1
|
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: 11] [Impact Index Per Article: 2.8] [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
|
2
|
Wulff H, Christophersen P, Colussi P, Chandy KG, Yarov-Yarovoy V. Antibodies and venom peptides: new modalities for ion channels. Nat Rev Drug Discov 2019; 18:339-357. [PMID: 30728472 PMCID: PMC6499689 DOI: 10.1038/s41573-019-0013-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Ion channels play fundamental roles in both excitable and non-excitable tissues and therefore constitute attractive drug targets for myriad neurological, cardiovascular and metabolic diseases as well as for cancer and immunomodulation. However, achieving selectivity for specific ion channel subtypes with small-molecule drugs has been challenging, and there currently is a growing trend to target ion channels with biologics. One approach is to improve the pharmacokinetics of existing or novel venom-derived peptides. In parallel, after initial studies with polyclonal antibodies demonstrated the technical feasibility of inhibiting channel function with antibodies, multiple preclinical programmes are now using the full spectrum of available technologies to generate conventional monoclonal and engineered antibodies or nanobodies against extracellular loops of ion channels. After a summary of the current state of ion channel drug discovery, this Review discusses recent developments using the purinergic receptor channel P2X purinoceptor 7 (P2X7), the voltage-gated potassium channel KV1.3 and the voltage-gated sodium channel NaV1.7 as examples of targeting ion channels with biologics.
Collapse
Affiliation(s)
- Heike Wulff
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | | | | | - K George Chandy
- Molecular Physiology Laboratory, Infection and Immunity Theme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Vladimir Yarov-Yarovoy
- Department of Physiology & Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
3
|
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
|
4
|
Li R, Yu H, Yue Y, Liu S, Xing R, Chen X, Li P. Combined proteomics and transcriptomics identifies sting-related toxins of jellyfish Cyanea nozakii. J Proteomics 2016; 148:57-64. [PMID: 27461980 DOI: 10.1016/j.jprot.2016.07.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/16/2016] [Accepted: 07/22/2016] [Indexed: 01/22/2023]
Abstract
UNLABELLED Jellyfish sting has become a worldwide issue of critical concern to human health and safety in coastal areas in recent decades. Cyanea nozakii is one of the dominant blooming species and dangerous stingers in China. However, it remains unclear how many and what types of toxins are present in the venom. So, we used a combined transcriptomics and proteomics approach to investigate the venom composition of jellyfish C. nozakii. In total 4,608,524 Illumina valid reads were obtained to de novo assemble to 40,434 unigenes in the transcriptomics analysis. And, a total of 311,635 MS/MS spectra with 12,247 unique MS/MS spectra were generated to 1556 homologous proteins in the proteomics analysis. 174 potential toxin proteins were identified, with 27 proteins homology to the toxins from venomous animals, including phospholipase A2, zinc metalloproteinase-disintegrin agkistin, serine protease inhibitor, plancitoxin-1, alpha-latrocrustotoxin-Lt1a, etc. This study described the transcriptomics and venom proteomics of jellyfish C. nozakii for the first time. Our findings provide a comprehensive understanding of the venom composition of C. nozakii. Furthermore, the results may also be very helpful for the discovery of novel bioactive proteins, as well as the development of effective treatments for jellyfish sting in the future. BIOLOGICAL SIGNIFICANCE Jellyfish Cyanea nozakii is one of the most dangerous stingers in the coast of china. Hundreds of thousands of people would be stung every year and victims suffered a severe pain, itch, swelling, inflammation, wheal and even more serious consequence. However, it remains unclear how many and what types of toxins are present as well as the relationship between the clinical symptoms and toxins. Our combined transcriptomics and proteomics findings can provide a comprehensive understanding of the venom composition of C. nozakii and will also be helpful for the development of effective treatments for jellyfish sting in the future.
