1
|
Primak AL, Orlov NA, Peigneur S, Tytgat J, Ignatova AA, Denisova KR, Yakimov SA, Kirpichnikov MP, Nekrasova OV, Feofanov AV. AgTx2-GFP, Fluorescent Blocker Targeting Pharmacologically Important K v1.x (x = 1, 3, 6) Channels. Toxins (Basel) 2023; 15:toxins15030229. [PMID: 36977120 PMCID: PMC10056440 DOI: 10.3390/toxins15030229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The growing interest in potassium channels as pharmacological targets has stimulated the development of their fluorescent ligands (including genetically encoded peptide toxins fused with fluorescent proteins) for analytical and imaging applications. We report on the properties of agitoxin 2 C-terminally fused with enhanced GFP (AgTx2-GFP) as one of the most active genetically encoded fluorescent ligands of potassium voltage-gated Kv1.x (x = 1, 3, 6) channels. AgTx2-GFP possesses subnanomolar affinities for hybrid KcsA-Kv1.x (x = 3, 6) channels and a low nanomolar affinity to KcsA-Kv1.1 with moderate dependence on pH in the 7.0-8.0 range. Electrophysiological studies on oocytes showed a pore-blocking activity of AgTx2-GFP at low nanomolar concentrations for Kv1.x (x = 1, 3, 6) channels and at micromolar concentrations for Kv1.2. AgTx2-GFP bound to Kv1.3 at the membranes of mammalian cells with a dissociation constant of 3.4 ± 0.8 nM, providing fluorescent imaging of the channel membranous distribution, and this binding depended weakly on the channel state (open or closed). AgTx2-GFP can be used in combination with hybrid KcsA-Kv1.x (x = 1, 3, 6) channels on the membranes of E. coli spheroplasts or with Kv1.3 channels on the membranes of mammalian cells for the search and study of nonlabeled peptide pore blockers, including measurement of their affinity.
Collapse
Affiliation(s)
- Alexandra L Primak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Nikita A Orlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Steve Peigneur
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium
| | - Jan Tytgat
- Toxicology and Pharmacology, Campus Gasthuisberg O&N2, University of Leuven (KU Leuven), Herestraat 49, P.O. Box 922, B-3000 Leuven, Belgium
| | - Anastasia A Ignatova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Kristina R Denisova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Sergey A Yakimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Mikhail P Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Oksana V Nekrasova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexey V Feofanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
2
|
sVmKTx, a transcriptome analysis-based synthetic peptide analogue of Vm24, inhibits Kv1.3 channels of human T cells with improved selectivity. Biochem Pharmacol 2022; 199:115023. [PMID: 35358481 DOI: 10.1016/j.bcp.2022.115023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/13/2022]
Abstract
Kv1.3 K+ channels play a central role in the regulation of T cell activation and Ca2+ signaling under physiological and pathophysiological conditions. Peptide toxins targeting Kv1.3 have a significant therapeutic potential in the treatment of autoimmune diseases; thus, the discovery of new toxins is highly motivated. Based on the transcriptome analysis of the venom gland of V. mexicanus smithi a novel synthetic peptide, sVmKTx was generated, containing 36 amino acid residues. sVmKTx shows high sequence similarity to Vm24, a previously characterized peptide from the same species, but contains a Glu at position 32 as opposed to Lys32 in Vm24. Vm24 inhibits Kv1.3 with high affinity (Kd = 2.9 pM). However, it has limited selectivity (~1,500-fold) for Kv1.3 over hKv1.2, hKCa3.1, and mKv1.1. sVmKTx displays reduced Kv1.3 affinity (Kd = 770 pM) but increased selectivity for Kv1.3 over hKv1.2 (~9,000-fold) as compared to Vm24, other channels tested in the panel (hKCa3.1, hKv1.1, hKv1.4, hKv1.5, rKv2.1, hKv11.1, hKCa1.1, hNav1.5) were practically insensitive to the toxin at 2.5 μM. Molecular dynamics simulations showed that introduction of a Glu instead of Lys at position 32 led to a decreased structural fluctuation of the N-terminal segment of sVmKTx, which may explain its increased selectivity for Kv1.3. sVmKTx at 100 nM concentration decreased the expression level of the Ca2+ -dependent T cell activation marker, CD40 ligand. The high affinity block of Kv1.3 and increased selectivity over the natural peptide makes sVmKTx a potential candidate for Kv1.3 blockade-mediated treatment of autoimmune diseases.
Collapse
|
3
|
Gubič Š, Hendrickx LA, Toplak Ž, Sterle M, Peigneur S, Tomašič T, Pardo LA, Tytgat J, Zega A, Mašič LP. Discovery of K V 1.3 ion channel inhibitors: Medicinal chemistry approaches and challenges. Med Res Rev 2021; 41:2423-2473. [PMID: 33932253 PMCID: PMC8252768 DOI: 10.1002/med.21800] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/03/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
The KV 1.3 voltage-gated potassium ion channel is involved in many physiological processes both at the plasma membrane and in the mitochondria, chiefly in the immune and nervous systems. Therapeutic targeting KV 1.3 with specific peptides and small molecule inhibitors shows great potential for treating cancers and autoimmune diseases, such as multiple sclerosis, type I diabetes mellitus, psoriasis, contact dermatitis, rheumatoid arthritis, and myasthenia gravis. However, no KV 1.3-targeted compounds have been approved for therapeutic use to date. This review focuses on the presentation of approaches for discovering new KV 1.3 peptide and small-molecule inhibitors, and strategies to improve the selectivity of active compounds toward KV 1.3. Selectivity of dalatazide (ShK-186), a synthetic derivate of the sea anemone toxin ShK, was achieved by chemical modification and has successfully reached clinical trials as a potential therapeutic for treating autoimmune diseases. Other peptides and small-molecule inhibitors are critically evaluated for their lead-like characteristics and potential for progression into clinical development. Some small-molecule inhibitors with well-defined structure-activity relationships have been optimized for selective delivery to mitochondria, and these offer therapeutic potential for the treatment of cancers. This overview of KV 1.3 inhibitors and methodologies is designed to provide a good starting point for drug discovery to identify novel effective KV 1.3 modulators against this target in the future.
