of functional complex-embedded P2X7 open probability, a result that is recapitulated by plasma membrane cholesterol depletion. We further show that macropore formation entails two distinct large molecule permeation components, one of which requires functional complexes featuring TMEM16F subtype, the other likely being direct permeation through the P2X7 pore itself. Such functional complexes can be considered to represent a regulatory hub that may orchestrate distinct P2X7 functionalities.


Subject(s)
Anoctamins/metabolism , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/metabolism , Algorithms , Animals , Anoctamins/chemistry , CRISPR-Cas Systems , Cell Membrane/metabolism , Cell Membrane Permeability , Cholesterol/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Models, Biological , Oocytes , Receptors, Purinergic P2X7/chemistry
3.
Proc Natl Acad Sci U S A ; 114(19): E3786-E3795, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28442564

ABSTRACT

Pore dilation is thought to be a hallmark of purinergic P2X receptors. The most commonly held view of this unusual process posits that under prolonged ATP exposure the ion pore expands in a striking manner from an initial small-cation conductive state to a dilated state, which allows the passage of larger synthetic cations, such as N-methyl-d-glucamine (NMDG+). However, this mechanism is controversial, and the identity of the natural large permeating cations remains elusive. Here, we provide evidence that, contrary to the time-dependent pore dilation model, ATP binding opens an NMDG+-permeable channel within milliseconds, with a conductance that remains stable over time. We show that the time course of NMDG+ permeability superimposes that of Na+ and demonstrate that the molecular motions leading to the permeation of NMDG+ are very similar to those that drive Na+ flow. We found, however, that NMDG+ "percolates" 10 times slower than Na+ in the open state, likely due to a conformational and orientational selection of permeating molecules. We further uncover that several P2X receptors, including those able to desensitize, are permeable not only to NMDG+ but also to spermidine, a large natural cation involved in ion channel modulation, revealing a previously unrecognized P2X-mediated signaling. Altogether, our data do not support a time-dependent dilation of the pore on its own but rather reveal that the open pore of P2X receptors is wide enough to allow the permeation of large organic cations, including natural ones. This permeation mechanism has considerable physiological significance.


Subject(s)
Cell Membrane Permeability , Glutamates/metabolism , Models, Biological , Receptors, Purinergic P2X/metabolism , Spermidine/metabolism , HEK293 Cells , Humans
4.
Elife ; 5: e11050, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26808983

ABSTRACT

P2X receptors function by opening a transmembrane pore in response to extracellular ATP. Recent crystal structures solved in apo and ATP-bound states revealed molecular motions of the extracellular domain following agonist binding. However, the mechanism of pore opening still remains controversial. Here we use photo-switchable cross-linkers as 'molecular tweezers' to monitor a series of inter-residue distances in the transmembrane domain of the P2X2 receptor during activation. These experimentally based structural constraints combined with computational studies provide high-resolution models of the channel in the open and closed states. We show that the extent of the outer pore expansion is significantly reduced compared to the ATP-bound structure. Our data further reveal that the inner and outer ends of adjacent pore-lining helices come closer during opening, likely through a hinge-bending motion. These results provide new insight into the gating mechanism of P2X receptors and establish a versatile strategy applicable to other membrane proteins.


Subject(s)
Adenosine Triphosphate/metabolism , Ion Channels/metabolism , Receptors, Purinergic P2X2/metabolism , Animals , Models, Biological , Molecular Dynamics Simulation , Optical Tweezers , Protein Conformation , Rats
5.
Proc Natl Acad Sci U S A ; 110(51): 20813-8, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297890

ABSTRACT

The powerful optogenetic pharmacology method allows the optical control of neuronal activity by photoswitchable ligands tethered to channels and receptors. However, this approach is technically demanding, as it requires the design of pharmacologically active ligands. The development of versatile technologies therefore represents a challenging issue. Here, we present optogating, a method in which the gating machinery of an ATP-activated P2X channel was reprogrammed to respond to light. We found that channels covalently modified by azobenzene-containing reagents at the transmembrane segments could be reversibly turned on and off by light, without the need of ATP, thus revealing an agonist-independent, light-induced gating mechanism. We demonstrate photocontrol of neuronal activity by a light-gated, ATP-insensitive P2X receptor, providing an original tool devoid of endogenous sensitivity to delineate P2X signaling in normal and pathological states. These findings open new avenues to specifically activate other ion channels independently of their natural stimulus.


