Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 16092, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997408

ABSTRACT

Thermally stable full-length scorpion toxin peptides and partially degraded peptides with complete disulfide bond pairing are valuable natural peptide resources in traditional Chinese scorpion medicinal material. However, their pharmacological activities are largely unknown. This study discovered BmKcug1a-P1, a novel N-terminal degraded peptide, in this medicinal material. BmKcug1a-P1 inhibited hKv1.2 and hKv1.3 potassium channels with IC50 values of 2.12 ± 0.27 µM and 1.54 ± 0.28 µM, respectively. To investigate the influence of N-terminal amino acid loss on the potassium channel inhibiting activities, three analogs (i.e., full-length BmKcug1a, BmKcug1a-P1-D2 and BmKcug1a-P1-D4) of BmKcug1a-P1 were prepared, and their potassium channel inhibiting activities on hKv1.3 channel were verified by whole-cell patch clamp technique. Interestingly, the potassium channel inhibiting activity of full-length BmKcug1a on the hKv1.3 channel was significantly improved compared to its N-terminal degraded form (BmKcug1a-P1), while the activities of two truncated analogs (i.e., BmKcug1a-P1-D2 and BmKcug1a-P1-D4) were similar to that of BmKcug1a-P1. Extensive alanine-scanning experiments identified the bonding interface (including two key functional residues, Asn30 and Arg34) of BmKcug1a-P1. Structural and functional dissection further elucidated whether N-terminal residues of the peptide are located at the bonding interface is important in determining whether the N-terminus significantly influences the potassium channel inhibiting activity of the peptide. Altogether, this research identified a novel N-terminal degraded active peptide, BmKcug1a-P1, from traditional Chinese scorpion medicinal material and elucidated how the N-terminus of peptides influences their potassium channel inhibiting activity, contributing to the functional identification and molecular truncation optimization of full-length and degraded peptides from traditional Chinese scorpion medicinal material Buthus martensii Karsch.


Subject(s)
Peptides , Potassium Channel Blockers , Scorpion Venoms , Scorpions , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Scorpions/chemistry , Scorpion Venoms/chemistry , Scorpion Venoms/pharmacology , Animals , Peptides/chemistry , Peptides/pharmacology , Humans , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/chemistry , Proteolysis , Kv1.2 Potassium Channel/metabolism , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/chemistry , Protein Stability , Amino Acid Sequence , Patch-Clamp Techniques , HEK293 Cells
2.
J Neurophysiol ; 125(5): 1954-1972, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33852808

ABSTRACT

Temporal coding precision of bushy cells in the ventral cochlear nucleus (VCN), critical for sound localization and communication, depends on the generation of rapid and temporally precise action potentials (APs). Voltage-gated potassium (Kv) channels are critically involved in this. The bushy cells in rat VCN express Kv1.1, 1.2, 1.3, 1.6, 3.1, 4.2, and 4.3 subunits. The Kv1.1 subunit contributes to the generation of a temporally precise single AP. However, the understanding of the functions of other Kv subunits expressed in the bushy cells is limited. Here, we investigated the functional diversity of Kv subunits concerning their contributions to temporal coding. We characterized the electrophysiological properties of the Kv channels with different subunits using whole cell patch-clamp recording and pharmacological methods. The neuronal firing pattern changed from single to multiple APs only when the Kv1.1 subunit was blocked. The Kv subunits, including the Kv1.1, 1.2, 1.6, or 3.1, were involved in enhancing temporal coding by lowering membrane excitability, shortening AP latencies, reducing jitter, and regulating AP kinetics. Meanwhile, all the Kv subunits contributed to rapid repolarization and sharpening peaks by narrowing half-width and accelerating fall rate, and the Kv1.1 subunit also affected the depolarization of AP. The Kv1.1, 1.2, and 1.6 subunits endowed bushy cells with a rapid time constant and a low input resistance of membrane for enhancing spike timing precision. The present results indicate that the Kv channels differentially affect intrinsic membrane properties to optimize the generation of rapid and reliable APs for temporal coding.NEW & NOTEWORTHY This study investigates the roles of Kv channels in effecting precision using electrophysiological and pharmacological methods in bushy cells. Different Kv channels have varying electrophysiological characteristics, which contribute to the interplay between changes in the membrane properties and regulation of neuronal excitability which then improve temporal coding. We conclude that the Kv channels are specialized to promote the precise and rapid coding of acoustic input by optimizing the generation of reliable APs.