Collapse
Affiliation(s)
- Rongfeng Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Huahua Yu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Yang Yue
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China; University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100039, China
| | - Song Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Ronge Xing
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Xiaolin Chen
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | - Pengcheng Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China.
| |
Collapse
|
5
|
Harjani JR, Yap BK, Leung EWW, Lucke A, Nicholson SE, Scanlon MJ, Chalmers DK, Thompson PE, Norton RS, Baell JB. Design, Synthesis, and Characterization of Cyclic Peptidomimetics of the Inducible Nitric Oxide Synthase Binding Epitope That Disrupt the Protein–Protein Interaction Involving SPRY Domain-Containing Suppressor of Cytokine Signaling Box Protein (SPSB) 2 and Inducible Nitric Oxide Synthase. J Med Chem 2016; 59:5799-809. [DOI: 10.1021/acs.jmedchem.6b00386] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jitendra R. Harjani
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Beow Keat Yap
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Eleanor W. W. Leung
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Andrew Lucke
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Sandra E. Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- The
Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Martin J. Scanlon
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - David K. Chalmers
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Philip E. Thompson
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Raymond S. Norton
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Jonathan B. Baell
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
6
|
Designer and natural peptide toxin blockers of the KcsA potassium channel identified by phage display. Proc Natl Acad Sci U S A 2015; 112:E7013-21. [PMID: 26627718 DOI: 10.1073/pnas.1514728112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Peptide neurotoxins are powerful tools for research, diagnosis, and treatment of disease. Limiting broader use, most receptors lack an identified toxin that binds with high affinity and specificity. This paper describes isolation of toxins for one such orphan target, KcsA, a potassium channel that has been fundamental to delineating the structural basis for ion channel function. A phage-display strategy is presented whereby ∼1.5 million novel and natural peptides are fabricated on the scaffold present in ShK, a sea anemone type I (SAK1) toxin stabilized by three disulfide bonds. We describe two toxins selected by sorting on purified KcsA, one novel (Hui1, 34 residues) and one natural (HmK, 35 residues). Hui1 is potent, blocking single KcsA channels in planar lipid bilayers half-maximally (Ki) at 1 nM. Hui1 is also specific, inhibiting KcsA-Shaker channels in Xenopus oocytes with a Ki of 0.5 nM whereas Shaker, Kv1.2, and Kv1.3 channels are blocked over 200-fold less well. HmK is potent but promiscuous, blocking KcsA-Shaker, Shaker, Kv1.2, and Kv1.3 channels with Ki of 1-4 nM. As anticipated, one Hui1 blocks the KcsA pore and two conserved toxin residues, Lys21 and Tyr22, are essential for high-affinity binding. Unexpectedly, potassium ions traversing the channel from the inside confer voltage sensitivity to the Hui1 off-rate via Arg23, indicating that Lys21 is not in the pore. The 3D structure of Hui1 reveals a SAK1 fold, rationalizes KcsA inhibition, and validates the scaffold-based approach for isolation of high-affinity toxins for orphan receptors.
Collapse
|
7
|
Bioactive Mimetics of Conotoxins and other Venom Peptides. Toxins (Basel) 2015; 7:4175-98. [PMID: 26501323 PMCID: PMC4626728 DOI: 10.3390/toxins7104175] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/08/2015] [Indexed: 11/17/2022] Open
Abstract
Ziconotide (Prialt®), a synthetic version of the peptide ω-conotoxin MVIIA found in the venom of a fish-hunting marine cone snail Conus magnus, is one of very few drugs effective in the treatment of intractable chronic pain. However, its intrathecal mode of delivery and narrow therapeutic window cause complications for patients. This review will summarize progress in the development of small molecule, non-peptidic mimics of Conotoxins and a small number of other venom peptides. This will include a description of how some of the initially designed mimics have been modified to improve their drug-like properties.
