1
|
Gubič Š, Montalbano A, Sala C, Becchetti A, Hendrickx LA, Van Theemsche KM, Pinheiro-Junior EL, Altadonna GC, Peigneur S, Ilaš J, Labro AJ, Pardo LA, Tytgat J, Tomašič T, Arcangeli A, Peterlin Mašič L. Immunosuppressive effects of new thiophene-based K V1.3 inhibitors. Eur J Med Chem 2023; 259:115561. [PMID: 37454520 DOI: 10.1016/j.ejmech.2023.115561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023]
Abstract
Voltage-gated potassium channel KV1.3 inhibitors have been shown to be effective in preventing T-cell proliferation and activation by affecting intracellular Ca2+ homeostasis. Here, we present the structure-activity relationship, KV1.3 inhibition, and immunosuppressive effects of new thiophene-based KV1.3 inhibitors with nanomolar potency on K+ current in T-lymphocytes and KV1.3 inhibition on Ltk- cells. The new KV1.3 inhibitor trans-18 inhibited KV1.3 -mediated current in phytohemagglutinin (PHA)-activated T-lymphocytes with an IC50 value of 26.1 nM and in mammalian Ltk- cells with an IC50 value of 230 nM. The KV1.3 inhibitor trans-18 also had nanomolar potency against KV1.3 in Xenopus laevis oocytes (IC50 = 136 nM). The novel thiophene-based KV1.3 inhibitors impaired intracellular Ca2+ signaling as well as T-cell activation, proliferation, and colony formation.
Collapse
Affiliation(s)
- Špela Gubič
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Alberto Montalbano
- University of Florence, Department of Experimental and Clinical Medicine, I-50134, Florence, Italy
| | - Cesare Sala
- University of Florence, Department of Experimental and Clinical Medicine, I-50134, Florence, Italy
| | - Andrea Becchetti
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, I-20126, Milano, Italy
| | - Louise Antonia Hendrickx
- University of Leuven, Toxicology and Pharmacology, Campus Gasthuisberg, Onderwijs en Navorsing 2, Herestraat 49, PO Box 922, 3000, Leuven, Belgium
| | - Kenny M Van Theemsche
- University of Antwerp, Department of Biomedical Sciences, Campus Drie Eiken, Universiteisplein 1, 2610, Wilrijk, Belgium; Ghent University, Department of Basic and Applied Medical Sciences, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Ernesto Lopes Pinheiro-Junior
- University of Leuven, Toxicology and Pharmacology, Campus Gasthuisberg, Onderwijs en Navorsing 2, Herestraat 49, PO Box 922, 3000, Leuven, Belgium
| | | | - Steve Peigneur
- University of Leuven, Toxicology and Pharmacology, Campus Gasthuisberg, Onderwijs en Navorsing 2, Herestraat 49, PO Box 922, 3000, Leuven, Belgium
| | - Janez Ilaš
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Alain J Labro
- Ghent University, Department of Basic and Applied Medical Sciences, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Luis A Pardo
- Max-Planck Institute for Experimental Medicine, AG Oncophysiology, Hermann-Rein-Str. 3, 37075, Göttingen, Germany
| | - Jan Tytgat
- University of Leuven, Toxicology and Pharmacology, Campus Gasthuisberg, Onderwijs en Navorsing 2, Herestraat 49, PO Box 922, 3000, Leuven, Belgium
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia
| | - Annarosa Arcangeli
- University of Florence, Department of Experimental and Clinical Medicine, I-50134, Florence, Italy.
| | - Lucija Peterlin Mašič
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Aškerčeva cesta 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Pharmacological Screening of Venoms from Five Brazilian Micrurus Species on Different Ion Channels. Int J Mol Sci 2022; 23:ijms23147714. [PMID: 35887062 PMCID: PMC9318628 DOI: 10.3390/ijms23147714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 12/05/2022] Open
Abstract
Coral snake venoms from the Micrurus genus are a natural library of components with multiple targets, yet are poorly explored. In Brazil, 34 Micrurus species are currently described, and just a few have been investigated for their venom activities. Micrurus venoms are composed mainly of phospholipases A2 and three-finger toxins, which are responsible for neuromuscular blockade—the main envenomation outcome in humans. Beyond these two major toxin families, minor components are also important for the global venom activity, including Kunitz-peptides, serine proteases, 5′ nucleotidases, among others. In the present study, we used the two-microelectrode voltage clamp technique to explore the crude venom activities of five different Micrurus species from the south and southeast of Brazil: M. altirostris, M. corallinus, M. frontalis, M. carvalhoi and M. decoratus. All five venoms induced full inhibition of the muscle-type α1β1δε nAChR with different levels of reversibility. We found M. altirostris and M. frontalis venoms acting as partial inhibitors of the neuronal-type α7 nAChR with an interesting subsequent potentiation after one washout. We discovered that M. altirostris and M. corallinus venoms modulate the α1β2 GABAAR. Interestingly, the screening on KV1.3 showed that all five Micrurus venoms act as inhibitors, being totally reversible after the washout. Since this activity seems to be conserved among different species, we hypothesized that the Micrurus venoms may rely on potassium channel inhibitory activity as an important feature of their envenomation strategy. Finally, tests on NaV1.2 and NaV1.4 showed that these channels do not seem to be targeted by Micrurus venoms. In summary, the venoms tested are multifunctional, each of them acting on at least two different types of targets.
Collapse
|
3
|
Design of New Potent and Selective Thiophene-Based K V1.3 Inhibitors and Their Potential for Anticancer Activity. Cancers (Basel) 2022; 14:cancers14112595. [PMID: 35681571 PMCID: PMC9179341 DOI: 10.3390/cancers14112595] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary In this article, we describe the discovery of a new class of potent and selective thiophene-based inhibitors of the voltage-gated potassium channel KV1.3 and their potential to induce apoptosis and inhibit proliferation. The KV1.3 channel has only recently emerged as a molecular target in cancer therapy. The most potent KV1.3 inhibitor 44 had an IC50 KV1.3 value of 470 nM (oocytes) and 950 nM (Ltk− cells) and appropriate selectivity for other KV channels. New KV1.3 inhibitors significantly inhibited proliferation of Panc-1 cells and KV1.3 inhibitor 4 induced significant apoptosis in tumor spheroids of Colo-357 cells. Abstract The voltage-gated potassium channel KV1.3 has been recognized as a tumor marker and represents a promising new target for the discovery of new anticancer drugs. We designed a novel structural class of KV1.3 inhibitors through structural optimization of benzamide-based hit compounds and structure-activity relationship studies. The potency and selectivity of the new KV1.3 inhibitors were investigated using whole-cell patch- and voltage-clamp experiments. 2D and 3D cell models were used to determine antiproliferative activity. Structural optimization resulted in the most potent and selective KV1.3 inhibitor 44 in the series with an IC50 value of 470 nM in oocytes and 950 nM in Ltk− cells. KV1.3 inhibitor 4 induced significant apoptosis in Colo-357 spheroids, while 14, 37, 43, and 44 significantly inhibited Panc-1 proliferation.
Collapse
|
4
|
Chokshi R, Bennett O, Zhelay T, Kozak JA. NSAIDs Naproxen, Ibuprofen, Salicylate, and Aspirin Inhibit TRPM7 Channels by Cytosolic Acidification. Front Physiol 2021; 12:727549. [PMID: 34733174 PMCID: PMC8558630 DOI: 10.3389/fphys.2021.727549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/10/2021] [Indexed: 01/23/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are used for relieving pain and inflammation accompanying numerous disease states. The primary therapeutic mechanism of these widely used drugs is the inhibition of cyclooxygenase 1 and 2 (COX1, 2) enzymes that catalyze the conversion of arachidonic acid into prostaglandins. At higher doses, NSAIDs are used for prevention of certain types of cancer and as experimental treatments for Alzheimer’s disease. In the immune system, various NSAIDs have been reported to influence neutrophil function and lymphocyte proliferation, and affect ion channels and cellular calcium homeostasis. Transient receptor potential melastatin 7 (TRPM7) cation channels are highly expressed in T lymphocytes and are inhibited by Mg2+, acidic pH, and polyamines. Here, we report a novel effect of naproxen, ibuprofen, salicylate, and acetylsalicylate on TRPM7. At concentrations of 3–30mM, they reversibly inhibited TRPM7 channel currents. By measuring intracellular pH with the ratiometric indicator BCECF, we found that at 300μM to 30mM, these NSAIDs reversibly acidified the cytoplasm in a concentration-dependent manner, and propose that TRPM7 channel inhibition is a consequence of cytosolic acidification, rather than direct. NSAID inhibition of TRPM7 channels was slow, voltage-independent, and displayed use-dependence, increasing in potency upon repeated drug applications. The extent of channel inhibition by salicylate strongly depended on cellular PI(4,5)P2 levels, as revealed when this phospholipid was depleted with voltage-sensitive lipid phosphatase (VSP). Salicylate inhibited heterologously expressed wildtype TRPM7 channels but not the S1107R variant, which is insensitive to cytosolic pH, Mg2+, and PI(4,5)P2 depletion. NSAID-induced acidification was also observed in Schneider 2 cells from Drosophila, an organism that lacks orthologous COX genes, suggesting that this effect is unrelated to COX enzyme activity. A 24-h exposure to 300μM–10mM naproxen resulted in a concentration-dependent reduction in cell viability. In addition to TRPM7, the described NSAID effect would be expected to apply to other ion channels and transporters sensitive to intracellular pH.
