Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 15: 1341799, 2024.
Article in English | MEDLINE | ID: mdl-38659572

ABSTRACT

Tetraethylammonium (TEA), a quaternary ammonium compound, is a well-known blocker of potassium channels belonging to various subfamilies, such as KV1-3, KCa1, 2 and prokaryotic KcsA. In many cases, TEA acts from the extracellular side by open pore blockade. TEA can also block transient receptor potential (TRP) cation channels, such as TRPM7, in a voltage-dependent manner. In human T lymphocytes, intracellular (cytosolic) TEA and its analog TMA (tetramethylammonium) inhibit TRPM7 channel currents in the outward but not inward direction. By contrast, intracellular Mg2+, protons and polyamines inhibit both outward and inward current components equally. Likewise, the majority of available pharmacological tools inhibit TRPM7 channels in a voltage-independent manner. Since TRPM7 is a steeply outwardly rectifying conductance, voltage-dependent blockers can be useful for studying the cellular functions of this channel. TRPM7 protein is endogenously expressed in diverse cell lines, including HEK, HeLa, CHO, RBL and Jurkat. Using patch-clamp electrophysiology, we found that incubating HEK293 and Jurkat T cells overnight in the presence of 20 mM TEA-Cl, resulted in the nearly complete blockade of whole-cell TRPM7 outward current, measured at break-in. By contrast, the inward current was unchanged in TEA-loaded cells. The blockade was fully reversible after washout of intracellular solution in whole-cell but not in perforated-patch recording configurations. Overnight incubation with 20 mM TMA-Cl resulted in a more modest blockade of the outward TRPM7 current. Internal 129 mM TMA and TEA eliminated most of the outward current. TEA uptake in transfected HEK293 cells led to blockade of recombinant murine TRPM7 and the Mg2+ and pH insensitive Ser1107Arg variant. Unexpectedly, Tris-HCl, a widely used pH buffer, could similarly be loaded into Jurkat and HEK cells, and preferentially blocked outward TRPM7 currents. 20 mM and 129 mM Tris in the internal solution blocked TRPM7 current in outward but not inward direction. Voltage-dependent channel blockade by TEA, TMA and Tris loading will be useful for studying the properties and functions of TRPM7-mediated ion transport in intact cells.

3.
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.

4.
FEBS J ; 288(11): 3585-3601, 2021 06.
Article in English | MEDLINE | ID: mdl-33354894

ABSTRACT

Transient receptor potential melastatin 7 (TRPM7) is a unique protein functioning as a cation channel as well as a serine/threonine kinase and is highly expressed in immune cells such as lymphocytes and macrophages. TRPM7 kinase-dead (KD) mouse model has been used to investigate the role of this protein in immune cells; these animals display moderate splenomegaly and ectopic hemopoiesis. The basal TRPM7 current magnitudes in peritoneal macrophages isolated from KD mice were higher; however, the maximum currents, achieved after cytoplasmic Mg2+ washout, were not different. In the present study, we investigated the consequences of TRPM7 kinase inactivation in splenic and peritoneal macrophages. We measured the basal phagocytic activity of splenic macrophages using fluorescent latex beads, pHrodo zymosan bioparticles, and opsonized red blood cells. KD macrophages phagocytized more efficiently and had slightly higher baseline calcium levels compared to WT cells. We found no obvious differences in store-operated Ca2+ entry between WT and KD macrophages. By contrast, the resting cytosolic pH in KD macrophages was significantly more alkaline than in WT. Pharmacological blockade of sodium hydrogen exchanger 1 (NHE1) reversed the cytosolic alkalinization and reduced phagocytosis in KD macrophages. Basal TRPM7 channel activity in KD macrophages was also reduced after NHE1 blockade. Cytosolic Mg2+ sensitivity of TRPM7 channels measured in peritoneal macrophages was similar in WT and KD mice. The higher basal TRPM7 channel activity in KD macrophages is likely due to alkalinization. Our results identify a novel role for TRPM7 kinase as a suppressor of basal phagocytosis and a regulator of cellular pH.