Collapse
Affiliation(s)
- Špela Gubič
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | - Louise A. Hendrickx
- Toxicology and PharmacologyUniversity of Leuven, Campus GasthuisbergLeuvenBelgium
| | - Žan Toplak
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | - Maša Sterle
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | - Steve Peigneur
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | | | - Luis A. Pardo
- AG OncophysiologyMax‐Planck Institute for Experimental MedicineGöttingenGermany
| | - Jan Tytgat
- Toxicology and PharmacologyUniversity of Leuven, Campus GasthuisbergLeuvenBelgium
| | - Anamarija Zega
- Faculty of PharmacyUniversity of LjubljanaLjubljanaSlovenia
| | | |
Collapse
|
4
|
Saikia C, Dym O, Altman-Gueta H, Gordon D, Reuveny E, Karbat I. A Molecular Lid Mechanism of K + Channel Blocker Action Revealed by a Cone Peptide. J Mol Biol 2021; 433:166957. [PMID: 33771569 DOI: 10.1016/j.jmb.2021.166957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/08/2021] [Accepted: 03/16/2021] [Indexed: 12/15/2022]
Abstract
Many venomous organisms carry in their arsenal short polypeptides that block K+ channels in a highly selective manner. These toxins may compete with the permeating ions directly via a "plug" mechanism or indirectly via a "pore-collapse" mechanism. An alternative "lid" mechanism was proposed but remained poorly defined. Here we study the Drosophila Shaker channel block by Conkunitzin-S1 and Conkunitzin-C3, two highly similar toxins derived from cone venom. Despite their similarity, the two peptides exhibited differences in their binding poses and biophysical assays, implying discrete action modes. We show that while Conkunitzin-S1 binds tightly to the channel turret and acts via a "pore-collapse" mechanism, Conkunitzin-C3 does not contact this region. Instead, Conk-C3 uses a non-conserved Arg to divert the permeant ions and trap them in off-axis cryptic sites above the SF, a mechanism we term a "molecular-lid". Our study provides an atomic description of the "lid" K+ blocking mode and offers valuable insights for the design of therapeutics based on venom peptides.
Collapse
Affiliation(s)
- Chandamita Saikia
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Orly Dym
- Structural Proteomic Unit, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hagit Altman-Gueta
- Department of Plant Molecular Biology and Ecology, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Dalia Gordon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eitan Reuveny
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Izhar Karbat
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
5
|
Qin C, Wan X, Li S, Yang F, Yang L, Zuo Z, Cao Z, Chen Z, Wu Y. Different pharmacological properties between scorpion toxin BmKcug2 and its degraded analogs highlight the diversity of K + channel blockers from thermally processed scorpions. Int J Biol Macromol 2021; 178:143-153. [PMID: 33636268 DOI: 10.1016/j.ijbiomac.2021.02.155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 11/16/2022]
Abstract
Novel degraded potassium channel-modulatory peptides were recently found in thermally processed scorpions, but their pharmacological properties remain unclear. Here, we identified a full-length scorpion toxin (i.e., BmKcug2) and its four truncated analogs (i.e., BmKcug2-P1, BmKcug2-P2, BmKcug2-P3 and BmKcug2-P4) with three conserved disulfide bonds in processed scorpion medicinal material by mass spectrometry. The pharmacological experiments revealed that the recombinant BmKcug2 and BmKcug2-P1 could selectively inhibit the human Kv1.2 and human Kv1.3 potassium channels, while the other three analogs showed a much weaker inhibitory effect on potassium channels. BmKcug2 inhibited hKv1.2 and hKv1.3 channels, with IC50 values of 45.6 ± 5.8 nM and 215.2 ± 39.7 nM, respectively, and BmKcug2-P1 inhibited hKv1.2 and hKv1.3, with IC50 values of 89.9 ± 9.6 nM and 1142.4 ± 64.5 nM, respectively. The chromatographic analysis and pharmacological properties of BmKcug2 and BmKcug2-P1 boiled in water for different times further strongly supported their good thermal stability. Structural and functional dissection indicated that one amino acid, i.e., Tyr36, determined the differential affinities of BmKcug2 and four BmKcug2 analogs. Altogether, this research investigated the different pharmacological properties of BmKcug2 and its truncated analogs, and the findings highlighted the diversity of K+ channel blockers from various scorpion species through thermal processing.
Collapse
Affiliation(s)
- Chenhu Qin
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiuping Wan
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Songryong Li
- College of Life Sciences, Wuhan University, Wuhan 430072, China; Department of Biotechnology, Institute for Life Science, Kim Hyong Jik University of Education, Pyongyang, Democratic People's Republic of Korea
| | - Fan Yang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Liuting Yang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zheng Zuo
- Center for BioDrug Research, Wuhan University, Wuhan 430072, China
| | - Zhijian Cao
- College of Life Sciences, Wuhan University, Wuhan 430072, China; Center for BioDrug Research, Wuhan University, Wuhan 430072, China
| | - Zongyun Chen
- College of Life Sciences, Wuhan University, Wuhan 430072, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hubei University of Medicine, Shiyan 442000, China
| | - Yingliang Wu
- College of Life Sciences, Wuhan University, Wuhan 430072, China; Center for BioDrug Research, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
6
|
Abe N, Toyama H, Saito K, Ejima Y, Yamauchi M, Mushiake H, Kazama I. Delayed Rectifier K +-Channel Is a Novel Therapeutic Target for Interstitial Renal Fibrosis in Rats with Unilateral Ureteral Obstruction. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7567638. [PMID: 31828127 PMCID: PMC6885154 DOI: 10.1155/2019/7567638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/15/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Delayed rectifier K+-channel, Kv1.3, is most predominantly expressed in T-lymphocytes and macrophages. In such leukocytes, Kv1.3-channels play pivotal roles in the activation and proliferation of cells, promoting cellular immunity. Since leukocyte-derived cytokines stimulate fibroblasts to produce collagen fibers in inflamed kidneys, Kv1.3-channels expressed in leukocytes would contribute to the progression of tubulointerstitial renal fibrosis. METHODS Male Sprague-Dawley rats that underwent unilateral ureteral obstruction (UUO) were used at 1, 2, or 3 weeks after the operation. We examined the histological features of the kidneys and the leukocyte expression of Kv1.3-channels. We also examined the therapeutic effects of a selective channel inhibitor, margatoxin, on the progression of renal fibrosis and the proliferation of leukocytes within the cortical interstitium. RESULTS In rat kidneys with UUO, progression of renal fibrosis and the infiltration of leukocytes became most prominent at 3 weeks after the operation, when Kv1.3-channels were overexpressed in proliferating leukocytes. In the cortical interstitium of margatoxin-treated UUO rat kidneys, immunohistochemistry revealed reduced expression of fibrosis markers. Additionally, margatoxin significantly decreased the numbers of leukocytes and suppressed their proliferation. CONCLUSIONS This study clearly demonstrated that the numbers of T-lymphocytes and macrophages were markedly increased in UUO rat kidneys with longer postobstructive days. The overexpression of Kv1.3-channels in leukocytes was thought to be responsible for the proliferation of these cells and the progression of renal fibrosis. This study strongly suggested the therapeutic usefulness of targeting lymphocyte Kv1.3-channels in the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Nozomu Abe
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Hiroaki Toyama
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Kazutomo Saito
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Yutaka Ejima
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Masanori Yamauchi
- Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Hajime Mushiake
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
| | - Itsuro Kazama
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan
- Miyagi University, School of Nursing, Gakuen, Taiwa-cho, Kurokawa-gun, Miyagi, Japan
| |
Collapse
|
7
|
Zhao Y, Chen Z, Cao Z, Li W, Wu Y. Diverse Structural Features of Potassium Channels Characterized by Scorpion Toxins as Molecular Probes. Molecules 2019; 24:molecules24112045. [PMID: 31146335 PMCID: PMC6600638 DOI: 10.3390/molecules24112045] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/15/2019] [Accepted: 05/26/2019] [Indexed: 12/21/2022] Open
Abstract
Scorpion toxins are well-known as the largest potassium channel peptide blocker family. They have been successfully proven to be valuable molecular probes for structural research on diverse potassium channels. The potassium channel pore region, including the turret and filter regions, is the binding interface for scorpion toxins, and structural features from different potassium channels have been identified using different scorpion toxins. According to the spatial orientation of channel turrets with differential sequence lengths and identities, conformational changes and molecular surface properties, the potassium channel turrets can be divided into the following three states: open state with less hindering effects on toxin binding, half-open state or half-closed state with certain effects on toxin binding, and closed state with remarkable effects on toxin binding. In this review, we summarized the diverse structural features of potassium channels explored using scorpion toxin tools and discuss future work in the field of scorpion toxin-potassium channel interactions.