Collapse
Affiliation(s)
- Rikki Chokshi
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| | - Orville Bennett
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| | - Tetyana Zhelay
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| | - J Ashot Kozak
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, College of Science and Mathematics, Wright State University, Dayton, OH, United States
| |
Collapse
|
5
|
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: 3.7] [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
|
6
|
Ryan RYM, Seymour J, Loukas A, Lopez JA, Ikonomopoulou MP, Miles JJ. Immunological Responses to Envenomation. Front Immunol 2021; 12:661082. [PMID: 34040609 PMCID: PMC8141633 DOI: 10.3389/fimmu.2021.661082] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/01/2021] [Indexed: 01/05/2023] Open
Abstract
Venoms are complex mixtures of toxic compounds delivered by bite or sting. In humans, the consequences of envenomation range from self-limiting to lethal. Critical host defence against envenomation comprises innate and adaptive immune strategies targeted towards venom detection, neutralisation, detoxification, and symptom resolution. In some instances, venoms mediate immune dysregulation that contributes to symptom severity. This review details the involvement of immune cell subtypes and mediators, particularly of the dermis, in host resistance and venom-induced immunopathology. We further discuss established venom-associated immunopathology, including allergy and systemic inflammation, and investigate Irukandji syndrome as a potential systemic inflammatory response. Finally, this review characterises venom-derived compounds as a source of immune modulating drugs for treatment of disease.
Collapse
Affiliation(s)
- Rachael Y. M. Ryan
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
- School of Environment and Sciences, Griffith University, Nathan, QLD, Australia
| | - Jamie Seymour
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Alex Loukas
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - J. Alejandro Lopez
- School of Environment and Sciences, Griffith University, Nathan, QLD, Australia
- QIMR Berghofer Medical Research Institute, The University of Queensland, Herston, QLD, Australia
| | - Maria P. Ikonomopoulou
- Translational Venomics Group, Madrid Institute for Advanced Studies (IMDEA) in Food, CEI UAM+CSIC, Madrid, Spain
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - John J. Miles
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
7
|
Dietrich M, Hartung HP, Albrecht P. Neuroprotective Properties of 4-Aminopyridine. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/3/e976. [PMID: 33653963 PMCID: PMC7931640 DOI: 10.1212/nxi.0000000000000976] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023]
Abstract
As an antagonist of voltage-gated potassium (Kv) channels, 4-aminopyridine (4-AP) is used as symptomatic therapy in several neurologic disorders. The improvement of visual function and motor skills and relieve of fatigue in patients with MS have been attributed to 4-AP. Its prolonged release formulation (fampridine) has been approved for the symptomatic treatment of walking disability in MS. The beneficial effects were explained by the blockade of axonal Kv channels, thereby enhancing conduction along demyelinated axons. However, an increasing body of evidence suggests that 4-AP may have additional properties beyond the symptomatic mode of action. In this review, we summarize preclinical and clinical data on possible neuroprotective features of 4-AP.
Collapse
Affiliation(s)
- Michael Dietrich
- From the Department of Neurology (M.D., H.-P.H., P.A.), Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; and Brain and Mind Center (H.-P.H.), University of Sydney, Australia
| | - Hans-Peter Hartung
- From the Department of Neurology (M.D., H.-P.H., P.A.), Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; and Brain and Mind Center (H.-P.H.), University of Sydney, Australia
| | - Philipp Albrecht
- From the Department of Neurology (M.D., H.-P.H., P.A.), Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; and Brain and Mind Center (H.-P.H.), University of Sydney, Australia.
| |
Collapse
|
8
|
Hartung HP, Dietrich M, Albrecht P. 4-Aminopyridine is not just a symptomatic therapy, it has a neuroprotective effect – Commentary. Mult Scler 2020; 26:1312-1314. [DOI: 10.1177/1352458520926458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
9
|
Heravi MM, Momeni T, Zadsirjan V, Mohammadi L. Application of The Dess-Martin Oxidation in Total Synthesis of Natural Products. Curr Org Synth 2020; 18:125-196. [PMID: 32940184 DOI: 10.2174/1570179417666200917102634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 11/22/2022]
Abstract
Dess-Martin periodinane (DMP), a commercially available chemical, is frequently utilized as a mild oxidative agent for the selective oxidation of primary and secondary alcohols to their corresponding aldehydes and ketones, respectively. DMP shows several merits over other common oxidative agents such as chromiumand DMSO-based oxidants; thus, it is habitually employed in the total synthesis of natural products. In this review, we try to underscore the applications of DMP as an effective oxidant in an appropriate step (steps) in the multi-step total synthesis of natural products.
Collapse
Affiliation(s)
- Majid M Heravi
- Department of Chemistry, School of Science, Alzahra University, POBox 1993891176, Vanak, Tehran, Iran
| | - Tayebe Momeni
- Department of Chemistry, School of Science, Alzahra University, POBox 1993891176, Vanak, Tehran, Iran
| | - Vahideh Zadsirjan
- Department of Chemistry, School of Science, Alzahra University, POBox 1993891176, Vanak, Tehran, Iran
| | - Leila Mohammadi
- Department of Chemistry, School of Science, Alzahra University, POBox 1993891176, Vanak, Tehran, Iran
| |
Collapse
|
10
|
Dietrich M, Koska V, Hecker C, Göttle P, Hilla AM, Heskamp A, Lepka K, Issberner A, Hallenberger A, Baksmeier C, Steckel J, Balk L, Knier B, Korn T, Havla J, Martínez-Lapiscina EH, Solà-Valls N, Manogaran P, Olbert ED, Schippling S, Cruz-Herranz A, Yiu H, Button J, Caldito NG, von Gall C, Mausberg AK, Stettner M, Zimmermann HG, Paul F, Brandt AU, Küry P, Goebels N, Aktas O, Berndt C, Saidha S, Green AJ, Calabresi PA, Fischer D, Hartung HP, Albrecht P. Protective effects of 4-aminopyridine in experimental optic neuritis and multiple sclerosis. Brain 2020; 143:1127-1142. [PMID: 32293668 DOI: 10.1093/brain/awaa062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/08/2019] [Accepted: 01/20/2020] [Indexed: 12/30/2022] Open
Abstract
Chronic disability in multiple sclerosis is linked to neuroaxonal degeneration. 4-aminopyridine (4-AP) is used and licensed as a symptomatic treatment to ameliorate ambulatory disability in multiple sclerosis. The presumed mode of action is via blockade of axonal voltage gated potassium channels, thereby enhancing conduction in demyelinated axons. In this study, we provide evidence that in addition to those symptomatic effects, 4-AP can prevent neuroaxonal loss in the CNS. Using in vivo optical coherence tomography imaging, visual function testing and histologic assessment, we observed a reduction in retinal neurodegeneration with 4-AP in models of experimental optic neuritis and optic nerve crush. These effects were not related to an anti-inflammatory mode of action or a direct impact on retinal ganglion cells. Rather, histology and in vitro experiments indicated 4-AP stabilization of myelin and oligodendrocyte precursor cells associated with increased nuclear translocation of the nuclear factor of activated T cells. In experimental optic neuritis, 4-AP potentiated the effects of immunomodulatory treatment with fingolimod. As extended release 4-AP is already licensed for symptomatic multiple sclerosis treatment, we performed a retrospective, multicentre optical coherence tomography study to longitudinally compare retinal neurodegeneration between 52 patients on continuous 4-AP therapy and 51 matched controls. In line with the experimental data, during concurrent 4-AP therapy, degeneration of the macular retinal nerve fibre layer was reduced over 2 years. These results indicate disease-modifying effects of 4-AP beyond symptomatic therapy and provide support for the design of a prospective clinical study using visual function and retinal structure as outcome parameters.
