Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 16092, 2024 07 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 Biol Chem ; 300(4): 107155, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479597

ABSTRACT

Despite significant advances in the development of therapeutic interventions targeting autoimmune diseases and chronic inflammatory conditions, lack of effective treatment still poses a high unmet need. Modulating chronically activated T cells through the blockade of the Kv1.3 potassium channel is a promising therapeutic approach; however, developing selective Kv1.3 inhibitors is still an arduous task. Phage display-based high throughput peptide library screening is a rapid and robust approach to develop promising drug candidates; however, it requires solid-phase immobilization of target proteins with their binding site preserved. Historically, the KcsA bacterial channel chimera harboring only the turret region of the human Kv1.3 channel was used for screening campaigns. Nevertheless, literature data suggest that binding to this type of chimera does not correlate well with blocking potency on the native Kv1.3 channels. Therefore, we designed and successfully produced advanced KcsA-Kv1.3, KcsA-Kv1.1, and KcsA-Kv1.2 chimeric proteins in which both the turret and part of the filter regions of the human Kv1.x channels were transferred. These T+F (turret-filter) chimeras showed superior peptide ligand-binding predictivity compared to their T-only versions in novel phage ELISA assays. Phage ELISA binding and competition results supported with electrophysiological data confirmed that the filter region of KcsA-Kv1.x is essential for establishing adequate relative affinity order among selected peptide toxins (Vm24 toxin, Hongotoxin-1, Kaliotoxin-1, Maurotoxin, Stichodactyla toxin) and consequently obtaining more reliable selectivity data. These new findings provide a better screening tool for future drug development efforts and offer insight into the target-ligand interactions of these therapeutically relevant ion channels.


Subject(s)
Kv1.3 Potassium Channel , Potassium Channel Blockers , Recombinant Fusion Proteins , Animals , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/chemistry , Ligands , Peptide Library , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Cell Line
3.
J Chem Inf Model ; 63(10): 3043-3053, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37143234

ABSTRACT

Peptide toxins that adopt the ShK fold can inhibit the voltage-gated potassium channel KV1.3 with IC50 values in the pM range and are therefore potential leads for drugs targeting autoimmune and neuroinflammatory diseases. Nuclear magnetic resonance (NMR) relaxation measurements and pressure-dependent NMR have shown that, despite being cross-linked by disulfide bonds, ShK itself is flexible in solution. This flexibility affects the local structure around the pharmacophore for the KV1.3 channel blockade and, in particular, the relative orientation of the key Lys and Tyr side chains (Lys22 and Tyr23 in ShK) and has implications for the design of KV1.3 inhibitors. In this study, we have performed molecular dynamics (MD) simulations on ShK and a close homologue, HmK, to probe the conformational space occupied by the Lys and Tyr residues, and docked the different conformations with a recently determined cryo-EM structure of the KV1.3 channel. Although ShK and HmK have 60% sequence identity, their dynamic behaviors are quite different, with ShK sampling a broad range of conformations over the course of a 5 µs MD simulation, while HmK is relatively rigid. We also investigated the importance of conformational dynamics, in particular the distance between the side chains of the key dyad Lys22 and Tyr23, for binding to KV1.3. Although these peptides have quite different dynamics, the dyad in both adopts a similar configuration upon binding, revealing a conformational selection upon binding to KV1.3 in the case of ShK. Both peptides bind to KV1.3 with Lys22 occupying the pore of the channel. Intriguingly, the more flexible peptide, ShK, binds with significantly higher affinity than HmK.