Subject(s)
Azo Compounds/chemistry , Ion Channel Gating/radiation effects , Light , Neurons/metabolism , Receptors, Purinergic P2X/chemistry , Receptors, Purinergic P2X/metabolism , Animals , HEK293 Cells , Humans , Ion Channel Gating/genetics , Rats
6.
Channels (Austin) ; 6(5): 398-402, 2012.
Article in English | MEDLINE | ID: mdl-22992569

ABSTRACT

The molecular mechanism underlying channel opening in response to agonist binding remains a challenging issue in neuroscience. In this regard, many efforts have been recently undertaken in ATP-gated P2X receptors. Among those efforts, we have provided evidence in the P2X2 receptor that tightening of ATP sites upon agonist binding induces opening of the ion channel. Here we extend our analysis to show that the sulfhydryl-reactive ATP analog 8-thiocyano-ATP (NCS-ATP), a potent P2X2 agonist, when covalently labeled in the ATP-binding site at position Leu186 likely favors the tightening mechanism, but not the channel opening mechanism. Our data predict the existence of intermediate or preactivation state(s) trapped by NCS-ATP, in which tightening of the binding site is favored while the channel is still closed. We propose that this (these) intermediate ATP-bound state(s) prime(s) channel gating in the P2X2 receptor.


Subject(s)
Adenosine Triphosphate/metabolism , Receptors, Purinergic P2X2/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Affinity Labels , Binding Sites , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Mutation , Purinergic P2X Receptor Agonists/pharmacology , Receptors, Purinergic P2X2/chemistry , Receptors, Purinergic P2X2/genetics
7.
EMBO J ; 31(9): 2134-43, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22473210

ABSTRACT

The opening of ligand-gated ion channels in response to agonist binding is a fundamental process in biology. In ATP-gated P2X receptors, little is known about the molecular events that couple ATP binding to channel opening. In this paper, we identify structural changes of the ATP site accompanying the P2X2 receptor activation by engineering extracellular zinc bridges at putative mobile regions as revealed by normal mode analysis. We provide evidence that tightening of the ATP sites shaped like open 'jaws' induces opening of the P2X ion channel. We show that ATP binding favours jaw tightening, whereas binding of a competitive antagonist prevents gating induced by this movement. Our data reveal the inherent dynamic of the binding jaw, and provide new structural insights into the mechanism of P2X receptor activation.


Subject(s)
Adenosine Triphosphate/physiology , Receptors, Purinergic P2X2/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Binding Sites , HEK293 Cells , Humans , Protein Binding , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Zinc/pharmacology
8.
Proc Natl Acad Sci U S A ; 108(22): 9066-71, 2011 May 31.
Article in English | MEDLINE | ID: mdl-21576497

ABSTRACT

ATP-gated P2X receptors are trimeric ion channels, as recently confirmed by X-ray crystallography. However, the structure was solved without ATP and even though extracellular intersubunit cavities surrounded by conserved amino acid residues previously shown to be important for ATP function were proposed to house ATP, the localization of the ATP sites remains elusive. Here we localize the ATP-binding sites by creating, through a proximity-dependent "tethering" reaction, covalent bonds between a synthesized ATP-derived thiol-reactive P2X2 agonist (NCS-ATP) and single cysteine mutants engineered in the putative binding cavities of the P2X2 receptor. By combining whole-cell and single-channel recordings, we report that NCS-ATP covalently and specifically labels two previously unidentified positions N140 and L186 from two adjacent subunits separated by about 18 Å in a P2X2 closed state homology model, suggesting the existence of at least two binding modes. Tethering reaction at both positions primes subsequent agonist binding, yet with distinct functional consequences. Labeling of one position impedes subsequent ATP function, which results in inefficient gating, whereas tethering of the other position, although failing to produce gating by itself, enhances subsequent ATP function. Our results thus define a large and dynamic intersubunit ATP-binding pocket and suggest that receptors trapped in covalently agonist-bound states differ in their ability to gate the ion channel.