Collapse
|
8
|
De novo design and synthesis of a μ-conotoxin KIIIA peptidomimetic. Bioorg Med Chem Lett 2013; 23:4892-5. [DOI: 10.1016/j.bmcl.2013.06.086] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/14/2013] [Accepted: 06/27/2013] [Indexed: 11/20/2022]
|
9
|
Abstract
Venomous animals use a highly complex cocktails of proteins, peptides and small molecules to subdue and kill their prey. As such, venoms represent highly valuable combinatorial peptide libraries, displaying an extensive range of pharmacological activities, honed by natural selection. Modern analytical technologies enable us to take full advantage of this vast pharmacological cornucopia in the hunt for novel drug leads. Spider venoms represent a resource of several million peptides, which selectively target specific subtypes of ion channels. Structure-function studies of spider toxins are leading not only to the discovery of novel molecules, but also to novel therapeutic routes for cardiovascular diseases, cancer, neuromuscular diseases, pain and to a variety of other pathological conditions. This review presents an overview of spider peptide toxins as candidates for therapeutics and focuses on their applications in the discovery of novel mechanisms of analgesia.
Collapse
Affiliation(s)
- Pierre Escoubas
- University of Nice - Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC) - CNRS UMR6097, 660 Route des Lucioles, 06560 Valbonne, France +33 04 93 95 77 35 ; +33 04 93 95 77 08 ;
| | | |
Collapse
|
10
|
Saez NJ, Senff S, Jensen JE, Er SY, Herzig V, Rash LD, King GF. Spider-venom peptides as therapeutics. Toxins (Basel) 2010; 2:2851-71. [PMID: 22069579 PMCID: PMC3153181 DOI: 10.3390/toxins2122851] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 12/17/2010] [Accepted: 12/17/2010] [Indexed: 01/01/2023] Open
Abstract
Spiders are the most successful venomous animals and the most abundant terrestrial predators. Their remarkable success is due in large part to their ingenious exploitation of silk and the evolution of pharmacologically complex venoms that ensure rapid subjugation of prey. Most spider venoms are dominated by disulfide-rich peptides that typically have high affinity and specificity for particular subtypes of ion channels and receptors. Spider venoms are conservatively predicted to contain more than 10 million bioactive peptides, making them a valuable resource for drug discovery. Here we review the structure and pharmacology of spider-venom peptides that are being used as leads for the development of therapeutics against a wide range of pathophysiological conditions including cardiovascular disorders, chronic pain, inflammation, and erectile dysfunction.
Collapse
Affiliation(s)
- Natalie J Saez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, 4072, Australia.
| | | | | | | | | | | | | |
Collapse
|
11
|
Caulfield MJ, Dudkin VY, Ottinger EA, Getty KL, Zuck PD, Kaufhold RM, Hepler RW, McGaughey GB, Citron M, Hrin RC, Wang YJ, Miller MD, Joyce JG. Small molecule mimetics of an HIV-1 gp41 fusion intermediate as vaccine leads. J Biol Chem 2010; 285:40604-11. [PMID: 20943652 DOI: 10.1074/jbc.m110.172197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We describe here a novel platform technology for the discovery of small molecule mimetics of conformational epitopes on protein antigens. As a model system, we selected mimetics of a conserved hydrophobic pocket within the N-heptad repeat region of the HIV-1 envelope protein, gp41. The human monoclonal antibody, D5, binds to this target and exhibits broadly neutralizing activity against HIV-1. We exploited the antigen-binding property of D5 to select complementary small molecules using a high throughput screen of a diverse chemical collection. The resulting small molecule leads were rendered immunogenic by linking them to a carrier protein and were shown to elicit N-heptad repeat-binding antibodies in a fraction of immunized mice. Plasma from HIV-1-infected subjects shown previously to contain broadly neutralizing antibodies was found to contain antibodies capable of binding to haptens represented in the benzylpiperidine leads identified as a result of the high throughput screen, further validating these molecules as vaccine leads. Our results suggest a new paradigm for vaccine discovery using a medicinal chemistry approach to identify lead molecules that, when optimized, could become vaccine candidates for infectious diseases that have been refractory to conventional vaccine development.
