1
|
Wong BHS, Shim H, Goay SSM, Ong ST, Muhammad Taib NAB, Chai KXY, Lim K, Huang D, Ong CK, Vaiyapuri TS, Cheah YC, Wang Y, Wulff H, Webster RD, Shelat VG, Verma NK. The novel quinoline derivative SKA-346 as a K Ca3.1 channel selective activator. RSC Adv 2024; 14:38364-38377. [PMID: 39635364 PMCID: PMC11615718 DOI: 10.1039/d4ra07330d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/17/2024] [Indexed: 12/07/2024] Open
Abstract
The calcium-activated KCa3.1 channel plays a crucial role in T-cell immune response. Genetic manipulation of T-cells to upregulate the expression of K+ channels has been shown to boost T-cell cytotoxicity in cancer. Here, we aimed to identify and characterize an activator that would augment KCa3.1 currents without affecting other channels. We synthesized five quinoline derivatives and used electrophysiology to screen them on KCa3.1 and a panel of 14 other ion channels. One quinoline derivative, SKA-346, activated KCa3.1 with an EC50 of 1.9 μM and showed selectivity against the other channels. In silico analysis using RosettaLigand and GLIDE demonstrated a well-converged pose of SKA-346 in a binding pocket at the interface between the calmodulin N-lobe and the S45A helix in the S4-S5 linker of the KCa3.1 channel. SKA-346 (30 mg kg-1), tolerated by mice after intra-peritoneal administration, exhibited a peak plasma concentration of 6.29 μg mL-1 (29.2 μM) at 15 min and a circulating half-life (t 1/2) of 2.8 h. SKA-346 could serve as a template for the development of more potent KCa3.1 activators to enhance T-cell cytotoxicity in cancer.
Collapse
Affiliation(s)
- Brandon Han Siang Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- Interdisciplinary Graduate Programme, NTU Institute for Health Technologies (HealthTech NTU), Nanyang Technological University Singapore Singapore
| | - Heesung Shim
- Physical and Life Sciences, Lawrence Livermore National Laboratory Livermore CA USA
| | - Stephanie Shee Min Goay
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Seow Theng Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- LKCMedicine-ICE Collaborative Platform, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Nur Ayuni Binte Muhammad Taib
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Kelila Xin Ye Chai
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Kerry Lim
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Dachuan Huang
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
- Duke-NUS Medical School Singapore
| | - Choon Kiat Ong
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore
- Duke-NUS Medical School Singapore
| | | | - Yeong Cheng Cheah
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Yulan Wang
- Singapore Phenome Center, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| | - Heike Wulff
- Department of Pharmacology, University of California Davis CA USA
| | - Richard D Webster
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University Singapore Singapore
| | - Vishalkumar G Shelat
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
- Department of General Surgery, Tan Tock Seng Hospital Singapore
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Singapore
| |
Collapse
|
2
|
Thi Hong Van N, Hyun Nam J. Intermediate conductance calcium-activated potassium channel (KCa3.1) in cancer: Emerging roles and therapeutic potentials. Biochem Pharmacol 2024; 230:116573. [PMID: 39396649 DOI: 10.1016/j.bcp.2024.116573] [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: 07/20/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
The KCa3.1 channel (also known as the KCNN4, IK1, or SK4 channel) is an intermediate-conductance calcium-activated potassium channel that regulates the membrane potential and maintains calcium homeostasis. Recently, KCa3.1 channels have attracted increasing attention because of their diverse roles in various types of cancers. In cancer cells, KCa3.1 channels regulate key processes, including cell proliferation, cell cycle, migration, invasion, tumor microenvironments, and therapy resistance. In addition, abnormal KCa3.1 expression in cancers is utilized to distinguish between tumor and normal tissues, classify cancer stages, and predict patient survival outcomes. This review comprehensively examines the current understanding of the contribution of KCa3.1 channels to tumor formation, metastasis, and its mechanisms. We evaluated the potential of KCa3.1 as a biomarker for cancer diagnosis and prognosis. Finally, we discuss the advances and challenges of applying KCa3.1 modulators in cancer treatment and propose approaches to overcome these obstacles. In summary, this review highlights the importance of this ion channel as a potent therapeutic target and prognostic biomarker of cancer.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea.