Subject(s)
Adenosine Triphosphate/chemistry , Receptors, Purinergic P2X2/chemistry , Amino Acid Sequence , Binding Sites , Biophysics/methods , Cell Line , Cell Membrane/metabolism , Cysteine/chemistry , DNA, Complementary/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Patch-Clamp Techniques , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid
9.
Br J Pharmacol ; 162(6): 1326-39, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21133889

ABSTRACT

BACKGROUND AND PURPOSE: Flavonoids, important plant pigments, have been shown to allosterically modulate brain GABA(A) receptors (GABA(A)Rs). We previously reported that trans-6,4'-dimethoxyretrochalcone (Rc-OMe), a hydrolytic derivative of the corresponding flavylium salt, displayed nanomolar affinity for the benzodiazepine binding site of GABA(A)Rs. Here, we evaluate the functional modulations of Rc-OMe, along with two other synthetic derivatives trans-6-bromo-4'-methoxyretrochalcone (Rc-Br) and 4,3'-dimethoxychalcone (Ch-OMe) on GABA(A)Rs. EXPERIMENTAL APPROACH: Whole-cell patch-clamp recordings were made to determine the effects of these derivatives on GABA(A)Rs expressed in HEK-293 cells and in hippocampal CA1 pyramidal and thalamic neurones from rat brain. KEY RESULTS: Rc-OMe strongly potentiated GABA-evoked currents at recombinant α(1-4)ß(2)γ(2s) and α(4)ß(3)δ receptors but much less at α(1)ß(2) and α(4)ß(3). Rc-Br and Ch-OMe potentiated GABA-evoked currents at α(1)ß(2)γ(2s). The potentiation by Rc-OMe was only reduced at α(1)H101Rß(2)γ(2s) and α(1)ß(2)N265Sγ(2s), mutations known to abolish the potentiation by diazepam and loreclezole respectively. The modulation of Rc-OMe and pentobarbital as well as by Rc-OMe and the neurosteroid 3α,21-dihydroxy-5α-pregnan-20-one was supra-additive. Rc-OMe modulation exhibited no apparent voltage-dependence, but was markedly dependent on GABA concentration. In neurones, Rc-Br slowed the decay of spontaneous inhibitory postsynaptic currents and both Rc-OMe and Rc-Br positively modulated synaptic and extrasynaptic diazepam-insensitive GABA(A)Rs. CONCLUSIONS AND IMPLICATIONS: The trans-retrochalcones are powerful positive allosteric modulators of synaptic and extrasynaptic GABA(A)Rs. These novel modulators act through an original mode, thus making them putative drug candidates in the treatment of GABA(A)-related disorders in vivo.


Subject(s)
CA1 Region, Hippocampal/drug effects , Chalcones/pharmacology , Pyramidal Cells/drug effects , Receptors, GABA-A/metabolism , Ventral Thalamic Nuclei/drug effects , Animals , Benzodiazepines/metabolism , Chalcones/chemical synthesis , HEK293 Cells , Humans , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Plasmids , Rats , Rats, Wistar , Stereoisomerism , gamma-Aminobutyric Acid/metabolism
10.
J Biol Chem ; 285(21): 15805-15, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20308075

ABSTRACT

The recent crystal structure of the ATP-gated P2X4 receptor revealed a static view of its architecture, but the molecular mechanisms underlying the P2X channels activation are still unknown. By using a P2X2 model based on the x-ray structure, we sought salt bridges formed between charged residues located in a region that directly connects putative ATP-binding sites to the ion channel. To reveal their significance for ion channel activation, we made systematic charge exchanges and measured the effects on ATP sensitivity. We found that charge reversals at the interfacial residues Glu(63) and Arg(274) produced gain-of-function phenotypes that were cancelled upon paired charge swapping. These results suggest that a putative intersubunit salt bridge formed between Glu(63) and Arg(274) contributes to the ion channel function. Engineered cysteines E63C and R274C formed redox-dependent cross-links in the absence of ATP. By contrast, the presence of ATP reduced the rate of disulfide bond formation, indicating that ATP binding might trigger relative movement of adjacent subunits at the level of Glu(63) and Arg(274), allowing the transmembrane helices to open the channel.