Collapse
Affiliation(s)
- Michael J Caulfield
- Department of Vaccine Basic Research, Merck Research Laboratories, West Point, Pennsylvania 19486, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Norton RS. Mu-conotoxins as leads in the development of new analgesics. Molecules 2010; 15:2825-44. [PMID: 20428082 PMCID: PMC6257286 DOI: 10.3390/molecules15042825] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/06/2010] [Accepted: 04/12/2010] [Indexed: 02/02/2023] Open
Abstract
Voltage-gated sodium channels (VGSCs) contain a specific binding site for a family of cone shell toxins known as mu-conotoxins. As some VGSCs are involved in pain perception and mu-conotoxins are able to block these channels, mu-conotoxins show considerable potential as analgesics. Recent studies have advanced our understanding of the three-dimensional structures and structure-function relationships of the mu-conotoxins, including their interaction with VGSCs. Truncated peptide analogues of the native toxins have been created in which secondary structure elements are stabilized by non-native linkers such as lactam bridges. Ultimately, it would be desirable to capture the favourable analgesic properties of the native toxins, in particular their potency and channel sub-type selectivity, in non-peptide mimetics. Such mimetics would constitute lead compounds in the development of new therapeutics for the treatment of pain.
Collapse
Affiliation(s)
- Raymond S Norton
- Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.
| |
Collapse
|
13
|
Lessene G, Smith BJ, Gable RW, Baell JB. Characterization of the Two Fundamental Conformations of Benzoylureas and Elucidation of the Factors That Facilitate Their Conformational Interchange. J Org Chem 2009; 74:6511-25. [DOI: 10.1021/jo900871a] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Guillaume Lessene
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | - Brian J. Smith
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | - Robert W. Gable
- School of Chemistry, The University of Melbourne, Victoria 3010, Australia
| | - Jonathan B. Baell
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
14
|
Wulff H, Zhorov BS. K+ channel modulators for the treatment of neurological disorders and autoimmune diseases. Chem Rev 2008; 108:1744-73. [PMID: 18476673 PMCID: PMC2714671 DOI: 10.1021/cr078234p] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA.
| | | |
Collapse
|
15
|
Green BR, Catlin P, Zhang MM, Fiedler B, Bayudan W, Morrison A, Norton RS, Smith BJ, Yoshikami D, Olivera BM, Bulaj G. Conotoxins containing nonnatural backbone spacers: cladistic-based design, chemical synthesis, and improved analgesic activity. ACTA ACUST UNITED AC 2007; 14:399-407. [PMID: 17462575 DOI: 10.1016/j.chembiol.2007.02.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 02/09/2007] [Accepted: 02/20/2007] [Indexed: 11/23/2022]
Abstract
Disulfide-rich neurotoxins from venomous animals continue to provide compounds with therapeutic potential. Minimizing neurotoxins often results in removal of disulfide bridges or critical amino acids. To address this drug-design challenge, we explored the concept of disulfide-rich scaffolds consisting of isostere polymers and peptidic pharmacophores. Flexible spacers, such as amino-3-oxapentanoic or 6-aminohexanoic acids, were used to replace conformationally constrained parts of a three-disulfide-bridged conotoxin, SIIIA. The peptide-polymer hybrids, polytides, were designed based on cladistic identification of nonconserved loci in related peptides. After oxidative folding, the polytides appeared to be better inhibitors of sodium currents in dorsal root ganglia and sciatic nerves in mice. Moreover, the polytides appeared to be significantly more potent and longer-lasting analgesics in the inflammatory pain model in mice, when compared to SIIIA. The resulting polytides provide a promising strategy for transforming disulfide-rich peptides into therapeutics.