Subject(s)
Phagocytosis/genetics , Sodium-Hydrogen Exchanger 1/genetics , Spleen/metabolism , TRPM Cation Channels/genetics , Animals , Antacids/pharmacology , Calcium Signaling/genetics , Cytosol/enzymology , Cytosol/metabolism , HEK293 Cells , Humans , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Magnesium/metabolism , Mice , Phagocytes/drug effects , Phagocytes/metabolism , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Spleen/drug effects
5.
Pflugers Arch ; 472(12): 1809-1810, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33006640

ABSTRACT

The original article was published with an error. In Figure 9b there are 3 typographical errors: instead of the Greek mu letter it shows the unconverted data.

6.
Pflugers Arch ; 472(11): 1589-1606, 2020 11.
Article in English | MEDLINE | ID: mdl-32964285

ABSTRACT

TRPM7 is a cation channel-protein kinase highly expressed in T lymphocytes and other immune cells. It has been proposed to constitute a cellular entry pathway for Mg2+ and divalent metal cations such as Ca2+, Zn2+, Cd2+, Mn2+, and Ni2+. TRPM7 channels are inhibited by cytosolic Mg2+, rendering them largely inactive in intact cells. The dependence of channel activity on extracellular Mg2+ is less well studied. Here, we measured native TRPM7 channel activity in Jurkat T cells maintained in external Mg2+ concentrations varying between 400 nM and 1.4 mM for 1-3 days, obtaining an IC50 value of 54 µM. Maintaining the cells in 400 nM or 8 µM [Mg2+]o resulted in almost complete activation of TRPM7 in intact cells, due to cytosolic Mg2+ depletion. A total of 1.4 mM [Mg2+]o was sufficient to fully eliminate the basal current. Submillimolar concentrations of amiloride prevented cellular Mg2+ depletion but not loading. We investigated whether the cytotoxicity of TRPM7 permeant metal ions Ni2+, Zn2+, Cd2+, Co2+, Mn2+, Sr2+, and Ba2+ requires TRPM7 channel activity. Mg2+ loading modestly reduced cytotoxicity of Zn2+, Co2+, Ni2+, and Mn2+ but not of Cd2+. Channel blocker NS8593 reduced Co2+ and Mn2+ but not Cd2+ or Zn2+ cytotoxicity and interfered with Mg2+ loading as evaluated by TRPM7 channel basal activity. Ba2+ and Sr2+ were neither detectably toxic nor permeant through the plasma membrane. These results indicate that in Jurkat T cells, entry of toxic divalent metal cations primarily occurs through pathways distinct from TRPM7. By contrast, we found evidence that Mg2+ entry requires TRPM7 channels.


Subject(s)
Magnesium/metabolism , Metals, Heavy/toxicity , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , Action Potentials , Humans , Inhibitory Concentration 50 , Ion Transport , Jurkat Cells , Metals, Heavy/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors
7.
J Biol Chem ; 293(47): 18151-18167, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30305398