Subject(s)
Adenosine Triphosphate/metabolism , Ion Channel Gating/physiology , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/chemistry , Amino Acid Substitution , Animals , Cell Line , Disulfides/metabolism , Humans , Mutation, Missense , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X2
11.
Biochem Biophys Res Commun ; 375(3): 405-9, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18718445

ABSTRACT

ATP-gated P2X receptors (P2XRs) are ligand-gated ion channels (LGICs) presumably trimeric. To date, no experimental high-resolution structures are available. Recent X-ray structure of the acid-sensing ion channel 1 (ASIC1) revealed an unexpected trimeric ion channel. Beside their quaternary structure, P2XR and ASIC1 share common membrane topologies, but no significant sequence similarity. In order to overcome this low sequence resemblance, we have developed comparative models of P2X(2)R based on secondary structure predictions using the crystal structure of ASIC1 as template. These models were constrained to be consistent with known arrangement of disulfide bridges. They agreed with cross-linking experiments and supported inter-subunit ATP-binding sites. One of our models reconciled most existing data and provides new structural insights for a plausible mechanism of gating, thus encouraging new experiments.


Subject(s)
Adenosine Triphosphate/chemistry , Ion Channel Gating , Models, Molecular , Receptors, Purinergic P2/chemistry , Acid Sensing Ion Channels , Binding Sites , Nerve Tissue Proteins/chemistry , Protein Folding , Protein Structure, Secondary , Receptors, Purinergic P2X2 , Sodium Channels/chemistry
12.
Bioorg Med Chem Lett ; 18(9): 2765-70, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18434149

ABSTRACT

To prepare thiol-reactive ifenprodil derivatives designed as potential probes for cysteine-substituted NR2B containing NMDA receptors, electrophilic centers were introduced in different areas of the ifenprodil structure. Intermediates and final compounds were evaluated by binding studies and by electrophysiology to determine the structural requirements for their selectivity. The reactive compounds were further tested for their stability and for their reactivity in model reactions; some were found suitable as structural probes to investigate the binding site and the docking mode of ifenprodil in the NR2B subunit.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Affinity Labels/chemistry , Brain/drug effects , Membrane Potentials/drug effects , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Adrenergic alpha-Antagonists/chemical synthesis , Animals , Binding Sites , Brain/metabolism , Cysteine/chemistry , Electrophysiology , Membrane Potentials/physiology , Models, Chemical , Piperidines/chemical synthesis , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Structure-Activity Relationship
13.
Nucleic Acids Res ; 34(1): 32-41, 2006.
Article in English | MEDLINE | ID: mdl-16397295

ABSTRACT

The two BRCT domains (BRCT1 and BRCT2) of XRCC1 mediate a network of protein-protein interactions with several key factors of the DNA single-strand breaks (SSBs) and base damage repair pathways. BRCT1 is required for the immediate poly(ADP-ribose)-dependent recruitment of XRCC1 to DNA breaks and is essential for survival after DNA damage. To better understand the biological role of XRCC1 in the processing of DNA ends, a search for the BRCT1 domain-associated proteins was performed by mass spectrometry of GST-BRCT1 pulled-down proteins from HeLa cell extracts. Here, we report that the double-strand break (DSB) repair heterotrimeric complex DNA-PK interacts with the BRCT1 domain of XRCC1 and phosphorylates this domain at serine 371 after ionizing irradiation. This caused XRCC1 dimer dissociation. The XRCC1 R399Q variant allele did not affect this phosphorylation. We also show that XRCC1 strongly stimulates the phosphorylation of p53-Ser15 by DNA-PK. The pseudo phosphorylated S371D mutant was a much weaker stimulator of DNA-PK activity whereas the non-phosphorylable mutant S371L endowed with a DNA-PK stimulating capacity failed to fully rescue the DSB repair defect of XRCC1-deficient EM9 rodent cells. The functional association between XRCC1 and DNA-PK in response to IR provides the first evidence for their involvement in a common DSB repair pathway.


Subject(s)
DNA Damage , DNA Repair , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Animals , Binding Sites , Cricetinae , DNA-Binding Proteins/chemistry , Dimerization , HeLa Cells , Humans , Mass Spectrometry , Phosphorylation , Protein Structure, Tertiary , Radiation, Ionizing , Serine/metabolism , X-ray Repair Cross Complementing Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...