Subject(s)
Action Potentials/physiology , Cochlear Nucleus/physiology , Neurons/physiology , Potassium Channels, Voltage-Gated/physiology , Action Potentials/drug effects , Animals , Female , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.1 Potassium Channel/physiology , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/physiology , Kv1.6 Potassium Channel/antagonists & inhibitors , Kv1.6 Potassium Channel/physiology , Male , Neurons/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
3.
Peptides ; 123: 170172, 2020 01.
Article in English | MEDLINE | ID: mdl-31626826

ABSTRACT

Scorpion venom contains diverse bioactive peptides that can recognize and interact with membrane proteins such as ion channels. These natural toxins are believed to be useful tools for exploring the structure and function of ion channels. In this study, we characterized a K+-channel toxin gene, ImKTx96, from the venom gland cDNA library of the scorpion Isometrus maculates. The peptide deduced from the ImKTx96 precursor nucleotide sequence contains a signal peptide of 27 amino acid residues and a mature peptide of 29 residues with three disulfide bridges. Multiple sequence alignment indicated that ImKTx96 is similar with the scorpion toxins that typically target K+-channels. The recombined ImKTx96 peptide (rImKTx96) was expressed in the Escherichia coli system, and purified by GST-affinity chromatography and RP-HPLC. Results from whole-cell patch-clamp experiments revealed that rImKTx96 can inhibit the current of the Kv1.2 ion channel expressed in HEK293 cells. The 3D structure of ImKTx96 was constructed by molecular modeling, and the complex formed by ImKTx96 interacting with the Kv1.2 ion channel was obtained by molecular docking. Based on its structural features and pharmacological functions, ImKTx96 was identified as one member of K+-channel scorpion toxin α-KTx10 group and may be useful as a molecular probe for investigating the structure and function of the Kv1.2 ion channel.


Subject(s)
Arthropod Proteins/chemistry , Kv1.2 Potassium Channel , Peptides/chemistry , Potassium Channel Blockers/chemistry , Scorpion Venoms/chemistry , Scorpions/chemistry , Animals , Humans , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/chemistry
4.
Proc Natl Acad Sci U S A ; 116(37): 18700-18709, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31444298

ABSTRACT

Voltage-dependent potassium channels (Kvs) gate in response to changes in electrical membrane potential by coupling a voltage-sensing module with a K+-selective pore. Animal toxins targeting Kvs are classified as pore blockers, which physically plug the ion conduction pathway, or as gating modifiers, which disrupt voltage sensor movements. A third group of toxins blocks K+ conduction by an unknown mechanism via binding to the channel turrets. Here, we show that Conkunitzin-S1 (Cs1), a peptide toxin isolated from cone snail venom, binds at the turrets of Kv1.2 and targets a network of hydrogen bonds that govern water access to the peripheral cavities that surround the central pore. The resulting ectopic water flow triggers an asymmetric collapse of the pore by a process resembling that of inherent slow inactivation. Pore modulation by animal toxins exposes the peripheral cavity of K+ channels as a novel pharmacological target and provides a rational framework for drug design.


Subject(s)
Cell Membrane/drug effects , Drosophila Proteins/antagonists & inhibitors , Ion Channel Gating/drug effects , Kv1.2 Potassium Channel/antagonists & inhibitors , Mollusk Venoms/toxicity , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Animals , Cell Membrane/metabolism , Crystallography, X-Ray , Drosophila Proteins/genetics , Drosophila Proteins/isolation & purification , Drosophila Proteins/metabolism , Drug Design , Female , Hydrogen Bonding/drug effects , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/isolation & purification , Kv1.2 Potassium Channel/metabolism , Lethal Dose 50 , Molecular Docking Simulation , Molecular Dynamics Simulation , Mollusk Venoms/chemistry , Mutation , Oocytes , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/isolation & purification , Shaker Superfamily of Potassium Channels/metabolism , Water/chemistry , Water/metabolism , Xenopus laevis
5.
Toxins (Basel) ; 11(6)2019 06 12.
Article in English | MEDLINE | ID: mdl-31212818

ABSTRACT

MiDCA1, a phospholipase A2 (PLA2) neurotoxin isolated from Micrurus dumerilii carinicauda coral snake venom, inhibited a major component of voltage-activated potassium (Kv) currents (41 ± 3% inhibition with 1 µM toxin) in mouse cultured dorsal root ganglion (DRG) neurons. In addition, the selective Kv2.1 channel blocker guangxitoxin (GxTx-1E) and MiDCA1 competitively inhibited the outward potassium current in DRG neurons. MiDCA1 (1 µM) reversibly inhibited the Kv2.1 current by 55 ± 8.9% in a Xenopus oocyte heterologous system. The toxin showed selectivity for Kv2.1 channels over all the other Kv channels tested in this study. We propose that Kv2.1 channel blockade by MiDCA1 underlies the toxin's action on acetylcholine release at mammalian neuromuscular junctions.