Collapse
Affiliation(s)
- Yonghui Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Zongyun Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Basic Medicine, Hubei University of Medicine, Shiyan 442000, China.
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
- Biodrug Research Center, Wuhan University, Wuhan 430072, China.
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
- Biodrug Research Center, Wuhan University, Wuhan 430072, China.
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
- Biodrug Research Center, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
8
|
Shen B, Cao Z, Li W, Sabatier JM, Wu Y. Treating autoimmune disorders with venom-derived peptides. Expert Opin Biol Ther 2017; 17:1065-1075. [DOI: 10.1080/14712598.2017.1346606] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Bingzheng Shen
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, China
- Department of Pharmacy, Renmin Hospital, Wuhan University, Wuhan, China
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, China
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, China
| | | | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Nekrasova OV, Volyntseva AD, Kudryashova KS, Novoseletsky VN, Lyapina EA, Illarionova AV, Yakimov SA, Korolkova YV, Shaitan KV, Kirpichnikov MP, Feofanov AV. Complexes of Peptide Blockers with Kv1.6 Pore Domain: Molecular Modeling and Studies with KcsA-Kv1.6 Channel. J Neuroimmune Pharmacol 2016; 12:260-276. [PMID: 27640211 DOI: 10.1007/s11481-016-9710-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/09/2016] [Indexed: 11/25/2022]
Abstract
Potassium voltage-gated Kv1.6 channel, which is distributed primarily in neurons of central and peripheral nervous systems, is of significant physiological importance. To date, several high-affinity Kv1.6-channel blockers are known, but the lack of selective ones among them hampers the studies of tissue localization and functioning of Kv1.6 channels. Here we present an approach to advanced understanding of interactions of peptide toxin blockers with a Kv1.6 pore. It combines molecular modeling studies and an application of a new bioengineering system based on a KcsA-Kv1.6 hybrid channel for the quantitative fluorescent analysis of blocker-channel interactions. Using this system we demonstrate that peptide toxins agitoxin 2, kaliotoxin1 and OSK1 have similar high affinity to the extracellular vestibule of the K+-conducting pore of Kv1.6, hetlaxin is a low-affinity ligand, whereas margatoxin and scyllatoxin do not bind to Kv1.6 pore. Binding of toxins to Kv1.6 pore has considerable inverse dependence on the ionic strength. Model structures of KcsA-Kv1.6 and Kv1.6 complexes with agitoxin 2, kaliotoxin 1 and OSK1 were obtained using homology modeling and molecular dynamics simulation. Interaction interfaces, which are formed by 15-19 toxin residues and 10 channel residues, are described and compared. Specific sites of Kv1.6 pore recognition are identified for targeting of peptide blockers. Analysis of interactions between agitoxin 2 derivatives with point mutations (S7K, S11G, L19S, R31G) and KcsA-Kv1.6 confirms reliability of the calculated complex structure.
Collapse
Affiliation(s)
- O V Nekrasova
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia
| | - A D Volyntseva
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia
| | - K S Kudryashova
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia
| | - V N Novoseletsky
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia
| | - E A Lyapina
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia
| | - A V Illarionova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia
| | - S A Yakimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia
| | - Yu V Korolkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia
| | - K V Shaitan
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia
| | - M P Kirpichnikov
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia
| | - A V Feofanov
- Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, Moscow, 119992, Russia. .,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| |
Collapse
|
10
|
Ye F, Hu Y, Yu W, Xie Z, Hu J, Cao Z, Li W, Wu Y. The Scorpion Toxin Analogue BmKTX-D33H as a Potential Kv1.3 Channel-Selective Immunomodulator for Autoimmune Diseases. Toxins (Basel) 2016; 8:115. [PMID: 27104568 PMCID: PMC4848641 DOI: 10.3390/toxins8040115] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 01/23/2023] Open
Abstract
The Kv1.3 channel-acting scorpion toxins usually adopt the conserved anti-parallel β-sheet domain as the binding interface, but it remains challenging to discover some highly selective Kv1.3 channel-acting toxins. In this work, we investigated the pharmacological profile of the Kv1.3 channel-acting BmKTX-D33H, a structural analogue of the BmKTX scorpion toxin. Interestingly, BmKTX-D33H, with its conserved anti-parallel β-sheet domain as a Kv1.3 channel-interacting interface, exhibited more than 1000-fold selectivity towards the Kv1.3 channel as compared to other K+ channels (including Kv1.1, Kv1.2, Kv1.7, Kv11.1, KCa2.2, KCa2.3, and KCa3.1). As expected, BmKTX-D33H was found to inhibit the cytokine production and proliferation of both Jurkat cells and human T cells in vitro. It also significantly improved the delayed-type hypersensitivity (DTH) responses, an autoreactive T cell-mediated inflammation in rats. Amino acid sequence alignment and structural analysis strongly suggest that the “evolutionary” Gly11 residue of BmKTX-D33H interacts with the turret domain of Kv1 channels; it appears to be a pivotal amino acid residue with regard to the selectivity of BmKTX-D33H towards the Kv1.3 channel (in comparison with the highly homologous scorpion toxins). Together, our data indicate that BmKTX-D33H is a Kv1.3 channel–specific blocker. Finally, the remarkable selectivity of BmKTX-D33H highlights the great potential of evolutionary-guided peptide drug design in future studies.
