Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Appl Environ Microbiol ; 87(20): e0096721, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34378994

ABSTRACT

The plant pathogen Fusarium graminearum contains two α-tubulin isotypes (α1 and α2) and two ß-tubulin isotypes (ß1 and ß2). The functional roles of these tubulins in microtubule assembly are not clear. Previous studies reported that α1- and ß2-tubulin deletion mutants showed severe growth defects and hypersensitivity to carbendazim, which have not been well explained. Here, we investigated the interaction between α- and ß-tubulin of F. graminearum. Colocalization experiments demonstrated that ß1- and ß2-tubulin are colocalized. Coimmunoprecipitation experiments suggested that ß1-tubulin binds to both α1- and α2-tubulin and that ß2-tubulin can also bind to α1- or α2-tubulin. Interestingly, deletion of α1-tubulin increased the interaction between ß2-tubulin and α2-tubulin. Microtubule observation assays showed that deletion of α1-tubulin completely disrupted ß1-tubulin-containing microtubules and significantly decreased ß2-tubulin-containing microtubules. Deletion of α2-, ß1-, or ß2-tubulin had no obvious effect on the microtubule cytoskeleton. However, microtubules in α1- and ß2-tubulin deletion mutants were easily depolymerized in the presence of carbendazim. The sexual reproduction assay indicates that α1- and ß1-tubulin deletion mutants could not produce asci and ascospores. These results implied that α1-tubulin may be essential for the microtubule cytoskeleton. However, our Δα1-2×α2 mutant (α1-tubulin deletion mutant containing two copies of α2-tubulin) exhibited normal microtubule network, growth, and sexual reproduction. Interestingly, the Δα1-2×α2 mutant was still hypersensitive to carbendazim. In addition, both ß1-tubulin and ß2-tubulin were found to bind the mitochondrial outer membrane voltage-dependent anion channel (VDAC), indicating that they could regulate the function of VDAC. IMPORTANCE In this study, we found that F. graminearum contains four different α-/ß-tubulin heterodimers (α1-/ß1-, α1-/ß2-, α2-/ß1-, and α2-/ß2-tubulin heterodimers), and they assemble together into a single microtubule. Moreover, α1- and α2-tubulins are functionally interchangeable in microtubule assembly, vegetative growth, and sexual reproduction. These results provide more insights into the functional roles of different tubulins of F. graminearum, which could be helpful for purification of tubulin heterodimers and development of new tubulin-binding agents.


Subject(s)
Fusarium/physiology , Microtubules/physiology , Tubulin/physiology , Fungal Proteins/physiology , Fusarium/genetics , Fusarium/growth & development , Voltage-Dependent Anion Channels/physiology
2.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298976

ABSTRACT

The voltage-dependent anion channel (VDAC) is the primary regulating pathway of water-soluble metabolites and ions across the mitochondrial outer membrane. When reconstituted into lipid membranes, VDAC responds to sufficiently large transmembrane potentials by transitioning to gated states in which ATP/ADP flux is reduced and calcium flux is increased. Two otherwise unrelated cytosolic proteins, tubulin, and α-synuclein (αSyn), dock with VDAC by a novel mechanism in which the transmembrane potential draws their disordered, polyanionic C-terminal domains into and through the VDAC channel, thus physically blocking the pore. For both tubulin and αSyn, the blocked state is observed at much lower transmembrane potentials than VDAC gated states, such that in the presence of these cytosolic docking proteins, VDAC's sensitivity to transmembrane potential is dramatically increased. Remarkably, the features of the VDAC gated states relevant for bioenergetics-reduced metabolite flux and increased calcium flux-are preserved in the blocked state induced by either docking protein. The ability of tubulin and αSyn to modulate mitochondrial potential and ATP production in vivo is now supported by many studies. The common physical origin of the interactions of both tubulin and αSyn with VDAC leads to a general model of a VDAC inhibitor, facilitates predictions of the effect of post-translational modifications of known inhibitors, and points the way toward the development of novel therapeutics targeting VDAC.


