51
|
Comes N, Bielanska J, Vallejo-Gracia A, Serrano-Albarrás A, Marruecos L, Gómez D, Soler C, Condom E, Ramón Y Cajal S, Hernández-Losa J, Ferreres JC, Felipe A. The voltage-dependent K(+) channels Kv1.3 and Kv1.5 in human cancer. Front Physiol 2013; 4:283. [PMID: 24133455 PMCID: PMC3794381 DOI: 10.3389/fphys.2013.00283] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 09/18/2013] [Indexed: 11/20/2022] Open
Abstract
Voltage-dependent K+ channels (Kv) are involved in a number of physiological processes, including immunomodulation, cell volume regulation, apoptosis as well as differentiation. Some Kv channels participate in the proliferation and migration of normal and tumor cells, contributing to metastasis. Altered expression of Kv1.3 and Kv1.5 channels has been found in several types of tumors and cancer cells. In general, while the expression of Kv1.3 apparently exhibits no clear pattern, Kv1.5 is induced in many of the analyzed metastatic tissues. Interestingly, evidence indicates that Kv1.5 channel shows inversed correlation with malignancy in some gliomas and non-Hodgkin's lymphomas. However, Kv1.3 and Kv1.5 are similarly remodeled in some cancers. For instance, expression of Kv1.3 and Kv1.5 correlates with a certain grade of tumorigenicity in muscle sarcomas. Differential remodeling of Kv1.3 and Kv1.5 expression in human cancers may indicate their role in tumor growth and their importance as potential tumor markers. However, despite of this increasing body of information, which considers Kv1.3 and Kv1.5 as emerging tumoral markers, further research must be performed to reach any conclusion. In this review, we summarize what it has been lately documented about Kv1.3 and Kv1.5 channels in human cancer.
Collapse
Affiliation(s)
- Núria Comes
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Vallejo-Gracia A, Bielanska J, Hernández-Losa J, Castellví J, Ruiz-Marcellan MC, Ramón y Cajal S, Condom E, Manils J, Soler C, Comes N, Ferreres JC, Felipe A. Emerging role for the voltage-dependent K+channel Kv1.5 in B-lymphocyte physiology: expression associated with human lymphoma malignancy. J Leukoc Biol 2013; 94:779-89. [DOI: 10.1189/jlb.0213094] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
53
|
Abstract
A recent patent application from Bionomics/Merck Serono describes novel compounds as blockers of the voltage-gated Kv1.3 ion channel. The blockade of this channel shows great promise as a new therapeutic target for the treatment of autoimmune disorders such as multiple sclerosis, psoriasis, diabetes and rheumatoid arthritis. The generic claim of this patent refers to a new chemotype of Kv1.3 blockers based on an amide core with potent IC50's which are potentially within the nanomolar range. This article briefly reviews the chemistry and biology found in the patent and compares it with previous discoveries in the field.
Collapse
Affiliation(s)
- William Nguyen
- The Scripps Research Institute, Department of Molecular Therapeutics , 130 Scripps Way, Jupiter, FL , USA +1 561 228 2210 ; +1 561 228 3092 ;
| |
Collapse
|
54
|
Marzian S, Stansfeld PJ, Rapedius M, Rinné S, Nematian-Ardestani E, Abbruzzese JL, Steinmeyer K, Sansom MSP, Sanguinetti MC, Baukrowitz T, Decher N. Side pockets provide the basis for a new mechanism of Kv channel-specific inhibition. Nat Chem Biol 2013; 9:507-13. [PMID: 23728494 PMCID: PMC4539245 DOI: 10.1038/nchembio.1271] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/29/2013] [Indexed: 01/09/2023]
Abstract
Most known small-molecule inhibitors of voltage-gated ion channels have poor subtype specificity because they interact with a highly conserved binding site in the central cavity. Using alanine-scanning mutagenesis, electrophysiological recordings and molecular modeling, we have identified a new drug-binding site in Kv1.x channels. We report that Psora-4 can discriminate between related Kv channel subtypes because, in addition to binding the central pore cavity, it binds a second, less conserved site located in side pockets formed by the backsides of S5 and S6, the S4-S5 linker, part of the voltage sensor and the pore helix. Simultaneous drug occupation of both binding sites results in an extremely stable nonconducting state that confers high affinity, cooperativity, use-dependence and selectivity to Psora-4 inhibition of Kv1.x channels. This new mechanism of inhibition represents a molecular basis for the development of a new class of allosteric and selective voltage-gated channel inhibitors.
Collapse
Affiliation(s)
- Stefanie Marzian
- Institute for Physiology and Pathophysiology, University of Marburg, Marburg, Germany
| | - Phillip J Stansfeld
- Structural Bioinformatics and Computational Biochemistry Unit, University of Oxford, Oxford, UK
| | | | - Susanne Rinné
- Institute for Physiology and Pathophysiology, University of Marburg, Marburg, Germany
| | | | - Jennifer L Abbruzzese
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - Klaus Steinmeyer
- Sanofi-Aventis, Therapeutic Strategic Unit–Aging, Frankfurt, Germany
| | - Mark S P Sansom
- Structural Bioinformatics and Computational Biochemistry Unit, University of Oxford, Oxford, UK
| | - Michael C Sanguinetti
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | | | - Niels Decher
- Institute for Physiology and Pathophysiology, University of Marburg, Marburg, Germany
| |
Collapse
|
55
|
Göbel K, Wedell JH, Herrmann AM, Wachsmuth L, Pankratz S, Bittner S, Budde T, Kleinschnitz C, Faber C, Wiendl H, Meuth SG. 4-Aminopyridine ameliorates mobility but not disease course in an animal model of multiple sclerosis. Exp Neurol 2013; 248:62-71. [PMID: 23748135 DOI: 10.1016/j.expneurol.2013.05.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 05/22/2013] [Accepted: 05/25/2013] [Indexed: 01/21/2023]
Abstract
Neuropathological changes following demyelination in multiple sclerosis (MS) lead to a reorganization of axolemmal channels that causes conduction changes including conduction failure. Pharmacological modulation of voltage-sensitive potassium channels (K(V)) has been found to improve conduction in experimentally induced demyelination and produces symptomatic improvement in MS patients. Here we used an animal model of autoimmune inflammatory neurodegeneration, namely experimental autoimmune encephalomyelitis (EAE), to test the influence of the K(V)-inhibitor 4-aminopyridine (4-AP) on various disease and immune parameters as well as mobility in MOG₃₅₋₅₅ immunized C57Bl/6 mice. We challenged the hypothesis that 4-AP exerts relevant immunomodulatory or neuroprotective properties. Neither prophylactic nor therapeutic treatment with 4-AP altered disease incidence or disease course of EAE. Histopathological signs of demyelination and neuronal damage as well as MRI imaging of brain volume changes were unaltered. While application of 4-AP significantly reduced the standing outward current of stimulated CD4(+) T cells compared to controls, it failed to impact intracellular calcium concentrations in these cells. Compatibly, KV channel inhibition neither influenced CD4(+) T cell effector functions (proliferation, IL17 or IFNγ production). Importantly however, despite equal disease severity scores 4-AP treated animals showed improved mobility as assessed by 2 independent methods, 1) foot print and 2) rotarod analysis (0.332 ± 0.03, n=7 versus 0.399 ± 0.08, n=14, p<0.001, respectively). Our data suggest that 4-AP while having no apparent immunomodulatory or direct neuroprotective effects, significantly ameliorates conduction abnormalities thereby improving gait and coordination. Improvement of mobility in this experimental model supports trial data and clinical experience with 4-AP in the symptomatic treatment of MS.