Collapse
Affiliation(s)
- Brad R Green
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
King GF. Modulation of insect Cav channels by peptidic spider toxins. Toxicon 2007; 49:513-30. [PMID: 17197008 DOI: 10.1016/j.toxicon.2006.11.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Accepted: 11/17/2006] [Indexed: 10/23/2022]
Abstract
Insects have a much smaller repertoire of voltage-gated calcium (Ca(V)) channels than vertebrates. Drosophila melanogaster harbors only a single ortholog of each of the vertebrate Ca(V)1, Ca(V)2, and Ca(V)3 subtypes, although its basal inventory is expanded by alternative splicing and editing of Ca(V) channel transcripts. Nevertheless, there appears to be little functional plasticity within this limited panel of insect Ca(V) channels, since severe loss-of-function mutations in genes encoding the pore-forming alpha1 subunits in Drosophila are embryonic lethal. Since the primary role of spider venom is to paralyze or kill insect prey, it is not surprising that most, if not all, spider venoms contain peptides that potently modify the activity of these functionally critical insect Ca(V) channels. Unfortunately, it has proven difficult to determine the precise ion channel subtypes recognized by these peptide toxins since insect Ca(V) channels have significantly different pharmacology to their vertebrate counterparts, and cloned insect Ca(V) channels are not available for electrophysiological studies. However, biochemical and genetic studies indicate that some of these spider toxins might ultimately become the defining pharmacology for certain subtypes of insect Ca(V) channels. This review focuses on peptidic spider toxins that specifically target insect Ca(V) channels. In addition to providing novel molecular tools for ion channel characterization, some of these toxins are being used as leads to develop new methods for controlling insect pests.
Collapse
Affiliation(s)
- Glenn F King
- Division of Chemical and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane Qld. 4072, Australia.
| |
Collapse
|
17
|
Baell JB, Duggan PJ, Forsyth SA, Lewis RJ, Lok YP, Schroeder CI, Shepherd NE. Synthesis and biological evaluation of anthranilamide-based non-peptide mimetics of ω-conotoxin GVIA. Tetrahedron 2006. [DOI: 10.1016/j.tet.2006.05.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
18
|
Harvey AJ, Baell JB, Toovey N, Homerick D, Wulff H. A new class of blockers of the voltage-gated potassium channel Kv1.3 via modification of the 4- or 7-position of khellinone. J Med Chem 2006; 49:1433-41. [PMID: 16480279 DOI: 10.1021/jm050839v] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The voltage-gated potassium channel Kv1.3 constitutes an attractive target for the selective suppression of effector memory T cells in autoimmune diseases. We have previously reported the natural product khellinone, 1a, as a versatile lead molecule and identified two new classes of Kv1.3 blockers: (i) chalcone derivatives of khellinone, and (ii) khellinone dimers linked through the 6-position. Here we describe the multiple parallel synthesis of a new class of khellinone derivatives selectively alkylated at either the 4- or 7-position via the phenolic OH and show that several chloro, bromo, methoxy, and nitro substituted benzyl derivatives inhibit Kv1.3 with submicromolar potencies. Representative examples of the most potent compounds from each subclass, 11m (5-acetyl-4-(4'-chloro)benzyloxy-6-hydroxy-7-methoxybenzofuran) and 14m (5-acetyl-7-(4'-bromo)benzyloxy-6-hydroxy-4-methoxybenzofuran), block Kv1.3 with EC50 values of 480 and 400 nM, respectively. Both compounds exhibit moderate selectivity over other Kv1-family channels and HERG, are not cytotoxic, and suppress human T cell proliferation at low micromolar concentrations.
Collapse
Affiliation(s)
- Andrew J Harvey
- The Walter and Eliza Hall Institute of Medical Research Biotechnology Centre, 4 Research Avenue, La Trobe R&D Park, Bundoora 3086, Australia.
| | | | | | | | | |
Collapse
|