Subject(s)
Anions/metabolism , Cell Respiration/physiology , Intrinsically Disordered Proteins/physiology , Mitochondrial Membranes/drug effects , Tubulin/physiology , Voltage-Dependent Anion Channels/antagonists & inhibitors , alpha-Synuclein/physiology , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Respiration/drug effects , Fluoresceins/chemistry , Humans , Intrinsically Disordered Proteins/chemistry , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Kinetics , Mitochondrial Membranes/metabolism , Models, Molecular , Osmolar Concentration , Potassium Chloride/pharmacology , Protein Conformation , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Transport , Sequence Alignment , Sulfonic Acids/chemistry , Tubulin/chemistry , Voltage-Dependent Anion Channels/chemistry , Voltage-Dependent Anion Channels/physiology , alpha-Synuclein/chemistry
3.
Plant Physiol ; 185(4): 1523-1541, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33598675

ABSTRACT

Membrane voltage arises from the transport of ions through ion-translocating ATPases, ion-coupled transport of solutes, and ion channels, and is an integral part of the bioenergetic "currency" of the membrane. The dynamics of membrane voltage-so-called action, systemic, and variation potentials-have also led to a recognition of their contributions to signal transduction, both within cells and across tissues. Here, we review the origins of our understanding of membrane voltage and its place as a central element in regulating transport and signal transmission. We stress the importance of understanding voltage as a common intermediate that acts both as a driving force for transport-an electrical "substrate"-and as a product of charge flux across the membrane, thereby interconnecting all charge-carrying transport across the membrane. The voltage interconnection is vital to signaling via second messengers that rely on ion flux, including cytosolic free Ca2+, H+, and the synthesis of reactive oxygen species generated by integral membrane, respiratory burst oxidases. These characteristics inform on the ways in which long-distance voltage signals and voltage oscillations give rise to unique gene expression patterns and influence physiological, developmental, and adaptive responses such as systemic acquired resistance to pathogens and to insect herbivory.


Subject(s)
Biological Transport/physiology , Cell Membrane/physiology , Ion Transport/physiology , Plant Development , Signal Transduction/physiology , Voltage-Dependent Anion Channels/physiology
4.
Proc Natl Acad Sci U S A ; 117(35): 21740-21746, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817533

ABSTRACT

The Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) anion channel is essential for epithelial salt-water balance. CFTR mutations cause cystic fibrosis, a lethal incurable disease. In cells CFTR is activated through the cAMP signaling pathway, overstimulation of which during cholera leads to CFTR-mediated intestinal salt-water loss. Channel activation is achieved by phosphorylation of its regulatory (R) domain by cAMP-dependent protein kinase catalytic subunit (PKA). Here we show using two independent approaches--an ATP analog that can drive CFTR channel gating but is unsuitable for phosphotransfer by PKA, and CFTR mutants lacking phosphorylatable serines--that PKA efficiently opens CFTR channels through simple binding, under conditions that preclude phosphorylation. Unlike when phosphorylation happens, CFTR activation by PKA binding is completely reversible. Thus, PKA binding promotes release of the unphosphorylated R domain from its inhibitory position, causing full channel activation, whereas phosphorylation serves only to maintain channel activity beyond termination of the PKA signal. The results suggest two levels of CFTR regulation in cells: irreversible through phosphorylation, and reversible through R-domain binding to PKA--and possibly also to other members of a large network of proteins known to interact with the channel.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Voltage-Dependent Anion Channels/metabolism , Adenosine Triphosphate/metabolism , Animals , Anions/metabolism , Biophysical Phenomena , Cyclic AMP-Dependent Protein Kinases/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Ion Channel Gating/physiology , Mutagenesis, Site-Directed , Nucleotides/metabolism , Oocytes/metabolism , Patch-Clamp Techniques/methods , Phosphorylation , Protein Binding/physiology , Serine/metabolism , Voltage-Dependent Anion Channels/physiology , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
5.
Cell Biol Int ; 44(11): 2178-2181, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32716117