Collapse
Affiliation(s)
- Kerstin Göbel
- University of Muenster, Department of Neurology, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Wang J, Xiang M. Targeting potassium channels Kv1.3 and KC a 3.1: routes to selective immunomodulators in autoimmune disorder treatment? Pharmacotherapy 2013; 33:515-28. [PMID: 23649812 DOI: 10.1002/phar.1236] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The Kv1.3 and KC a 3.1 potassium channels are promising targets for the treatment of autoimmune disorders. Many Kv1.3 and KC a 3.1 blockers have a more favorable adverse event profiles than existing immunosuppressants, suggesting the selectivity of Kv1.3 and KC a 3.1 blockade. The Kv1.3 and KC a 3.1 blockers exert differential effects in different autoimmune diseases. The Kv1.3 inhibitors or gene deletion have been shown to have benefits in multiple sclerosis, type 1 diabetes, rheumatoid arthritis, psoriasis, and rapidly progressive glomerulonephritis. The KC a 3.1 blockers have demonstrated efficacy in human primary biliary cirrhosis and showed protective effects in animal models of severe colitis, allergic encephalomyelitis, inflammatory bowel disease, and multiple sclerosis. The KC a 3.1 blockers are not considered candidates for treatment of multiple sclerosis. The selective immunosuppressive effects of the Kv1.3 and KC a 3.1 blockers are due to the differences in their distribution on autoimmune-related immune cells and tissues and β1 integrin (very late activating antigen)-Kv1.3 channel cross-talk.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | |
Collapse
|
57
|
Decher N, Netter MF, Streit AK. Putative Impact of RNA Editing on Drug Discovery. Chem Biol Drug Des 2012; 81:13-21. [DOI: 10.1111/cbdd.12045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
58
|
Bagal SK, Brown AD, Cox PJ, Omoto K, Owen RM, Pryde DC, Sidders B, Skerratt SE, Stevens EB, Storer RI, Swain NA. Ion Channels as Therapeutic Targets: A Drug Discovery Perspective. J Med Chem 2012; 56:593-624. [DOI: 10.1021/jm3011433] [Citation(s) in RCA: 227] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sharan K. Bagal
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Alan D. Brown
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Peter J. Cox
- Pfizer Neusentis, The
Portway Building, Granta Park, Great Abington, Cambridge, CB21
6GS, U.K
| | - Kiyoyuki Omoto
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Robert M. Owen
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - David C. Pryde
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Benjamin Sidders
- Pfizer Neusentis, The
Portway Building, Granta Park, Great Abington, Cambridge, CB21
6GS, U.K
| | - Sarah E. Skerratt
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Edward B. Stevens
- Pfizer Neusentis, The
Portway Building, Granta Park, Great Abington, Cambridge, CB21
6GS, U.K
| | - R. Ian Storer
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Nigel A. Swain
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| |
Collapse
|
59
|
Li Z, Liu WH, Han S, Peng BW, Yin J, Wu YL, He XH, Li WX. Selective inhibition of CCR7(-) effector memory T cell activation by a novel peptide targeting Kv1.3 channel in a rat experimental autoimmune encephalomyelitis model. J Biol Chem 2012; 287:29479-94. [PMID: 22761436 DOI: 10.1074/jbc.m112.379594] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The voltage-gated Kv1.3 K(+) channel in effector memory T cells serves as a new therapeutic target for multiple sclerosis. In our previous studies, the novel peptide ADWX-1 was designed and synthesized as a specific Kv1.3 blocker. However, it is unclear if and how ADWX-1 alleviates experimental autoimmune encephalomyelitis, a model for multiple sclerosis. In this study, the administration of ADWX-1 significantly ameliorated the rat experimental autoimmune encephalomyelitis model by selectively inhibiting CD4(+)CCR7(-) phenotype effector memory T cell activation. In contrast, the Kv1.3-specific peptide had little effect on CD4(+)CCR7(+) cells, thereby limiting side effects. Furthermore, we determined that ADWX-1 is involved in the regulation of NF-κB signaling through upstream protein kinase C-θ (PKCθ) in the IL-2 pathway of CD4(+)CCR7(-) cells. The elevated expression of Kv1.3 mRNA and protein in activated CD4(+)CCR7(-) cells was reduced by ADWX-1 engagement; however, an apparent alteration in CD4(+)CCR7(+) cells was not observed. Moreover, the selective regulation of the Kv1.3 channel gene expression pattern by ADWX-1 provided a further and sustained inhibition of the CD4(+)CCR7(-) phenotype, which depends on the activity of Kv1.3 to modulate its activation signal. In addition, ADWX-1 mediated the activation of differentiated Th17 cells through the CCR7(-) phenotype. The efficacy of ADWX-1 is supported by multiple functions, which are based on a Kv1.3(high) CD4(+)CCR7(-) T cell selectivity through two different pathways, including the classic channel activity-associated IL-2 pathway and the new Kv1.3 channel gene expression pathway.
Collapse
Affiliation(s)
- Zhi Li
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | | | | | | | | | | | | | | |
Collapse
|
60
|
Feske S, Skolnik EY, Prakriya M. Ion channels and transporters in lymphocyte function and immunity. Nat Rev Immunol 2012; 12:532-47. [PMID: 22699833 DOI: 10.1038/nri3233] [Citation(s) in RCA: 350] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lymphocyte function is regulated by a network of ion channels and transporters in the plasma membrane of B and T cells. These proteins modulate the cytoplasmic concentrations of diverse cations, such as calcium, magnesium and zinc ions, which function as second messengers to regulate crucial lymphocyte effector functions, including cytokine production, differentiation and cytotoxicity. The repertoire of ion-conducting proteins includes calcium release-activated calcium (CRAC) channels, P2X receptors, transient receptor potential (TRP) channels, potassium channels, chloride channels and magnesium and zinc transporters. This Review discusses the roles of ion conduction pathways in lymphocyte function and immunity.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, New York University Langone Medical Center, New York, New York 10016, USA.
| | | | | |
Collapse
|
61
|
Yang XF, Yang Y, Lian YT, Wang ZH, Li XW, Cheng LX, Liu JP, Wang YF, Gao X, Liao YH, Wang M, Zeng QT, Liu K. The antibody targeting the E314 peptide of human Kv1.3 pore region serves as a novel, potent and specific channel blocker. PLoS One 2012; 7:e36379. [PMID: 22558454 PMCID: PMC3338681 DOI: 10.1371/journal.pone.0036379] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 04/04/2012] [Indexed: 01/08/2023] Open
Abstract
Selective blockade of Kv1.3 channels in effector memory T (T(EM)) cells was validated to ameliorate autoimmune or autoimmune-associated diseases. We generated the antibody directed against one peptide of human Kv1.3 (hKv1.3) extracellular loop as a novel and possible Kv1.3 blocker. One peptide of hKv1.3 extracellular loop E3 containing 14 amino acids (E314) was chosen as an antigenic determinant to generate the E314 antibody. The E314 antibody specifically recognized 63.8KD protein stably expressed in hKv1.3-HEK 293 cell lines, whereas it did not recognize or cross-react to human Kv1.1(hKv1.1), Kv1.2(hKv1.2), Kv1.4(hKv1.4), Kv1.5(hKv1.5), KCa3.1(hKCa3.1), HERG, hKCNQ1/hKCNE1, Nav1.5 and Cav1.2 proteins stably expressed in HEK 293 cell lines or in human atrial or ventricular myocytes by Western blotting analysis and immunostaining detection. By the technique of whole-cell patch clamp, the E314 antibody was shown to have a directly inhibitory effect on hKv1.3 currents expressed in HEK 293 or Jurkat T cells and the inhibition showed a concentration-dependence. However, it exerted no significant difference on hKv1.1, hKv1.2, hKv1.4, hKv1.5, hKCa3.1, HERG, hKCNQ1/hKCNE1, L-type Ca(2+) or voltage-gated Na(+) currents. The present study demonstrates that the antibody targeting the E314 peptide of hKv1.3 pore region could be a novel, potent and specific hKv1.3 blocker without affecting a variety of closely related K(v)1 channels, KCa3.1 channels and functional cardiac ion channels underlying central nervous system (CNS) disorders or drug-acquired arrhythmias, which is required as a safe clinic-promising channel blocker.