Subject(s)
Cnidarian Venoms , Sea Anemones , Animals , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/metabolism , Cnidarian Venoms/chemistry , Cnidarian Venoms/metabolism , Cnidarian Venoms/pharmacology , Sea Anemones/chemistry , Sea Anemones/metabolism , Peptides/chemistry , Molecular Conformation , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Kv1.2 Potassium Channel/metabolism
4.
Toxins (Basel) ; 14(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36548755

ABSTRACT

The voltage-gated potassium Kv1.3 channel is an essential component of vital cellular processes which is also involved in the pathogenesis of some autoimmune, neuroinflammatory and oncological diseases. Pore blockers of the Kv1.3 channel are considered as potential drugs and are used to study Kv1 channels' structure and functions. Screening and study of the blockers require the assessment of their ability to bind the channel. Expanding the variety of methods used for this, we report on the development of the fluorescent competitive binding assay for measuring affinities of pore blockers to Kv1.3 at the membrane of mammalian cells. The assay constituents are hongotoxin 1 conjugated with Atto488, fluorescent mKate2-tagged Kv1.3 channel, which was designed to improve membrane expression of the channel in mammalian cells, confocal microscopy, and a special protocol of image processing. The assay is implemented in the "mix and measure", format and allows the screening of Kv1.3 blockers, such as peptide toxins, that bind to the extracellular vestibule of the K+-conducting pore, and analyzing their affinity.


Subject(s)
Eukaryotic Cells , Potassium Channels, Voltage-Gated , Animals , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Kv1.3 Potassium Channel/chemistry , Mammals
5.
Bioconjug Chem ; 33(11): 2197-2212, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36330854

ABSTRACT

Upregulation of the voltage-gated potassium channel KV1.3 is implicated in a range of autoimmune and neuroinflammatory diseases, including rheumatoid arthritis, psoriasis, multiple sclerosis, and type I diabetes. Understanding the expression, localization, and trafficking of KV1.3 in normal and disease states is key to developing targeted immunomodulatory therapies. HsTX1[R14A], an analogue of a 34-residue peptide toxin from the scorpion Heterometrus spinifer, binds KV1.3 with high affinity (IC50 of 45 pM) and selectivity (2000-fold for KV1.3 over KV1.1). We have synthesized a fluorescent analogue of HsTX1[R14A] by N-terminal conjugation of a Cy5 tag. Electrophysiology assays show that Cy5-HsTX1[R14A] retains activity against KV1.3 (IC50 ∼ 0.9 nM) and selectivity over a range of other potassium channels (KV1.2, KV1.4, KV1.5, KV1.6, KCa1.1 and KCa3.1), as well as selectivity against heteromeric channels assembled from KV1.3/KV1.5 tandem dimers. Live imaging of CHO cells expressing green fluorescent protein-tagged KV1.3 shows co-localization of Cy5-HsTX1[R14A] and KV1.3 fluorescence signals at the cell membrane. Moreover, flow cytometry demonstrated that Cy5-HsTX1[R14A] can detect KV1.3-expressing CHO cells. Stimulation of mouse microglia by lipopolysaccharide, which enhances membrane expression of KV1.3, was associated with increased staining by Cy5-HsTX1[R14A], demonstrating that it can be used to identify KV1.3 in disease-relevant models of inflammation. Furthermore, the biodistribution of Cy5-HsTX1[R14A] could be monitored using ex vivo fluorescence imaging of organs in mice dosed subcutaneously with the peptide. These results illustrate the utility of Cy5-HsTX1[R14A] as a tool for visualizing KV1.3, with broad applicability in fundamental investigations of KV1.3 biology, and the validation of novel disease indications where KV1.3 inhibition may be of therapeutic value.


Subject(s)
Kv1.3 Potassium Channel , Scorpion Venoms , Mice , Animals , Cricetinae , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/metabolism , Scorpion Venoms/pharmacology , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Cricetulus , Tissue Distribution , Peptides/chemistry
6.
Nat Commun ; 13(1): 3854, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35788586

ABSTRACT

The Kv1.3 potassium channel is expressed abundantly on activated T cells and mediates the cellular immune response. This role has made the channel a target for therapeutic immunomodulation to block its activity and suppress T cell activation. Here, we report structures of human Kv1.3 alone, with a nanobody inhibitor, and with an antibody-toxin fusion blocker. Rather than block the channel directly, four copies of the nanobody bind the tetramer's voltage sensing domains and the pore domain to induce an inactive pore conformation. In contrast, the antibody-toxin fusion docks its toxin domain at the extracellular mouth of the channel to insert a critical lysine into the pore. The lysine stabilizes an active conformation of the pore yet blocks ion permeation. This study visualizes Kv1.3 pore dynamics, defines two distinct mechanisms to suppress Kv1.3 channel activity with exogenous inhibitors, and provides a framework to aid development of emerging T cell immunotherapies.