ABSTRACT

A recent study suggests that voltage-dependent anion channel (VDAC) oligomer pores promote mitochondrial outer membrane permeabilization and allow mitochondrial DNA (mtDNA) to be released into the cytosol in live cells. It challenges the notion that only occurs in apoptotic cells via BAX/BAK macropores. Cytosolic mtDNA activates cyclic GMP-AMP synthase (cGAS)-stimulator of IFN gene (STING) pathway and triggers type I interferon (IFN) response thereafter, which ultimately causes systemic lupus erythematosus. Mechanistically, mtDNA can interact with three positively charged residues (Lys12, Arg15, and Lys20) at the N-terminus of VDAC1, thereby strengthening VDAC1 oligomerization and facilitating mtDNA release. In addition, there are other pathways that can mediate mtDNA release, such as BAX/BAK macropores and virus-derived pores. The mtDNA released into the cytosol also triggers type I IFN response via the generally accepted cGAS-STING-TANK-binding kinase 1-IFN regulatory factor 3 axis. Collectively, VDAC oligomer pores provide us an attractive direction for us to understand mtDNA release-related diseases.


Subject(s)
DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Voltage-Dependent Anion Channels/metabolism , Apoptosis , Cytosol/metabolism , DNA, Mitochondrial/genetics , Mitochondrial Membranes/metabolism , Signal Transduction , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channel 1/physiology , Voltage-Dependent Anion Channels/physiology , bcl-2-Associated X Protein/metabolism
6.
Article in English | MEDLINE | ID: mdl-32428575

ABSTRACT

Olesoxime is a cholesterol-like neuroprotective compound that targets to mitochondrial voltage dependent anion channels (VDACs). VDACs were also found in the plasma membrane and highly expressed in the presynaptic compartment. Here, we studied the effects of olesoxime and VDAC inhibitors on neurotransmission in the mouse neuromuscular junction. Electrophysiological analysis revealed that olesoxime suppressed selectively evoked neurotransmitter release in response to a single stimulus and 20 Hz activity. Also olesoxime decreased the rate of FM1-43 dye loss (an indicator of synaptic vesicle exocytosis) at low frequency stimulation and 20 Hz. Furthermore, an increase in extracellular Cl- enhanced the action of olesoxime on the exocytosis and olesoxime increased intracellular Cl- levels. The effects of olesoxime on the evoked synaptic vesicle exocytosis and [Cl-]i were blocked by membrane-permeable and impermeable VDAC inhibitors. Immunofluorescent labeling pointed on the presence of VDACs on the synaptic membranes. Rotenone-induced mitochondrial dysfunction perturbed the exocytotic release of FM1-43 and cell-permeable VDAC inhibitor (but not olesoxime or impermeable VDAC inhibitor) partially mitigated the rotenone-driven alterations in the FM1-43 unloading and mitochondrial superoxide production. Thus, olesoxime restrains neurotransmission by acting on plasmalemmal VDACs whose activation can limit synaptic vesicle exocytosis probably via increasing anion flux into the nerve terminals.


Subject(s)
Cholestenones/pharmacology , Neuroprotective Agents/pharmacology , Phrenic Nerve/drug effects , Synaptic Vesicles/drug effects , Voltage-Dependent Anion Channels/physiology , Animals , Cholesterol , Diaphragm/drug effects , Diaphragm/innervation , Diaphragm/physiology , Exocytosis/drug effects , Mice , Phrenic Nerve/physiology , Synaptic Potentials/drug effects , Voltage-Dependent Anion Channels/antagonists & inhibitors
7.
J Pharmacol Sci ; 143(3): 176-181, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32386905

ABSTRACT

The volume-regulated anion channel (VRAC) plays a central role in maintaining cell volume in response to osmotic stress. Leucine-rich repeat-containing 8A (LRRC8A) was recently identified as an essential component of VRAC although other Cl- channels were also suggested to contribute to VRAC. VRAC is activated when a cell is challenged with a hypotonic environment or even in isotonic conditions challenged with different stimuli. It is not clear how VRAC is activated and whether activation of VRAC in hypotonic and isotonic conditions share the same mechanism. In this present study, we investigated relative contribution of LRRC8A and anoctamin 1(ANO1) to VRAC currents activated by fetal bovine serum (FBS) in isotonic condition, and studied the role of intracellular Ca2+ in this activation. We used CRISPR/Cas9 gene editing approach, electrophysiology, and pharmacology approaches to show that VRAC currents induced by FBS is mostly mediated by LRRC8A in HEK293 cells, but also with significant contribution from ANO1. FBS induces Ca2+ transients and these Ca2+ signals are required for the activation of VRAC by serum. These findings will help to further understand the mechanism in activation of VRAC.