Collapse
Affiliation(s)
- Xiao-Fang Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Tian Lian
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Wang
- Department of Geriatrics, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Wei Li
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Long-Xian Cheng
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Ping Liu
- Department of Cardiovascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Fu Wang
- Department of Cardiology, Affiliated Hospital, Jining Medical College, Shandong, China
| | - Xiang Gao
- Department of Geriatrics, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Hua Liao
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu-Tang Zeng
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Cardiology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
62
|
Leanza L, Henry B, Sassi N, Zoratti M, Chandy KG, Gulbins E, Szabò I. Inhibitors of mitochondrial Kv1.3 channels induce Bax/Bak-independent death of cancer cells. EMBO Mol Med 2012; 4:577-93. [PMID: 22496117 PMCID: PMC3407946 DOI: 10.1002/emmm.201200235] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 02/17/2012] [Accepted: 02/21/2012] [Indexed: 02/01/2023] Open
Abstract
Overcoming the resistance of tumours to chemotherapy, often due to downregulation of Bax and Bak, represents a significant clinical challenge. It is therefore important to identify novel apoptosis inducers that bypass Bax and Bak. Potassium channels are emerging as oncological targets and a crucial role of mitochondrial Kv1.3 in apoptosis has been demonstrated. Here we report for the first time that Psora-4, PAP-1 and clofazimine, three distinct membrane-permeant inhibitors of Kv1.3, induce death by directly targeting the mitochondrial channel in multiple human and mouse cancer cell lines. Importantly, these drugs activated the intrinsic apoptotic pathway also in the absence of Bax and Bak, a result in agreement with the current mechanistic model for mitochondrial Kv1.3 action. Genetic deficiency or short interfering RNA (siRNA)-mediated downregulation of Kv1.3 abrogated the effects of the drugs. Intraperitoneal injection of clofazimine reduced tumour size by 90% in an orthotopic melanoma B16F10 mouse model in vivo, while no adverse effects were observed in several healthy tissues. The study indicates that inhibition of mitochondrial Kv1.3 might be a novel therapeutic option for the induction of cancer cell death independent of Bax and Bak.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
63
|
Streit AK, Decher N. A-to-I RNA editing modulates the pharmacology of neuronal ion channels and receptors. BIOCHEMISTRY (MOSCOW) 2012; 76:890-9. [PMID: 22022962 DOI: 10.1134/s0006297911080049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The regulation of neuronal excitability is complex, as ion channels and neurotransmitter receptors are underlying a large variety of modulating effects. Alterations in the expression patterns of receptors or channel subunits as well as differential splicing contribute to the regulation of neuronal excitability. RNA editing is another and more recently explored mechanism to increase protein diversity, as the genomic recoding leads to new gene products with novel functional and pharmacological properties. In humans A-to-I RNA editing targets several neuronal receptors and channels, including GluR2/5/6 subunits, the Kv1.1 channel, and the 5-HT(2C) receptor. Our review summarizes that RNA editing of these proteins does not only change protein function, but also the pharmacology and presumably the drug therapy in human diseases.
Collapse
Affiliation(s)
- A K Streit
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, 35037, Germany
| | | |
Collapse
|
64
|
Lee WG, Kim WS, Park SG, Kim H, Hong J, Ko H, Kim YC. Immunosuppressive effects of subglutinol derivatives. ChemMedChem 2011; 7:218-22. [PMID: 22114006 DOI: 10.1002/cmdc.201100409] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 10/28/2011] [Indexed: 11/08/2022]
Affiliation(s)
- Won-Gil Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), 261 Cheomdan-gwagiro(Oryong- dong), Buk-gu, Gwangju 500-712, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
65
|
Wu SN, Chen BS, Lo YC. Evidence for aconitine-induced inhibition of delayed rectifier K(+) current in Jurkat T-lymphocytes. Toxicology 2011; 289:11-18. [PMID: 21782880 DOI: 10.1016/j.tox.2011.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 07/04/2011] [Accepted: 07/06/2011] [Indexed: 02/07/2023]
Abstract
Aconitine (ACO) is a highly toxic diterpenoid alkaloid and known to exert the immunomodulatory action. However, whether it has any effects on ion currents in immune cells remains unknown. The effects of ACO and other related compounds on ion currents in Jurkat T-lymphocytes were investigated in this study. ACO suppressed the amplitude of delayed-rectifier K(+) current (I(K(DR))) in a time- and concentration-dependent manner. Margatoxin (100 nM), a specific blocker of K(V)1.3-encoded current, decreased the I(K(DR)) amplitude in these cells and the ACO-induced inhibition of I(K(DR)) was not reversed by 1-ethyl-2-benzimidazolinone (30 μM) or nicotine (10 μM). The IC(50) value for ACO-mediated inhibition of I(K(DR)) was 5.6 μM. ACO accelerated the inactivation of I(K(DR)) with no change in the activation rate of this current. Increasing the ACO concentration not only reduced the I(K(DR)) amplitude, but also accelerated the inactivation time course of the current. With the aid of minimal binding scheme, the inhibitory action of ACO on I(K(DR)) was estimated with a dissociation constant of 6.8 μM. ACO also shifted the inactivation curve of I(K(DR)) to a hyperpolarized potential with no change in the slope factor. Cumulative inactivation for I(K(DR)) was enhanced in the presence of ACO. In Jurkat cells incubated with amiloride (30 μM), the ACO-induced inhibition of I(K(DR)) remained unaltered. In RAW 264.7 murine macrophages, ACO did not modify the kinetics of I(K(DR)), although it suppressed I(K(DR)) amplitude. Taken together, these effects can significantly contribute to its action on functional activity of immune cells if similar results are found in vivo.
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan.
| | | | | |
Collapse
|
66
|
Chi V, Pennington MW, Norton RS, Tarcha EJ, Londono LM, Sims-Fahey B, Upadhyay SK, Lakey JT, Iadonato S, Wulff H, Beeton C, Chandy KG. Development of a sea anemone toxin as an immunomodulator for therapy of autoimmune diseases. Toxicon 2011; 59:529-46. [PMID: 21867724 DOI: 10.1016/j.toxicon.2011.07.016] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/16/2011] [Accepted: 07/20/2011] [Indexed: 01/05/2023]
Abstract
Electrophysiological and pharmacological studies coupled with molecular identification have revealed a unique network of ion channels--Kv1.3, KCa3.1, CRAC (Orai1 + Stim1), TRPM7, Cl(swell)--in lymphocytes that initiates and maintains the calcium signaling cascade required for activation. The expression pattern of these channels changes during lymphocyte activation and differentiation, allowing the functional network to adapt during an immune response. The Kv1.3 channel is of interest because it plays a critical role in subsets of T and B lymphocytes implicated in autoimmune disorders. The ShK toxin from the sea anemone Stichodactyla helianthus is a potent blocker of Kv1.3. ShK-186, a synthetic analog of ShK, is being developed as a therapeutic for autoimmune diseases, and is scheduled to begin first-in-man phase-1 trials in 2011. This review describes the journey that has led to the development of ShK-186.