Collapse
Affiliation(s)
- Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Valeria Koska
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexander M Hilla
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Annemarie Heskamp
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Klaudia Lepka
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Angelika Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Christine Baksmeier
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Steckel
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Lisanne Balk
- Department of Neurology, Amsterdam Neuroscience, MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Benjamin Knier
- Department of Experimental Neuroimmunology, Technische Universität München, Munich, Germany
| | - Thomas Korn
- Department of Experimental Neuroimmunology, Technische Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians University, Munich, Germany.,Data Integration for Future Medicine consortium (DIFUTURE), Ludwig-Maximilians University, Munich, Germany
| | - Elena H Martínez-Lapiscina
- Service of Neurology, Hospital Clinic, University of Barcelona, Spain Neuroimmunology Program, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Solà-Valls
- Service of Neurology, Hospital Clinic, University of Barcelona, Spain Neuroimmunology Program, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Praveena Manogaran
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland.,Department of Information Technology and Electrical Engineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Elisabeth D Olbert
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Sven Schippling
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Andrés Cruz-Herranz
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Hao Yiu
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Julia Button
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | | | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Anne K Mausberg
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Mark Stettner
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Hannah G Zimmermann
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Alexander U Brandt
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany.,Department of Neurology, University of California, Irvine, USA
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Norbert Goebels
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Shiv Saidha
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | - Ari J Green
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA.,Department of Ophthalmology, University of California San Francisco, San Francisco, USA
| | - Peter A Calabresi
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
11
|
Strijbis EM, Nij Bijvank JA, Killestein J. 4-aminopyridine is not just a symptomatic therapy, it has a neuroprotective effect - Yes. Mult Scler 2020; 26:1309-1310. [PMID: 32628062 PMCID: PMC7543014 DOI: 10.1177/1352458520923951] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Eva Mm Strijbis
- Department of Neurology, MS Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Jenny A Nij Bijvank
- Department of Neurology, MS Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands/Department of Ophthalmology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Joep Killestein
- Department of Neurology, MS Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Leussink VI, Montalban X, Hartung HP. Restoring Axonal Function with 4-Aminopyridine: Clinical Efficacy in Multiple Sclerosis and Beyond. CNS Drugs 2018; 32:637-651. [PMID: 29992409 DOI: 10.1007/s40263-018-0536-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The oral potassium channel blocker 4-aminopyridine has been used in various neurological conditions for decades. Numerous case reports and studies have supported its clinical efficacy in ameliorating the clinical presentation of certain neurological disorders. However, its short half-life, erratic drug levels, and safety-related dose restrictions limited its use as a self-compounded drug in clinical practice. This changed with the introduction of a prolonged-release formulation, which was successfully tested in patients with multiple sclerosis. It was fully approved by the US FDA in January 2010 but initially received only conditional approval from the European Medicines Agency (EMA) in July 2011. After additional clinical studies, this conditional approval was changed to unrestricted approval in August 2017. This article reviews and discusses these recent studies and places aminopyridines and their clinical utility into the context of a broader spectrum of neurological disorders, where clinical efficacy has been suggested. In 2010, prolonged-release 4-aminopyridine became the first drug specifically licensed to improve walking in patients with multiple sclerosis. About one-third of patients across disease courses benefit from this treatment. In addition, various reports indicate clinical efficacy beyond multiple sclerosis, which may broaden its use in clinical practice.
Collapse
Affiliation(s)
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Hospital Universitario Vall d'Hebron, Barcelona, Spain.,St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
13
|
Pereira G, Szwarc B, Mondragão MA, Lima PA, Pereira F. A Ligand-Based Approach to the Discovery of Lead-Like Potassium Channel KV
1.3 Inhibitors. ChemistrySelect 2018. [DOI: 10.1002/slct.201702977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Gilberto Pereira
- LAQV and REQUIMTE; Departamento de Química; Faculdade de Ciências e Tecnologia; Universidade Nova de Lisboa; 2829-516 Caparica Portugal
- NOVA Medical School; Laboratório de Fisiologia; Faculdade de Ciências Médicas; Universidade Nova de Lisboa; Campo dos Mártires da Pátria, 130 1169-056 Lisboa PORTUGAL
| | - Beatriz Szwarc
- Sea4Us - Biotecnologia e Recursos Marinhos, Lda; Rua do Poente S/N 8650-378 Sagres Portugal
- NOVA Medical School; Laboratório de Fisiologia; Faculdade de Ciências Médicas; Universidade Nova de Lisboa; Campo dos Mártires da Pátria, 130 1169-056 Lisboa PORTUGAL
| | - Miguel A. Mondragão
- Sea4Us - Biotecnologia e Recursos Marinhos, Lda; Rua do Poente S/N 8650-378 Sagres Portugal
- NOVA Medical School; Laboratório de Fisiologia; Faculdade de Ciências Médicas; Universidade Nova de Lisboa; Campo dos Mártires da Pátria, 130 1169-056 Lisboa PORTUGAL
| | - Pedro A. Lima
- Sea4Us - Biotecnologia e Recursos Marinhos, Lda; Rua do Poente S/N 8650-378 Sagres Portugal
- NOVA Medical School; Laboratório de Fisiologia; Faculdade de Ciências Médicas; Universidade Nova de Lisboa; Campo dos Mártires da Pátria, 130 1169-056 Lisboa PORTUGAL
| | - Florbela Pereira
- LAQV and REQUIMTE; Departamento de Química; Faculdade de Ciências e Tecnologia; Universidade Nova de Lisboa; 2829-516 Caparica Portugal
| |
Collapse
|
14
|
Valverde P, Kawai T, Taubman MA. Potassium Channel-blockers as Therapeutic Agents to Interfere with Bone Resorption of Periodontal Disease. J Dent Res 2016; 84:488-99. [PMID: 15914584 DOI: 10.1177/154405910508400603] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Inflammatory lesions of periodontal disease contain all the cellular components, including abundant activated/memory T- and B-cells, necessary to control immunological interactive networks and to accelerate bone resorption by RANKL-dependent and -independent mechanisms. Blockade of RANKL function has been shown to ameliorate periodontal bone resorption and other osteopenic disorders without affecting inflammation. Development of therapies aimed at decreasing the expression of RANKL and pro-inflammatory cytokines by T-cells constitutes a promising strategy to ameliorate not only bone resorption, but also inflammation. Several reports have demonstrated that the potassium channels Kv1.3 and IKCa1, through the use of selective blockers, play important roles in T-cell-mediated events, including T-cell proliferation and the production of pro-inflammatory cytokines. More recently, a potassium channel-blocker for Kv1.3 has been shown to down-regulate bone resorption by decreasing the ratio of RANKL-to-OPG expression by memory-activated T-cells. In this article, we first summarize the mechanisms by which chronically activated/memory T-cells, in concert with B-cells and macrophages, trigger inflammatory bone resorption. Then, we describe the main structural and functional characteristics of potassium channels Kv1.3 and IKCa1 in some of the cells implicated in periodontal disease progression. Finally, this review elucidates some recent advances in the use of potassium channel-blockers of Kv1.3 and IKCa1 to ameliorate the clinical signs or side-effects of several immunological disorders and to decrease inflammatory bone resorption in periodontal disease. ABBREVIATIONS: AICD, activation-induced cell death; APC, antigen-presenting cells; B(K), large conductance; CRAC, calcium release-activated calcium channels; DC, dendritic cell; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; IFN-γ, interferon-γ; IP3, inositol (1,4,5)-triphosphate; (K)ir, inward rectifier; JNK, c-Jun N-terminal kinase; I(K), intermediate conductance; LPS, lipopolysaccharide; L, ligand; MCSF, macrophage colony-stimulating factor; MHC, major histocompatibility complex; NFAT, nuclear factor of activated T-cells; RANK, receptor activator of nuclear factor-κB; TCM, central memory T-cells; TEM, effector memory T-cells; TNF, tumor necrosis factor; TRAIL, TNF-related apoptosis-inducing ligand; OPG, osteoprotegerin; Omp29, 29-kDa outer membrane protein; PKC, protein kinase C; PLC, phospholipase C; RT-PCR, reverse-transcriptase polymerase chain-reaction; S(K), small conductance; TCR, T-cell receptor; and (K)v, voltage-gated.
