Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Physiol ; 12: 727549, 2021.
Article in English | MEDLINE | ID: mdl-34733174

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are used for relieving pain and inflammation accompanying numerous disease states. The primary therapeutic mechanism of these widely used drugs is the inhibition of cyclooxygenase 1 and 2 (COX1, 2) enzymes that catalyze the conversion of arachidonic acid into prostaglandins. At higher doses, NSAIDs are used for prevention of certain types of cancer and as experimental treatments for Alzheimer's disease. In the immune system, various NSAIDs have been reported to influence neutrophil function and lymphocyte proliferation, and affect ion channels and cellular calcium homeostasis. Transient receptor potential melastatin 7 (TRPM7) cation channels are highly expressed in T lymphocytes and are inhibited by Mg2+, acidic pH, and polyamines. Here, we report a novel effect of naproxen, ibuprofen, salicylate, and acetylsalicylate on TRPM7. At concentrations of 3-30mM, they reversibly inhibited TRPM7 channel currents. By measuring intracellular pH with the ratiometric indicator BCECF, we found that at 300µM to 30mM, these NSAIDs reversibly acidified the cytoplasm in a concentration-dependent manner, and propose that TRPM7 channel inhibition is a consequence of cytosolic acidification, rather than direct. NSAID inhibition of TRPM7 channels was slow, voltage-independent, and displayed use-dependence, increasing in potency upon repeated drug applications. The extent of channel inhibition by salicylate strongly depended on cellular PI(4,5)P2 levels, as revealed when this phospholipid was depleted with voltage-sensitive lipid phosphatase (VSP). Salicylate inhibited heterologously expressed wildtype TRPM7 channels but not the S1107R variant, which is insensitive to cytosolic pH, Mg2+, and PI(4,5)P2 depletion. NSAID-induced acidification was also observed in Schneider 2 cells from Drosophila, an organism that lacks orthologous COX genes, suggesting that this effect is unrelated to COX enzyme activity. A 24-h exposure to 300µM-10mM naproxen resulted in a concentration-dependent reduction in cell viability. In addition to TRPM7, the described NSAID effect would be expected to apply to other ion channels and transporters sensitive to intracellular pH.

2.
Mol Pharmacol ; 88(5): 926-34, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26268529

ABSTRACT

Compounds PKTHPP (1-{1-[6-(biphenyl-4-ylcarbonyl)-5,6,7,8-tetrahydropyrido[4,3-d]-pyrimidin-4-yl]piperidin-4-yl}propan-1-one), A1899 (2''-[(4-methoxybenzoylamino)methyl]biphenyl-2-carboxylic acid 2,4-difluorobenzylamide), and doxapram inhibit TASK-1 (KCNK3) and TASK-3 (KCNK9) tandem pore (K2P) potassium channel function and stimulate breathing. To better understand the molecular mechanism(s) of action of these drugs, we undertook studies to identify amino acid residues in the TASK-3 protein that mediate this inhibition. Guided by homology modeling and molecular docking, we hypothesized that PKTHPP and A1899 bind in the TASK-3 intracellular pore. To test our hypothesis, we mutated each residue in or near the predicted PKTHPP and A1899 binding site (residues 118-128 and 228-248), individually, to a negatively charged aspartate. We quantified each mutation's effect on TASK-3 potassium channel concentration response to PKTHPP. Studies were conducted on TASK-3 transiently expressed in Fischer rat thyroid epithelial monolayers; channel function was measured in an Ussing chamber. TASK-3 pore mutations at residues 122 (L122D, E, or K) and 236 (G236D) caused the IC50 of PKTHPP to increase more than 1000-fold. TASK-3 mutants L122D, G236D, L239D, and V242D were resistant to block by PKTHPP, A1899, and doxapram. Our data are consistent with a model in which breathing stimulant compounds PKTHPP, A1899, and doxapram inhibit TASK-3 function by binding at a common site within the channel intracellular pore region, although binding outside the channel pore cannot yet be excluded.


Subject(s)
Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Respiratory System Agents/pharmacology , Amino Acid Sequence , Animals , Benzamides/pharmacology , Benzeneacetamides/pharmacology , Binding Sites , Cells, Cultured , Doxapram/pharmacology , Molecular Docking Simulation , Molecular Sequence Data , Mutagenesis , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/physiology , Rats , Rats, Inbred F344 , Respiratory System Agents/metabolism , Structure-Activity Relationship
3.
Channels (Austin) ; 6(5): 362-9, 2012.
Article in English | MEDLINE | ID: mdl-22922232

ABSTRACT

2-APB is a widely used compound in ion channel research. It affects numerous channels including inositol 1,4,5-trisphosphate receptors, store-operated calcium channels and TRP channels, TRPV3 and TRPM7 among them. A characteristic property of TRPM7 channels is their sensitivity to intracellular Mg ( 2+) and pH. Using patch clamp electrophysiology we find that in Jurkat T lymphocytes, 100-300 µM extracellular 2-APB reversibly inhibits TRPM7 channels when internal HEPES concentration is low (1 mM). Increasing the concentration to 140 mM abolishes the 2-APB effect. Using single-cell fluorescence pH video imaging, we show that at concentrations of 100 µM and higher, 2-APB potently acidifies the cytoplasm. We conclude that TRPM7 sensitivity to 2-APB is not direct but rather, can be explained by cytoplasmic acidification and a resulting channel inhibition.