Collapse
Affiliation(s)
- Victor Chi
- Department of Physiology and Biophysics, UC Irvine, Irvine, CA 92697, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Cheong A, Li J, Sukumar P, Kumar B, Zeng F, Riches K, Munsch C, Wood IC, Porter KE, Beech DJ. Potent suppression of vascular smooth muscle cell migration and human neointimal hyperplasia by KV1.3 channel blockers. Cardiovasc Res 2011; 89:282-9. [PMID: 20884640 PMCID: PMC3020133 DOI: 10.1093/cvr/cvq305] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 09/16/2010] [Accepted: 09/20/2010] [Indexed: 11/14/2022] Open
Abstract
AIM The aim of the study was to determine the potential for K(V)1 potassium channel blockers as inhibitors of human neoinitimal hyperplasia. METHODS AND RESULTS Blood vessels were obtained from patients or mice and studied in culture. Reverse transcriptase-polymerase chain reaction and immunocytochemistry were used to detect gene expression. Whole-cell patch-clamp, intracellular calcium measurement, cell migration assays, and organ culture were used to assess channel function. K(V)1.3 was unique among the K(V)1 channels in showing preserved and up-regulated expression when the vascular smooth muscle cells switched to the proliferating phenotype. There was strong expression in neointimal formations. Voltage-dependent potassium current in proliferating cells was sensitive to three different blockers of K(V)1.3 channels. Calcium entry was also inhibited. All three blockers reduced vascular smooth muscle cell migration and the effects were non-additive. One of the blockers (margatoxin) was highly potent, suppressing cell migration with an IC(50) of 85 pM. Two of the blockers were tested in organ-cultured human vein samples and both inhibited neointimal hyperplasia. CONCLUSION K(V)1.3 potassium channels are functional in proliferating mouse and human vascular smooth muscle cells and have positive effects on cell migration. Blockers of the channels may be useful as inhibitors of neointimal hyperplasia and other unwanted vascular remodelling events.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Calcium/metabolism
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Ficusin/pharmacology
- Humans
- Hyperplasia
- Immunohistochemistry
- Kv1.3 Potassium Channel/antagonists & inhibitors
- Kv1.3 Potassium Channel/genetics
- Kv1.3 Potassium Channel/metabolism
- Male
- Membrane Potentials
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Organ Culture Techniques
- Patch-Clamp Techniques
- Potassium Channel Blockers/pharmacology
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Saphenous Vein/drug effects
- Saphenous Vein/metabolism
- Scorpion Venoms/pharmacology
- Time Factors
- Triterpenes/pharmacology
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Alex Cheong
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- Faculty of Biological Sciences, Institute of Membrane and Systems Biology, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Jing Li
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- Faculty of Biological Sciences, Institute of Membrane and Systems Biology, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Piruthivi Sukumar
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- Faculty of Biological Sciences, Institute of Membrane and Systems Biology, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Bhaskar Kumar
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- Faculty of Biological Sciences, Institute of Membrane and Systems Biology, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Fanning Zeng
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- Faculty of Biological Sciences, Institute of Membrane and Systems Biology, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Kirsten Riches
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Christopher Munsch
- Department of Cardiac Surgery, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | - Ian C. Wood
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- Faculty of Biological Sciences, Institute of Membrane and Systems Biology, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| | - Karen E. Porter
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - David J. Beech
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK
- Faculty of Biological Sciences, Institute of Membrane and Systems Biology, Garstang Building, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
68
|
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]
|
69
|
Felipe A, Soler C, Comes N. Kv1.5 in the immune system: the good, the bad, or the ugly? Front Physiol 2010; 1:152. [PMID: 21423392 PMCID: PMC3059964 DOI: 10.3389/fphys.2010.00152] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 10/28/2010] [Indexed: 11/13/2022] Open
Abstract
For the last 20 years, knowledge of the physiological role of voltage-dependent potassium channels (Kv) in the immune system has grown exponentially. Leukocytes express a limited repertoire of Kv channels, which contribute to the membrane potential. These proteins are involved in the immune response and are therefore considered good pharmacological targets. Although there is a clear consensus about the physiological relevance of Kv1.3, the expression and the role of Kv1.5 are controversial. However, recent reports indicate that certain heteromeric Kv1.3/Kv1.5 associations may provide insight on Kv1.5. Here, we summarize what is known about this issue and highlight the role of Kv1.5 partnership interactions that could be responsible for this debate. The Kv1.3/Kv1.5 heterotetrameric composition of the channel and their possible differential associations with accessory regulatory proteins warrant further investigation.
Collapse
Affiliation(s)
- Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona Barcelona, Spain.
| | | | | |
Collapse
|
70
|
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 PMCID: PMC2981387 DOI: 10.1124/mol.110.064014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 06/30/2010] [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
|
71
|
Hyodo T, Oda T, Kikuchi Y, Higashi K, Kushiyama T, Yamamoto K, Yamada M, Suzuki S, Hokari R, Kinoshita M, Seki S, Fujinaka H, Yamamoto T, Miura S, Kumagai H. Voltage-gated potassium channel Kv1.3 blocker as a potential treatment for rat anti-glomerular basement membrane glomerulonephritis. Am J Physiol Renal Physiol 2010; 299:F1258-69. [PMID: 20810612 DOI: 10.1152/ajprenal.00374.2010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The voltage-gated potassium channel Kv1.3 has been recently identified as a molecular target that allows the selective pharmacological suppression of effector memory T cells (T(EM)) without affecting the function of naïve T cells (T(N)) and central memory T cells (T(CM)). We found that Kv1.3 was expressed on glomeruli and some tubules in rats with anti-glomerular basement membrane glomerulonephritis (anti-GBM GN). A flow cytometry analysis using kidney cells revealed that most of the CD4(+) T cells and some of the CD8(+) T cells had the T(EM) phenotype (CD45RC(-)CD62L(-)). Double immunofluorescence staining using mononuclear cell suspensions isolated from anti-GBM GN kidney showed that Kv1.3 was expressed on T cells and some macrophages. We therefore investigated whether the Kv1.3 blocker Psora-4 can be used to treat anti-GBM GN. Rats that had been given an injection of rabbit anti-rat GBM antibody were also injected with Psora-4 or the vehicle intraperitoneally. Rats given Psora-4 showed less proteinuria and fewer crescentic glomeruli than rats given the vehicle. These results suggest that T(EM) and some macrophages expressing Kv1.3 channels play a critical role in the pathogenesis of crescentic GN and that Psora-4 will be useful for the treatment of rapidly progressive glomerulonephritis.
Collapse
Affiliation(s)
- Toshitake Hyodo
- Dept. of Nephrology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
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: 18] [Impact Index Per Article: 1.2] [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
|
73
|
RNA editing modulates the binding of drugs and highly unsaturated fatty acids to the open pore of Kv potassium channels. EMBO J 2010; 29:2101-13. [PMID: 20461057 DOI: 10.1038/emboj.2010.88] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 04/14/2010] [Indexed: 12/23/2022] Open
Abstract
The time course of inactivation of voltage-activated potassium (Kv) channels is an important determinant of the firing rate of neurons. In many Kv channels highly unsaturated lipids as arachidonic acid, docosahexaenoic acid and anandamide can induce fast inactivation. We found that these lipids interact with hydrophobic residues lining the inner cavity of the pore. We analysed the effects of these lipids on Kv1.1 current kinetics and their competition with intracellular tetraethylammonium and Kvbeta subunits. Our data suggest that inactivation most likely represents occlusion of the permeation pathway, similar to drugs that produce 'open-channel block'. Open-channel block by drugs and lipids was strongly reduced in Kv1.1 channels whose amino acid sequence was altered by RNA editing in the pore cavity, and in Kv1.x heteromeric channels containing edited Kv1.1 subunits. We show that differential editing of Kv1.1 channels in different regions of the brain can profoundly alter the pharmacology of Kv1.x channels. Our findings provide a mechanistic understanding of lipid-induced inactivation and establish RNA editing as a mechanism to induce drug and lipid resistance in Kv channels.
Collapse
|
74
|
Varga Z, Hajdu P, Panyi G. Ion channels in T lymphocytes: An update on facts, mechanisms and therapeutic targeting in autoimmune diseases. Immunol Lett 2010; 130:19-25. [DOI: 10.1016/j.imlet.2009.12.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 12/08/2009] [Accepted: 12/10/2009] [Indexed: 12/31/2022]
|
75
|
Rangaraju S, Chi V, Pennington MW, Chandy KG. Kv1.3 potassium channels as a therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2010; 13:909-24. [PMID: 19538097 DOI: 10.1517/14728220903018957] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We discuss the potential use of inhibitors of Kv1.3 potassium channels in T lymphocytes as therapeutics for multiple sclerosis. Current treatment strategies target the immune system in a non-selective manner. The resulting general immunosuppression, toxic side-effects and increased risk of opportunistic infections create the need for more selective therapeutics. Autoreactive effector-memory T (T(EM)) cells, considered to be major mediators of autoimmunity, express large numbers of Kv1.3 channels. Selective blockers of Kv1.3 inhibit calcium signaling, cytokine production and proliferation of T(EM) cells in vitro, and T(EM) cell-motility in vivo. Kv1.3 blockers ameliorate disease in animal models of multiple sclerosis, rheumatoid arthritis, type 1 diabetes mellitus and contact dermatitis without compromising the protective immune response to acute infections. Kv1.3 blockers have a good safety profile in rodents and primates.