Subject(s)
Kv1.3 Potassium Channel/chemistry , T-Lymphocytes , Humans , Immunoglobulins/metabolism , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Lysine , T-Lymphocytes/chemistry
7.
Mol Pharmacol ; 102(3): 150-160, 2022 09.
Article in English | MEDLINE | ID: mdl-35764383

ABSTRACT

Voltage-gated KV1.3 channel has been reported to be a drug target for the treatment of autoimmune diseases, and specific inhibitors of Kv1.3 are potential therapeutic drugs for multiple diseases. The scorpions could produce various bioactive peptides that could inhibit KV1.3 channel. Here, we identified a new scorpion toxin polypeptide gene ImKTX58 from the venom gland cDNA library of the Chinese scorpion Isometrus maculatus Sequence alignment revealed high similarities between ImKTX58 mature peptide and previously reported KV1.3 channel blockers-LmKTX10 and ImKTX88-suggesting that ImKTX58 peptide might also be a KV1.3 channel blocker. By using electrophysiological recordings, we showed that recombinant ImKTX58 prepared by genetic engineering technologies had a highly selective inhibiting effect on KV1.3 channel. Further alanine scanning mutagenesis and computer simulation identified four amino acid residues in ImKTX58 peptide as key binding sites to KV1.3 channel by forming hydrogen bonds, salt bonds, and hydrophobic interactions. Among these four residues, 28th lysine of the ImKTX58 mature peptide was found to be the most critical amino acid residue for blocking KV1.3 channel. SIGNIFICANCE STATEMENT: In this study, we discovered a scorpion toxin gene ImKTX58 that has not been reported before in Hainan Isometrus maculatus and successfully used the prokaryotic expression system to express and purify the polypeptides encoded by this gene. Electrophysiological experiments on ImKTX58 showed that ImKTX58 has a highly selective blocking effect on KV1.3 channel over Kv1.1, Kv1.2, Kv1.5, SK2, SK3, and BK channels. These findings provide a theoretical basis for designing highly effective KV1.3 blockers to treat autoimmune and other diseases.


Subject(s)
Scorpion Venoms , Amino Acid Sequence , Amino Acids , Animals , Computer Simulation , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Peptides/chemistry , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Scorpion Venoms/chemistry , Scorpion Venoms/metabolism , Scorpion Venoms/pharmacology , Scorpions/chemistry , Scorpions/genetics , Scorpions/metabolism
8.
Biol Futur ; 72(1): 75-83, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34554500

ABSTRACT

Since the discovery of the Kv1.3 voltage-gated K+ channel in human T cells in 1984, ion channels are considered crucial elements of the signal transduction machinery in the immune system. Our knowledge about Kv1.3 and its inhibitors is outstanding, motivated by their potential application in autoimmune diseases mediated by Kv1.3 overexpressing effector memory T cells (e.g., Multiple Sclerosis). High affinity Kv1.3 inhibitors are either small organic molecules (e.g., Pap-1) or peptides isolated from venomous animals. To date, the highest affinity Kv1.3 inhibitors with the best Kv1.3 selectivity are the engineered analogues of the sea anemone peptide ShK (e.g., ShK-186), the engineered scorpion toxin HsTx1[R14A] and the natural scorpion toxin Vm24. These peptides inhibit Kv1.3 in picomolar concentrations and are several thousand-fold selective for Kv1.3 over other biologically critical ion channels. Despite the significant progress in the field of Kv1.3 molecular pharmacology several progressive questions remain to be elucidated and discussed here. These include the conjugation of the peptides to carriers to increase the residency time of the peptides in the circulation (e.g., PEGylation and engineering the peptides into antibodies), use of rational drug design to create novel peptide inhibitors and understanding the potential off-target effects of Kv1.3 inhibition.