Collapse
Affiliation(s)
- Fang Ye
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Youtian Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Weiwei Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Zili Xie
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Jun Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
11
|
Zhang L, Shi W, Zeng XC, Ge F, Yang M, Nie Y, Bao A, Wu S, E G. Unique diversity of the venom peptides from the scorpion Androctonus bicolor revealed by transcriptomic and proteomic analysis. J Proteomics 2015; 128:231-50. [DOI: 10.1016/j.jprot.2015.07.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/21/2015] [Accepted: 07/24/2015] [Indexed: 12/22/2022]
|
12
|
Kazama I, Tamada T, Tachi M. Usefulness of targeting lymphocyte Kv1.3-channels in the treatment of respiratory diseases. Inflamm Res 2015. [PMID: 26206235 DOI: 10.1007/s00011-015-0855-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
T lymphocytes predominantly express delayed rectifier K(+)-channels (Kv1.3) in their plasma membranes. Patch-clamp studies revealed that the channels play crucial roles in facilitating the calcium influx necessary to trigger lymphocyte activation and proliferation. Using selective channel inhibitors in experimental animal models, in vivo studies further revealed the clinically relevant relationship between the channel expression and the development of chronic respiratory diseases, in which chronic inflammation or the overstimulation of cellular immunity in the airways is responsible for the pathogenesis. In chronic respiratory diseases, such as chronic obstructive pulmonary disease, asthma, diffuse panbronchiolitis and cystic fibrosis, in addition to the supportive management for the symptoms, the anti-inflammatory effects of macrolide antibiotics were shown to be effective against the over-activation or proliferation of T lymphocytes. Recently, we provided physiological and pharmacological evidence that macrolide antibiotics, together with calcium channel blockers, HMG-CoA reductase inhibitors, and nonsteroidal anti-inflammatory drugs, effectively suppress the Kv1.3-channel currents in lymphocytes, and thus exert anti-inflammatory or immunomodulatory effects. In this review article, based on the findings obtained from recent in vivo and in vitro studies, we address the novel therapeutic implications of targeting the lymphocyte Kv1.3-channels for the treatment of chronic or acute respiratory diseases.
Collapse
Affiliation(s)
- Itsuro Kazama
- Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan.
| | - Tsutomu Tamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masahiro Tachi
- Department of Plastic and Reconstructive Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
13
|
Roles of lymphocyte Kv1.3-channels in gut mucosal immune system: Novel therapeutic implications for inflammatory bowel disease. Med Hypotheses 2015; 85:61-3. [DOI: 10.1016/j.mehy.2015.03.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/25/2015] [Indexed: 01/16/2023]
|
14
|
Chen Z, Hu Y, Hong J, Hu J, Yang W, Xiang F, Yang F, Xie Z, Cao Z, Li W, Lin D, Wu Y. Toxin acidic residue evolutionary function-guided design of de novo peptide drugs for the immunotherapeutic target, the Kv1.3 channel. Sci Rep 2015; 5:9881. [PMID: 25955787 PMCID: PMC4424837 DOI: 10.1038/srep09881] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/24/2015] [Indexed: 12/25/2022] Open
Abstract
During the long-term evolution of animal toxins acting on potassium channels, the acidic residues can orientate the toxin binding interfaces by adjusting the molecular polarity. Based on the evolutionary function of toxin acidic residues, de novo peptide drugs with distinct binding interfaces were designed for the immunotherapeutic target, the Kv1.3 channel. Using a natural basic toxin, BmKTX, as a template, which contains 2 acidic residues (Asp19 and Asp33), we engineered two new peptides BmKTX-19 with 1 acidic residue (Asp33), and BmKTX-196 with 2 acidic residues (Asp6 and Asp33) through only adjusting acidic residue distribution for reorientation of BmKTX binding interface. Pharmacological experiments indicated that BmKTX-19 and BmKTX-196 peptides were specific inhibitors of the Kv1.3 channel and effectively suppressed cytokine secretion. In addition to the structural similarity between the designed and native peptides, both experimental alanine-scanning mutagenesis and computational simulation further indicated that the binding interface of wild-type BmKTX was successfully reoriented in BmKTX-19 and BmKTX-196, which adopted distinct toxin surfaces as binding interfaces. Together, these findings indicate not only the promising prospect of BmKTX-19 and BmKTX-196 as drug candidates but also the desirable feasibility of the evolution-guided peptide drug design for discovering numerous peptide drugs for the Kv1.3 channel.
Collapse
Affiliation(s)
- Zongyun Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Youtian Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jing Hong
- College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
| | - Jun Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Weishan Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Fang Xiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Fan Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zili Xie
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhijian Cao
- 1] State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China [2] Center for BioDrug Research, Wuhan University, Wuhan 430072, China
| | - Wenxin Li
- 1] State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China [2] Center for BioDrug Research, Wuhan University, Wuhan 430072, China
| | - Donghai Lin
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yingliang Wu
- 1] State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China [2] Center for BioDrug Research, Wuhan University, Wuhan 430072, China
| |
Collapse
|
15
|
Roles of lymphocyte kv1.3-channels in the pathogenesis of renal diseases and novel therapeutic implications of targeting the channels. Mediators Inflamm 2015; 2015:436572. [PMID: 25866450 PMCID: PMC4381730 DOI: 10.1155/2015/436572] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/25/2015] [Indexed: 12/04/2022] Open
Abstract
Delayed rectifier K+-channels (Kv1.3) are predominantly expressed in T lymphocytes. Based on patch-clamp studies, the channels play crucial roles in facilitating the calcium influx necessary to trigger lymphocyte activation and proliferation. Using selective channel inhibitors in experimental animal models, in vivo studies then revealed the clinically relevant relationship between the channel expression and the pathogenesis of autoimmune diseases. In renal diseases, in which “chronic inflammation” or “the overstimulation of cellular immunity” is responsible for the pathogenesis, the overexpression of Kv1.3-channels in lymphocytes promotes their cellular proliferation and thus contributes to the progression of tubulointerstitial fibrosis. We recently demonstrated that benidipine, a potent dihydropyridine calcium channel blocker, which also strongly and persistently inhibits the lymphocyte Kv1.3-channel currents, suppressed the proliferation of kidney lymphocytes and actually ameliorated the progression of renal fibrosis. Based on the recent in vitro evidence that revealed the pharmacological properties of the channels, the most recent studies have revealed novel therapeutic implications of targeting the lymphocyte Kv1.3-channels for the treatment of renal diseases.