Subject(s)
Coral Snakes , Elapid Venoms/toxicity , Kv1.2 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/toxicity , Animals , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/physiology , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Oocytes/physiology , Phospholipases A2 , Xenopus
6.
CNS Neurol Disord Drug Targets ; 18(5): 382-404, 2019.
Article in English | MEDLINE | ID: mdl-30892167

ABSTRACT

BACKGROUND & OBJECTIVE: Kunitz-type venoms are bioactive proteins isolated from a wide variety of venomous animals. These venoms are involved in protease inhibitory activity or potassium channel blocking activity. Therefore, they are reported as an important source for lead drug candidates towards protease or channel associated diseases like neurological, metabolic and cardiovascular disorders. METHODS: This study aimed to check the inhibitory action of Kunitz-type venoms against potassium channels using computational tools. RESULTS: Among potassium channels, Human Voltage-Gated Potassium Channel 1.2 (hKv1.2) was used as a receptor whereas Kunitz-type peptides from the venoms of various species were selected as ligand dataset. CONCLUSION: This study helped in finding the binding interface between the receptor and ligand dataset for their potential therapeutic use in treating potassium channelopathies.


Subject(s)
Kv1.2 Potassium Channel/antagonists & inhibitors , Molecular Docking Simulation , Protein Interaction Mapping , Serine Proteinase Inhibitors/pharmacology , Venoms/pharmacology , Animals , Binding Sites/drug effects , Humans , Kv1.2 Potassium Channel/chemistry , Ligands , Molecular Structure , Protein Interaction Domains and Motifs/drug effects , Rats
7.
J Mol Graph Model ; 87: 197-203, 2019 03.
Article in English | MEDLINE | ID: mdl-30553160

ABSTRACT

The Tityus stigmurus scorpion is widely distributed in the Northeast of Brazil and is the main causal agent of human envenoming. The venom produced by this scorpion includes neurotoxins, which are peptides belonging to Family 2 toxins and are able to interact with ion channels. The KTx subfamily displays selectivity and affinity for Kv channel subtypes and the result of this interaction is the blockade of potassium channels, impairing vital functions. We report the optimized structural model of a transcript encoding a potassium channel blocker toxin from T. stigmurus. LC-MS analysis confirmed the presence of the toxin in the venom and the three-dimensional structure was obtained by computational homology modeling and refined by molecular dynamic simulations. Furthermore, docking simulations were performed using a Shaker kV-1.2 potassium channel from rats as receptor model and the contacts were identified revealing which amino acid residues and interactions could be involved in its blockade. These residues were mapped and their contact and electrostatic interactions were evaluated revealing the influence of positive lysine residues and the additional contribution of an asparagine to the stabilization of the complex, bringing new insights into the mechanism of action of this toxin.


Subject(s)
Kv1.2 Potassium Channel/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Scorpions/chemistry , Toxins, Biological/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Chromatography, Liquid , Humans , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/genetics , Mass Spectrometry , Molecular Conformation , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Protein Binding , Scorpions/genetics , Toxins, Biological/genetics , Toxins, Biological/pharmacology
8.
J Neurosci ; 38(46): 9883-9899, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30266739