Subject(s)
Immune System/drug effects , Kv1.3 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , T-Lymphocytes/drug effects , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/prevention & control , Humans , Immune System/cytology , Immune System/metabolism , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/metabolism , Peptides/pharmacology , Proteins/pharmacology , Scorpion Venoms/pharmacology , T-Lymphocytes/metabolism
9.
J Cell Physiol ; 236(6): 4330-4347, 2021 06.
Article in English | MEDLINE | ID: mdl-33230847

ABSTRACT

The voltage-dependent potassium channel Kv1.3 has been implicated in proliferation in many cell types, based on the observation that Kv1.3 blockers inhibited proliferation. By modulating membrane potential, cell volume, and/or Ca2+ influx, K+  channels can influence cell cycle progression. Also, noncanonical channel functions could contribute to modulate cell proliferation independent of K+ efflux. The specificity of the requirement of Kv1.3 channels for proliferation suggests the involvement of molecule-specific interactions, but the underlying mechanisms are poorly identified. Heterologous expression of Kv1.3 channels in HEK cells has been shown to increase proliferation independently of K+ fluxes. Likewise, some of the molecular determinants of Kv1.3-induced proliferation have been located in the C-terminus region, where individual point mutations of putative phosphorylation sites (Y447A and S459A) abolished Kv1.3-induced proliferation. Here, we investigated the mechanisms linking Kv1.3 channels to proliferation exploring the correlation between Kv1.3 voltage-dependent molecular dynamics and cell cycle progression. Using transfected HEK cells, we analyzed both the effect of changes in resting membrane potential on Kv1.3-induced proliferation and the effect of mutated Kv1.3 channels with altered voltage dependence of gating. We conclude that voltage-dependent transitions of Kv1.3 channels enable the activation of proliferative pathways. We also found that Kv1.3 associated with IQGAP3, a scaffold protein involved in proliferation, and that membrane depolarization facilitates their interaction. The functional contribution of Kv1.3-IQGAP3 interplay to cell proliferation was demonstrated both in HEK cells and in vascular smooth muscle cells. Our data indicate that voltage-dependent conformational changes of Kv1.3 are an essential element in Kv1.3-induced proliferation.


Subject(s)
Cell Proliferation , Ion Channel Gating , Kv1.3 Potassium Channel/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , HEK293 Cells , Humans , KATP Channels/genetics , KATP Channels/metabolism , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/genetics , Membrane Potentials , Mutation , Protein Conformation , Signal Transduction , Structure-Activity Relationship
10.
Toxins (Basel) ; 12(12)2020 12 16.
Article in English | MEDLINE | ID: mdl-33339256

ABSTRACT

Recently developed fluorescent protein-scorpion toxin chimeras (FP-Tx) show blocking activities for potassium voltage-gated channels of Kv1 family and retain almost fully pharmacological profiles of the parental peptide toxins (Kuzmenkov et al., Sci Rep. 2016, 6, 33314). Here we report on N-terminally green fluorescent protein (GFP)-tagged agitoxin 2 (GFP-L2-AgTx2) with high affinity and selectivity for the binding site of Kv1.3 channel involved in the pathogenesis of various (primarily of autoimmune origin) diseases. The basis for this selectivity relates to N-terminal location of GFP, since transposition of GFP to the C-terminus of AgTx2 recovered specific interactions with the Kv1.1 and Kv1.6 binding sites. Competitive binding experiments revealed that the binding site of GFP-L2-AgTx2 overlaps that of charybdotoxin, kaliotoxin 1, and agitoxin 2, the known Kv1.3-channel pore blockers. GFP-L2-AgTx2 was demonstrated to be applicable as a fluorescent probe to search for Kv1.3 pore blockers among individual compounds and in complex mixtures, to measure blocker affinities, and to visualize Kv1.3 distribution at the plasma membrane of Kv1.3-expressing HEK293 cells. Our studies show that definite combinations of fluorescent proteins and peptide blockers can result in considerable modulation of the natural blocker-channel binding profile yielding selective fluorescent ligands of certain channels.