Collapse
Affiliation(s)
- Srikant Rangaraju
- University of California, Department of Physiology and Biophysics, Irvine, California 92697, USA
| | | | | | | |
Collapse
|
76
|
Liu K, Samuel M, Tillett J, Hennan JK, Mekonnen B, Soloveva V, Harrison RK, Paslay JW, Larocque J. High-Throughput Screening for Kv1.3 Channel Blockers Using an Improved FLIPR-Based Membrane-Potential Assay. ACTA ACUST UNITED AC 2009; 15:185-95. [DOI: 10.1177/1087057109356209] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Voltage-gated K+ channels are potential drug targets for an increasing number of disease indications. Searching for compounds that modulate K+ channel activities by high-throughput screening (HTS) is becoming a standard approach in the drug discovery effort. Here the authors report an improved fluorometric imaging plate reader (FLIPR) membrane potential assay for Kv1.3 K+ channel HTS. They have found that the Chinese hamster ovary (CHO) cells have endogenous membrane electrogenic transporters that contribute to maintaining membrane potential. Blocking the recombinant K+ channels in the overexpressing CHO cell line hardly changed the membrane potential. Inhibition of the endogenous transporters is essential to achieve the required assay robustness. The authors identified the optimal assay conditions and designed a simple assay format. After an HTS campaign using this assay, various chemical series of Kv1.3 channel blockers have been identified and confirmed by the automated electrophysiological IonWorks assay. The correlation in dose response between FLIPR and IonWorks was established by biophysical modeling and experimental data. After characterization using patch-clamp recording, both use-dependent and use-independent compounds were identified. Some compounds possess nanomolar potency, indicating that the FLIPR assay is effective for successfully identifying K+ channel blockers as novel drug candidates.
Collapse
Affiliation(s)
- Kun Liu
- Department of Screening Sciences
| | | | - Jeff Tillett
- Department of Cardiovascular and Metabolic Diseases
| | | | - Belew Mekonnen
- Department of Medicinal Chemistry, Wyeth Research, Collegeville, Pennsylvania
| | | | | | | | | |
Collapse
|
77
|
Viola G, Salvador A, Vedaldi D, Dall'Acqua F, Bianchi N, Zuccato C, Borgatti M, Lampronti I, Gambari R. Differentiation and apoptosis in UVA-irradiated cells treated with furocoumarin derivatives. Ann N Y Acad Sci 2009; 1171:334-44. [PMID: 19723073 DOI: 10.1111/j.1749-6632.2009.04894.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this review we summarize the structure and biological effects of linear and angular psoralens. These compounds exhibit interesting biological effects on the cell cycle, apoptosis and differentiation. These molecules should be considered promising drugs in the therapy of several diseases, including psoriasis, mycosis fungoides and cancer. Also, preclinical data demonstrate a possible use of these molecules for the treatment of beta-thalassemia and other hematological disorders.
Collapse
Affiliation(s)
- Giampietro Viola
- Department of Pharmaceutical Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
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 2009; 44:1838-52. [PMID: 19056148 PMCID: PMC2662044 DOI: 10.1016/j.ejmech.2008.10.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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 & Biomedical Sciences Facility, 451 Health Sciences Drive, Davis CA 95616, USA
| | - Cédrick Mahieux
- Department of Pharmacology, University of California, Davis, Genome & Biomedical Sciences Facility, 451 Health Sciences Drive, Davis CA 95616, USA
| | - Wolfram Hänsel
- Institute of Pharmaceutical Chemistry, University of Kiel, Gutenbergstr. 76, 24118 Kiel, Germany
| | - Heike Wulff
- Department of Pharmacology, University of California, Davis, Genome & Biomedical Sciences Facility, 451 Health Sciences Drive, Davis CA 95616, USA
| |
Collapse
|
79
|
Tobin AA, Joseph BK, Al-Kindi HN, Albarwani S, Madden JA, Nemetz LT, Rusch NJ, Rhee SW. Loss of cerebrovascular Shaker-type K(+) channels: a shared vasodilator defect of genetic and renal hypertensive rats. Am J Physiol Heart Circ Physiol 2009; 297:H293-303. [PMID: 19411284 DOI: 10.1152/ajpheart.00991.2008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cerebral arteries of hypertensive rats are depolarized and highly myogenic, suggesting a loss of K(+) channels in the vascular smooth muscle cells (VSMCs). The present study evaluated whether the dilator function of the prominent Shaker-type voltage-gated K(+) (K(V)1) channels is attenuated in middle cerebral arteries from two rat models of hypertension. Block of K(V)1 channels by correolide (1 micromol/l) or psora-4 (100 nmol/l) reduced the resting diameter of pressurized (80 mmHg) cerebral arteries from normotensive rats by an average of 28 +/- 3% or 26 +/- 3%, respectively. In contrast, arteries from spontaneously hypertensive rats (SHR) and aortic-banded (Ao-B) rats with chronic hypertension showed enhanced Ca(2+)-dependent tone and failed to significantly constrict to correolide or psora-4, implying a loss of K(V)1 channel-mediated vasodilation. Patch-clamp studies in the VSMCs of SHR confirmed that the peak K(+) current density attributed to K(V)1 channels averaged only 5.47 +/- 1.03 pA/pF, compared with 9.58 +/- 0.82 pA/pF in VSMCs of control Wistar-Kyoto rats. Subsequently, Western blots revealed a 49 +/- 7% to 66 +/- 7% loss of the pore-forming alpha(1.2)- and alpha(1.5)-subunits that compose K(V)1 channels in cerebral arteries of SHR and Ao-B rats compared with control animals. In each case, the deficiency of K(V)1 channels was associated with reduced mRNA levels encoding either or both alpha-subunits. Collectively, these findings demonstrate that a deficit of alpha(1.2)- and alpha(1.5)-subunits results in a reduced contribution of K(V)1 channels to the resting diameters of cerebral arteries from two rat models of hypertension that originate from different etiologies.
Collapse
Affiliation(s)
- Ann A Tobin
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Effect of psoralen on the cloned Kv3.1 currents. Arch Pharm Res 2009; 32:407-12. [PMID: 19387585 DOI: 10.1007/s12272-009-1314-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 03/04/2009] [Accepted: 03/06/2009] [Indexed: 10/20/2022]
Abstract
The psoralen, a furocoumarin derivative, on the cloned neuronal rat Kv3.1 channels stably expressed in Chinese hamster ovary cells was investigated using the whole-cell patch-clamp technique. Psoralen reduced Kv3.1 whole-cell currents in a reversible concentration-dependent manner, with an IC50 value and a Hill coefficient of 2.3 +/- 0.03 microM and 0.9 +/- 0.08, respectively. Psoralen accelerated the decay rate of inactivation of Kv3.1 currents without modifying the kinetics of current activation. The psoralen-induced inhibition of Kv3.1 channels was voltage-dependent, with a steep increase over the voltage range of channel opening. However, the inhibition exhibited voltage independence over the voltage range in which channels are fully activated. Psoralen slowed the deactivation time course, resulting in a tail crossover phenomenon when the tail currents, recorded in the presence and absence of psoralen, were superimposed. Inhibition of Kv3.1 by psoralen was use-dependent at a frequency of 1 Hz. The present results suggest that psoralen acts on Kv3.1 currents as an open-channel blocker.