ABSTRACT

The transmission of normal sensory and/or acute noxious information requires intact expression of pain-associated genes within the pain pathways of nervous system. Expressional changes of these genes after peripheral nerve injury are also critical for neuropathic pain induction and maintenance. Methyl-CpG-binding domain protein 1 (MBD1), an epigenetic repressor, regulates gene transcriptional activity. We report here that MBD1 in the primary sensory neurons of DRG is critical for the genesis of acute pain and neuropathic pain as DRG MBD1-deficient mice exhibit the reduced responses to acute mechanical, heat, cold, and capsaicin stimuli and the blunted nerve injury-induced pain hypersensitivities. Furthermore, DRG overexpression of MBD1 leads to spontaneous pain and evoked pain hypersensitivities in the WT mice and restores acute pain sensitivities in the MBD1-deficient mice. Mechanistically, MDB1 represses Oprm1 and Kcna2 gene expression by recruiting DNA methyltransferase DNMT3a into these two gene promoters in the DRG neurons. DRG MBD1 is likely a key player under the conditions of acute pain and neuropathic pain.SIGNIFICANCE STATEMENT In the present study, we revealed that the mice with deficiency of methyl-CpG-binding domain protein 1 (MBD1), an epigenetic repressor, in the DRG displayed the reduced responses to acute noxious stimuli and the blunted neuropathic pain. We also showed that DRG overexpression of MBD1 produced the hypersensitivities to noxious stimuli in the WT mice and rescued acute pain sensitivities in the MBD1-deficient mice. We have also provided the evidence that MDB1 represses Oprm1 and Kcna2 gene expression by recruiting DNA methyltransferase DNMT3a into these two gene promoters in the DRG neurons. DRG MBD1 may participate in the genesis of acute pain and neuropathic pain likely through regulating DNMT3a-controlled Oprm1 and Kcna2 gene expression in the DRG neurons.


Subject(s)
Acute Pain/metabolism , DNA-Binding Proteins/biosynthesis , Epigenesis, Genetic/physiology , Kv1.2 Potassium Channel/biosynthesis , Neuralgia/metabolism , Receptors, Opioid, mu/biosynthesis , Acute Pain/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Ganglia, Spinal/chemistry , Ganglia, Spinal/metabolism , Gene Silencing/physiology , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/genetics , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Sensory Receptor Cells/chemistry , Sensory Receptor Cells/metabolism
9.
Neuropharmacology ; 143: 228-238, 2018 12.
Article in English | MEDLINE | ID: mdl-30248306

ABSTRACT

Scorpion venom is an unmatched source of selective high-affinity ligands of potassium channels. There is a high demand for such compounds to identify and manipulate the activity of particular channel isoforms. The objective of this study was to obtain and characterize a specific ligand of voltage-gated potassium channel KV1.2. As a result, we report the remarkable selectivity of the peptide MeKTx11-1 (α-KTx 1.16) from Mesobuthus eupeus scorpion venom to this channel isoform. MeKTx11-1 is a high-affinity blocker of KV1.2 (IC50 ∼0.2 nM), while its activity against KV1.1, KV1.3, and KV1.6 is 10 000, 330 and 45 000 fold lower, respectively, as measured using the voltage-clamp technique on mammalian channels expressed in Xenopus oocytes. Two substitutions, G9V and P37S, convert MeKTx11-1 to its natural analog MeKTx11-3 (α-KTx 1.17) having 15 times lower activity and reduced selectivity to KV1.2. We produced MeKTx11-1 and MeKTx11-3 as well as their mutants MeKTx11-1(G9V) and MeKTx11-1(P37S) recombinantly and demonstrated that point mutations provide an intermediate effect on selectivity. Key structural elements that explain MeKTx11-1 specificity were identified by molecular modeling of the toxin-channel complexes. Confirming our molecular modeling predictions, site-directed transfer of these elements from the pore region of KV1.2 to KV1.3 resulted in the enhanced sensitivity of mutant KV1.3 channels to MeKTx11-1. We conclude that MeKTx11-1 may be used as a selective tool in neurobiology.


Subject(s)
Kv1.2 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Amino Acid Sequence , Animals , Blattellidae , Humans , Kv1.2 Potassium Channel/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Neurotoxins/chemistry , Neurotoxins/pharmacology , Oocytes , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , Rats , Recombinant Proteins , Scorpions , Structure-Activity Relationship , Xenopus laevis
10.
Peptides ; 108: 14-18, 2018 10.
Article in English | MEDLINE | ID: mdl-30121363