Subject(s)
Green Fluorescent Proteins/metabolism , Kv1.3 Potassium Channel/metabolism , Potassium Channel Blockers/metabolism , Scorpion Venoms/metabolism , Amino Acid Sequence , Binding Sites/physiology , Green Fluorescent Proteins/chemistry , HEK293 Cells , Humans , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/chemistry , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Protein Structure, Secondary , Scorpion Venoms/analysis , Scorpion Venoms/chemistry
11.
Sci Adv ; 6(10): eaaz3439, 2020 03.
Article in English | MEDLINE | ID: mdl-32181366

ABSTRACT

We show here that membrane-tethered toxins facilitate the biophysical study of the roles of toxin residues in K+ channel blockade to reveal two blocking mechanisms in the K+ channel pore. The structure of the sea anemone type I (SAK1) toxin HmK is determined by NMR. T-HmK residues are scanned by point mutation to map the toxin surface, and seven residues are identified to be critical to occlusion of the KcsA channel pore. T-HmK-Lys22 is shown to interact with K+ ions traversing the KcsA pore from the cytoplasm conferring voltage dependence on the toxin off rate, a classic mechanism that we observe as well with HmK in solution and for Kv1.3 channels. In contrast, two related SAK1 toxins, Hui1 and ShK, block KcsA and Kv1.3, respectively, via an arginine rather than the canonical lysine, when tethered and as free peptides.


Subject(s)
Bacterial Proteins/chemistry , Cnidarian Venoms/pharmacology , Kv1.3 Potassium Channel/chemistry , Neurotoxins/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cations, Monovalent , Cnidarian Venoms/chemistry , Cnidarian Venoms/genetics , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurotoxins/chemistry , Neurotoxins/genetics , Nuclear Magnetic Resonance, Biomolecular , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Point Mutation , Potassium/chemistry , Potassium/metabolism , Potassium Channel Blockers/chemistry , Potassium Channels/genetics , Potassium Channels/metabolism , Sea Anemones , Xenopus laevis
12.
Sci Rep ; 9(1): 19307, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848433

ABSTRACT

ShK is a 35-residue disulfide-linked polypeptide produced by the sea anemone Stichodactyla helianthus, which blocks the potassium channels Kv1.1 and Kv1.3 with pM affinity. An analogue of ShK has been developed that blocks Kv1.3 > 100 times more potently than Kv1.1, and has completed Phase 1b clinical trials for the treatment of autoimmune diseases such as psoriasis and rheumatoid arthritis. Previous studies have indicated that ShK undergoes a conformational exchange that is critical to its function, but this has proved difficult to characterise. Here, we have used high hydrostatic pressure as a tool to increase the population of the alternative state, which is likely to resemble the active form that binds to the Kv1.3 channel. By following changes in chemical shift with pressure, we have derived the chemical shift values of the low- and high-pressure states, and thus characterised the locations of structural changes. The main difference is in the conformation of the Cys17-Cys32 disulfide, which is likely to affect the positions of the critical Lys22-Tyr23 pair by twisting the 21-24 helix and increasing the solvent exposure of the Lys22 sidechain, as indicated by molecular dynamics simulations.