Subject(s)
Boron Compounds/pharmacology , TRPM Cation Channels/antagonists & inhibitors , Humans , Hydrogen-Ion Concentration , Ion Channel Gating/drug effects , Jurkat Cells , Magnesium/metabolism , Patch-Clamp Techniques , Protein Serine-Threonine Kinases , TRPM Cation Channels/metabolism
4.
Am J Physiol Cell Physiol ; 302(11): C1642-51, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22460708

ABSTRACT

Transient receptor potential melastatin 7 (TRPM7) channels were originally identified electrophysiologically when depletion of cytosolic Mg(2+) resulted in the gradual development of an outwardly rectifying cation current. Conversely, inclusion of millimolar Mg(2+) in internal solutions prevented activation of these channels in whole cell patch clamp. We recently demonstrated that the Jurkat T-cell whole cell TRPM7 channels are inhibited by internal Mg(2+) in a biphasic manner, displaying high [IC(50(1)) ≈ 10 µM] and low [IC(50(2)) ≈ 165 µM] affinity inhibitor sites. In that study, we had characterized the dependence of the maximum cell current density on intracellular Mg(2+) concentration. To characterize Mg(2+) inhibition in Jurkat T cells in more detail and compare it to whole cell results, we recorded single TRPM7 channels in cell-free membrane patches and investigated the dependence of their activity on Mg(2+) added on the cytoplasmic side. We systematically varied free Mg(2+) from 265 nM to 407 µM and evaluated the extent of channel inhibition in inside-out patch for 58 patches. We found that the TRPM7 channel shows two conductance levels of 39.0 pS (γ(1)) and 18.6 pS (γ(2)) and that both are reversibly inhibited by internal Mg(2+). The 39.0-pS conductance is the dominant state of the channel, observed most frequently in this recording configuration. The dose-response relation in inside-out patches shows a steeper Mg(2+) dependence than in whole cell, yielding IC(50(1)) of 25.1 µM and IC(50(2)) of 91.2 µM.. Single-channel analysis shows that the primary effect of Mg(2+) in multichannel patches is a reversible reduction of the number of conducting channels (N(o)). Additionally, at high Mg(2+) concentrations, we observed a saturating 20% reduction in unitary conductance (γ(1)). Thus Mg(2+) inhibition in whole cell can be explained by a drop in individual participating channels and a modest reduction in conductance. We also found that TRPM7 channels in some patches were not sensitive to this ion at submaximal Mg(2+) concentrations. Interestingly, Mg(2+) inhibition showed the property of use dependence: with repeated applications, Mg(2+) effect became gradually more potent, which suggests that Mg(2+) sensitivity of the channel is a dynamic characteristic that depends on other membrane factors.


Subject(s)
Magnesium/pharmacology , Membrane Potentials/physiology , T-Lymphocytes/metabolism , TRPM Cation Channels/metabolism , Cell Line , Electrophysiology , Humans , Jurkat Cells , Membrane Potentials/drug effects , Patch-Clamp Techniques , Protein Serine-Threonine Kinases
5.
Am J Physiol Cell Physiol ; 302(7): C1004-11, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22301056

ABSTRACT

TRPM7 channel kinase is a protein highly expressed in cells of hematopoietic lineage, such as lymphocytes. Studies performed in native and heterologous expression systems have shown that TRPM7 forms nonselective cation channels functional in the plasma membrane and activated on depletion of cellular Mg(2+). In addition to internal Mg(2+), cytosolic pH and the phospholipid phosphatidylinositol-(4,5)-bisphosphate [PI(4,5)P(2)] are potent physiological regulators of this channel: protons inhibit, while PI(4,5)P(2) is required for TRPM7 channel activity. These channels are also inhibited from inside by other metal cations and polyamines. While the regulation of TRPM7 channels by internal metal ions, acidic pH, and PI(4,5)P(2) is voltage independent, extracellular metal cations and polyamines block voltage dependently at micromolar concentrations and appear to occupy a distinct blocking site. In the present study we investigated intracellular Mg(2+) and pH dependence of native TRPM7 currents using whole cell patch-clamp electrophysiology in human Jurkat T lymphocytes and HEK293 cells. Our main findings are 1) Mg(2+) inhibition involves not one but two separate sites of high (∼10 µM) and low (∼165 µM) affinity; and 2) while sharing certain characteristics with Mg(2+) inhibition, protons most likely inhibit through one inhibitory site, corresponding to the low-affinity Mg(2+) site, with an estimated IC(50) of pH 6.3. Additionally, we present data on amplitude distribution of preactivated TRPM7 currents in Jurkat T lymphocytes in the absence of prior Mg(2+) or proton depletion.


Subject(s)
Magnesium/metabolism , TRPM Cation Channels/metabolism , Cations/metabolism , Cell Line, Tumor , Cell Membrane/genetics , Cell Membrane/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Jurkat Cells , Membrane Potentials/physiology , Patch-Clamp Techniques/methods , Polyamines/metabolism , Protein Serine-Threonine Kinases , Protons
SELECTION OF CITATIONS
SEARCH DETAIL
...