ABSTRACT

Recent reports have identified defensins as a new type of potassium channel inhibitors; differential binding mechanisms of human ß-defensins hBD1 and hBD2 point to complex interactions between human ß-defensins and potassium channels. We investigated the inhibitory effects of human defensins hBD3 and hBD4 on potassium channels. The data indicate that hBD3 is a voltage-gated channel subfamily A member 3 (Kv1.3) inhibitor with an IC50 value of 187.6 ±â€¯25.7 nM; 1 µM hBD4 inhibited 34.0 ±â€¯0.2% of Kv1.3 channel currents. Moreover, 1 µM hBD3 inhibited 50.6 ±â€¯3.6% of Kv1.2 channel currents and had smaller effects on Kv1.1, SKCa3, and IKCa channel currents; these effects differed from the Kv1.3 channel-specific inhibitors hBD1 and hBD2. Similar to the pharmacological profiles of hBD1 and hBD2, hBD4 had lower inhibitory effects on Kv1.1, Kv1.2, SKCa3, and IKCa channels. Subsequent mutagenesis and channel activation experiments confirmed that hBD3 binds in a manner similar to that of hBD1, interacting with the outer pore region of the Kv1.3 channel without affecting Kv1.3 channel activation. Thus, the data indicate that the human ß-defensin family is a novel group of potassium channel inhibitors with diverse types of human ß-defensin-potassium channel interactions.


Subject(s)
Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , beta-Defensins/pharmacology , HEK293 Cells , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kinetics , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/metabolism , Potassium Channels/metabolism , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/metabolism , beta-Defensins/metabolism
12.
J Neurosci ; 37(23): 5648-5658, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28483976

ABSTRACT

The voltage-gated K+ channel Kv2.1 has been intimately linked with neuronal apoptosis. After ischemic, oxidative, or inflammatory insults, Kv2.1 mediates a pronounced, delayed enhancement of K+ efflux, generating an optimal intracellular environment for caspase and nuclease activity, key components of programmed cell death. This apoptosis-enabling mechanism is initiated via Zn2+-dependent dual phosphorylation of Kv2.1, increasing the interaction between the channel's intracellular C-terminus domain and the SNARE (soluble N-ethylmaleimide-sensitive factor activating protein receptor) protein syntaxin 1A. Subsequently, an upregulation of de novo channel insertion into the plasma membrane leads to the critical enhancement of K+ efflux in damaged neurons. Here, we investigated whether a strategy designed to interfere with the cell death-facilitating properties of Kv2.1, specifically its interaction with syntaxin 1A, could lead to neuroprotection following ischemic injury in vivo The minimal syntaxin 1A-binding sequence of Kv2.1 C terminus (C1aB) was first identified via a far-Western peptide screen and used to create a protherapeutic product by conjugating C1aB to a cell-penetrating domain. The resulting peptide (TAT-C1aB) suppressed enhanced whole-cell K+ currents produced by a mutated form of Kv2.1 mimicking apoptosis in a mammalian expression system, and protected cortical neurons from slow excitotoxic injury in vitro, without influencing NMDA-induced intracellular calcium responses. Importantly, intraperitoneal administration of TAT-C1aB in mice following transient middle cerebral artery occlusion significantly reduced ischemic stroke damage and improved neurological outcome. These results provide strong evidence that targeting the proapoptotic function of Kv2.1 is an effective and highly promising neuroprotective strategy.SIGNIFICANCE STATEMENT Kv2.1 is a critical regulator of apoptosis in central neurons. It has not been determined, however, whether the cell death-enabling function of this K+ channel can be selectively targeted to improve neuronal survival following injury in vivo The experiments presented here demonstrate that the cell death-specific role of Kv2.1 can be uniquely modulated to provide neuroprotection in an animal model of acute ischemic stroke. We thus reveal a novel therapeutic strategy for neurological disorders that are accompanied by Kv2.1-facilitated forms of cell death.


Subject(s)
Apoptosis/drug effects , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/metabolism , Neuroprotective Agents/administration & dosage , Stroke/drug therapy , Stroke/physiopathology , Animals , Cells, Cultured , Drug Delivery Systems/methods , Female , Male , Potassium Channel Blockers/administration & dosage , Qa-SNARE Proteins/antagonists & inhibitors , Qa-SNARE Proteins/metabolism , Rats , Stroke/pathology , Treatment Outcome
13.
Physiol Res ; 66(3): 539-544, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28248535