Subject(s)
Cnidarian Venoms/chemistry , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/chemistry , Amino Acid Sequence/genetics , Animals , Autoimmune Diseases/drug therapy , Cnidarian Venoms/genetics , Cnidarian Venoms/pharmacology , Humans , Kv1.1 Potassium Channel/chemistry , Kv1.1 Potassium Channel/ultrastructure , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/ultrastructure , Molecular Conformation , Molecular Dynamics Simulation , Peptides/chemistry , Peptides/genetics , Potassium Channel Blockers/pharmacology , Sea Anemones/chemistry
13.
J Biol Chem ; 294(48): 18349-18359, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31533989

ABSTRACT

Tk-hefu is an artificial peptide designed based on the α-hairpinin scaffold, which selectively blocks voltage-gated potassium channels Kv1.3. Here we present its spatial structure resolved by NMR spectroscopy and analyze its interaction with channels using computer modeling. We apply protein surface topography to suggest mutations and increase Tk-hefu affinity to the Kv1.3 channel isoform. We redesign the functional surface of Tk-hefu to better match the respective surface of the channel pore vestibule. The resulting peptide Tk-hefu-2 retains Kv1.3 selectivity and displays ∼15 times greater activity compared with Tk-hefu. We verify the mode of Tk-hefu-2 binding to the channel outer vestibule experimentally by site-directed mutagenesis. We argue that scaffold engineering aided by protein surface topography represents a reliable tool for design and optimization of specific ion channel ligands.


Subject(s)
Kv1.3 Potassium Channel/chemistry , Peptides/chemistry , Potassium Channel Blockers/chemistry , Proteins/chemistry , Amino Acid Sequence , Animals , Humans , Kv1.3 Potassium Channel/metabolism , Ligands , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Dynamics Simulation , Mutation , Peptides/genetics , Peptides/metabolism , Potassium Channel Blockers/metabolism , Protein Binding , Protein Conformation , Proteins/metabolism , Surface Properties
14.
J Biochem Mol Toxicol ; 33(2): e22244, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30381903

ABSTRACT

Fungus defensin is a kind of important natural peptide resource, such as plectasin from the soil fungus Pseudoplectania nigrella with potential application in the antimicrobial peptide lead drug discovery. Here, a fungus defensin named Bldesin with Kv1.3 channel and serine protease inhibitory activities was first explored. By GST-Bldesin fusion expression and enterokinase cleaving strategy, recombinant Bldesin was obtained successfully. Antimicrobial assays showed that Bldesin had potent activity against Gram-positive Staphylococcus aureus, but had no effect on Gram-negative Escherichia coli. Electrophysiological experiments showed that Bldesin had Kv1.3 channel inhibitory activity. Serine protease inhibitory associated experiments showed that Bldesin had unique chymotrypsin protease inhibitory, elastase protease inhibitory, and serine protease-associated coagulation inhibitory activities. To the best of our knowledge, Bldesin is the first functionally characterized pathogenic fungus defensin with Kv1.3 channel and chymotrypsin inhibitory activities and highlighted novel pharmacological effects of fungus-derived defensin peptides.


Subject(s)
Ascomycota/chemistry , Chymotrypsin/antagonists & inhibitors , Defensins/chemistry , Fungal Proteins/chemistry , Kv1.3 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/chemistry , Serpins/chemistry , Anti-Bacterial Agents/chemistry , Chymotrypsin/chemistry , HEK293 Cells , Humans , Kv1.3 Potassium Channel/chemistry
15.
Anal Biochem ; 556: 70-77, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29936096