ABSTRACT

Transient receptor potential cation channel subfamily M member 7 (TRPM7) is an ion channel/protein kinase belonging to the TRP melastatin and eEF2 kinase families. Under physiological conditions, most native TRPM7 channels are inhibited by cytoplasmic Mg2+, protons, and polyamines. Currents through these channels (ITRPM7) are robustly potentiated when the cell interior is exchanged with low Mg2+-containing buffers. ITRPM7 is also potentiated by phosphatidyl inositol bisphosphate (PI(4,5)P2) and suppressed by its hydrolysis. Here we characterized internal Mg2+- and pH-mediated inhibition of TRPM7 channels in HEK293 cells overexpressing WT voltage-sensing phospholipid phosphatase (VSP) or its catalytically inactive variant VSP-C363S. VSP-mediated depletion of membrane phosphoinositides significantly increased channel sensitivity to Mg2+ and pH. Proton concentrations that were too low to inhibit ITRPM7 when the VSP-C363S variant was expressed (pH 8.2) became inhibitory in WT VSP-expressing cells. At pH 6.5, protons inhibited ITRPM7 both in WT and VSP C363S-expressing cells but with a faster time course in the WT VSP-expressing cells. Inhibition by 150 µm Mg2+ was also significantly faster in the WT VSP-expressing cells. Cellular PI(4,5)P2 depletion increased the sensitivity of TRPM7 channels to the inhibitor 2-aminoethyl diphenyl borinate, which acidifies the cytosol. Single substitutions at Ser-1107 of TRPM7, reducing its sensitivity to Mg2+, also decreased its inhibition by spermine and acidic pH. Furthermore, these channel variants were markedly less sensitive to VSP-mediated PI(4,5)P2 depletion than the WT. We conclude that the internal Mg2+-, polyamine-, and pH-mediated inhibition of TRPM7 channels is not direct but, rather, reflects electrostatic screening and resultant disruption of PI(4,5)P2-channel interactions.


Subject(s)
Cell Membrane/metabolism , Cytosol/metabolism , Magnesium/metabolism , Phosphatidylinositols/metabolism , Spermine/metabolism , TRPM Cation Channels/metabolism , Animals , Biological Transport , Cell Membrane/genetics , Hydrogen-Ion Concentration , Mice , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Polyamines/metabolism , Protons , TRPM Cation Channels/genetics
8.
Sci Rep ; 8(1): 3023, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445164

ABSTRACT

T lymphocytes enlarge (blast) and proliferate in response to antigens in a multistep program that involves obligatory cytosolic calcium elevations. Store-operated calcium entry (SOCE) pathway is the primary source of Ca2+ in these cells. Here, we describe a novel modulator of blastogenesis, proliferation and SOCE: the TRPM7 channel kinase. TRPM7 kinase-dead (KD) K1646R knock-in mice exhibited splenomegaly and impaired blastogenic responses elicited by PMA/ionomycin or anti-CD3/CD28 antibodies. Splenic T-cell proliferation in vitro was weaker in the mutant compared to wildtype littermates. TRPM7 current magnitudes in WT and KD mouse T cells were, however, similar. We tested the dependence of T-cell proliferation on external Ca2+ and Mg2+ concentrations. At a fixed [Mg2+o] of ~0.4 mM, Ca2+o stimulated proliferation with a steep concentration dependence and vice versa, at a fixed [Ca2+o] of ~0.4 mM, Mg2+o positively regulated proliferation but with a shallower dependence. Proliferation was significantly lower in KD mouse than in wildtype at all Ca2+ and Mg2+ concentrations. Ca2+ elevations elicited by anti-CD3 antibody were diminished in KD mutant T cells and SOCE measured in activated KD splenocytes was reduced. These results demonstrate that a functional TRPM7 kinase supports robust SOCE, blastogenesis and proliferation, whereas its inactivation suppresses these cellular events.


Subject(s)
Calcium/metabolism , TRPM Cation Channels/metabolism , Animals , Calcium Channels/metabolism , Calcium Signaling/physiology , Cell Proliferation/physiology , Gene Knock-In Techniques/methods , Lymphocyte Activation/physiology , Magnesium/metabolism , Mice , Mice, Inbred C57BL , Spleen/pathology , Splenomegaly/metabolism , Stromal Interaction Molecule 1/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , TRPM Cation Channels/genetics
9.
Sci Rep ; 7(1): 1732, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28496181