Subject(s)
Anoctamin-1/physiology , Calcium/metabolism , Cell Size , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Voltage-Dependent Anion Channels/metabolism , Voltage-Dependent Anion Channels/physiology , Animals , CRISPR-Associated Protein 9/genetics , Cattle , Chloride Channels/metabolism , Chloride Channels/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing , HEK293 Cells , Humans , Osmotic Pressure/physiology , Serum
8.
Biomolecules ; 10(1)2020 01 03.
Article in English | MEDLINE | ID: mdl-31947864

ABSTRACT

The bacterial channel SecYEG efficiently translocates both hydrophobic and hydrophilic proteins across the plasma membrane. Translocating polypeptide chains may dislodge the plug, a half helix that blocks the permeation of small molecules, from its position in the middle of the aqueous translocation channel. Instead of the plug, six isoleucines in the middle of the membrane supposedly seal the channel, by forming a gasket around the translocating polypeptide. However, this hypothesis does not explain how the tightness of the gasket may depend on membrane potential. Here, we demonstrate voltage-dependent closings of the purified and reconstituted channel in the presence of ligands, suggesting that voltage sensitivity may be conferred by motor protein SecA, ribosomes, signal peptides, and/or translocating peptides. Yet, the presence of a voltage sensor intrinsic to SecYEG was indicated by voltage driven closure of pores that were forced-open either by crosslinking the plug to SecE or by plug deletion. We tested the involvement of SecY's half-helix 2b (TM2b) in voltage sensing, since clearly identifiable gating charges are missing. The mutation L80D accelerated voltage driven closings by reversing TM2b's dipolar orientation. In contrast, the L80K mutation decelerated voltage induced closings by increasing TM2b's dipole moment. The observations suggest that TM2b is part of a larger voltage sensor. By partly aligning the combined dipole of this sensor with the orientation of the membrane-spanning electric field, voltage may drive channel closure.


Subject(s)
Protein Transport/physiology , SEC Translocation Channels/metabolism , Voltage-Dependent Anion Channels/metabolism , Bacteria/metabolism , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/metabolism , Peptides/metabolism , SEC Translocation Channels/physiology , Voltage-Dependent Anion Channels/physiology
9.
Cells ; 9(1)2019 Dec 24.
Article in English | MEDLINE | ID: mdl-31878344

ABSTRACT

Transient receptor potential ankyrin 1 channel (TRPA1) serves as a key sensor for reactive electrophilic compounds across all species. Its sensitivity to temperature, however, differs among species, a variability that has been attributed to an evolutionary divergence. Mouse TRPA1 was implicated in noxious cold detection but was later also identified as one of the prime noxious heat sensors. Moreover, human TRPA1, originally considered to be temperature-insensitive, turned out to act as an intrinsic bidirectional thermosensor that is capable of sensing both cold and heat. Using electrophysiology and modeling, we compare the properties of human and mouse TRPA1, and we demonstrate that both orthologues are activated by heat, and their kinetically distinct components of voltage-dependent gating are differentially modulated by heat and cold. Furthermore, we show that both orthologues can be strongly activated by cold after the concurrent application of voltage and heat. We propose an allosteric mechanism that could account for the variability in TRPA1 temperature responsiveness.