| |
Collapse
|
3
|
Chen C, Guo L, Shen Y, Hu J, Gu J, Ji G. Oxidative damage and cardiotoxicity induced by 2-aminobenzothiazole in zebrafish (Danio rerio). JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135032. [PMID: 38959826 DOI: 10.1016/j.jhazmat.2024.135032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
There is limited information available on cardiovascular toxicity of 2-Aminobenzothiazole (NTH), a derivative of benzothiazole (BTH) commonly used in tire production, in aquatic organisms. In the present study, the zebrafish embryos were exposed to varying concentrations of NTH (0, 0.05, 0.5, and 5 mg/L) until adulthood and the potential cardiovascular toxicity was assessed. NTH exposure resulted in striking aberrations in cardiac development, including heart looping failure and interference with atrioventricular canal differentiation. RNA-sequencing analysis indicated that NTH causes oxidative damage to the heart via ferroptosis, leading to oxygen supply disruption, cardiac malformation, and ultimately, zebrafish death. Quantitative real-time polymerase chain reaction (qPCR) analysis demonstrated the dysregulation of genes associated with early heart development, contraction, and oxidative stress. Additionally, reactive oxygen species accumulation and glutathione/malondialdehyde levels changes suggested a potential link between cardiac developmental toxicity and oxidative stress. In adult zebrafish, NTH exposure led to ventricular enlargement, decreased heart rate, reduced blood flow, and prolonged RR, QRS, and QTc intervals. To the best of our knowledge, this study is the first to provide evidence of cardiac toxicity and the adverse effects of ontogenetic NTH exposure in zebrafish, revealing the underlying toxic mechanisms connected with oxidative stress damage. These findings may provide crucial insights into the environmental risks associated with NTH and other BTHs.
Collapse
Affiliation(s)
- Chen Chen
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Liguo Guo
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Yuehong Shen
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Jun Hu
- School of Environmental Science and Engineering, Nanjing Tech University, Jiangsu 211816, China
| | - Jie Gu
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China.
| | - Guixiang Ji
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China.
| |
Collapse
|
4
|
Zhang Y, Shaabani S, Vowinkel K, Trombetta-Lima M, Sabogal-Guáqueta AM, Chen T, Hoekstra J, Lembeck J, Schmidt M, Decher N, Dömling A, Dolga AM. Novel SK channel positive modulators prevent ferroptosis and excitotoxicity in neuronal cells. Biomed Pharmacother 2024; 171:116163. [PMID: 38242037 DOI: 10.1016/j.biopha.2024.116163] [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: 10/23/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Small conductance calcium-activated potassium (SK) channel activity has been proposed to play a role in the pathology of several neurological diseases. Besides regulating plasma membrane excitability, SK channel activation provides neuroprotection against ferroptotic cell death by reducing mitochondrial Ca2+ uptake and reactive oxygen species (ROS). In this study, we employed a multifaceted approach, integrating structure-based and computational techniques, to strategically design and synthesize an innovative class of potent small-molecule SK2 channel modifiers through highly efficient multicomponent reactions (MCRs). The compounds' neuroprotective activity was compared with the well-studied SK positive modulator, CyPPA. Pharmacological SK channel activation by selected compounds confers neuroprotection against ferroptosis at low nanomolar ranges compared to CyPPA, that mediates protection at micromolar concentrations, as shown by an MTT assay, real-time cell impedance measurements and propidium iodide staining (PI). These novel compounds suppress increased mitochondrial ROS and Ca2+ level induced by ferroptosis inducer RSL3. Moreover, axonal degeneration was rescued by these novel SK channel activators in primary mouse neurons and they attenuated glutamate-induced neuronal excitability, as shown via microelectrode array. Meanwhile, functional afterhyperpolarization of the novel SK2 channel modulators was validated by electrophysiological measurements showing more current change induced by the novel modulators than the reference compound, CyPPA. These data support the notion that SK2 channel activation can represent a therapeutic target for brain diseases in which ferroptosis and excitotoxicity contribute to the pathology.