Collapse
Affiliation(s)
- P Valverde
- Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA.
| | | | | |
Collapse
|
15
|
Lenaeus MJ, Burdette D, Wagner T, Focia PJ, Gross A. Structures of KcsA in complex with symmetrical quaternary ammonium compounds reveal a hydrophobic binding site. Biochemistry 2014; 53:5365-73. [PMID: 25093676 PMCID: PMC4139162 DOI: 10.1021/bi500525s] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
Potassium channels allow for the
passive movement of potassium
ions across the cell membrane and are instrumental in controlling
the membrane potential in all cell types. Quaternary ammonium (QA)
compounds block potassium channels and have long been used to study
the functional and structural properties of these channels. Here we
describe the interaction between three symmetrical hydrophobic QAs
and the prokaryotic potassium channel KcsA. The structures demonstrate
the presence of a hydrophobic pocket between the inner helices of
KcsA and provide insight into the binding site and blocking mechanism
of hydrophobic QAs. The structures also reveal a structurally hidden
pathway between the central cavity and the outside membrane environment
reminiscent of the lateral fenestration observed in sodium channels
that can be accessed through small conformational changes in the pore
wall. We propose that the hydrophobic binding pocket stabilizes the
alkyl chains of long-chain QA molecules and may play a key role in
hydrophobic drug binding in general.
Collapse
Affiliation(s)
- Michael J Lenaeus
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School , 303 East Chicago Avenue, Chicago, Illinois 60611, United States
| | | | | | | | | |
Collapse
|
16
|
Siewert B, Csuk R. Membrane damaging activity of a maslinic acid analog. Eur J Med Chem 2014; 74:1-6. [PMID: 24440377 DOI: 10.1016/j.ejmech.2013.12.031] [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: 10/03/2013] [Revised: 11/28/2013] [Accepted: 12/22/2013] [Indexed: 02/06/2023]
Abstract
Close inspection of human ovarian cancer cells A2780 in the course of an antitumor screening using maslinic acid analogs revealed for one of the compounds, 4-oxa-4-phenyl-butyl 2,3-dihydroxy-olean-12-en-28-oate (1), an unusual behavior. During the incubation of the cells with 1, at the perimeter of the cells or close by crystals were formed consisting of cholesterol and excess 1. Compound 1 was incorporated into the cell's membrane followed by an extrusion of cholesterol from the lipid rafts. As a consequence of the alterations of the cell membrane, a volume decrease was initiated that triggered apoptosis; this extends previous models on apoptosis initiating mechanisms.
Collapse
Affiliation(s)
- Bianka Siewert
- Bereich Organische Chemie, Martin-Luther-Universität Halle-Wittenberg, Kurt-Mothes-Straße 2, D-06120 Halle (Saale), Germany
| | - René Csuk
- Bereich Organische Chemie, Martin-Luther-Universität Halle-Wittenberg, Kurt-Mothes-Straße 2, D-06120 Halle (Saale), Germany.
| |
Collapse
|
17
|
Tikhonov DB, Zhorov BS. Homology modeling of Kv1.5 channel block by cationic and electroneutral ligands. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:978-87. [PMID: 24316168 DOI: 10.1016/j.bbamem.2013.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 11/12/2013] [Accepted: 11/26/2013] [Indexed: 01/05/2023]
Abstract
The inner pore of potassium channels is targeted by many ligands of intriguingly different chemical structures. Previous studies revealed common and diverse characteristics of action of ligands including cooperativity of ligand binding, voltage- and use-dependencies, and patterns of ligand-sensing residues. Not all these data are rationalized in published models of ligand-channel complexes. Here we have used energy calculations with experimentally defined constraints to dock flecainide, ICAGEN-4, benzocaine, vernakalant, and AVE0118 into the inner pore of Kv1.5 channel. We arrived at ligand-binding models that suggest possible explanations for different values of the Hill coefficient, different voltage dependencies of ligands action, and effects of mutations of residues in subunit interfaces. Two concepts were crucial to build the models. First, the inner-pore block of a potassium channel requires a cationic "blocking particle". A ligand, which lacks a positively charged group, blocks the channel in a complex with a permeant ion. Second, hydrophobic moieties of a flexible ligand have a tendency to bind in hydrophobic subunit interfaces.
Collapse
Affiliation(s)
- Denis B Tikhonov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.
| | - Boris S Zhorov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
18
|
Lam J, Wulff H. The Lymphocyte Potassium Channels Kv1.3 and KCa3.1 as Targets for Immunosuppression. Drug Dev Res 2011; 72:573-584. [PMID: 22241939 PMCID: PMC3253536 DOI: 10.1002/ddr.20467] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The voltage-gated Kv1.3 and the calcium-activated KCa3.1 potassium channel modulate many calcium-dependent cellular processes in immune cells, including T-cell activation and proliferation, and have therefore been proposed as novel therapeutic targets for immunomodulation. Kv1.3 is highly expressed in CCR7(-) effector memory T cells and is emerging as a target for T-cell mediated diseases like multiple sclerosis, rheumatoid arthritis, type-1 diabetes mellitus, allergic contact dermatitis, and psoriasis. KCa3.1 in contrast is expressed in CCR7(+) naïve and central memory T cells, as well as in mast cells, macrophages, dedifferentiated vascular smooth muscle cells, fibroblasts, vascular endothelium, and airway epithelium. Given this expression pattern, KCa3.1 is a potential therapeutic target for conditions ranging from inflammatory bowel disease, multiple sclerosis, arthritis, and asthma to cardiovascular diseases like atherosclerosis and post-angioplasty restenosis. Results from animal studies have been supportive of the therapeutic potential of both Kv1.3 and KCa3.1 blockers and have also not shown any toxicities associated with pharmacological Kv1.3 and KCa3.1 blockade. To date, two compounds targeting Kv1.3 are in preclinical development but, so far, no Kv1.3 blocker has advanced into clinical trials. KCa3.1 blockers, on the other hand, have been evaluated in clinical trials for sickle cell anemia and exercise-induced asthma, but have so far not shown efficacy. However, the trial results support KCa3.1 as a safe therapeutic target, and will hopefully help enable clinical trials for other medical conditions that might benefit from KCa3.1 blockade.
Collapse
Affiliation(s)
- Jenny Lam
- Department of Pharmacology, University of California Davis, Davis, CA 95616, USA
| | | |
Collapse
|
19
|
Lee JH, Choi SH, Shin TJ, Lee BH, Hwang SH, Kim HC, Nah SY. Effect of dextromethorphan on human Kv1.3 channel activity: Involvement of C-type inactivation. Eur J Pharmacol 2011; 651:122-7. [DOI: 10.1016/j.ejphar.2010.10.091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 10/12/2010] [Accepted: 10/31/2010] [Indexed: 10/18/2022]
|
20
|
Haffner CD, Thomson SA, Guo Y, Petrov K, Larkin A, Banker P, Schaaf G, Dickerson S, Gobel J, Gillie D, Condreay JP, Poole C, Carpenter T, Ulrich J. Substituted N-{3-[(1,1-dioxido-1,2-benzothiazol-3-yl)(phenyl)amino]propyl}benzamide analogs as potent Kv1.3 ion channel blockers. Part 2. Bioorg Med Chem Lett 2010; 20:6989-92. [PMID: 20974533 DOI: 10.1016/j.bmcl.2010.09.131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/23/2010] [Accepted: 09/24/2010] [Indexed: 10/19/2022]
Abstract
We report the synthesis and in vitro activity of a series of novel substituted N-{3-[(1,1-dioxido-1,2-benzothiazol-3-yl)(phenyl)amino]propyl}benzamide analogs. These analogs showed potent inhibitory activity against Kv1.3. Several demonstrated similar potency to the known Kv1.3 inhibitor PAP-1 when tested under the IonWorks patch clamp assay conditions. Two compounds 13i and 13rr were advanced further as potential tool compounds for in vivo validation studies.
Collapse
Affiliation(s)
- Curt D Haffner
- Department of Medicinal Chemistry, GlaxoSmithKline Research and Development, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wulff H. Spiro azepane-oxazolidinones as Kv1.3 potassium channel blockers: WO2010066840. Expert Opin Ther Pat 2010; 20:1759-65. [PMID: 20954790 DOI: 10.1517/13543776.2010.528392] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This article evaluates a patent application from Solvay Pharmaceuticals, which claims spiro azepane-oxazolidinones as novel blockers of the voltage-gated potassium channel Kv1.3 for the treatment of diabetes, psoriasis, obesity, transplant rejection and T-cell mediated autoimmune diseases such as rheumatoid arthritis and MS. The patent describes a new chemotype of Kv1.3 blockers and thus illustrates the growing interest of the pharmaceutical industry in Kv1.3 as a target of immunosuppression and metabolic disorders. This article briefly summarizes the chemistry and biological data provided in the patent and then compares the new compounds to Kv1.3 blockers previously disclosed by both academia and pharmaceutical companies.