Subject(s)
TRPA1 Cation Channel/metabolism , Amino Acid Sequence , Animals , Cold Temperature , Electrophysiology/methods , HEK293 Cells , Hot Temperature , Humans , Mice , Models, Biological , Species Specificity , Voltage-Dependent Anion Channels/metabolism , Voltage-Dependent Anion Channels/physiology
10.
Sci Rep ; 9(1): 5392, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30931966

ABSTRACT

Regulation of cellular volume is an essential process to balance volume changes during cell proliferation and migration or when intracellular osmolality increases due to transepithelial transport. We previously characterized the key role of volume-regulated anion channels (VRAC) in the modulation of the volume of trabecular meshwork (TM) cells and, in turn, the aqueous humour (AH) outflow from the eye. The balance between the secretion and the drainage of AH determines the intraocular pressure (IOP) that is the major casual risk factor for glaucoma. Glaucoma is an ocular disease that causes irreversible blindness due to the degeneration of retinal ganglion cells. The recent identification of Leucine-Rich Repeat-Containing 8 (LRRC8A-E) proteins as the molecular components of VRAC opens the field to elucidate their function in the physiology of TM and glaucoma. Human TM cells derived from non-glaucomatous donors and from open-angle glaucoma patients were used to determine the expression and the functional activity of LRRC8-mediated channels. Expression levels of LRRC8A-E subunits were decreased in HTM glaucomatous cells compared to normotensive HTM cells. Consequently, the activity of VRAC currents and volume regulation of TM cells were significantly affected. Impaired cell volume regulation will likely contribute to altered aqueous outflow and intraocular pressure.


Subject(s)
Glaucoma, Open-Angle/genetics , Membrane Proteins/genetics , Trabecular Meshwork/metabolism , Voltage-Dependent Anion Channels/genetics , Aged , Aqueous Humor/cytology , Aqueous Humor/metabolism , Aqueous Humor/physiology , Cell Line , Cell Size , Cells, Cultured , Female , Gene Expression Profiling/methods , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/physiopathology , Humans , Intraocular Pressure/physiology , Male , Membrane Proteins/metabolism , Middle Aged , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology , Trabecular Meshwork/cytology , Voltage-Dependent Anion Channels/metabolism , Voltage-Dependent Anion Channels/physiology
11.
Biochim Biophys Acta Biomembr ; 1859(11): 2213-2223, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28888364

ABSTRACT

Inhibition of cell respiration by high concentrations of glucose (glucose repression), known as "Crabtree effect", has been demonstrated for various cancerous strains, highly proliferating cells and yeast lines. Although significant progress in understanding metabolic events associated with the glucose repression of cell respiration has been achieved, it is not yet clear whether the Crabtree effect is the result of a limited activity of the respiratory chain, or of some glucose-mediated regulation of mitochondrial metabolic state. In this work we propose an electrical mechanism of glucose repression of the yeast S. cerevisiae, resulting from generation of the mitochondrial outer membrane potential (OMP) coupled to the direct oxidation of cytosolic NADH in mitochondria. This yeast-type mechanism of OMP generation is different from the earlier proposed VDAC-hexokinase-mediated voltage generation of cancer-type, associated with the mitochondrial outer membrane. The model was developed assuming that VDAC is more permeable to NADH than to NAD+. Thermodynamic estimations of OMP, generated as a result of NADH(2-)/NAD+(1-) turnover through the outer membrane, demonstrated that the values of calculated negative OMP match the known range of VDAC voltage sensitivity, thus suggesting a possibility of OMP-dependent VDAC-mediated regulation of cell energy metabolism. According to the proposed mechanism, we suggest that the yeast-type Crabtree effect is the result of a fast VDAC-mediated electrical repression of mitochondria due to a decrease in the outer membrane permeability to charged metabolites and owing their redistribution between the mitochondrial intermembrane space and the cytosol, both controlled by metabolically-derived OMP.