Collapse
Affiliation(s)
- Yuequ Zhang
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Shabnam Shaabani
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Kirsty Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technologies and Biopharmacy, Research Institute of Pharmacy, University of Groningen, the Netherlands
| | | | - Tingting Chen
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Hoekstra
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Lembeck
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| |
Collapse
|
5
|
Nam YW, Downey M, Rahman MA, Cui M, Zhang M. Channelopathy of small- and intermediate-conductance Ca 2+-activated K + channels. Acta Pharmacol Sin 2023; 44:259-267. [PMID: 35715699 PMCID: PMC9889811 DOI: 10.1038/s41401-022-00935-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Small- and intermediate-conductance Ca2+-activated K+ (KCa2.x/KCa3.1 also called SK/IK) channels are gated exclusively by intracellular Ca2+. The Ca2+ binding protein calmodulin confers sub-micromolar Ca2+ sensitivity to the channel-calmodulin complex. The calmodulin C-lobe is constitutively associated with the proximal C-terminus of the channel. Interactions between calmodulin N-lobe and the channel S4-S5 linker are Ca2+-dependent, which subsequently trigger conformational changes in the channel pore and open the gate. KCNN genes encode four subtypes, including KCNN1 for KCa2.1 (SK1), KCNN2 for KCa2.2 (SK2), KCNN3 for KCa2.3 (SK3), and KCNN4 for KCa3.1 (IK). The three KCa2.x channel subtypes are expressed in the central nervous system and the heart. The KCa3.1 subtype is expressed in the erythrocytes and the lymphocytes, among other peripheral tissues. The impact of dysfunctional KCa2.x/KCa3.1 channels on human health has not been well documented. Human loss-of-function KCa2.2 mutations have been linked with neurodevelopmental disorders. Human gain-of-function mutations that increase the apparent Ca2+ sensitivity of KCa2.3 and KCa3.1 channels have been associated with Zimmermann-Laband syndrome and hereditary xerocytosis, respectively. This review article discusses the physiological significance of KCa2.x/KCa3.1 channels, the pathophysiology of the diseases linked with KCa2.x/KCa3.1 mutations, the structure-function relationship of the mutant KCa2.x/KCa3.1 channels, and potential pharmacological therapeutics for the KCa2.x/KCa3.1 channelopathy.
Collapse
Affiliation(s)
- Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Myles Downey
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University School of Pharmacy, Boston, MA, 02115, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA.
| |
Collapse
|
6
|
Wrzosek A, Gałecka S, Żochowska M, Olszewska A, Kulawiak B. Alternative Targets for Modulators of Mitochondrial Potassium Channels. Molecules 2022; 27:299. [PMID: 35011530 PMCID: PMC8746388 DOI: 10.3390/molecules27010299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial potassium channels control potassium influx into the mitochondrial matrix and thus regulate mitochondrial membrane potential, volume, respiration, and synthesis of reactive oxygen species (ROS). It has been found that pharmacological activation of mitochondrial potassium channels during ischemia/reperfusion (I/R) injury activates cytoprotective mechanisms resulting in increased cell survival. In cancer cells, the inhibition of these channels leads to increased cell death. Therefore, mitochondrial potassium channels are intriguing targets for the development of new pharmacological strategies. In most cases, however, the substances that modulate the mitochondrial potassium channels have a few alternative targets in the cell. This may result in unexpected or unwanted effects induced by these compounds. In our review, we briefly present the various classes of mitochondrial potassium (mitoK) channels and describe the chemical compounds that modulate their activity. We also describe examples of the multidirectional activity of the activators and inhibitors of mitochondrial potassium channels.