ABSTRACT

Intracellular chloride concentration ([Cl-]i) in pancreatic ß-cells is kept above electrochemical equilibrium due to the predominant functional presence of Cl- loaders such as the Na+K+2Cl- co-transporter 1 (Slc12a2) over Cl-extruders of unidentified nature. Using molecular cloning, RT-PCR, Western blotting, immunolocalization and in vitro functional assays, we establish that the "neuron-specific" K+Cl- co-transporter 2 (KCC2, Slc12a5) is expressed in several endocrine cells of the pancreatic islet, including glucagon secreting α-cells, but particularly in insulin-secreting ß-cells, where we provide evidence for its role in the insulin secretory response. Three KCC2 splice variants were identified: the formerly described KCC2a and KCC2b along with a novel one lacking exon 25 (KCC2a-S25). This new variant is undetectable in brain or spinal cord, the only and most abundant known sources of KCC2. Inhibition of KCC2 activity in clonal MIN6 ß-cells increases basal and glucose-stimulated insulin secretion and Ca2+ uptake in the presence of glibenclamide, an inhibitor of the ATP-dependent potassium (KATP)-channels, thus suggesting a possible mechanism underlying KCC2-dependent insulin release. We propose that the long-time considered "neuron-specific" KCC2 co-transporter is expressed in pancreatic islet ß-cells where it modulates Ca2+-dependent insulin secretion.


Subject(s)
Insulin Secretion , Neurons/metabolism , Symporters/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , Cell Line , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , KATP Channels/metabolism , Mice , Pyridazines , Symporters/chemistry , Symporters/genetics , Thiazoles , K Cl- Cotransporters
10.
J Vis Exp ; (118)2016 12 27.
Article in English | MEDLINE | ID: mdl-28060354

ABSTRACT

Lymphocyte proliferation in response to antigenic or mitogenic stimulation is a readily quantifiable phenomenon useful for testing immunomodulatory (i.e., immunosuppressive or immunostimulatory) chemical compounds and biologics. One of the earliest steps during mitogenesis is cell enlargement or blastogenic transformation, whereupon the cell volume increases before division. It is usually detectable in the first several hours of T-lymphocyte stimulation. Here, we describe a rapid method to quantify blastogenesis in T lymphocytes isolated from mouse spleens and human peripheral blood mononuclear cells (PBMCs) using an automated cell counter. Various commonly used proliferation assays for the most part are laborious and only reflect the overall population effect rather than individual cellular effects within a population. In contrast, the presented automated cell counter assay provides rapid, direct, and precise measurements of cell diameters that can be used for assessing the effectiveness of various mitogens and immunomodulatory drugs in vitro.


Subject(s)
Lymphocyte Activation , Mitogens/pharmacology , T-Lymphocytes/drug effects , Animals , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , T-Lymphocytes/cytology
11.
Sci Rep ; 4: 5718, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25030553

ABSTRACT

Transient receptor potential (TRP) family channels are involved in sensory pathways and respond to various environmental stimuli. Among the members of this family, TRPM7 is a unique fusion of an ion channel and a C-terminus kinase domain that is highly expressed in immune cells. TRPM7 serves as a key molecule governing cellular Mg(2+) homeostasis in mammals since its channel pore is permeable to Mg(2+) ions and can act as a Mg(2+) influx pathway. However, mechanistic links between its kinase activity and channel function have remained uncertain. In this study, we generated kinase inactive knock-in mutant mice by mutagenesis of a key lysine residue involved in Mg(2+)-ATP binding. These mutant mice were normal in development and general locomotor activity. In peritoneal macrophages isolated from adult animals the basal activity of TRPM7 channels prior to cytoplasmic Mg(2+) depletion was significantly potentiated, while maximal current densities measured after Mg(2+) depletion were unchanged in the absence of detectable kinase function. Serum total Ca(2+) and Mg(2+) levels were not significantly altered in kinase-inactive mutant mice. Our findings suggest that abolishing TRPM7 kinase activity does not impair its channel activity and kinase activity is not essential for regulation of mammalian Mg(2+) homeostasis.