Collapse
|
16
|
Yang W, Feng J, Xiang F, Xie Z, Zhang G, Sabatier JM, Cao Z, Li W, Chen Z, Wu Y. Endogenous animal toxin-like human β-defensin 2 inhibits own K(+) channels through interaction with channel extracellular pore region. Cell Mol Life Sci 2015; 72:845-53. [PMID: 25238780 PMCID: PMC11113244 DOI: 10.1007/s00018-014-1715-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 11/25/2022]
Abstract
Human potassium channels are widely inhibited by peptide toxins from venomous animals. However, no human endogenous peptide inhibitor has been discovered so far. In this study, we demonstrate for the first time using electrophysiological techniques, that endogenous human β-defensin 2 (hBD2) is able to selectively and dose-dependently inhibit the human voltage-gated Kv1.3 channel at picomolar peptide concentration. The co-immunoprecipitation assays further supported the selective binding of hBD2 to Kv1.3 channel. Using mutagenesis experiments, we found that the outer pore domain of Kv1.3 channel was the binding site of hBD2, which is similar to the interacting site of Kv1.3 channel recognized by animal toxin inhibitors. The hBD2 was able to suppress IL-2 production through inhibition of Kv1.3 channel currents in human Jurkat cells, which was further confirmed by the lack of hBD2 activity on IL-2 production after Kv1.3 knockdown in these cells. More interestingly, hBD2 was also found to efficiently inhibit Kv1.3 channel currents and suppress IL-2 production in both human primary CD3(+) T cells and peripheral mononuclear cells from either healthy donors or psoriasis patients. Our findings not only evidenced hBD2 as the first characterized endogenous peptide inhibitor of human potassium channels, but also paved a promising avenue to investigate newly discovered function of hBD2 as Kv1.3 channel inhibitor in the immune system and other fields.
Collapse
Affiliation(s)
- Weishan Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Jing Feng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Fang Xiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Zili Xie
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Guoyi Zhang
- Institute of Dermatology, Chinese Academy of Medicine Sciences, Nanjing, 210042 China
| | - Jean-Marc Sabatier
- Laboratoire INSERM UMR 1097, Université d’Aix-Marseille, 163, Avenue de Luminy, Case 939, TPR2 INSERM, 13288 Marseille Cedex 09, France
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
- Center for BioDrug Research, Wuhan University, Wuhan, 430072 China
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
- Center for BioDrug Research, Wuhan University, Wuhan, 430072 China
| | - Zongyun Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072 China
- Center for BioDrug Research, Wuhan University, Wuhan, 430072 China
| |
Collapse
|
17
|
Kazama I. Physiological significance of delayed rectifier K(+) channels (Kv1.3) expressed in T lymphocytes and their pathological significance in chronic kidney disease. J Physiol Sci 2015; 65:25-35. [PMID: 25096892 PMCID: PMC10717717 DOI: 10.1007/s12576-014-0331-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/14/2014] [Indexed: 11/24/2022]
Abstract
T lymphocytes predominantly express delayed rectifier K(+) channels (Kv1.3) in their plasma membranes. More than 30 years ago, patch-clamp studies revealed that the channels play crucial roles in facilitating the calcium influx necessary to trigger lymphocyte activation and proliferation. In addition to selective channel inhibitors that have been developed, we recently showed physiological evidence that drugs such as nonsteroidal anti-inflammatory drugs, antibiotics, and anti-hypertensives effectively suppress the channel currents in lymphocytes, and thus exert immunosuppressive effects. Using experimental animal models, previous studies revealed the pathological relevance between the expression of ion channels and the progression of renal diseases. As an extension, we recently demonstrated that the overexpression of lymphocyte Kv1.3 channels contributed to the progression of chronic kidney disease (CKD) by promoting cellular proliferation and interstitial fibrosis. Together with our in-vitro results, the studies indicated the therapeutic potency of Kv1.3-channel inhibitors in the treatment or the prevention of CKD.
Collapse
Affiliation(s)
- Itsuro Kazama
- Department of Physiology I, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi, Japan,
| |
Collapse
|
18
|
Quintero-Hernández V, Jiménez-Vargas J, Gurrola G, Valdivia H, Possani L. Scorpion venom components that affect ion-channels function. Toxicon 2013; 76:328-42. [PMID: 23891887 PMCID: PMC4089097 DOI: 10.1016/j.toxicon.2013.07.012] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 07/16/2013] [Indexed: 12/31/2022]
Abstract
The number and types of venom components that affect ion-channel function are reviewed. These are the most important venom components responsible for human intoxication, deserving medical attention, often requiring the use of specific anti-venoms. Special emphasis is given to peptides that recognize Na(+)-, K(+)- and Ca(++)-channels of excitable cells. Knowledge generated by direct isolation of peptides from venom and components deduced from cloned genes, whose amino acid sequences are deposited into databanks are nowadays in the order of 1.5 thousands, out of an estimate biodiversity closed to 300,000. Here the diversity of components is briefly reviewed with mention to specific references. Structural characteristic are discussed with examples taken from published work. The principal mechanisms of action of the three different types of peptides are also reviewed. Na(+)-channel specific venom components usually are modifier of the open and closing kinetic mechanisms of the ion-channels, whereas peptides affecting K(+)-channels are normally pore blocking agents. The Ryanodine Ca(++)-channel specific peptides are known for causing sub-conducting stages of the channels conductance and some were shown to be able to internalize penetrating inside the muscle cells.
Collapse
Affiliation(s)
- V. Quintero-Hernández
- Department of Molecular Medicine and Bioprocesses, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Avenida Universidad, 2001, Apartado Postal 510-3, Cuernavaca 62210, Morelos, Mexico
| | - J.M. Jiménez-Vargas
- Department of Molecular Medicine and Bioprocesses, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Avenida Universidad, 2001, Apartado Postal 510-3, Cuernavaca 62210, Morelos, Mexico
| | - G.B. Gurrola
- Department of Molecular Medicine and Bioprocesses, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Avenida Universidad, 2001, Apartado Postal 510-3, Cuernavaca 62210, Morelos, Mexico
- Cardiovascular Center 2139, Michigan University, Ann Arbor, MI 48109-5644, U.S.A
| | - H.H.F. Valdivia
- Cardiovascular Center 2139, Michigan University, Ann Arbor, MI 48109-5644, U.S.A
| | - L.D. Possani
- Department of Molecular Medicine and Bioprocesses, Instituto de Biotecnologia, Universidad Nacional Autonoma de Mexico, Avenida Universidad, 2001, Apartado Postal 510-3, Cuernavaca 62210, Morelos, Mexico
| |
Collapse
|
19
|
Ali SA, Alam M, Abbasi A, Kalbacher H, Schaechinger TJ, Hu Y, Zhijian C, Li W, Voelter W. Structure–Activity Relationship of a Highly Selective Peptidyl Inhibitor of Kv1.3 Voltage-Gated K+-Channel from Scorpion (B. sindicus) Venom. Int J Pept Res Ther 2013. [DOI: 10.1007/s10989-013-9362-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
20
|
Gordon D, Chen R, Chung SH. Computational methods of studying the binding of toxins from venomous animals to biological ion channels: theory and applications. Physiol Rev 2013; 93:767-802. [PMID: 23589832 PMCID: PMC3768100 DOI: 10.1152/physrev.00035.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The discovery of new drugs that selectively block or modulate ion channels has great potential to provide new treatments for a host of conditions. One promising avenue revolves around modifying or mimicking certain naturally occurring ion channel modulator toxins. This strategy appears to offer the prospect of designing drugs that are both potent and specific. The use of computational modeling is crucial to this endeavor, as it has the potential to provide lower cost alternatives for exploring the effects of new compounds on ion channels. In addition, computational modeling can provide structural information and theoretical understanding that is not easily derivable from experimental results. In this review, we look at the theory and computational methods that are applicable to the study of ion channel modulators. The first section provides an introduction to various theoretical concepts, including force-fields and the statistical mechanics of binding. We then look at various computational techniques available to the researcher, including molecular dynamics, brownian dynamics, and molecular docking systems. The latter section of the review explores applications of these techniques, concentrating on pore blocker and gating modifier toxins of potassium and sodium channels. After first discussing the structural features of these channels, and their modes of block, we provide an in-depth review of past computational work that has been carried out. Finally, we discuss prospects for future developments in the field.