Collapse
Affiliation(s)
- Antoni Wrzosek
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| | - Shur Gałecka
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| | - Monika Żochowska
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| | - Anna Olszewska
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland;
| | - Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| |
Collapse
|
7
|
Abstract
Potassium channels are the most diverse and ubiquitous family of ion channels found in cells. The Ca2+ and voltage gated members form a subfamily that play a variety of roles in both excitable and non-excitable cells and are further classified on the basis of their single channel conductance to form the small conductance (SK), intermediate conductance (IK) and big conductance (BK) K+ channels.In this chapter, we will focus on the mechanisms underlying the gating of BK channels, whose function is modified in different tissues by different splice variants as well as the expanding array of regulatory accessory subunits including β, γ and LINGO subunits. We will examine how BK channels are modified by these regulatory subunits and describe how the channel gating is altered by voltage and Ca2+ whilst setting this in context with the recently published structures of the BK channel. Finally, we will discuss how BK and other calcium-activated channels are modulated by novel ion channel modulators and describe some of the challenges associated with trying to develop compounds with sufficient efficacy, potency and selectivity to be of therapeutic benefit.
Collapse
|
8
|
Tiffner A, Derler I. Molecular Choreography and Structure of Ca 2+ Release-Activated Ca 2+ (CRAC) and K Ca2+ Channels and Their Relevance in Disease with Special Focus on Cancer. MEMBRANES 2020; 10:E425. [PMID: 33333945 PMCID: PMC7765462 DOI: 10.3390/membranes10120425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Ca2+ ions play a variety of roles in the human body as well as within a single cell. Cellular Ca2+ signal transduction processes are governed by Ca2+ sensing and Ca2+ transporting proteins. In this review, we discuss the Ca2+ and the Ca2+-sensing ion channels with particular focus on the structure-function relationship of the Ca2+ release-activated Ca2+ (CRAC) ion channel, the Ca2+-activated K+ (KCa2+) ion channels, and their modulation via other cellular components. Moreover, we highlight their roles in healthy signaling processes as well as in disease with a special focus on cancer. As KCa2+ channels are activated via elevations of intracellular Ca2+ levels, we summarize the current knowledge on the action mechanisms of the interplay of CRAC and KCa2+ ion channels and their role in cancer cell development.
Collapse
Affiliation(s)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
9
|
Catacuzzeno L, Sforna L, Esposito V, Limatola C, Franciolini F. Ion Channels in Glioma Malignancy. Rev Physiol Biochem Pharmacol 2020; 181:223-267. [DOI: 10.1007/112_2020_44] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
10
|
Hsieh JY, Ulrich BN, Issa FA, Lin MCA, Brown B, Papazian DM. Infant and adult SCA13 mutations differentially affect Purkinje cell excitability, maturation, and viability in vivo. eLife 2020; 9:57358. [PMID: 32644043 PMCID: PMC7386905 DOI: 10.7554/elife.57358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/08/2020] [Indexed: 12/23/2022] Open
Abstract
Mutations in KCNC3, which encodes the Kv3.3 K+ channel, cause spinocerebellar ataxia 13 (SCA13). SCA13 exists in distinct forms with onset in infancy or adulthood. Using zebrafish, we tested the hypothesis that infant- and adult-onset mutations differentially affect the excitability and viability of Purkinje cells in vivo during cerebellar development. An infant-onset mutation dramatically and transiently increased Purkinje cell excitability, stunted process extension, impaired dendritic branching and synaptogenesis, and caused rapid cell death during cerebellar development. Reducing excitability increased early Purkinje cell survival. In contrast, an adult-onset mutation did not significantly alter basal tonic firing in Purkinje cells, but reduced excitability during evoked high frequency spiking. Purkinje cells expressing the adult-onset mutation matured normally and did not degenerate during cerebellar development. Our results suggest that differential changes in the excitability of cerebellar neurons contribute to the distinct ages of onset and timing of cerebellar degeneration in infant- and adult-onset SCA13.