Subject(s)
Electric Conductivity , Glucose/pharmacology , Saccharomyces cerevisiae , Thermodynamics , Voltage-Dependent Anion Channels/physiology , Cell Membrane Permeability/drug effects , Cell Respiration/drug effects , Electrophysiological Phenomena/drug effects , Glucose/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Oxidation-Reduction/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/physiology , Voltage-Dependent Anion Channels/metabolism
12.
Neurochem Res ; 42(9): 2551-2559, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28401401

ABSTRACT

Anion channels and connexin hemichannels are permeable to amino acid neurotransmitters. It is hypothesized that these conductive pathways release GABA, thereby influencing ambient GABA levels and tonic GABAergic inhibition. To investigate this, we measured the effects of anion channel/hemichannel antagonists on tonic GABA currents of rat hippocampal neurons. In contrast to predictions, blockade of anion channels and hemichannels with NPPB potentiated tonic GABA currents of neurons in culture and acute hippocampal slices. In contrast, the anion channel/hemichannel antagonist carbenoxolone (CBX) inhibited tonic currents. These findings could result from alterations of ambient GABA concentration or direct effects on GABAA receptors. To test for effects on GABAA receptors, we measured currents evoked by exogenous GABA. Coapplication of NPPB with GABA potentiated GABA-evoked currents. CBX dose-dependently inhibited GABA-evoked currents. These results are consistent with direct effects of NPPB and CBX on GABAA receptors. GABA release from hippocampal cell cultures was directly measured using HPLC. Inhibition of anion channels with NPPB or CBX did not affect GABA release from cultured hippocampal neurons. NPPB reduced GABA release from pure astrocytic cultures by 21%, but the total GABA release from astrocytes was small compared to that of mixed cultures. These data indicate that drugs commonly used to antagonize anion channels and connexin hemichannels may affect tonic currents via direct effects on GABAA receptors and have negligible effects on ambient GABA concentrations. Interpretation of experiments using NPPB or CBX should include consideration of their effects on tonic GABA currents.


Subject(s)
Connexins/antagonists & inhibitors , Connexins/physiology , GABA-A Receptor Antagonists/pharmacology , Receptors, GABA-A/physiology , Voltage-Dependent Anion Channels/antagonists & inhibitors , Voltage-Dependent Anion Channels/physiology , Aminobenzoates/pharmacology , Animals , Animals, Newborn , Carbenoxolone/pharmacology , Cells, Cultured , Female , Hippocampus/drug effects , Hippocampus/physiology , Male , Nitrobenzoates/pharmacology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacology
13.
Neuropharmacology ; 127: 224-242, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28396143

ABSTRACT

κ-Hexatoxins (κ-HXTXs) are a family of excitotoxic insect-selective neurotoxins from Australian funnel-web spiders that are lethal to a wide range of insects, but display no toxicity towards vertebrates. The prototypic κ-HXTX-Hv1c selectively blocks native and expressed cockroach large-conductance calcium-activated potassium (BKCa or KCa1.1) channels, but not their mammalian orthologs. Despite this potent and selective action on insect KCa1.1 channels, we found that the classical KCa1.1 blockers paxilline, charybdotoxin and iberiotoxin, which all block insect KCa1.1 channels, are not lethal in crickets. We therefore used whole-cell patch-clamp analysis of cockroach dorsal unpaired median (DUM) neurons to study the effects of κ-HXTX-Hv1c on sodium-activated (KNa), delayed-rectifier (KDR) and 'A-type' transient (KA) K+ channels. 1 µM κ-HXTX-Hv1c failed to significantly inhibit cockroach KNa and KDR channels, but did cause a 30 ± 7% saturating inhibition of KA channel currents, possibly via a Kv4 (Shal-like) action. However, this modest action at such a high concentration of κ-HXTX-Hv1c would indicate a different lethal target. Accordingly, we assessed the actions of κ-HXTX-Hv1c on neurotransmitter-gated ion channels in cockroach DUM neurons. We found that κ-HXTX-Hv1c failed to produce any major effects on GABAA or glutamate-Cl receptors but dramatically slowed nicotine-evoked ACh receptor (nAChR) current decay and reversed nAChR desensitization. These actions occurred without any alterations to nAChR current amplitude or the nicotine concentration-response curve, and are consistent with a positive allosteric modulation of nAChRs. κ-HXTX-Hv1c therefore represents the first venom peptide that selectively modulates insect nAChRs with a mode of action similar to the excitotoxic insecticide spinosyn A. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'