Collapse
Affiliation(s)
- Heike Wulff
- University of California, Department of Pharmacology, Davis, Davis, CA 95616, USA.
| |
Collapse
|
22
|
Haffner CD, Thomson SA, Guo Y, Schaller LT, Boggs S, Dickerson S, Gobel J, Gillie D, Condreay JP. N-{3-[(1,1-dioxido-1,2-benzothiazol-3-yl)(phenyl)amino]propyl}benzamide analogs as potent Kv1.3 inhibitors. Part 1. Bioorg Med Chem Lett 2010; 20:6983-8. [PMID: 20971642 DOI: 10.1016/j.bmcl.2010.09.132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/23/2010] [Accepted: 09/24/2010] [Indexed: 10/19/2022]
Abstract
We report the synthesis and in vitro activity of a series of novel N-{3-[(1,1-dioxido-1,2-benzothiazol-3-yl)(phenyl)amino]propyl}benzamide analogs. These analogs showed potent inhibitory activity against Kv1.3. Several compounds, including compound 8b, showed similar potency to the known Kv1.3 inhibitor PAP-1 when tested under the IonWorks patch clamp assay conditions.
Collapse
Affiliation(s)
- Curt D Haffner
- Department of Medicinal Chemistry, GlaxoSmithKline Research and Development, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Castle NA. Pharmacological modulation of voltage-gated potassium channels as a therapeutic strategy. Expert Opin Ther Pat 2010; 20:1471-503. [PMID: 20726689 DOI: 10.1517/13543776.2010.513384] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE OF THE FIELD The human genome encodes at least 40 distinct voltage-gated potassium channel subtypes, which vary in regional expression, pharmacological and biophysical properties. Voltage-dependent potassium (Kv) channels help orchestrate many of the physiological and pathophysiological processes that promote and sometimes hinder the healthy functioning of our bodies. AREAS COVERED IN THIS REVIEW This review summarizes patent and scientific literature reports from the past decade highlighting the opportunities that Kv channels offer for the development of new therapeutic interventions for a wide variety of disorders. WHAT THE READER WILL GAIN The reader will gain an insight from an analysis of the associations of different Kv family members with disease processes, summary and evaluation of the development of therapeutically relevant pharmacological modulators of these channels, particularly focusing on proprietary agents being developed. TAKE HOME MESSAGE Development of new drugs that target Kv channels continue to be of great interest but is proving to be challenging. Nevertheless, opportunities for Kv channel modulators to have an impact on a wide range of disorders in the future remain an exciting prospect.
Collapse
|
24
|
Zimin PI, Garic B, Bodendiek SB, Mahieux C, Wulff H, Zhorov BS. Potassium channel block by a tripartite complex of two cationophilic ligands and a potassium ion. Mol Pharmacol 2010; 78:588-99. [PMID: 20601455 DOI: 10.1124/mol.110.064014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Voltage-gated potassium channels (Kv) are targets for drugs of large chemical diversity. Although hydrophobic cations block Kv channels with Hill coefficients of 1, uncharged electron-rich (cationophilic) molecules often display Hill coefficients of 2. The mechanism of the latter block is unknown. Using a combination of computational and experimental approaches, we mapped the receptor for the immunosuppressant PAP-1 (5-(4-phenoxybutoxy)psoralen), a high-affinity blocker of Kv1.3 channels in lymphocytes. Ligand-docking using Monte Carlo minimizations suggested a model in which two cationophilic PAP-1 molecules coordinate a K(+) ion in the pore with their coumarin moieties, whereas the hydrophobic phenoxyalkoxy side chains extend into the intrasubunit interfaces between helices S5 and S6. We tested the model by generating 58 point mutants involving residues in and around the predicted receptor and then determined their biophysical properties and sensitivity to PAP-1 by whole-cell patch-clamp. The model correctly predicted the key PAP-1-sensing residues in the outer helix, the P-loop, and the inner helix and explained the Hill coefficient of 2 by demonstrating that the Kv1.3 pore can accommodate two or even four PAP-1 molecules. The model further explained the voltage-dependence of block by PAP-1 and its thousand-fold selectivity for Kv1.3 over non-Kv1 channels. The 23- to 125-fold selectivity of PAP-1 for Kv1.3 over other Kv1 channels is probably due to its preferential affinity to the C-type inactivated state, in which cessation of K(+) flux stabilizes the tripartite PAP-1:K(+):PAP-1 complex in the pore. Our study provides a new concept for potassium channel block by cationophilic ligands.
Collapse
Affiliation(s)
- Pavel I Zimin
- Department of Pharmacology, University of California, Davis, California, USA
| | | | | | | | | | | |
Collapse
|
25
|
Husseini L, Leussink VI, Kieseier BC, Hartung HP. [4-Aminopyridine (Fampridine). A new attempt for the symptomatic treatment of multiple sclerosis]. DER NERVENARZT 2010; 81:203-211. [PMID: 20112006 DOI: 10.1007/s00115-009-2902-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Mobility limitation is a frequent clinical symptom of multiple sclerosis (MS) that poses a therapeutic challenge. For years results of animal experiments and clinical experience have indicated that the potassium channel blocker 4-aminopyridine improves axonal excitatory circuits and thus muscular strength in demyelinating diseases. A recently conducted randomized, placebo-controlled, multicenter phase 3 clinical trial in MS patients was able to show that an oral sustained-release formulation of 4-aminopyridine (Fampridine-SR) represents a suitable agent for treatment of walking disability in MS patients.This overview presents the study data and discusses the value of 4-aminopyridine for the symptomatic treatment of MS as a neurofunctional modifier of this disabling disease.
Collapse
Affiliation(s)
- L Husseini
- Neurologische Klinik, Heinrich-Heine-Universität Düsseldorf, Moorenstrasse 5, Düsseldorf, Germany
| | | | | | | |
Collapse
|
26
|
Oguchi T, Watanabe K, Ohkubo K, Abe H, Katoh T. Enantioselective total synthesis of (-)-candelalides A, B and C: potential Kv1.3 blocking immunosuppressive agents. Chemistry 2009; 15:2826-45. [PMID: 19191240 DOI: 10.1002/chem.200802122] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Novel Kv1.3 blocking immunosuppressants, (-)-candelalides A, B and C, were efficiently synthesized for the first time in a convergent and unified manner starting from (+)-5-methyl-Wieland-Miescher ketone. The synthetic method involved the following key steps: i) a strategic [2,3]-Wittig rearrangement of a stannylmethyl ether to install the stereogenic center at C9 and the exo-methylene function at C8 present in the decalin portion; ii) a straightforward coupling of a trans-decalin portion (BC ring) and a gamma-pyrone moiety through the C16-C3' bond to assemble the requisite carbon framework; and iii) a construction of a characteristic di or tetrahydropyran ring (A ring) by internal nucleophilic ring closure of a hydroxy aldehyde or a hydroxy epoxide. The present total synthesis has fully established the absolute configuration of these natural products.
Collapse
Affiliation(s)
- Takamasa Oguchi
- Laboratory of Synthetic Medicinal Chemistry, Department of Chemical Pharmaceutical Science, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan
| | | | | | | | | |
Collapse
|
27
|
Inhibitors of potassium channels KV1.3 and IK-1 as immunosuppressants. Bioorg Med Chem Lett 2009; 19:2299-304. [PMID: 19282171 DOI: 10.1016/j.bmcl.2009.02.077] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 02/19/2009] [Accepted: 02/20/2009] [Indexed: 11/22/2022]
Abstract
New structural classes of K(V)1.3 and IK-1 ion channel blockers have been identified based on a virtual high throughput screening approach using a homology model of KcsA. These compounds display inhibitory effects on T-cell and/or keratinocyte proliferation and immunosuppressant activity within a DTH animal model.