Collapse
Affiliation(s)
- Dan Gordon
- Research School of Biology, The Australian National University, Acton, ACT 0200, Australia.
| | | | | |
Collapse
|
21
|
Feng J, Yu C, Wang M, Li Z, Wu Y, Cao Z, Li W, He X, Han S. Expression and characterization of a novel scorpine-like peptide Ev37, from the scorpion Euscorpiops validus. Protein Expr Purif 2012; 88:127-33. [PMID: 23262394 DOI: 10.1016/j.pep.2012.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 12/10/2012] [Accepted: 12/12/2012] [Indexed: 11/17/2022]
Abstract
Scorpion venom contains a group of two-domain peptides that function to block potassium channels or have cytolytic activities. These peptides, whose functions are poorly studied, are named β-KTx or scorpine-like peptides. Ev37, the first identified gene in the Euscorpiidae family, which encoded a novel scorpine-like peptide, was cloned from the venom cDNA library of scorpion Euscorpiops validus. Sequence analysis showed that the mature Ev37 peptide contained 78 amino acid residues, which formed two structural domains: a putative α-helical N-terminus and a C-terminus with the cysteine-stabilized α/β motif. The peptide rEv37 and two truncated peptides representing the individual structural domains (Ev37-N and Ev37-C) were expressed in Escherichia coli and purified for functional study. Unlike classic scorpine-like peptides, rEv37 and truncated peptides showed no cytolytic activity against bacteria or eukaryotic cells. Interestingly, rEv37 selectively inhibited Kv1.3 channel without effectively blocking Kv1.1 and Kv1.2 channels. Neither Ev37-N nor Ev37-C blocked Kv1.3 channel, suggesting that both the N-terminal and C-terminal domain of Ev37 are likely involved in the interaction with Kv1.3 channel. These results not only enrich our knowledge of scorpion toxins from scorpine-like subfamily but also provide a novel template with unique structure for designing new types of selective Kv1.3 blockers.
Collapse
Affiliation(s)
- Jing Feng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Xie S, Feng J, Yu C, Li Z, Wu Y, Cao Z, Li W, He X, Xiang M, Han S. Identification of a new specific Kv1.3 channel blocker, Ctri9577, from the scorpion Chaerilus tricostatus. Peptides 2012; 36:94-9. [PMID: 22580271 DOI: 10.1016/j.peptides.2012.04.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 04/26/2012] [Accepted: 04/26/2012] [Indexed: 01/04/2023]
Abstract
Scorpion toxins are valuable resources for discovering new ion channel modulators and drug candidates. Potassium channel Kv1.3 is an important pharmacological target of T cell-mediated autoimmune diseases, which are encouraging the screening and design of the specific peptide blockers for Kv1.3 channel. Ctri9577, the first neurotoxin gene of Chaerilidae family was cloned from the venom of the scorpion Chaerilus tricostatus through the constructing its cDNA library. The sequence analysis showed that the mature peptide of Ctri9577 contained 39 amino acid residues including six conserved cysteines, whose low sequence similarity indicated that it was a new member of α-KTx15 subfamily. By using expression and purification technology, the recombinant peptide was obtained. Subsequently, the electrophysiological experiments indicated that the Ctri9577 peptide selectively inhibited Kv1.3 channel current with an IC(50) of 0.49±0.45 nM without effectively blocking potassium channels Kv1.1, Kv1.2, hERG and SK3. All these findings not only enrich the knowledge of toxins from the Chaerilidae family, but also present a novel potential drug candidate targeting Kv1.3 channels for the therapy of autoimmune diseases.
Collapse
Affiliation(s)
- Shujun Xie
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chen R, Robinson A, Gordon D, Chung SH. Modeling the binding of three toxins to the voltage-gated potassium channel (Kv1.3). Biophys J 2012; 101:2652-60. [PMID: 22261053 DOI: 10.1016/j.bpj.2011.10.029] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 10/20/2011] [Accepted: 10/24/2011] [Indexed: 11/24/2022] Open
Abstract
The conduction properties of the voltage-gated potassium channel Kv1.3 and its modes of interaction with several polypeptide venoms are examined using Brownian dynamics simulations and molecular dynamics calculations. Employing an open-state homology model of Kv1.3, we first determine current-voltage and current-concentration curves and ascertain that simulated results accord with experimental measurements. We then investigate, using a molecular docking method and molecular dynamics simulations, the complexes formed between the Kv1.3 channel and several Kv-specific polypeptide toxins that are known to interfere with the conducting mechanisms of several classes of voltage-gated K(+) channels. The depths of potential of mean force encountered by charybdotoxin, α-KTx3.7 (also known as OSK1) and ShK are, respectively, -19, -27, and -25 kT. The dissociation constants calculated from the profiles of potential of mean force correspond closely to the experimentally determined values. We pinpoint the residues in the toxins and the channel that are critical for the formation of the stable venom-channel complexes.