Subject(s)
Neurotoxins/toxicity , Receptors, Nicotinic/drug effects , Spider Venoms/toxicity , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Analysis of Variance , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Electric Stimulation , Gryllidae , Humans , Indoles/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Neuroblastoma/pathology , Neurons/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Receptors, Nicotinic/chemistry , Voltage-Dependent Anion Channels/physiology
14.
Biochim Biophys Acta Bioenerg ; 1858(8): 665-673, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28283400

ABSTRACT

The voltage-dependent anion channel (VDAC) is a pore located at the outer membrane of the mitochondrion. It allows the entry and exit of numerous ions and metabolites between the cytosol and the mitochondrion. Flux through the pore occurs in an active way: first, it depends on the open or closed state and second, on the negative or positive charges of the different ion species passing through the pore. The flux of essential metabolites, such as ATP, determines the functioning of the mitochondria to a noxious stimulus. Moreover, VDAC acts as a platform for many proteins and in so doing supports glycolysis and prevents apoptosis by interacting with hexokinase, or members of the Bcl-2 family, respectively. VDAC is thus involved in the choice the cells make to survive or die, which is particularly relevant to cancer cells. For these reasons, VDAC has become a potential therapeutic target to fight cancer but also other diseases in which mitochondrial metabolism is modified. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.


Subject(s)
Mitochondrial Membranes/metabolism , Mitochondrial Proteins/physiology , Neoplasm Proteins/physiology , Neoplasms/metabolism , Voltage-Dependent Anion Channels/physiology , Animals , Apoptosis/physiology , Biological Transport , Calcium Signaling , Energy Metabolism , Hexokinase/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitophagy/physiology , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Structure-Activity Relationship , Substrate Specificity , Voltage-Dependent Anion Channels/antagonists & inhibitors , Voltage-Dependent Anion Channels/chemistry , Voltage-Dependent Anion Channels/genetics
15.
Nat Methods ; 14(3): 271-274, 2017 03.
Article in English | MEDLINE | ID: mdl-28114289

ABSTRACT

Optogenetics uses light exposure to manipulate physiology in genetically modified organisms. Abundant tools for optogenetic excitation are available, but the limitations of current optogenetic inhibitors present an obstacle to demonstrating the necessity of neuronal circuits. Here we show that anion channelrhodopsins can be used to specifically and rapidly inhibit neural systems involved in Drosophila locomotion, wing expansion, memory retrieval and gustation, thus demonstrating their broad utility in the circuit analysis of behavior.


Subject(s)
Behavior, Animal/drug effects , Drosophila/physiology , Neural Pathways/physiology , Optogenetics/methods , Rhodopsin/pharmacology , Action Potentials/physiology , Animals , Behavior, Animal/physiology , Light , Locomotion/physiology , Neurons/physiology , Organisms, Genetically Modified , Taste Perception/physiology , Voltage-Dependent Anion Channels/physiology
17.
Physiol Res ; 66(1): 63-73, 2017 03 31.
Article in English | MEDLINE | ID: mdl-27782747

ABSTRACT

Patch clamp recordings carried out in the inside-out configuration revealed activity of three kinds of channels: nonselective cation channels, small-conductance K(+) channels, and large-conductance anion channels. The nonselective cation channels did not distinguish between Na(+) and K(+). The unitary conductance of these channels reached 28 pS in a symmetrical concentration of 200 mM NaCl. A lower value of this parameter was recorded for the small-conductance K(+) channels and in a 50-fold gradient of K(+) (200 mM/4 mM) it reached 8 pS. The high selectivity of these channels to potassium was confirmed by the reversal potential (-97 mV), whose value was close to the equilibrium potential for potassium (-100 mV). One of the features of the largeconductance anion channels was high conductance amounting to 493 pS in a symmetrical concentration of 200 mM NaCl. The channels exhibited three subconductance levels. Moreover, an increase in the open probability of the channels at voltages close to zero was observed. The anion selectivity of the channels was low, because the channels were permeable to both Cl(-) and gluconate - a large anion. Research on the calcium dependence revealed that internal calcium activates nonselective cation channels and small-conductance K(+) channels, but not largeconductance anion channels.