Subject(s)
TRPM Cation Channels/genetics , Animals , Cells, Cultured , Female , Gene Knock-In Techniques , Homeostasis , Macrophages/metabolism , Magnesium/metabolism , Male , Membrane Potentials , Mice, 129 Strain , Mice, Inbred C57BL , Patch-Clamp Techniques , TRPM Cation Channels/metabolism
12.
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
13.
PLoS One ; 7(4): e35090, 2012.
Article in English | MEDLINE | ID: mdl-22563377

ABSTRACT

The impact of synthetic amyloid ß (1-42) (Aß(1-42)) oligomers on biophysical properties of voltage-gated potassium channels Kv 1.3 and lipid bilayer membranes (BLMs) was quantified for protocols using hexafluoroisopropanol (HFIP) or sodium hydroxide (NaOH) as solvents prior to initiating the oligomer formation. Regardless of the solvent used Aß(1-42) samples contained oligomers that reacted with the conformation-specific antibodies A11 and OC and had similar size distributions as determined by dynamic light scattering. Patch-clamp recordings of the potassium currents showed that synthetic Aß(1-42) oligomers accelerate the activation and inactivation kinetics of Kv 1.3 current with no significant effect on current amplitude. In contrast to oligomeric samples, freshly prepared, presumably monomeric, Aß(1-42) solutions had no effect on Kv 1.3 channel properties. Aß(1-42) oligomers had no effect on the steady-state current (at -80 mV) recorded from Kv 1.3-expressing cells but increased the conductance of artificial BLMs in a dose-dependent fashion. Formation of amyloid channels, however, was not observed due to conditions of the experiments. To exclude the effects of HFIP (used to dissolve lyophilized Aß(1-42) peptide), and trifluoroacetic acid (TFA) (used during Aß(1-42) synthesis), we determined concentrations of these fluorinated compounds in the stock Aß(1-42) solutions by (19)F NMR. After extensive evaporation, the concentration of HFIP in the 100× stock Aß(1-42) solutions was ∼1.7 µM. The concentration of residual TFA in the 70× stock Aß(1-42) solutions was ∼20 µM. Even at the stock concentrations neither HFIP nor TFA alone had any effect on potassium currents or BLMs. The Aß(1-42) oligomers prepared with HFIP as solvent, however, were more potent in the electrophysiological tests, suggesting that fluorinated compounds, such as HFIP or structurally-related inhalational anesthetics, may affect Aß(1-42) aggregation and potentially enhance ability of oligomers to modulate voltage-gated ion channels and biological membrane properties.


Subject(s)
Amyloid beta-Peptides/pharmacology , Electric Conductivity , Kv1.3 Potassium Channel/metabolism , Lipid Bilayers/metabolism , Peptide Fragments/pharmacology , Solvents/chemistry , Amyloid beta-Peptides/chemical synthesis , Halogenation , Kinetics , Light , Membranes, Artificial , Patch-Clamp Techniques , Peptide Fragments/chemical synthesis , Propanols/chemistry , Scattering, Radiation , Sodium Hydroxide/chemistry
14.
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
15.
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
16.
J Biol Chem ; 283(25): 17662-71, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18420579

ABSTRACT

We evaluated currents induced by expression of human homologs of Orai together with STIM1 in human embryonic kidney cells. When co-expressed with STIM1, Orai1 induced a large inwardly rectifying Ca(2+)-selective current with Ca(2+)-induced slow inactivation. A point mutation of Orai1 (E106D) altered the ion selectivity of the induced Ca(2+) release-activated Ca(2+) (CRAC)-like current while retaining an inwardly rectifying I-V characteristic. Expression of the C-terminal portion of STIM1 with Orai1 was sufficient to generate CRAC current without store depletion. 2-APB activated a large relatively nonselective current in STIM1 and Orai3 co-expressing cells. 2-APB also induced Ca(2+) influx in Orai3-expressing cells without store depletion or co-expression of STIM1. The Orai3 current induced by 2-APB exhibited outward rectification and an inward component representing a mixed calcium and monovalent current. A pore mutant of Orai3 inhibited store-operated Ca(2+) entry and did not carry significant current in response to either store depletion or addition of 2-APB. Analysis of a series of Orai1-3 chimeras revealed the structural determinant responsible for 2-APB-induced current within the sequence from the second to third transmembrane segment of Orai3. The Orai3 current induced by 2-APB may reflect a store-independent mode of CRAC channel activation that opens a relatively nonselective cation pore.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Gene Expression Regulation , Boron Compounds/pharmacology , Cloning, Molecular , Genes, Dominant , Humans , Membrane Proteins/metabolism , Models, Biological , Mutation , Neoplasm Proteins/metabolism , ORAI1 Protein , Patch-Clamp Techniques , Point Mutation , Protein Conformation , Protein Structure, Tertiary , Stromal Interaction Molecule 1
17.
Proc Natl Acad Sci U S A ; 105(6): 2011-6, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18250319