Collapse
|
28
|
Bodendiek SB, Mahieux C, Hänsel W, Wulff H. 4-Phenoxybutoxy-substituted heterocycles--a structure-activity relationship study of blockers of the lymphocyte potassium channel Kv1.3. Eur J Med Chem 2008; 44:1838-52. [PMID: 19056148 DOI: 10.1016/j.ejmech.2008.10.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 10/25/2008] [Accepted: 10/30/2008] [Indexed: 10/21/2022]
Abstract
The voltage-gated potassium channel Kv1.3 constitutes an attractive pharmacological target for the treatment of effector memory T cell-mediated autoimmune diseases such as multiple sclerosis and psoriasis. Using 5-methoxypsoralen (5-MOP, 1), a compound isolated from Ruta graveolens, as a template we previously synthesized 5-(4-phenoxybutoxy)psoralen (PAP-1, 2) which inhibits Kv1.3 with an IC(50) of 2nM. Since PAP-1 is more than 1000-fold more potent than 5-MOP, we here investigated whether attaching a 4-phenoxybutoxy side chain to other heterocyclic systems would also produce potent Kv1.3 blockers. While 4-phenoxybutoxy-substituted quinolines, quinazolines and phenanthrenes were inactive, 4-phenoxybutoxy-substituted quinolinones, furoquinolines, coumarins or furochromones inhibited Kv1.3 with IC(50)s of 150 nM to 10 microM in whole-cell patch-clamp experiments. Our most potent new compound is 4-(4-phenoxybutoxy)-7H-furo[3,2-g]chromene-7-thione (73, IC(50) 17 nM), in which the carbonyl oxygen of PAP-1 is replaced by sulfur. Taken together, our results demonstrate that the psoralen system is a crucial part of the pharmacophore of phenoxyalkoxypsoralen-type Kv1.3 blockers.
Collapse
Affiliation(s)
- Silke B Bodendiek
- Department of Pharmacology, University of California, Davis, Genome and Biomedical Sciences Facility, 451 Health Sciences Drive, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
29
|
Karczewski J, Kiss L, Kane SA, Koblan KS, Lynch RJ, Spencer RH. High-throughput analysis of drug binding interactions for the human cardiac channel, Kv1.5. Biochem Pharmacol 2008; 77:177-85. [PMID: 18955031 DOI: 10.1016/j.bcp.2008.09.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 09/17/2008] [Accepted: 09/25/2008] [Indexed: 10/21/2022]
Abstract
The voltage-gated potassium channel Kv1.5 is one of the key regulators of membrane potential repolarization in human atrial myocytes and is considered a potential drug target to treat atrial fibrillation. In this study we sought to determine molecular mechanism of action of DPO-1, a diphenylphosphine oxide derivative recently shown to terminate experimental atrial arrhythmia without affecting ventricular refractory period. In addition, we provided similar analysis for additional two small molecule blockers, representing different structural classes: cyclohexanones (PAC) and nor-triterpenoids (correolide). To rapidly identify the residues within the Kv1.5 channel critical for blocking activity of these molecules, two functional high-throughput ion channel assays were employed together with site-directed mutagenesis. Our study revealed that the residues critical for blocking activity of for DPO-1 include T480, localized at the outer mouth of the pore, and two residues along S6 helix: V505 and I508. The overlapping site was identified for PAC and included residues T480 and V505. In contrast to DPO-1, the I508A mutation resulted in only a modest reduction in the block of Kv1.5 by PAC (9-fold). Correolide, the largest molecule examined, made widespread interactions along the entire length of the pore (from T480 to V516). In summary, we have identified multiple residues involved in forming high affinity binding site for Kv1.5 blockers. Similar approaches of high-throughput ion channel technologies, combined with site-directed mutagenesis, may allow for parallel, rapid and accurate analysis of ion channel interactions with multiple compounds and could facilitate the design of more potent and selective ion channel blockers.
Collapse
Affiliation(s)
- Jerzy Karczewski
- Department of Pain Research, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Ratliff KS, Petrov A, Eiermann GJ, Deng Q, Green MD, Kaczorowski GJ, McManus OB, Herrington J. An automated electrophysiology serum shift assay for K(V) channels. Assay Drug Dev Technol 2008; 6:243-53. [PMID: 18471078 DOI: 10.1089/adt.2008.128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The presence of serum in biological samples often negatively impacts the quality of in vitro assays. However, assays tolerant of serum are useful for assessing the in vivo availability of a small molecule for its target. Electrophysiology assays of ion channels are notoriously sensitive to serum because of their reliance on the interaction of the plasma membrane with a recording electrode. Here we investigate the tolerance of an automated electrophysiology assay for a voltage-gated potassium (K(V)) channel to serum and purified plasma proteins. The delayed rectifier channel, K(V)2.1, stably expressed in Chinese hamster ovary cells produces large, stable currents on the IonWorks Quattro platform (MDS Analytical Technologies, Sunnyvale, CA), making it an ideal test case. K(V)2.1 currents recorded on this platform are highly resistant to serum, allowing recordings in as high as 33% serum. Using a set of compounds related to the K(V) channel blocker, 4-phenyl-4-[3-(2-methoxyphenyl)-3-oxo-2-azaprop-1-yl]cyclohexanone, we show that shifts in compound potency with whole serum or isolated serum proteins can be reliably measured with this assay. Importantly, this assay is also relatively insensitive to plasma, allowing the creation of a bioassay for inhibitors of K(V)2.1 channel activity. Here we show that such a bioassay can quantify the levels of the gating modifier, guangxitoxin-1E, in plasma samples from mice dosed with the peptide. This study demonstrates the utility of using an automated electrophysiology platform for measuring serum shifts and for bioassays of ion channel modulators.
Collapse
Affiliation(s)
- Kevin S Ratliff
- Department of Ion Channels, Merck Research Laboratories, Rahway, NJ 07065-0900, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abe T, Iwasaki K, Inoue M, Suzuki T, Watanabe K, Katoh T. Convergent and enantioselective total synthesis of (−)-nalanthalide, a potential Kv1.3 blocking immunosuppressant. Tetrahedron Lett 2006. [DOI: 10.1016/j.tetlet.2006.03.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
32
|
Al-Rawi S, Ahlborn C, Richert C. 3'-immobilized probes with 2'-caps: synthesis of oligonucleotides with 2'-N-methyl-2'-(anthraquinone carboxamido)uridine residues. Org Lett 2006; 7:1569-72. [PMID: 15816754 DOI: 10.1021/ol0502432] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
[reaction: see text] A synthesis for oligodeoxynucleotides with a 3'-terminal 2'-N-methyl-2'-acylamido-2'-deoxyuridine residue was developed. Unlike their unmethylated counterparts, these oligodeoxynucleotides can be stably immobilized on aldehyde-displaying glass surfaces to provide DNA microarrays. An anthraquinone carboxamido group as a 2'-substituent doubled the capture efficiency of an immobilized tetradecamer.
Collapse
Affiliation(s)
- Samy Al-Rawi
- Institute for Organic Chemistry, Universität Karlsruhe (TH), D-76131 Karlsruhe, Germany
| | | | | |
Collapse
|
33
|
Slack M, Kirchhoff C, Moller C, Winkler D, Netzer R. Identification of novel Kv1.3 blockers using a fluorescent cell-based ion channel assay. ACTA ACUST UNITED AC 2005; 11:57-64. [PMID: 16314404 DOI: 10.1177/1087057105282712] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A functional cell-based assay was developed using a generic proprietary assay protocol, based on a membrane-potential sensitive dye, for the identification of small-molecule antagonists against the Kv1.3 potassium ion channel. A high-throughput screen (HTS) was subsequently performed with 20,000 compounds from the Evotec library, preselected using known small molecule antagonists for both sodium and potassium ion channels. Following data analysis, the hit rate was measured at 1.72%, and subsequent dose-response analysis of selected hits showed a high hit confirmation rate yielding approximately 50 compounds with an apparent IC50 value lower than 10 microM. Subsequent electrophysiological characterization of selected hits confirmed the initial activity and potency of the identified hits on the Kv1.3 target and also selectivity toward Kv1.3 through measurements on HERG as well as Kv1.3-expressing cell lines. Follow-up structure-activity relationship analysis revealed a variety of different clusters distributed throughout the library as well as several singlicates. In comparison to known Kv1.3 blockers, new chemical entities and scaffolds showing potency and selectivity against the Kv1.3 ion channel were detected. In addition, a screening strategy for ion channel drug discovery HTS, medicinal chemistry, and electrophysiology is presented.