Subject(s)
Cell Membrane/physiology , Fibroblasts/physiology , Ion Channels/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Voltage-Dependent Anion Channels/physiology , Animals , Cell Line , Mice
18.
Handb Exp Pharmacol ; 240: 71-101, 2017.
Article in English | MEDLINE | ID: mdl-27783269

ABSTRACT

Mitochondria are the "power house" of a cell continuously generating ATP to ensure its proper functioning. The constant production of ATP via oxidative phosphorylation demands a large electrochemical force that drives protons across the highly selective and low-permeable mitochondrial inner membrane. Besides the conventional role of generating ATP, mitochondria also play an active role in calcium signaling, generation of reactive oxygen species (ROS), stress responses, and regulation of cell-death pathways. Deficiencies in these functions result in several pathological disorders like aging, cancer, diabetes, neurodegenerative and cardiovascular diseases. A plethora of ion channels and transporters are present in the mitochondrial inner and outer membranes which work in concert to preserve the ionic equilibrium of a cell for the maintenance of cell integrity, in physiological as well as pathophysiological conditions. For, e.g., mitochondrial cation channels KATP and BKCa play a significant role in cardioprotection from ischemia-reperfusion injury. In addition to the cation channels, mitochondrial anion channels are equally essential, as they aid in maintaining electro-neutrality by regulating the cell volume and pH. This chapter focusses on the information on molecular identity, structure, function, and physiological relevance of mitochondrial chloride channels such as voltage dependent anion channels (VDACs), uncharacterized mitochondrial inner membrane anion channels (IMACs), chloride intracellular channels (CLIC) and the aspects of forthcoming chloride channels.


Subject(s)
Chloride Channels/physiology , Mitochondria/metabolism , Voltage-Dependent Anion Channels/physiology , Animals , Humans , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Permeability Transition Pore
19.
Biochem Biophys Res Commun ; 477(3): 490-4, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27318085

ABSTRACT

Dietary trans fatty acids (TFAs) are known to increase the risk of cardiovascular diseases by altering plasma lipid profile and activating various inflammatory signaling pathways. Here we show that elaidic acid (EA), the most abundant TFA in diet, alters the electrophysiological properties of voltage-dependent anion channel (VDAC) of mitochondria. Purified bovine brain VDAC, when incorporated in the planar lipid bilayer (PLB) composed of 1,2-diphytanoyl-sn-glycero-3 phosphatidyl choline (DPhPC) and EA in a 9 to 1 ratio (wt/wt), exhibited complete closing events at different voltages. The closing events were observed at even -10 mV, a voltage at which VDAC usually remains fully open all the time. Additionally, the voltage sensitivity of VDAC was lost in presence of EA; the channel conductance did not decrease with increasing voltages. In identical experimental conditions, membrane containing oleic acid (OA), the cis isomer of EA did not produce any such effect. We propose that EA possibly exerts its adverse effect by modulating VDAC.


Subject(s)
Mitochondria/physiology , Oleic Acid/pharmacology , Voltage-Dependent Anion Channels/drug effects , Animals , Mitochondria/drug effects , Oleic Acids , Voltage-Dependent Anion Channels/physiology
20.
Nat Rev Mol Cell Biol ; 17(5): 293-307, 2016 05.
Article in English | MEDLINE | ID: mdl-27033257

ABSTRACT

Cells need to regulate their volume to counteract osmotic swelling or shrinkage, as well as during cell division, growth, migration and cell death. Mammalian cells adjust their volume by transporting potassium, sodium, chloride and small organic osmolytes using plasma membrane channels and transporters. This generates osmotic gradients, which drive water in and out of cells. Key players in this process are volume-regulated anion channels (VRACs), the composition of which has recently been identified and shown to encompass LRRC8 heteromers. VRACs also transport metabolites and drugs and function in extracellular signal transduction, apoptosis and anticancer drug resistance.


Subject(s)
Cell Size , Voltage-Dependent Anion Channels/physiology , Animals , Apoptosis , Cell Membrane , Cell Membrane Permeability , Humans , Ion Transport , Membrane Potentials , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...