ABSTRACT

Bis(3)-tacrine is a dimeric AChE inhibitor derived from tacrine with a potential to treat Alzheimer's disease. It was recently been reported to act as a fast off-rate antagonist of NMDA receptors with moderate affinity. In the present study, we aimed to explore whether bis(3)-tacrine could modulate the function of native sustained potassium current in cultured rat hippocampal neurons using whole-cell patch-clamp technique. We found that bis(3)-tacrine inhibited the amplitude of sustained potassium current in a reversible and concentration-dependent manner, with a potency two orders of magnitude higher than that of tacrine. The inhibition was voltage-independent between 0 to +60 mV. The IC(50) values for bis(3)-tacrine and tacrine inhibition of sustained potassium current were 0.45+/-0.07 and 50.5+/-4.8 microM, respectively. I-V curves showed a more potent inhibition of sustained potassium current by bis(3)-tacrine (1 microM) compared to tacrine at the same concentration. Bis(3)-tacrine hyperpolarized the activation curve of the current by 11.2 mV, albeit leaving the steady-state inactivation of the current unaffected.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Hippocampus/physiology , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/physiology , Neurons/physiology , Tacrine/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Electric Stimulation/methods , Hippocampus/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Rats , Rats, Sprague-Dawley
14.
Biochim Biophys Acta Proteins Proteom ; 1865(5): 465-472, 2017 May.
Article in English | MEDLINE | ID: mdl-28179135

ABSTRACT

We report isolation, sequencing, and electrophysiological characterization of OSK3 (α-KTx 8.8 in Kalium and Uniprot databases), a potassium channel blocker from the scorpion Orthochirus scrobiculosus venom. Using the voltage clamp technique, OSK3 was tested on a wide panel of 11 voltage-gated potassium channels expressed in Xenopus oocytes, and was found to potently inhibit Kv1.2 and Kv1.3 with IC50 values of ~331nM and ~503nM, respectively. OdK1 produced by the scorpion Odontobuthus doriae differs by just two C-terminal residues from OSK3, but shows marked preference to Kv1.2. Based on the charybdotoxin-potassium channel complex crystal structure, a model was built to explain the role of the variable residues in OdK1 and OSK3 selectivity.


Subject(s)
Potassium Channel Blockers/chemistry , Protein Conformation , Scorpion Venoms/metabolism , Amino Acid Sequence/genetics , Animals , Crystallography, X-Ray , Electrophysiology , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/chemistry , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/chemistry , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/chemistry , Potassium/metabolism , Potassium Channel Blockers/isolation & purification , Potassium Channel Blockers/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpion Venoms/isolation & purification , Scorpions/chemistry , Scorpions/metabolism , Xenopus/genetics
15.
Biochem Biophys Res Commun ; 482(4): 1135-1140, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27916464

ABSTRACT

The neurotoxic cone snail peptide µ-GIIIA specifically blocks skeletal muscle voltage-gated sodium (NaV1.4) channels. The related conopeptides µ-PIIIA and µ-SIIIA, however, exhibit a wider activity spectrum by also inhibiting the neuronal NaV channels NaV1.2 and NaV1.7. Here we demonstrate that those µ-conopeptides with a broader target range also antagonize select subtypes of voltage-gated potassium channels of the KV1 family: µ-PIIIA and µ-SIIIA inhibited KV1.1 and KV1.6 channels in the nanomolar range, while being inactive on subtypes KV1.2-1.5 and KV2.1. Construction and electrophysiological evaluation of chimeras between KV1.5 and KV1.6 revealed that these toxins block KV channels involving their pore regions; the subtype specificity is determined in part by the sequence close to the selectivity filter but predominantly by the so-called turret domain, i.e. the extracellular loop connecting the pore with transmembrane segment S5. Conopeptides µ-SIIIA and µ-PIIIA, thus, are not specific for NaV channels, and the known structure of some KV channel subtypes may provide access to structural insight into the molecular interaction between µ-conopeptides and their target channels.


Subject(s)
Conotoxins/chemistry , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.4 Potassium Channel/antagonists & inhibitors , Kv1.6 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/chemistry , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Electrophysiology , HEK293 Cells , Humans , Neurons/metabolism , Peptides/chemistry , Protein Domains
16.
Sci Rep ; 6: 32121, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27573545

ABSTRACT

Scorpion toxins that block potassium channels and antimicrobial plant defensins share a common structural CSαß-motif. These toxins contain a toxin signature (K-C4-X-N) in their amino acid sequence, and based on in silico analysis of 18 plant defensin sequences, we noted the presence of a toxin signature (K-C5-R-G) in the amino acid sequence of the Arabidopsis thaliana defensin AtPDF2.3. We found that recombinant (r)AtPDF2.3 blocks Kv1.2 and Kv1.6 potassium channels, akin to the interaction between scorpion toxins and potassium channels. Moreover, rAtPDF2.3[G36N], a variant with a KCXN toxin signature (K-C5-R-N), is more potent in blocking Kv1.2 and Kv1.6 channels than rAtPDF2.3, whereas rAtPDF2.3[K33A], devoid of the toxin signature, is characterized by reduced Kv channel blocking activity. These findings highlight the importance of the KCXN scorpion toxin signature in the plant defensin sequence for blocking potassium channels. In addition, we found that rAtPDF2.3 inhibits the growth of Saccharomyces cerevisiae and that pathways regulating potassium transport and/or homeostasis confer tolerance of this yeast to rAtPDF2.3, indicating a role for potassium homeostasis in the fungal defence response towards rAtPDF2.3. Nevertheless, no differences in antifungal potency were observed between the rAtPDF2.3 variants, suggesting that antifungal activity and Kv channel inhibitory function are not linked.