ABSTRACT

For efficient development of an immune response, T lymphocytes require long-lasting calcium influx through calcium release-activated calcium (CRAC) channels and the formation of a stable immunological synapse (IS) with the antigen-presenting cell (APC). Recent RNAi screens have identified Stim and Orai in Drosophila cells, and their corresponding mammalian homologs STIM1 and Orai1 in T cells, as essential for CRAC channel activation. Here, we show that STIM1 and Orai1 are recruited to the immunological synapse between primary human T cells and autologous dendritic cells. Both STIM1 and Orai1 accumulated in the area of contact between either resting or super-antigen (SEB)-pretreated T cells and SEB-pulsed dendritic cells, where they were colocalized with T cell receptor (TCR) and costimulatory molecules. In addition, imaging of intracellular calcium signaling in T cells loaded with EGTA revealed significantly higher Ca2+ concentration near the interface, indicating Ca2+ influx localized at the T cell/dendritic cell contact area. Expression of a dominant-negative Orai1 mutant blocked T cell Ca2+ signaling but did not interfere with the initial accumulation of STIM1, Orai1, and CD3 in the contact zone. In activated T cell blasts, mRNA expression for endogenous STIM1 and all three human homologs of Orai was up-regulated, accompanied by a marked increase in Ca2+ influx through CRAC channels. These results imply a positive feedback loop in which an initial TCR signal favors up-regulation of STIM1 and Orai proteins that would augment Ca2+ signaling during subsequent antigen encounter.


Subject(s)
Calcium Channels/physiology , Lymphocyte Activation , Membrane Proteins/physiology , Neoplasm Proteins/physiology , T-Lymphocytes/immunology , Up-Regulation , Calcium/metabolism , Cell Line , Humans , Ion Transport , ORAI1 Protein , Reverse Transcriptase Polymerase Chain Reaction , Stromal Interaction Molecule 1
18.
J Gen Physiol ; 128(6): 637-47, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17101816

ABSTRACT

The amyloid hypothesis of Alzheimer's toxicity has undergone a resurgence with increasing evidence that it is not amyloid fibrils but a smaller oligomeric species that produces the deleterious results. In this paper we address the mechanism of this toxicity. Only oligomers increase the conductance of lipid bilayers and patch-clamped mammalian cells, producing almost identical current-voltage curves in both preparations. Oligomers increase the conductance of the bare bilayer, the cation conductance induced by nonactin, and the anion conductance induced by tetraphenyl borate. Negative charge reduces the sensitivity of the membrane to amyloid, but cholesterol has little effect. In contrast, the area compressibility of the lipid has a very large effect. Membranes with a large area compressibility modulus are almost insensitive to amyloid oligomers, but membranes formed from soft, highly compressible lipids are highly susceptible to amyloid oligomer-induced conductance changes. Furthermore, membranes formed using the solvent decane (instead of squalane) are completely insensitive to the presence of oligomers. One simple explanation for these effects on bilayer conductance is that amyloid oligomers increase the area per molecule of the membrane-forming lipids, thus thinning the membrane, lowering the dielectric barrier, and increasing the conductance of any mechanism sensitive to the dielectric barrier.