Collapse
Affiliation(s)
- Jui-Yi Hsieh
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Brittany N Ulrich
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Fadi A Issa
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Meng-Chin A Lin
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Brandon Brown
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Diane M Papazian
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Brain Research Institute, UCLA, Los Angeles, United States.,Molecular Biology Institute, UCLA, Los Angeles, United States
| |
Collapse
|
11
|
K + and Ca 2+ Channels Regulate Ca 2+ Signaling in Chondrocytes: An Illustrated Review. Cells 2020; 9:cells9071577. [PMID: 32610485 PMCID: PMC7408816 DOI: 10.3390/cells9071577] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
An improved understanding of fundamental physiological principles and progressive pathophysiological processes in human articular joints (e.g., shoulders, knees, elbows) requires detailed investigations of two principal cell types: synovial fibroblasts and chondrocytes. Our studies, done in the past 8–10 years, have used electrophysiological, Ca2+ imaging, single molecule monitoring, immunocytochemical, and molecular methods to investigate regulation of the resting membrane potential (ER) and intracellular Ca2+ levels in human chondrocytes maintained in 2-D culture. Insights from these published papers are as follows: (1) Chondrocyte preparations express a number of different ion channels that can regulate their ER. (2) Understanding the basis for ER requires knowledge of (a) the presence or absence of ligand (ATP/histamine) stimulation and (b) the extraordinary ionic composition and ionic strength of synovial fluid. (3) In our chondrocyte preparations, at least two types of Ca2+-activated K+ channels are expressed and can significantly hyperpolarize ER. (4) Accounting for changes in ER can provide insights into the functional roles of the ligand-dependent Ca2+ influx through store-operated Ca2+ channels. Some of the findings are illustrated in this review. Our summary diagram suggests that, in chondrocytes, the K+ and Ca2+ channels are linked in a positive feedback loop that can augment Ca2+ influx and therefore regulate lubricant and cytokine secretion and gene transcription.
Collapse
|
12
|
Manfroni G, Ragonese F, Monarca L, Astolfi A, Mancinelli L, Iannitti RG, Bastioli F, Barreca ML, Cecchetti V, Fioretti B. New Insights on KCa3.1 Channel Modulation. Curr Pharm Des 2020; 26:2096-2101. [PMID: 32175839 DOI: 10.2174/1381612826666200316152645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/11/2020] [Indexed: 11/22/2022]
Abstract
The human intermediate conductance calcium-activated potassium channel, KCa3.1, is involved in several pathophysiological conditions playing a critical role in cell secretory machinery and calcium signalling. The recent cryo-EM analysis provides new insights for understanding the modulation by both endogenous and pharmacological agents. A typical feature of this channel is the low open probability in saturating calcium concentrations and its modulation by potassium channel openers (KCOs), such as benzo imidazolone 1-EBIO, without changing calcium-dependent activation. In this paper, we proposed a model of KCOs action in the modulation of channel activity. The KCa3.1 channel has a very rich pharmacological profile with several classes of molecules that selectively interact with different binding sites of the channel. Among them, benzo imidazolones can be openers (positive modulators such as 1-EBIO, DC-EBIO) or blockers (negative modulators such as NS1619). Through computation modelling techniques, we identified the 1,4-benzothiazin-3-one as a promising scaffold to develop new KCa3.1 channel modulators. Further studies are needed to explore the potential use of 1-4 benzothiazine- 3-one in KCa3.1 modulation and its pharmacological application.