Subject(s)
Antifungal Agents/pharmacology , Arabidopsis Proteins/pharmacology , Homeodomain Proteins/pharmacology , Kv1.2 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Antifungal Agents/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Kv1.6 Potassium Channel , Potassium Channel Blockers/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/metabolism
17.
J Biol Chem ; 291(13): 7097-106, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26817841

ABSTRACT

The structural similarity between defensins and scorpion neurotoxins suggests that they might have evolved from a common ancestor. However, there is no direct experimental evidence demonstrating a functional link between scorpion neurotoxins and defensins. The scorpion defensin BmKDfsin4 from Mesobuthus martensiiKarsch contains 37 amino acid residues and a conserved cystine-stabilized α/ß structural fold. The recombinant BmKDfsin4, a classical defensin, has been found to have inhibitory activity against Gram-positive bacteria such as Staphylococcus aureus, Bacillus subtilis, and Micrococcus luteusas well as methicillin-resistant Staphylococcus aureus Interestingly, electrophysiological experiments showed that BmKDfsin4,like scorpion potassium channel neurotoxins, could effectively inhibit Kv1.1, Kv1.2, and Kv1.3 channel currents, and its IC50value for the Kv1.3 channel was 510.2 nm Similar to the structure-function relationships of classical scorpion potassium channel-blocking toxins, basic residues (Lys-13 and Arg-19) of BmKDfsin4 play critical roles in peptide-Kv1.3 channel interactions. Furthermore, mutagenesis and electrophysiological experiments demonstrated that the channel extracellular pore region is the binding site of BmKDfsin4, indicating that BmKDfsin4 adopts the same mechanism for blocking potassium channel currents as classical scorpion toxins. Taken together, our work identifies scorpion BmKDfsin4 as the first invertebrate defensin to block potassium channels. These findings not only demonstrate that defensins from invertebrate animals are a novel type of potassium channel blockers but also provide evidence of a functional link between defensins and neurotoxins.


Subject(s)
Anti-Bacterial Agents/chemistry , Defensins/chemistry , Neurotoxins/chemistry , Potassium Channel Blockers/chemistry , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Defensins/genetics , Defensins/metabolism , Defensins/pharmacology , Gene Expression , Humans , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/metabolism , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Micrococcus luteus/drug effects , Micrococcus luteus/growth & development , Models, Molecular , Molecular Sequence Data , Neurotoxins/genetics , Neurotoxins/metabolism , Neurotoxins/pharmacology , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Scorpion Venoms/biosynthesis , Scorpions/chemistry , Scorpions/physiology , Sequence Alignment , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Structural Homology, Protein , Structure-Activity Relationship
18.
Peptides ; 80: 9-17, 2016 06.
Article in English | MEDLINE | ID: mdl-26116782

ABSTRACT

Ts19 Fragment II (Ts19 Frag-II) was first isolated from the venom of the scorpion Tityus serrulatus (Ts). It is a protein presenting 49 amino acid residues, three disulfide bridges, Mr 5534Da and was classified as a new member of class (subfamily) 2 of the ß-KTxs, the second one described for Ts scorpion. The ß-KTx family is composed by two-domain peptides: N-terminal helical domain (NHD), with cytolytic activity, and a C-terminal CSαß domain (CCD), with Kv blocking activity. The extensive electrophysiological screening (16 Kv channels and 5 Nav channels) showed that Ts19 Frag-II presents a specific and significant blocking effect on Kv1.2 (IC50 value of 544±32nM). However, no cytolytic activity was observed with this toxin. We conclude that the absence of 9 amino acid residues from the N-terminal sequence (compared to Ts19 Frag-I) is responsible for the absence of cytolytic activity. In order to prove this hypothesis, we synthesized the peptide with these 9 amino acid residues, called Ts19 Frag-III. As expected, Ts19 Frag-III showed to be cytolytic and did not block the Kv1.2 channel. The post-translational modifications of Ts19 and its fragments (I-III) are also discussed here. A mechanism of post-translational processing (post-splitting) is suggested to explain Ts19 fragments production. In addition to the discovery of this new toxin, this report provides further evidence for the existence of several compounds in the scorpion venom contributing to the diversity of the venom arsenal.