Collapse
Affiliation(s)
- Rong Chen
- Research School of Biology, Australian National University, Canberra, Australia
| | | | | | | |
Collapse
|
24
|
Chen ZY, Hu YT, Yang WS, He YW, Feng J, Wang B, Zhao RM, Ding JP, Cao ZJ, Li WX, Wu YL. Hg1, novel peptide inhibitor specific for Kv1.3 channels from first scorpion Kunitz-type potassium channel toxin family. J Biol Chem 2012; 287:13813-21. [PMID: 22354971 DOI: 10.1074/jbc.m112.343996] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The potassium channel Kv1.3 is an attractive pharmacological target for autoimmune diseases. Specific peptide inhibitors are key prospects for diagnosing and treating these diseases. Here, we identified the first scorpion Kunitz-type potassium channel toxin family with three groups and seven members. In addition to their function as trypsin inhibitors with dissociation constants of 140 nM for recombinant LmKTT-1a, 160 nM for LmKTT-1b, 124 nM for LmKTT-1c, 136 nM for BmKTT-1, 420 nM for BmKTT-2, 760 nM for BmKTT-3, and 107 nM for Hg1, all seven recombinant scorpion Kunitz-type toxins could block the Kv1.3 channel. Electrophysiological experiments showed that six of seven scorpion toxins inhibited ~50-80% of Kv1.3 channel currents at a concentration of 1 μM. The exception was rBmKTT-3, which had weak activity. The IC(50) values of rBmKTT-1, rBmKTT-2, and rHg1 for Kv1.3 channels were ~129.7, 371.3, and 6.2 nM, respectively. Further pharmacological experiments indicated that rHg1 was a highly selective Kv1.3 channel inhibitor with weak affinity for other potassium channels. Different from classical Kunitz-type potassium channel toxins with N-terminal regions as the channel-interacting interfaces, the channel-interacting interface of Hg1 was in the C-terminal region. In conclusion, these findings describe the first scorpion Kunitz-type potassium channel toxin family, of which a novel inhibitor, Hg1, is specific for Kv1.3 channels. Their structural and functional diversity strongly suggest that Kunitz-type toxins are a new source to screen and design potential peptides for diagnosing and treating Kv1.3-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Zong-Yun Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen R, Chung SH. Engineering a potent and specific blocker of voltage-gated potassium channel Kv1.3, a target for autoimmune diseases. Biochemistry 2012; 51:1976-82. [PMID: 22352687 DOI: 10.1021/bi201811j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A polypeptide toxin extracted from scorpion venom, OSK1, is modified such that its potency is drastically enhanced in blocking one class of voltage-gated potassium channels, Kv1.3, which is a pharmacological target for immunosuppressive therapy. The bound complex of Kv1.3 and OSK1 reveals that one lysine residue of the toxin is in the proximity of another lysine residue on the external vestibule of the channel, just outside of the selectivity filter. This unfavorable electrostatic interaction is eliminated by interchanging the positions of two amino acids in the toxin. The potentials of mean force of the wild-type and mutant OSK1 bound to Kv1.1-Kv1.3 channels are constructed using molecular dynamics, and the half-maximal inhibitory concentration (IC(50)) of each toxin-channel complex is computed. We show that the IC(50) values predicted for three toxins and three channels match closely with experiment. Kv1.3 is half-blocked by 0.2 pM mutant OSK1; it is >10000-fold more specific for this channel than for Kv1.1 and Kv1.2.
Collapse
Affiliation(s)
- Rong Chen
- Research School of Biology, Australian National University, Canberra, ACT 0200, Australia.
| | | |
Collapse
|
26
|
Chen Z, Hu Y, Han S, Yin S, He Y, Wu Y, Cao Z, Li W. ImKTx1, a new Kv1.3 channel blocker with a unique primary structure. J Biochem Mol Toxicol 2011; 25:244-51. [DOI: 10.1002/jbt.20382] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 05/13/2010] [Accepted: 06/03/2010] [Indexed: 12/25/2022]
|
27
|
Han S, Hu Y, Zhang R, Yi H, Wei J, Wu Y, Cao Z, Li W, He X. ImKTx88, a novel selective Kv1.3 channel blocker derived from the scorpion Isometrus maculates. Toxicon 2010; 57:348-55. [PMID: 21194541 DOI: 10.1016/j.toxicon.2010.12.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 12/20/2010] [Accepted: 12/20/2010] [Indexed: 01/15/2023]
Abstract
Scorpion toxins are useful in the structure-function research of ion channels and valuable resources for drug design. The Kv1.3 channel is an important pharmacological target for the therapy of T cell-mediated autoimmune diseases, and many toxin peptides targeting Kv1.3 have been identified as good drug candidates in recent years. In this study, a novel toxin gene ImKTx88 was isolated from the venom of the scorpion Isometrus maculates through the construction of the cDNA library method, and the recombinant toxin peptide was purified and characterized physiologically. The mature peptide of ImKTx88 contained 39 amino acid residues including six cysteines and was predicted to be a new member of α-KTx scorpion family by sequence analysis. The electrophysiological experiments further indicated that the rImKTx88 peptide had a novel pharmacological profile: it inhibited Kv1.3 channel current with an IC₅₀ of 91 ± 42 pM, and exhibited very good selectivity for Kv1.3 over Kv1.1 (4200-fold) and Kv1.2 (93000-fold) channels, respectively. All these results suggested that, as a new selective Kv1.3 channel blocker, the ImKTx88 peptide may serve as a potential drug candidate in the therapy of autoimmune diseases.
Collapse
Affiliation(s)
- Song Han
- School of Medicine, Wuhan University, Wuhan 430071, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Han S, Yin S, Yi H, Mouhat S, Qiu S, Cao Z, Sabatier JM, Wu Y, Li W. Protein−Protein Recognition Control by Modulating Electrostatic Interactions. J Proteome Res 2010; 9:3118-25. [DOI: 10.1021/pr100027k] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Song Han
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Shijin Yin
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Hong Yi
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Stéphanie Mouhat
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Su Qiu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Jean-Marc Sabatier
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P. R. China, and ERT 62 ≪Ingénierie des peptides à visée thérapeutique≫, Université de la Méditerranée - Ambrilia Biopharma S.A., Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille Cédex 20, France
| |
Collapse
|
29
|
Abstract
For more than 25 years, it has been widely appreciated that Ca2+ influx is essential to trigger T-lymphocyte activation. Patch clamp analysis, molecular identification, and functional studies using blockers and genetic manipulation have shown that a unique contingent of ion channels orchestrates the initiation, intensity, and duration of the Ca2+ signal. Five distinct types of ion channels--Kv1.3, KCa3.1, Orai1+ stromal interacting molecule 1 (STIM1) [Ca2+-release activating Ca2+ (CRAC) channel], TRPM7, and Cl(swell)--comprise a network that performs functions vital for ongoing cellular homeostasis and for T-cell activation, offering potential targets for immunomodulation. Most recently, the roles of STIM1 and Orai1 have been revealed in triggering and forming the CRAC channel following T-cell receptor engagement. Kv1.3, KCa3.1, STIM1, and Orai1 have been found to cluster at the immunological synapse following contact with an antigen-presenting cell; we discuss how channels at the synapse might function to modulate local signaling. Immuno-imaging approaches are beginning to shed light on ion channel function in vivo. Importantly, the expression pattern of Ca2+ and K+ channels and hence the functional network can adapt depending upon the state of differentiation and activation, and this allows for different stages of an immune response to be targeted specifically.