Collapse
|
81
|
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: 43] [Impact Index Per Article: 2.7] [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
|
82
|
Bao K, Fan A, Dai Y, Zhang L, Zhang W, Cheng M, Yao X. Selective demethylation and debenzylation of aryl ethers by magnesium iodide under solvent-free conditions and its application to the total synthesis of natural products. Org Biomol Chem 2009; 7:5084-90. [DOI: 10.1039/b916969e] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
83
|
Ren YR, Pan F, Parvez S, Fleig A, Chong CR, Xu J, Dang Y, Zhang J, Jiang H, Penner R, Liu JO. Clofazimine inhibits human Kv1.3 potassium channel by perturbing calcium oscillation in T lymphocytes. PLoS One 2008; 3:e4009. [PMID: 19104661 PMCID: PMC2602975 DOI: 10.1371/journal.pone.0004009] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Accepted: 11/08/2008] [Indexed: 01/03/2023] Open
Abstract
The Kv1.3 potassium channel plays an essential role in effector memory T cells and has been implicated in several important autoimmune diseases including multiple sclerosis, psoriasis and type 1 diabetes. A number of potent small molecule inhibitors of Kv1.3 channel have been reported, some of which were found to be effective in various animal models of autoimmune diseases. We report herein the identification of clofazimine, a known anti-mycobacterial drug, as a novel inhibitor of human Kv1.3. Clofazimine was initially identified as an inhibitor of intracellular T cell receptor-mediated signaling leading to the transcriptional activation of human interleukin-2 gene in T cells from a screen of the Johns Hopkins Drug Library. A systematic mechanistic deconvolution revealed that clofazimine selectively blocked the Kv1.3 channel activity, perturbing the oscillation frequency of the calcium-release activated calcium channel, which in turn led to the inhibition of the calcineurin-NFAT signaling pathway. These effects of clofazimine provide the first line of experimental evidence in support of a causal relationship between Kv1.3 and calcium oscillation in human T cells. Furthermore, clofazimine was found to be effective in blocking human T cell-mediated skin graft rejection in an animal model in vivo. Together, these results suggest that clofazimine is a promising immunomodulatory drug candidate for treating a variety of autoimmune disorders.
Collapse
Affiliation(s)
- Yunzhao R. Ren
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Program in Biochemistry, Cellular and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Fan Pan
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Suhel Parvez
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Curtis R. Chong
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jing Xu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yongjun Dang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jin Zhang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hongsi Jiang
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, Illinois, United States of America
| | - Reinhold Penner
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine at the University of Hawaii, Honolulu, Hawaii, United States of America
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Program in Biochemistry, Cellular and Molecular Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
84
|
Mitochondrial potassium channel Kv1.3 mediates Bax-induced apoptosis in lymphocytes. Proc Natl Acad Sci U S A 2008; 105:14861-6. [PMID: 18818304 DOI: 10.1073/pnas.0804236105] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The potassium channel Kv1.3 has recently been located to the inner mitochondrial membrane of lymphocytes. Here, we show that mouse and human cells either genetically deficient in Kv1.3 or transfected with siRNA to suppress Kv1.3-expression resisted apoptosis induced by several stimuli, including Bax over-expression [corrected]. Retransfection of either Kv1.3 or a mitochondrial-targeted Kv1.3 restored cell death . Bax interacted with and functionally inhibited mitochondrial Kv1.3. Incubation of isolated Kv1.3-positive mitochondria with recombinant Bax, t-Bid, or toxins that bind to and inhibit Kv1.3 successively triggered hyperpolarization, formation of reactive oxygen species, release of cytochrome c, and marked depolarization. Kv1.3-deficient mitochondria were resistant to Bax, t-Bid, and the toxins. Mutation of Bax at K128, which corresponds to a conserved lysine in Kv1.3-inhibiting toxins, abrogated its effects on both Kv1.3 and mitochondria. These findings suggest that Bax mediates cytochrome c release and mitochondrial depolarization in lymphocytes, at least in part, via its interaction with mitochondrial Kv1.3.
Collapse
|
85
|
Stokely ME, Garg P, Bhat MA, Koulen P. Transient 5-(4-phenylbutoxy)psoralen (PAP-1) treatment dissociates developing pathologies in autoimmune optic neuritis into two distinct pathology profiles. J Neurosci Res 2008; 86:2111-24. [PMID: 18335521 DOI: 10.1002/jnr.21645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Discovery of treatments to protect axonal function of neurons and prevent permanent disability associated with progressive multiple sclerosis (MS) has faced the uphill challenge of assessing relatively small changes in accumulated axon damage within a background environment that is disorganized by CNS inflammation. We hypothesized that transient immunosuppression after initiation of MS-like autoimmune mechanisms would disassociate development of MS-like myelinated axon pathology from development of CNS inflammation in a rat model of autoimmune optic neuritis (AON). A rat model of myelin oligodendrocyte glycoprotein peptide-induced AON was transiently treated (on days 3-7 after antigen exposure) with 5-(4-phenylbutoxy)psoralen (PAP-1), an immunomodulatory drug previously shown specifically to suppress proliferation of effector memory T-cells and immunoglobulin class-switched B-cells. Thirteen days after antigen exposure, optic nerves were harvested for quantitative assessment of 12 MS-associated pathologies using microfluorimetry. With one exception, the immunoreactivities (-ir) for eight markers of MS-like neuroinflammation and immune infiltration were significantly reduced (P < 0.05) by transient PAP-1 treatment, often to levels significantly below those detected in normal control rat optic nerves. With one exception, four immunoreactive markers of MS-like myelinated axon pathology were detected at levels indicating increased axon/myelin pathology compared with vehicle-treated rats with AON (P < 0.05). These data suggest the conclusion that early causative mechanisms in CNS autoimmunity initiate signaling mechanisms that diverge into two separate pathways, one that is strongly associated with inflammatory responses and one that is associated predominantly with disturbed axon-myelin interactions and impaired fast axonal transport.
Collapse
Affiliation(s)
- Martha E Stokely
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas 76107-2699, USA
| | | | | | | |
Collapse
|
86
|
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: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA.
| | | |
Collapse
|
87
|
OdK2, a Kv1.3 channel-selective toxin from the venom of the Iranian scorpion Odonthobuthus doriae. Toxicon 2008; 51:1424-30. [PMID: 18471844 DOI: 10.1016/j.toxicon.2008.03.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Revised: 03/17/2008] [Accepted: 03/18/2008] [Indexed: 01/06/2023]
Abstract
The first Kv1.3 channel-selective toxin from the venom of the Iranian scorpion Odonthobuthus doriae (OdK2) was purified, sequenced and characterized physiologically. OdK2 consists of 38 amino acids, including six conserved cysteine and a C-terminal lysine residue, as revealed by the unique use of a quadrupole ion cyclotron resonance Fourier-transform mass spectrometer. Based on multiple sequence alignments, OdK2 was classified as alpha-KTX3.11. The pharmacological effects of OdK2 were studied on a panel of eight different cloned K(+) channels (vertebrate Kv1.1-Kv1.6, Shaker IR and hERG) expressed in Xenopus laevis oocytes. Interestingly, OdK2 selectively inhibits the currents through Kv1.3 channels with an IC50 value of 7.2+/-2.7nM.
Collapse
|
88
|
Meuth SG, Bittner S, Meuth P, Simon OJ, Budde T, Wiendl H. TWIK-related acid-sensitive K+ channel 1 (TASK1) and TASK3 critically influence T lymphocyte effector functions. J Biol Chem 2008; 283:14559-70. [PMID: 18375952 DOI: 10.1074/jbc.m800637200] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Two major K(+) channels are expressed in T cells, (i) the voltage-dependent K(V)1.3 channel and (ii) the Ca(2+)-activated K(+) channel KCa 3.1 (IKCa channel). Both critically influence T cell effector functions in vitro and animal models in vivo. Here we identify and characterize TWIK-related acid-sensitive potassium channel 1 (TASK1) and TASK3 as an important third K(+) conductance on T lymphocytes. T lymphocytes constitutively express TASK1 and -3 protein. Application of semi-selective TASK blockers resulted in a significant reduction of cytokine production and cell proliferation. Interference with TASK channels on CD3(+) T cells revealed a dose-dependent reduction ( approximately 40%) of an outward current in patch clamp recordings indicative of TASK channels, a finding confirmed by computational modeling. In vivo relevance of our findings was addressed in an experimental model of multiple sclerosis, adoptive transfer experimental autoimmune encephalomyelitis. Pretreatment of myelin basic protein-specific encephalitogenic T lymphocytes with TASK modulators was associated with significant amelioration of the disease course in Lewis rats. These data introduce K(2)P channels as novel potassium conductance on T lymphocytes critically influencing T cell effector function and identify a possible molecular target for immunomodulation in T cell-mediated autoimmune disorders.