Subject(s)
Peptides/chemistry , Peptides/pharmacology , Scorpion Venoms/chemistry , Animals , Electrophysiology/methods , Erythrocytes/drug effects , Female , Kv1.2 Potassium Channel/antagonists & inhibitors , Mice , Oocytes/drug effects , Peptides/isolation & purification , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Protein Processing, Post-Translational , Scorpion Venoms/isolation & purification , Scorpions/chemistry , Voltage-Gated Sodium Channels/metabolism , Xenopus laevis
19.
Bioorg Med Chem Lett ; 25(3): 514-8, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25556093

ABSTRACT

Gambierol and its heptacyclic and tetracyclic analogs were tested for inhibitory activity against the human voltage-gated potassium channel Kv1.2 (hKv1.2), which was stably expressed in Chinese hamster ovary (CHO) cells. Gambierol, the heptacyclic analog, and the tetracyclic analog inhibited the potassium current evoked by a step pulse from -80mV to 40mV. The IC50 values for the three compounds were 0.75±0.15nM, 7.6±1.2nM, and 28±4.0nM (the mean±SEM, n=3), respectively. The cytotoxic activity was examined in order to assess a relationship between cytotoxicity and inhibition of the hKv1.2. The IC50 values for gambierol, the heptacyclic analog, and the tetracyclic analog in the wild-type CHO cells were 95±7.1µM, 6.5±0.8µM (the mean±SEM, n=3), and >100µM (n=3), respectively, whereas those in the CHO cells stably expressing hKv1.2 were 78±5.8µM, 6.0±1.0µM (the mean±SEM, n=3), and >100µM (n=3). These results suggested that cytotoxicity is not triggered by inhibition of the human Kv1.2. The electrophysiological recording at the resting potential in the presence of gambierol, the heptacyclic analog, and the tetracyclic analog revealed the dose-dependent leak current, which was largest when the heptacyclic analog was administered to the cells. We thus propose that the leak current induced by these compounds might cause a fatal effect on the cultured cells.


Subject(s)
Ciguatoxins/chemistry , Kv1.2 Potassium Channel/antagonists & inhibitors , Animals , CHO Cells , Cell Proliferation/drug effects , Ciguatoxins/metabolism , Ciguatoxins/toxicity , Cricetinae , Cricetulus , HEK293 Cells , Humans , Kv1.2 Potassium Channel/genetics , Kv1.2 Potassium Channel/metabolism , Membrane Potentials/drug effects , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
20.
Biochemistry ; 53(43): 6786-92, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25300013

ABSTRACT

Many drug molecules inhibit the conduction of several families of cation channels by binding to a small cavity just below the selectivity filter of the channel protein. The exact mechanisms governing drug-channel binding and the subsequent inhibition of conduction are not well understood. Here the inhibition of two K(+) channel isoforms, Kv1.2 and KCa3.1, by two drug molecules, lidocaine and TRAM-34, is examined in atomic detail using molecular dynamics simulations. A conserved valine-alanine-valine motif in the inner cavity is found to be crucial for drug binding in both channels, consistent with previous studies of similar systems. Potential of mean force calculations show that lidocaine in its charged form creates an energy barrier of ∼6 kT for a permeating K(+) ion when the ion is crossing over the drug, while the neutral form of lidocaine has no significant effect on the energetics of ion permeation. On the other hand, TRAM-34 in the neutral form is able to create a large energy barrier of ∼10 kT by causing the permeating ion to dehydrate. Our results suggest that TRAM-34 analogues that remain neutral and permeable to membranes under acidic conditions common to inflammation may act as possible drug scaffolds for combating local anesthetic failure in inflammation.


Subject(s)
Anesthetics, Local/chemistry , Antifungal Agents/chemistry , Intermediate-Conductance Calcium-Activated Potassium Channels , Kv1.2 Potassium Channel , Lidocaine/chemistry , Molecular Dynamics Simulation , Pyrazoles/chemistry , Amino Acid Motifs , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/chemistry , Kv1.2 Potassium Channel/antagonists & inhibitors , Kv1.2 Potassium Channel/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...