Collapse
Affiliation(s)
- Mark Slack
- Evotec AG Schnackenburgallee 114 22525 Hamburg, Germany.
| | | | | | | | | |
Collapse
|
34
|
Watanabe K, Iwasaki K, Abe T, Inoue M, Ohkubo K, Suzuki T, Katoh T. Enantioselective Total Synthesis of (−)-Candelalide A, a Novel Blocker of the Voltage-Gated Potassium Channel Kv1.3 for an Immunosuppressive Agent. Org Lett 2005; 7:3745-8. [PMID: 16092865 DOI: 10.1021/ol051398c] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A convergent route to (-)-candelalide A involved the union of a trans-decalin portion (AB ring) and a gamma-pyrone moiety through the C16-C3' bond to assemble the whole carbon framework and subsequent formation of the dihydropyran ring (C ring) as the crucial steps. A strategic [2,3]-Wittig rearrangement was employed for establishing the stereogenic center at C9 and an exo-methylene function at C8 present in the decalin portion. [reaction: see text]
Collapse
Affiliation(s)
- Kazuhiro Watanabe
- Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | | | | | | | | | | | | |
Collapse
|
35
|
Dreker T, Grissmer S. Investigation of the phenylalkylamine binding site in hKv1.3 (H399T), a mutant with a reduced C-type inactivated state. Mol Pharmacol 2005; 68:966-73. [PMID: 16000530 DOI: 10.1124/mol.105.012401] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
To screen for residues of hKv1.3 important for current block by the phenylalkylamine verapamil, the inactivated-state-reduced H399T mutant was used as a background for mutagenesis studies. This approach was applied mainly to abolish the accumulation in the inactivated blocked state, recovery from which in the wild type is normally slow. Substitution of amino acids in the S6 transmembrane helix indicated a heavy disruption of verapamil block by the A413C mutation, reducing the IC(50) from 2.4 to 267 microM. Subsequent scanning for verapamil moieties essential for current block was performed by application of derivatives with altered side groups. Neither the removal of the nitrile or the methyl group nor the addition of a methoxy group resulted in major variations of IC(50) values for hKv1.3 (H399T) current block. However, disruption of current block by A413C was 4- to 10-fold less pronounced for derivatives lacking the 4-methoxy group of the (3,4-dimethoxyphenyl)ethylmethyl-amino part (devapamil) or all four methoxy groups (emopamil), respectively. Emopamil displayed a Hill coefficient of 2 for hKv1.3 (H399T/A413C) instead of 1 for hKv1.3 (H399T) current block. These results might indicate that the alteration of Ala413 modulates the access of phenylalkylamines to their binding site depending on the occupancy of the phenyl rings with methoxy groups. A computer-based docking model shows a subset of docked PAA conformations, with a spatial proximity between the (4-methoxyphenyl)ethyl-methyl-amino group and Ala413. The PAA binding site might therefore include a binding pocket for the aromatic ring of the ethyl-methyl-amino part in an S6-S6 interface gap.
Collapse
Affiliation(s)
- Tobias Dreker
- Department of Applied Physiology, University of Ulm, Germany
| | | |
Collapse
|
36
|
Bao J, Miao S, Kayser F, Kotliar AJ, Baker RK, Doss GA, Felix JP, Bugianesi RM, Slaughter RS, Kaczorowski GJ, Garcia ML, Ha SN, Castonguay L, Koo GC, Shah K, Springer MS, Staruch MJ, Parsons WH, Rupprecht KM. Potent Kv1.3 inhibitors from correolide-modification of the C18 position. Bioorg Med Chem Lett 2005; 15:447-51. [PMID: 15603971 DOI: 10.1016/j.bmcl.2004.10.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Revised: 10/15/2004] [Accepted: 10/18/2004] [Indexed: 11/28/2022]
Abstract
Kv1.3, the voltage-gated potassium channel in human T cells, represents a new target for treating immunosuppression and autoimmune diseases. Correolide (1), a pentacyclic natural product, is a potent and selective Kv1.3 channel blocker. Simplification of correolide via removal of its E-ring generates enone 4, whose modification produced a new series of tetracyclic Kv1.3 blockers. The structure-activity relationship for this class of compounds in two functional assays, Rb_Kv and human T cell proliferation, is presented herein. The most potent analog 43 is 15-fold more potent than correolide as inhibitor of human T cell proliferation.
Collapse
Affiliation(s)
- Jianming Bao
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 2000, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Herrington J, Sanchez M, Wunderler D, Yan L, Bugianesi RM, Dick IE, Clark SA, Brochu RM, Priest BT, Kohler MG, McManus OB. Biophysical and pharmacological properties of the voltage-gated potassium current of human pancreatic beta-cells. J Physiol 2005; 567:159-75. [PMID: 15932888 PMCID: PMC1474166 DOI: 10.1113/jphysiol.2005.089375] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Voltage-gated potassium (Kv) currents of human pancreatic islet cells were studied by whole-cell patch clamp recording. On average, 75% of the cells tested were identified as beta-cells by single cell, post-recording RT-PCR for insulin mRNA. In most cells, the dominant Kv current was a delayed rectifier. The delayed rectifier activated at potentials above -20 mV and had a V(1/2) for activation of -5.3 mV. Onset of inactivation was slow for a major component (tau = 3.2 s at +20 mV) observed in all cells; a smaller component (tau = 0.30 s) with an amplitude of approximately 25% was seen in some cells. Recovery from inactivation had a tau of 2.5 s at -80 mV and steady-state inactivation had a V(1/2) of -39 mV. In 12% of cells (21/182) a low-threshold, transient Kv current (A-current) was present. The A-current activated at membrane potentials above -40 mV, inactivated with a time constant of 18.5 ms at -20 mV, and had a V(1/2) for steady-state inactivation of -52 mV. TEA inhibited total Kv current with an IC50 = 0.54 mm and PAC, a disubstituted cyclohexyl Kv channel inhibitor, inhibited with an IC50 = 0.57 microm. The total Kv current was insensitive to margatoxin (100 nm), agitoxin-2 (50 nm), kaliotoxin (50 nm) and ShK (50 nm). Hanatoxin (100 nm) inhibited total Kv current by 65% at +20 mV. Taken together, these data provide evidence of at least two distinct types of Kv channels in human pancreatic beta-cells and suggest that more than one type of Kv channel may be involved in the regulation of glucose-dependent insulin secretion.
Collapse
Affiliation(s)
- James Herrington
- Department of Ion Channels, Merck Research Laboratories, PO Box 2000, RY-80N-C31, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Beeton C, Pennington MW, Wulff H, Singh S, Nugent D, Crossley G, Khaytin I, Calabresi PA, Chen CY, Gutman GA, Chandy KG. Targeting effector memory T cells with a selective peptide inhibitor of Kv1.3 channels for therapy of autoimmune diseases. Mol Pharmacol 2005; 67:1369-81. [PMID: 15665253 PMCID: PMC4275123 DOI: 10.1124/mol.104.008193] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The voltage-gated Kv1.3 K(+) channel is a novel target for immunomodulation of autoreactive effector memory T (T(EM)) cells that play a major role in the pathogenesis of autoimmune diseases. We describe the characterization of the novel peptide ShK(L5) that contains l-phosphotyrosine linked via a nine-atom hydrophilic linker to the N terminus of the ShK peptide from the sea anemone Stichodactyla helianthus. ShK(L5) is a highly specific Kv1.3 blocker that exhibits 100-fold selectivity for Kv1.3 (K(d) = 69 pM) over Kv1.1 and greater than 250-fold selectivity over all other channels tested. ShK(L5) suppresses the proliferation of human and rat T(EM) cells and inhibits interleukin-2 production at picomolar concentrations. Naive and central memory human T cells are initially 60-fold less sensitive than T(EM) cells to ShK(L5) and then become resistant to the peptide during activation by up-regulating the calcium-activated K(Ca)3.1 channel. ShK(L5) does not exhibit in vitro cytotoxicity on mammalian cell lines and is negative in the Ames test. It is stable in plasma and when administered once daily by subcutaneous injection (10 mug/kg) attains "steady state" blood levels of approximately 300 pM. This regimen does not cause cardiac toxicity assessed by continuous EKG monitoring and does not alter clinical chemistry and hematological parameters after 2-week therapy. ShK(L5) prevents and treats experimental autoimmune encephalomyelitis and suppresses delayed type hypersensitivity in rats. ShK(L5) might prove useful for therapy of autoimmune disorders.
Collapse
Affiliation(s)
- Christine Beeton
- Department of Physiology and Biophysics, 291 Irvine Hall, Medical School, University of California-Irvine, Irvine, CA 92697-4561, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Monte M Winslow
- Immunology Program, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
40
|
Abstract
New immunosuppressants are consistently developed to treat autoimmune diseases and some of them might have implications in multiple sclerosis (MS). A new antiproliferative agent, pixantrone, an analogue of mitoxantrone (MX), has a much lower cardiotoxicity and exerts the same potent immunosuppressive effects in experimental allergic encephalomyelitis (EAE). A phase I trial in MS patients is planned in the next future. New monoclonal antibodies (mAb) and other biological constructs containing foreign proteins are developed but their potential immunogenicity is a considerable drawback to their long-term administration. In addition, their beneficial effects in MS are not evident so far. Small molecules targeting the voltage-gated Kv1.3K+ channel regulating CA2+ signaling in T lymphocytes, specifically target activated, pathogenic T cells. Already found effective in EAE, those agents would be easier to handle than T-cell vaccination. Two new immunosuppressants with a unique mechanism of action (FTY720 and Epomycine M) selectively impair autoreactive T-cell homing, without affecting the other components of the immune response. The potent protective effect of TRY720 has been demonstrated in EAE and a phase I trial in MS appears warranted. Finally, a new concept about immunosuppressive treatments in organ transplantation, "tolerogenic immunosuppression", may have potential in MS.