Collapse
Affiliation(s)
- Sven G Meuth
- Department of Neurology, University of Wuerzburg, Wuerzburg, Germany.
| | | | | | | | | | | |
Collapse
|
89
|
Kleinschnitz C, Meuth SG, Kieseier BC, Wiendl H. [Update on pathophysiologic and immunotherapeutic approaches for the treatment of multiple sclerosis]. DER NERVENARZT 2007; 78:883-911. [PMID: 17551708 DOI: 10.1007/s00115-007-2261-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis (MS) is a chronic disabling disease with significant implications for patients and society. The individual disease course is difficult to predict due to the heterogeneity of clinical presentation and of radiologic and pathologic findings. Although its etiology still remains unknown, the last decade has brought considerable understanding of the underlying pathophysiology of MS. In addition to its acceptance as a prototypic inflammatory autoimmune disorder, recent data reveal the importance of primary and secondary neurodegenerative mechanisms such as oligodendrocyte death, axonal loss, and ion channel dysfunction. The deepened understanding of its immunopathogenesis and the limited effectiveness of currently approved disease-modifying therapies have led to a tremendous number of trials investigating potential new drugs. Emerging treatments take into account the different immunopathological mechanisms and strategies, to protect against axonal damage and promote remyelination. This review provides a compilation of novel immunotherapeutic strategies and recently uncovered aspects of known immunotherapeutic agents. The pathogenetic rationale of these novel drugs for the treatment of MS and accompanying preclinical and clinical data are highlighted.
Collapse
Affiliation(s)
- C Kleinschnitz
- Neurologische Klinik und Poliklinik, Universitätsklinikum, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany
| | | | | | | |
Collapse
|
90
|
Eun JS, Kim KS, Kim HN, Park SA, Ma TZ, Lee KA, Kim DK, Kim HK, Kim IS, Jung YH, Zee OP, Yoo DJ, Kwak YG. Synthesis of psoralen derivatives and their blocking effect of hKv1.5 channel. Arch Pharm Res 2007; 30:155-60. [PMID: 17366735 DOI: 10.1007/bf02977688] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Previously, we found that a furocoumarin derivative, psoralen (7H-furo[3,2-g][1]benzopyran-7-one), blocked a human Kv1.5 potassium channel (hKv1.5) and has a potential antiarrhythmic effect. In the present study, to develop more potent hKv1.5 blockers or antiarrhythmic drugs, we synthesized ten psoralen derivatives and examined their blocking effects on hKv1.5 stably expressed in Ltk cells. Among the newly synthesized psoralen derivatives, three derivatives (Compounds 5, 9 and 10) showed the open channel-blocking effect. Compound 9 among them was the most potent in blocking hKv1.5. We found that compound 9, one of the psoralen derivatives, inhibited the hKv1.5 current in a concentration-, use- and voltage-dependent manner with an IC50 value of 27.4 +/- 5.1 nM at +60 mV. Compound 9 accelerated the inactivation kinetics of the hKv1.5 channel, slowed the deactivation kinetics of hKv1.5 current resulting in a tail crossover phenomenon. Compound 9 inhibited hKv1.5 current in a use-dependent manner. These results indicate that compound 9, one of psoralen derivatives, acts on hKv1.5 channel as an open channel blocker and is much more potent than psoralen in blocking hKv1.5 channel. If further studies were done, compound 9 might be an ideal antiarrhythmic drug for atrial fibrillation.
Collapse
Affiliation(s)
- Jae Soon Eun
- College of Pharmacy, Woosuk University, Samrye 565-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Liebau S, Pröpper C, Böckers T, Lehmann-Horn F, Storch A, Grissmer S, Wittekindt OH. Selective blockage of Kv1.3 and Kv3.1 channels increases neural progenitor cell proliferation. J Neurochem 2007; 99:426-37. [PMID: 17029597 DOI: 10.1111/j.1471-4159.2006.03967.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The modulation of cell proliferation in neural progenitor cells (NPCs) is believed to play a role in neuronal regeneration. Recent studies showed that K(+) channel activity influenced cell proliferation. Therefore, we examined NPCs for K(+) channels and tested whether NPC self renewing can be modulated by synthetic K(+) channel modulators. The whole-cell K(+) current was partly K(+) dependent and showed a cumulative inactivating component. Two tetra-ethyl-ammonium ion (TEA)-sensitive K(+) currents with different voltage dependencies ( = 65 microm, E(50) = -0.3 +/- 1.3 mV and = 8 mm, E(50) = -15.2 +/- 2.8 mV) and an almost TEA-insensitive current were identified. Kaliotoxin blocked approximately 50% of the entire K(+) currents (IC(50) = 0.25 nm). These properties resembled functional characteristics of K(v)1.4, K(v)1.3 and K(v)3.1 channels. Transcripts for these channels, as well as proteins for K(v)1.3 and K(v)3.1, were identified. Immunocytochemical staining revealed K(v)1.3 and K(v)3.1 K(+) channel expression in almost all NPCs. The blockage of K(v)3.1 by low concentrations of TEA, as well as the blockage of K(v)1.3 by Psora-4, increased NPC proliferation. These findings underline the regulatory role of K(+) channels on the cell cycle and imply that K(+) channel modulators might be used therapeutically to activate endogenous NPCs.
Collapse
Affiliation(s)
- Stefan Liebau
- Department of Anatomy and Cell Biology, University of Ulm, Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
92
|
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: 21] [Impact Index Per Article: 1.1] [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
|
93
|
Sauviat MP, Vercauteren J, Grimaud N, Jugé M, Nabil M, Petit JY, Biard JF. Sensitivity of cardiac background inward rectifying K+ outward current (IK1) to the alkaloids lepadiformines A, B, and C. JOURNAL OF NATURAL PRODUCTS 2006; 69:558-62. [PMID: 16643025 DOI: 10.1021/np050215s] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Three marine alkaloids, purified from Clavelina moluccensis, were structurally identified as lepadiformines A, B, and C and studied on frog atrial myocytes I(K1), using the patch-clamp technique. Lepadiformine A (0.4 to 3.3 microM) blocked I(K1) dose-dependently with an apparent dissociation constant (K(D)) equal to 1.42 microM and a stoichiometry of 0.77. The block is voltage-dependent, suggesting that lepadiformine A occupies a receptor site located at about two-thirds of the membrane depth. The shortening of the aliphatic chain at position C13 of lepadiformine B decreased the potency of the molecule to block I(K1) but not the affinity (K(D) = 1.56 microM) and stoichiometry (0.72). Additional deletion of the oxygenated side chain at C2 in lepadiformine C markedly decreased the inhibitory effect of the molecule. In conclusion, lepadiformine modulates I(K1) response in cardiac muscle. The oxygenated side chain in C2 is implicated in the affinity of lepadiformine, which behaved as an amine, for a receptor located near or inside the I(K1) pore, and the aliphatic chain length at position C13 is involved in the degree of I(K1) blockage.