ABSTRACT

Despite the significant role integral membrane proteins (IMPs) play in the drug discovery process, it remains extremely challenging to express, purify, and in vitro stabilize them for detailed biophysical analyses. Cell-free transcription-translation systems have emerged as a promising alternative for producing complex proteins, but they are still not a viable option for expressing IMPs due to improper post-translational folding of these proteins. We have studied key factors influencing in vitro folding of cell-free-expressed IMPs, particularly oligomeric proteins (i.e., ion channels). Using a chimeric ion channel, KcsA-Kv1.3 (K-K), as a model IMP, we have investigated several physiochemical determinants including artificial bilayer environments (i.e., lipid, detergent) for K-K in vitro stabilization. We observed that fusion of a 'superfolder' green fluorescent protein (sfGFP) to K-K as a protein expression reporter not only improves the protein yield, but surprisingly facilitates the K-K tetramer formation, probably by enhancing the solubility of monomeric K-K. Additionally, anionic lipids (i.e., DMPG) were found to be essential for the correct folding of cell-free-expressed monomeric K-K into tetramer, underscoring the importance of lipid-protein interaction in maintaining structural-functional integrity of ion channels. We further developed methods to integrate cell-free-expressed IMPs directly onto a biosensor chip. We employed a solid-supported lipid bilayer onto the surface plasmon resonance (SPR) chip to insert nascent K-K in a membrane. In a different approach, an anti-GFP-functionalized surface was used to capture in situ expressed K-K via its sfGFP tag. Interestingly, only the K-K-functionalized capture surface prepared by the latter strategy was able to interact with K-K's small binding partners. This generalizable approach can be further extended to other membrane proteins for developing direct binding assays involving small ligands.


Subject(s)
Biosensing Techniques/methods , Kv1.3 Potassium Channel , Lab-On-A-Chip Devices , Lipid Bilayers , Protein Biosynthesis , Cell-Free System/chemistry , Cell-Free System/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Humans , Kv1.3 Potassium Channel/blood , Kv1.3 Potassium Channel/chemistry , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Protein Binding
16.
Am J Physiol Cell Physiol ; 314(1): C27-C42, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28931540

ABSTRACT

Kv1.3 channels are involved in the switch to proliferation of normally quiescent cells, being implicated in the control of cell cycle in many different cell types and in many different ways. They modulate membrane potential controlling K+ fluxes, sense changes in potential, and interact with many signaling molecules through their intracellular domains. From a mechanistic point of view, we can describe the role of Kv1.3 channels in proliferation with at least three different models. In the "membrane potential model," membrane hyperpolarization resulting from Kv1.3 activation provides the driving force for Ca2+ influx required to activate Ca2+-dependent transcription. This model explains most of the data obtained from several cells from the immune system. In the "voltage sensor model," Kv1.3 channels serve mainly as sensors that transduce electrical signals into biochemical cascades, independently of their effect on membrane potential. Kv1.3-dependent proliferation of vascular smooth muscle cells (VSMCs) could fit this model. Finally, in the "channelosome balance model," the master switch determining proliferation may be related to the control of the Kv1.3 to Kv1.5 ratio, as described in glial cells and also in VSMCs. Since the three mechanisms cannot function independently, these models are obviously not exclusive. Nevertheless, they could be exploited differentially in different cells and tissues. This large functional flexibility of Kv1.3 channels surely gives a new perspective on their functions beyond their elementary role as ion channels, although a conclusive picture of the mechanisms involved in Kv1.3 signaling to proliferation is yet to be reached.


Subject(s)
Cell Proliferation , Kv1.3 Potassium Channel/metabolism , Potassium/metabolism , Animals , Calcium Signaling , Cell Proliferation/drug effects , Humans , Ion Channel Gating , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/genetics , Membrane Potentials , Models, Biological , Potassium Channel Blockers/pharmacology , Protein Conformation , Structure-Activity Relationship
17.
J Biol Chem ; 292(30): 12599-12605, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28596383