Subject(s)
Amyloid beta-Peptides/physiology , Electric Conductivity , Lipid Bilayers/metabolism , Animals , Benzyl Alcohol/pharmacology , Cells, Cultured , Cholesterol/pharmacology , Ion Channels/physiology , Macrolides/metabolism , Protein Structure, Quaternary , Rats , Solvents/pharmacology , Sphingomyelins/chemistry , Tetraphenylborate/metabolism
19.
J Gen Physiol ; 126(5): 499-514, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16260839

ABSTRACT

The Mg2+-inhibited cation (MIC) current, believed to represent activity of TRPM7 channels, is found in lymphocytes and mast cells, cardiac and smooth muscle, and several other eukaryotic cell types. MIC current is activated during whole-cell dialysis with divalent-free internal solutions. Millimolar concentrations of intracellular Mg2+ (or other divalent metal cations) inhibit the channels in a voltage-independent manner. The nature of divalent inhibition and the mechanism of channel activation in an intact cell remain unknown. We show that the polyamines (spermine, spermidine, and putrescine) inhibit the MIC current, also in a voltage-independent manner, with a potency that parallels the number of charges. Neomycin and poly-lysine also potently inhibited MIC current in the absence of Mg2+. These same positively charged ions inhibited IRK1 current in parallel with MIC current, suggesting that they probably act by screening the head group phosphates on PIP2 and other membrane phospholipids. In agreement with this hypothesis, internal protons also inhibited MIC current. By contrast, tetramethylammonium, tetraethylammonium, and hexamethonium produced voltage-dependent block but no inhibition. We show that inhibition by internal polyvalent cations can be relieved by alkalinizing the cytosol using externally applied ammonium or by increasing pH in inside-out patches. Furthermore, in perforated-patch and cell-attached recordings, when intracellular Mg2+ is not depleted, endogenous MIC or recombinant TRPM7 currents are activated by cytosolic alkalinization and inhibited by acidification; and they can be reactivated by PIP2 following rundown in inside-out patches. We propose that MIC (TRPM7) channels are regulated by a charge screening mechanism and may function as sensors of intracellular pH.


Subject(s)
Cations/pharmacology , Hydrogen-Ion Concentration , Magnesium/pharmacology , TRPM Cation Channels/drug effects , Animals , CHO Cells/drug effects , CHO Cells/physiology , Cations/metabolism , Cricetinae , Cytosol/drug effects , Cytosol/physiology , Magnesium/antagonists & inhibitors , Magnesium/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Phosphatidylinositol 4,5-Diphosphate/physiology , Phospholipids/physiology , Polyamines/pharmacology , Potassium Channels, Inwardly Rectifying/physiology , Recombinant Fusion Proteins , Second Messenger Systems/physiology , TRPM Cation Channels/genetics , TRPM Cation Channels/physiology , Transfection
20.
Nature ; 437(7060): 902-5, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16208375

ABSTRACT

As the sole Ca2+ entry mechanism in a variety of non-excitable cells, store-operated calcium (SOC) influx is important in Ca2+ signalling and many other cellular processes. A calcium-release-activated calcium (CRAC) channel in T lymphocytes is the best-characterized SOC influx channel and is essential to the immune response, sustained activity of CRAC channels being required for gene expression and proliferation. The molecular identity and the gating mechanism of SOC and CRAC channels have remained elusive. Previously we identified Stim and the mammalian homologue STIM1 as essential components of CRAC channel activation in Drosophila S2 cells and human T lymphocytes. Here we show that the expression of EF-hand mutants of Stim or STIM1 activates CRAC channels constitutively without changing Ca2+ store content. By immunofluorescence, EM localization and surface biotinylation we show that STIM1 migrates from endoplasmic-reticulum-like sites to the plasma membrane upon depletion of the Ca2+ store. We propose that STIM1 functions as the missing link between Ca2+ store depletion and SOC influx, serving as a Ca2+ sensor that translocates upon store depletion to the plasma membrane to activate CRAC channels.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Animals , Biotinylation , Calcium Signaling , Cell Line , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , EF Hand Motifs/genetics , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , Humans , Ion Transport , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microscopy, Immunoelectron , Models, Biological , Mutation/genetics , Protein Transport , Rats , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL
...