Collapse
Affiliation(s)
- Martin-Pierre Sauviat
- Laboratoire d'Optique et Biosciences, INSERM U 696, CNRS UMR 7645, X, Ecole Polytechnique, 91128 Palaiseau Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
94
|
Li Y, Wang P, Xu J, Desir GV. Voltage-gated potassium channel Kv1.3 regulates GLUT4 trafficking to the plasma membrane via a Ca2+-dependent mechanism. Am J Physiol Cell Physiol 2006; 290:C345-51. [PMID: 16403947 DOI: 10.1152/ajpcell.00091.2005] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Kv1.3 is a voltage-gated K(+) channel expressed in insulin-sensitive tissues. We previously showed that gene inactivation or pharmacological inhibition of Kv1.3 channel activity increased peripheral insulin sensitivity independently of body weight by augmenting the amount of GLUT4 at the plasma membrane. In the present study, we further examined the effect Kv1.3 on GLUT4 trafficking and tested whether it occurred via an insulin-dependent pathway. We found that Kv1.3 inhibition by margatoxin (MgTX) stimulated glucose uptake in adipose tissue and skeletal muscle and that the effect of MgTX on glucose transport was additive to that of insulin. Furthermore, whereas the increase in uptake was wortmannin insensitive, it was completely inhibited by dantrolene, a blocker of Ca(2+) release from intracellular Ca(2+) stores. In white adipocytes in primary culture, channel inhibition by Psora-4 increased GLUT4 translocation to the plasma membrane. In these cells, GLUT4 protein translocation was unaffected by the addition of wortmannin but was significantly inhibited by dantrolene. Channel inhibition depolarized the membrane voltage and led to sustained, dantrolene-sensitive oscillations in intracellular Ca(2+) concentration. These results indicate that the apparent increase in insulin sensitivity observed in association with inhibition of Kv1.3 channel activity is mediated by an increase in GLUT4 protein at the plasma membrane, which occurs largely through a Ca(2+)-dependent process.
Collapse
Affiliation(s)
- Yanyan Li
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520-8029, USA
| | | | | | | |
Collapse
|
95
|
Abstract
Multiple sclerosis is a chronic inflammatory autoimmune disease of the central nervous system characterized by demyelination and axonal damage that result in disabling neurological deficits. Here the authors explain the rationale for the use of inhibitors of the Kv1.3 K+ channel in immune cells as a therapy for multiple sclerosis and other autoimmune disorders.
Collapse
Affiliation(s)
- Christine Beeton
- Department of Physiology and Biophysics, Medical School, University of California, Irvine, 92697, USA
| | | |
Collapse
|
96
|
Abstract
The development of our knowledge of the function, structure and pharmacology of K(+) channels is briefly outlined. This is the most diverse of all the ion channel families with at least 75 coding genes in mammals. Alternative splicing as well as variations in the channel subunits and accessory proteins that co-assemble to form the functional channel add to the multiplicity. Whereas diversity of this order suggests that it may be possible to develop new classes of drug, for example, for immunomodulation and some diseases of the central nervous system, the ubiquity of K(+) channels imposes stringent requirements for selectivity. Animal toxins from the snake, bee and scorpion provide useful leads, though only in a few instances (e.g. with apamin) it has been possible to produce non-peptidic analogues of high potency. The scale of the resources needed to identify, and characterize fully, specific K(+) channel as targets and then develop modulators with the required selectivity presents a challenge to both academic and applied pharmacologists.
Collapse
Affiliation(s)
- Donald H Jenkinson
- Department of Pharmacology, University College London, Gower St., London WC1E 6BT.
| |
Collapse
|
97
|
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
|
98
|
Schmitz A, Sankaranarayanan A, Azam P, Schmidt-Lassen K, Homerick D, Hänsel W, Wulff H. Design of PAP-1, a selective small molecule Kv1.3 blocker, for the suppression of effector memory T cells in autoimmune diseases. Mol Pharmacol 2005; 68:1254-70. [PMID: 16099841 DOI: 10.1124/mol.105.015669] [Citation(s) in RCA: 182] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The lymphocyte K+ channel Kv1.3 constitutes an attractive pharmacological target for the selective suppression of terminally differentiated effector memory T (TEM) cells in T cell-mediated autoimmune diseases, such as multiple sclerosis and type 1 diabetes. Unfortunately, none of the existing small-molecule Kv1.3 blockers is selective, and many of them, such as correolide, 4-phenyl-4-[3-(methoxyphenyl)-3-oxo-2-azapropyl]cyclohexanone, and our own compound Psora-4 inhibit the cardiac K+ channel Kv1.5. By further exploring the structure-activity relationship around Psora-4 through a combination of traditional medicinal chemistry and whole-cell patch-clamp, we identified a series of new phenoxyalkoxypsoralens that exhibit 2- to 50-fold selectivity for Kv1.3 over Kv1.5, depending on their exact substitution pattern. The most potent and "drug-like" compound of this series, 5-(4-phenoxybutoxy)psoralen (PAP-1), blocks Kv1.3 in a use-dependent manner, with a Hill coefficient of 2 and an EC50 of 2 nM, by preferentially binding to the C-type inactivated state of the channel. PAP-1 is 23-fold selective over Kv1.5, 33- to 125-fold selective over other Kv1-family channels, and 500- to 7500-fold selective over Kv2.1, Kv3.1, Kv3.2, Kv4.2, HERG, calcium-activated K+ channels, Na+,Ca2+, and Cl- channels. PAP-1 does not exhibit cytotoxic or phototoxic effects, is negative in the Ames test, and affects cytochrome P450-dependent enzymes only at micromolar concentrations. PAP-1 potently inhibits the proliferation of human TEM cells and suppresses delayed type hypersensitivity, a TEM cell-mediated reaction, in rats. PAP-1 and several of its derivatives therefore constitute excellent new tools to further explore Kv1.3 as a target for immunosuppression and could potentially be developed into orally available immunomodulators.
Collapse
Affiliation(s)
- Alexander Schmitz
- Department of Medical Pharmacology and Toxicology, Genome and Biomedical Sciences Facility, Room 3502, 451 East Health Sciences Drive, University of California, Davis, Davis, CA 95616, USA
| | | | | | | | | | | | | |
Collapse
|
99
|
Decher N, Kumar P, Gonzalez T, Renigunta V, Sanguinetti MC. Structural basis for competition between drug binding and Kvbeta 1.3 accessory subunit-induced N-type inactivation of Kv1.5 channels. Mol Pharmacol 2005; 68:995-1005. [PMID: 16024663 DOI: 10.1124/mol.105.011668] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Kvbeta subunits are accessory proteins that modify gating of Kv1 channels. Kvbeta1.3 subunits bind to the N termini of Kv1.5 alpha-subunits and induce fast N-type inactivation, slow the rate of deactivation, and alter the voltage dependence and kinetics of channel activation. The N terminus of a Kvbeta subunit and quaternary ammonium compounds bind to the inner pore of Kv1 channels; however, it is unknown to what extent the pore binding sites for drugs and Kvbeta subunits overlap. Here, we used site-directed Ala mutagenesis to scan residues of the Kv1.5 pore to define the binding site for Kvbeta1.3 subunits. Individual mutations of five residues in the S6 domain (Val505, Ile508, Leu510, Val512, and Val516) greatly retarded or prevented Kvbeta1.3 induced inactivation, and reduced effects on Kv1.5 deactivation. Mutation of Thr479 and Thr480 enhanced Kvbeta1.3-induced N-type inactivation. None of the Ala mutations prevented the Kvbeta1.3-induced negative shifts in the voltage dependence of activation or slow C-type inactivation, suggesting that these gating effects are mediated by an interaction other than the one for N-type inactivation. Thr479, Thr480, Val505, Ile508, and Val512, of Kv1.5 channels are also important interaction sites for the anthranilic acid S0100176 (N-benzyl-N-pyridin-3-ylmethyl-2-(toluene-4-sulfonylamino)-benzamide hydrochloride). Leu510 and V516A prevented Kvbeta1.3-induced inactivation but did not alter drug block. Block of Kv1.5 by S0100176 was reduced and voltage-dependent in the presence of Kvbeta1.3 but not in the presence of an N-truncated form of the Kvbeta subunit. Thus, residues in the pore of Kv1.5 required for N-type inactivation overlap with but are not identical to the drug binding site.
Collapse
Affiliation(s)
- Niels Decher
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | |
Collapse
|
100
|
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
|