Collapse
Affiliation(s)
- R E Gonsette
- National Center for Multiple Sclerosis, Vanheylenstraat 16, Melsbroek B-1820, Belgium.
| |
Collapse
|
41
|
Chandy KG, Wulff H, Beeton C, Pennington M, Gutman GA, Cahalan MD. K+ channels as targets for specific immunomodulation. Trends Pharmacol Sci 2004; 25:280-9. [PMID: 15120495 PMCID: PMC2749963 DOI: 10.1016/j.tips.2004.03.010] [Citation(s) in RCA: 351] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The voltage-gated Kv1.3 channel and the Ca(2+)-activated IKCa1 K(+) channel are expressed in T cells in a distinct pattern that depends on the state of lymphocyte activation and differentiation. The channel phenotype changes during the progression from the resting to the activated cell state and from naïve to effector memory cells, affording promise for specific immunomodulatory actions of K(+) channel blockers. In this article, we review the functional roles of these channels in both naïve cells and memory cells, describe the development of selective inhibitors of Kv1.3 and IKCa1 channels, and provide a rationale for the potential therapeutic use of these inhibitors in immunological disorders.
Collapse
Affiliation(s)
- K George Chandy
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| | | | | | | | | | | |
Collapse
|
42
|
High-throughput technologies for studying potassium channels – progresses and challenges. ACTA ACUST UNITED AC 2004. [DOI: 10.1016/s1741-8372(04)02394-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
43
|
Abstract
Human peripheral blood T lymphocytes possess two types of K(+) channels: the voltage-gated Kv1.3 and the calcium-activated IKCa1 channels. The use of peptidyl inhibitors of Kv1.3 and IKCa1 indicated that these channels are involved in the maintenance of membrane potential and that they play a crucial role in Ca(2+) signaling during T-cell activation. Thus, in vitro blockade of Kv1.3 and IKCa1 leads to inhibition of cytokine production and lymphocyte proliferation. These observations prompted several groups of investigators in academia and pharmaceutical companies to characterize the expression of Kv1.3 and IKCa1 in different subsets of human T lymphocytes and to evaluate their potential as novel targets for immunosuppression. Recent in vivo studies showed that chronically activated T lymphocytes involved in the pathogenesis of multiple sclerosis present unusually high expression of Kv1.3 channels and that the treatment with selective Kv1.3 inhibitors can either prevent or ameliorate the symptoms of the disease. In this model of multiple sclerosis, blockade of IKCa1 channels had no effect alone, but improved the response to Kv1.3 inhibitors. In addition, the expression of Kv1.3 and IKCa1 channels in human cells is very restricted, which makes them attractive targets for a more cell-specific and less harmful action than what is typically obtained with classical immunosuppressants. Studies using high-throughput toxin displacement, (86)Rb-efflux screening or membrane potential assays led to the identification of non-peptidyl small molecules with high affinity for Kv1.3 or IKCa1 channels. Analysis of structure-function relationships in Kv1.3 and IKCa1 channels helped define the binding sites for channel blockers, allowing the design of a new generation of small molecules with selectivity for either Kv1.3 or IKCa1, which could help the development of new drugs for safer treatment of auto-immune diseases.
Collapse
Affiliation(s)
- Rosane Vianna-Jorge
- Divisão de Farmacologia, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil.
| | | |
Collapse
|
44
|
Mori Y, Inagaki C, Kuno M, Inoue R, Okada Y, Imaizumi Y. [Ionic mechanisms underlying the regulation of cell proliferation, differentiation and death]. Nihon Yakurigaku Zasshi 2003; 122:201-14. [PMID: 12939538 DOI: 10.1254/fpj.122.201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Ion channels and transporters act as major components that regulate membrane excitability in neurons, muscles, and some secretory glands, but may also contribute to the regulation of proliferation, differentiation, and death in a greater variety of cells including non-excitable ones. The molecular basis of ionic mechanisms underlying the later regulation has been partly identified in the last several years and is a hot issue now. In this short review, some of the molecular mechanisms underlying these regulations and novel compounds acting on the mechanisms were introduced as exciting topics in this area. Several types of transient receptor potential (TRP), identified as Ca(2+)-permeable, non-selective cation channels, may play obligatory roles in functional complexes, which regulate multiple signal transduction pathways triggering proliferation, differentiation, or death of many cell types. In addition, the relation between Cl(-) pump activity and the induction of beta-amyloid protein toxicity for neuronal cell death in Alzheimer disease was described. Unique functions of H(+) channel and pump in osteoclasts in bone mineral homeostasis and remodeling were also discussed. Finally, topics about activation of specific types of Cl(-) channels and K(+) channels, which are responsible for the induction of apoptosis or proliferation in several types of cells, were introduced.
Collapse
Affiliation(s)
- Yasuo Mori
- Ctr. Integ. Biosci., Okazaki Natl. Res. Inst., Japan
| | | | | | | | | | | |
Collapse
|
45
|
Wulff H, Calabresi PA, Allie R, Yun S, Pennington M, Beeton C, Chandy KG. The voltage-gated Kv1.3 K(+) channel in effector memory T cells as new target for MS. J Clin Invest 2003; 111:1703-13. [PMID: 12782673 PMCID: PMC156104 DOI: 10.1172/jci16921] [Citation(s) in RCA: 339] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Through a combination of fluorescence microscopy and patch-clamp analysis we have identified a striking alteration in K(+) channel expression in terminally differentiated human CCR7(-)CD45RA(-) effector memory T lymphocytes (T(EM)). Following activation, T(EM) cells expressed significantly higher levels of the voltage-gated K(+) channel Kv1.3 and lower levels of the calcium-activated K(+) channel IKCa1 than naive and central memory T cells (T(CM)). Upon repeated in vitro antigenic stimulation, naive cells differentiated into Kv1.3(high)IKCa1(low) T(EM) cells, and the potent Kv1.3-blocking sea anemone Stichodactyla helianthus peptide (ShK) suppressed proliferation of T(EM) cells without affecting naive or T(CM) lymphocytes. Thus, the Kv1.3(high)IKCa1(low) phenotype is a functional marker of activated T(EM) lymphocytes. Activated myelin-reactive T cells from patients with MS exhibited the Kv1.3(high)IKCa1(low) T(EM) phenotype, suggesting that they have undergone repeated stimulation during the course of disease; these cells may contribute to disease pathogenesis due to their ability to home to inflamed tissues and exhibit immediate effector function. The Kv1.3(high)IKCa1(low) phenotype was not seen in glutamic acid decarboxylase, insulin-peptide or ovalbumin-specific and mitogen-activated T cells from MS patients, or in myelin-specific T cells from healthy controls. Selective targeting of Kv1.3 in T(EM) cells may therefore hold therapeutic promise for MS and other T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Heike Wulff
- Department of Physiology and Biophysics, University of California Irvine, College of Medicine, Irvine, California 92697, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Miao S, Bao J, Garcia ML, Goulet JL, Hong XJ, Kaczorowski GJ, Kayser F, Koo GC, Kotliar A, Schmalhofer WA, Shah K, Sinclair PJ, Slaughter RS, Springer MS, Staruch MJ, Tsou NN, Wong F, Parsons WH, Rupprecht KM. Benzamide derivatives as blockers of Kv1.3 ion channel. Bioorg Med Chem Lett 2003; 13:1161-4. [PMID: 12643934 DOI: 10.1016/s0960-894x(03)00014-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The voltage-gated potassium channel, Kv1.3, is present in human T-lymphocytes. Blockade of Kv1.3 results in T-cell depolarization, inhibition of T-cell activation, and attenuation of immune responses in vivo. A class of benzamide Kv1.3 channel inhibitors has been identified. The structure-activity relationship within this class of compounds in two functional assays, Rb_Kv and T-cell proliferation, is presented. In in vitro assays, trans isomers display moderate selectivity for binding to Kv1.3 over other Kv1.x channels present in human brain.
Collapse
Affiliation(s)
- Shouwu Miao
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 2000, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|