ABSTRACT

ShK toxin from the sea anemone Stichodactyla helianthus is a 35-residue protein that binds to the Kv1.3 ion channel with high affinity. Recently we determined the X-ray structure of ShK toxin by racemic crystallography, in the course of which we discovered that d-ShK has a near-background IC50 value ∼50,000 times lower than that of the l-ShK toxin. This lack of activity was at odds with previously reported results for an ShK diastereomer designated d-allo-ShK, for which significant biological activity had been observed in a similar receptor-blocking assay. As reported, d-allo-ShK was made up of d-amino acids, but with retention of the natural stereochemistry of the chiral side chains of the Ile and Thr residues, i.e. containing d-allo-Ile and d-allo-Thr along with d-amino acids and glycine. To understand its apparent biological activity, we set out to chemically synthesize d-allo-ShK and determine its X-ray structure by racemic crystallography. Using validated allo-Thr and allo-Ile, both l-allo-ShK and d-allo-ShK polypeptide chains were prepared by total chemical synthesis. Neither the l-allo-ShK nor the d-allo-ShK polypeptides folded, whereas both l-ShK and d-ShK folded smoothly under the same conditions. Re-examination of NMR spectra of the previously reported d-allo-ShK protein revealed that diagnostic Thr and Ile signals were the same as for authentic d-ShK. On the basis of these results, we conclude that the previously reported d-allo-ShK was in fact d-ShK, the true enantiomer of natural l-ShK toxin, and that the apparent biological activity may have arisen from inadvertent contamination with trace amounts of l-ShK toxin.


Subject(s)
Cnidarian Venoms/metabolism , Sea Anemones/chemistry , Animals , Cnidarian Venoms/chemistry , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/metabolism , Molecular Conformation , Nuclear Magnetic Resonance, Biomolecular , Sea Anemones/metabolism
18.
J Mol Biol ; 429(11): 1722-1732, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28478285

ABSTRACT

Proteins begin to fold in the ribosome, and misfolding has pathological consequences. Among the earliest folding events in biogenesis is the formation of a helix, an elementary structure that is ubiquitously present and required for correct protein folding in all proteomes. The determinants underlying helix formation in the confined space of the ribosome exit tunnel are relatively unknown. We chose the second transmembrane segment, S2, of a voltage-gated potassium channel, Kv1.3, as a model to probe this issue. Since the N terminus of S2 is initially in an extended conformation in the folding vestibule of the ribosome yet ultimately emerges at the exit port as a helix, S2 is ideally suited for delineating sequential events and folding determinants of helix formation inside the ribosome. We show that S2's extended N terminus inside the tunnel is converted into a helix by a single, distant mutation in the nascent peptide. This transition depends on nascent peptide sequence at specific tunnel locations. Co-translational secondary folding of nascent chains inside the ribosome has profound physiological consequences that bear on correct membrane insertion, tertiary folding, oligomerization, and biochemical modification of the newborn protein during biogenesis.


Subject(s)
Kv1.3 Potassium Channel/biosynthesis , Kv1.3 Potassium Channel/chemistry , Protein Folding , Ribosomes/metabolism , Protein Conformation, alpha-Helical
19.
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
20.
J Cell Sci ; 129(22): 4265-4277, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27802162

ABSTRACT

The voltage-dependent K+ channel Kv1.3 (also known as KCNA3), which plays crucial roles in leukocytes, physically interacts with KCNE4. This interaction inhibits the K+ currents because the channel is retained within intracellular compartments. Thus, KCNE subunits are regulators of K+ channels in the immune system. Although the canonical interactions of KCNE subunits with Kv7 channels are under intensive investigation, the molecular determinants governing the important Kv1.3- KCNE4 association in the immune system are unknown. Our results suggest that the tertiary structure of the C-terminal domain of Kv1.3 is necessary and sufficient for such an interaction. However, this element is apparently not involved in modulating Kv1.3 gating. Furthermore, the KCNE4-dependent intracellular retention of the channel, which negatively affects the activity of Kv1.3, is mediated by two independent and additive mechanisms. First, KCNE4 masks the YMVIEE signature at the C-terminus of Kv1.3, which is crucial for the surface targeting of the channel. Second, we identify a potent endoplasmic reticulum retention motif in KCNE4 that further limits cell surface expression. Our results define specific molecular determinants that play crucial roles in the physiological function of Kv1.3 in leukocytes.


Subject(s)
Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Subunits/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COP-Coated Vesicles/metabolism , Dendritic Cells/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Ion Channel Gating , Jurkat Cells , Leukocytes , Mice , Potassium Channels, Voltage-Gated/chemistry , Protein Binding , Protein Domains , Rats
SELECTION OF CITATIONS
SEARCH DETAIL