Collapse
Affiliation(s)
- Michael D Cahalan
- Department of Physiology and Biophysics, and the Institute for Immunology, University of California, Irvine, Irvine, CA 92697-4561, USA.
| | | |
Collapse
|
30
|
Liu J, Ma Y, Yin S, Zhao R, Fan S, Hu Y, Wu Y, Cao Z, Li W. Molecular cloning and functional identification of a new K(+) channel blocker, LmKTx10, from the scorpion Lychas mucronatus. Peptides 2009; 30:675-80. [PMID: 19103241 DOI: 10.1016/j.peptides.2008.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 11/21/2008] [Accepted: 11/21/2008] [Indexed: 12/31/2022]
Abstract
Scorpions have a venom gland which is an important determinant in contributing to their successful survival for more than 400 million years. Their venoms contain a diversity of neurotoxins, which represent a tremendous hitherto partially unexplored resource not only for understanding ion channels but also for use in drug design and development. In this investigation, LmKTx10, a new toxin gene was identified from the venom of the scorpion Lychas mucronatus by constructing cDNA library method, and its product was expressed and characterized physiologically. The mature peptide has 38 residues including six conserved cysteines. The electrophysiological experiments further indicated that the recombinant LmKTx10 peptide has an interesting pharmacological profile: it blocks Kv1.3 channel with IC(50)=28nM which is moderate Kv1.3 channel blocking activity compared to the other a-KTxs toxins, and exhibits good selectivity on Kv1.3 over Kv1.1 and Kv1.2, about 60 folds and 450 folds, respectively. These data not only enrich the family of K(+) channel toxins from scorpion venoms but also present a potential drug template for selectively targeting the Kv1.3 channel.
Collapse
Affiliation(s)
- Jun Liu
- Wuhan University, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yin SJ, Jiang L, Yi H, Han S, Yang DW, Liu ML, Liu H, Cao ZJ, Wu YL, Li WX. Different Residues in Channel Turret Determining the Selectivity of ADWX-1 Inhibitor Peptide between Kv1.1 and Kv1.3 Channels. J Proteome Res 2008; 7:4890-7. [DOI: 10.1021/pr800494a] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Shi-Jin Yin
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Ling Jiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Hong Yi
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Song Han
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Dai-Wen Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Mai-Li Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Hui Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Zhi-Jian Cao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Ying-Liang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| | - Wen-Xin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P. R. China, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, P. R. China, and Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore 117543
| |
Collapse
|
32
|
Corzo G, Papp F, Varga Z, Barraza O, Espino-Solis PG, Rodríguez de la Vega RC, Gaspar R, Panyi G, Possani LD. A selective blocker of Kv1.2 and Kv1.3 potassium channels from the venom of the scorpion Centruroides suffusus suffusus. Biochem Pharmacol 2008; 76:1142-54. [DOI: 10.1016/j.bcp.2008.08.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Revised: 07/29/2008] [Accepted: 08/04/2008] [Indexed: 11/30/2022]
|
33
|
Recombinant Kv channels at the membrane of Escherichia coli bind specifically agitoxin2. J Neuroimmune Pharmacol 2008; 4:83-91. [PMID: 18649142 DOI: 10.1007/s11481-008-9116-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2008] [Accepted: 06/24/2008] [Indexed: 01/08/2023]
Abstract
Potassium voltage-gated channels (Kv) are considered as molecular targets in a number of serious neuronal, immune, and cardiac disorders. Search for efficient low-molecular weight modulators of Kv channel function provides a basis for the development of an appropriate therapy for various Kv-mediated diseases. We report here on a new bacterial cell-based system, which is suitable for study of interactions between ligands and ligand-binding sites of eukaryotic Kv1.3 and Kv1.1 channels. To create this system, high-level expression of KcsA-Kv1.3 and KcsA-Kv1.1 hybrid proteins (ligand-binding sites of Kv1.3 or Kv1.1 fused with prokaryotic KcsA potassium channel) was achieved in the plasma membrane of Escherichia coli. An efficient procedure of E. coli conversion to intact spheroplasts was developed. We demonstrate that fluorescently labeled agitoxin 2 binds specifically to high-affinity and lower-affinity sites of KcsA-Kv1.3 and KcsA-Kv1.1, respectively, at the membrane of spheroplasts. Number of binding sites per cell is estimated to be (1.0 +/- 0.6) x 10(5) and (0.3 +/- 0.2) x 10(5) for KcsA-Kv1.3- and KcsA-Kv1.1-presenting cells, respectively, that allows reliable detection of ligand-receptor interactions by confocal laser scanning microscopy. This bacterial cell-based system is intended for screening of ligands to membrane-embedded pharmaceutical targets.
Collapse
|
34
|
Han S, Yi H, Yin SJ, Chen ZY, Liu H, Cao ZJ, Wu YL, Li WX. Structural Basis of a Potent Peptide Inhibitor Designed for Kv1.3 Channel, a Therapeutic Target of Autoimmune Disease. J Biol Chem 2008; 283:19058-65. [DOI: 10.1074/jbc.m802054200] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
35
|
Jin L, Wu Y. Molecular mechanism of the sea anemone toxin ShK recognizing the Kv1.3 channel explored by docking and molecular dynamic simulations. J Chem Inf Model 2007; 47:1967-72. [PMID: 17718553 DOI: 10.1021/ci700178w] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Computational methods are employed to simulate the interaction of the sea anemone toxin ShK in complex with the voltage-gated potassium channel Kv1.3 from mice. All of the available 20 structures of ShK in the Protein Data Bank were considered for improving the performance of the rigid protein docking of ZDOCK. The traditional and novel binding modes were obtained among a large number of predicted complexes by using clustering analysis, screening with expert knowledge, energy minimization, and molecular dynamic simulations. The quality and validity of the resulting complexes were further evaluated to identify a favorable complex structure by 500 ps molecular dynamic simulations and the change of binding free energies with a computational alanine scanning technique. The novel and reasonable ShK-Kv1.3 complex structure was found to be different from the traditional model by using the Lys22 residue to block the channel pore. From the resulting structure of the ShK-Kv1.3 complex, ShK mainly associates the channel outer vestibule with its second helical segment. Structural analysis first revealed that the Lys22 residue side chain of the ShK peptide just hangs between C and D chains of the Kv1.3 channel instead of physically blocking the channel pore. The obvious loss of the ShK Ser20Ala and Tyr23Ala mutant binding ability to the Kv1.3 channel is caused by the conformational change. The five hydrogen bonds between Arg24 in ShK and H404(A) and D402(D) in Kv1.3 make Arg24 the most crucial for its binding to the Kv1.3 channel. Besides the detailed interaction between ShK and Kv1.3 at the atom level, the significant conformational change induced by the interaction between the ShK peptide and the Kv1.3 channel, accompanied by the gradual decrease of binding free energies, strongly implies that the binding of the ShK peptide toward the Kv1.3 channel is a dynamic process of conformational rearrangement and energy stabilization. All of these can accelerate the development of ShK structure-based immunosuppressants.
Collapse
Affiliation(s)
- Ling Jin
- Department of Applied Chemistry, School of Natural Science, Wuhan University of Technology, Wuhan, Hubei 430070, PR China.
| | | |
Collapse
|