Collapse
Affiliation(s)
- Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy.,Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy.,Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Astolfi
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Loretta Mancinelli
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | | | | | - Maria L Barreca
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
13
|
Chimote AA, Gawali VS, Newton HS, Wise-Draper TM, Conforti L. A Compartmentalized Reduction in Membrane-Proximal Calmodulin Reduces the Immune Surveillance Capabilities of CD8 + T Cells in Head and Neck Cancer. Front Pharmacol 2020; 11:143. [PMID: 32184726 PMCID: PMC7059094 DOI: 10.3389/fphar.2020.00143] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/04/2020] [Indexed: 01/28/2023] Open
Abstract
The limited ability of cytotoxic CD8+ T cells to infiltrate solid tumors and function within the tumor microenvironment presents a major roadblock to effective immunotherapy. Ion channels and Ca2+-dependent signaling events control the activity of T cells and are implicated in the failure of immune surveillance in cancer. Reduced KCa3.1 channel activity mediates the heightened inhibitory effect of adenosine on the chemotaxis of circulating T cells from head and neck squamous cell carcinoma (HNSCC) patients. Herein, we conducted experiments that elucidate the mechanisms of KCa3.1 dysfunction and impaired chemotaxis in HNSCC CD8+ T cells. The Ca2+ sensor calmodulin (CaM) controls multiple cellular functions including KCa3.1 activation. Our data showed that CaM expression is lower in HNSCC than healthy donor (HD) T cells. This reduction was due to an intrinsic decrease in the genes encoding CaM combined to the failure of HNSCC T cells to upregulate CaM upon activation. Furthermore, the reduction in CaM was confined to the plasma membrane and resulted in decreased CaM-KCa3.1 association and KCa3.1 activity (which was rescued by the delivery of CaM). IFNγ production, also Ca2+- and CaM-dependent, was instead not reduced in HNSCC T cells, which maintained intact cytoplasmic CaM and Ca2+ fluxing ability. Knockdown of CaM in HD T cells decreased KCa3.1 activity, but not IFNγ production, and reduced their chemotaxis in the presence of adenosine, thus recapitulating HNSCC T cell dysfunction. Activation of KCa3.1 with 1-EBIO restored the ability of CaM knockdown HD T cells to chemotax in the presence of adenosine. Additionally, 1-EBIO enhanced INFγ production. Our data showed a localized downregulation of membrane-proximal CaM that suppressed KCa3.1 activity in HNSCC circulating T cells and limited their ability to infiltrate adenosine-rich tumor-like microenvironments. Furthermore, they indicate that KCa3.1 activators could be used as positive CD8+ T cell modulators in cancers.
Collapse
Affiliation(s)
- Ameet A. Chimote
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Vaibhavkumar S. Gawali
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Hannah S. Newton
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Trisha M. Wise-Draper
- Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Laura Conforti
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
14
|
Kouba S, Braire J, Félix R, Chantôme A, Jaffrès PA, Lebreton J, Dubreuil D, Pipelier M, Zhang X, Trebak M, Vandier C, Mathé-Allainmat M, Potier-Cartereau M. Lipidic synthetic alkaloids as SK3 channel modulators. Synthesis and biological evaluation of 2-substituted tetrahydropyridine derivatives with potential anti-metastatic activity. Eur J Med Chem 2020; 186:111854. [PMID: 31753515 DOI: 10.1016/j.ejmech.2019.111854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/04/2019] [Indexed: 11/18/2022]
Abstract
Small Conductance Calcium (Ca2+)-activated potassium (K+) channels (SKCa) are now proved to be involved in many cancer cell behaviors such as proliferation or migration. The SK3 channel isoform was particularly described in breast cancer where it can be associated with the Orai1 Ca2+ channel to form a complex that regulates the Ca2+ homeostasis during tumor development and acts as a potent mediator of bone metastases development in vivo. Until now, very few specific blockers of Orai1 and/or SK3 have been developed as potential anti-metastatic compounds. In this study, we illustrated the synthesis of new families of lipophilic pyridine and tetrahydropyridine derivatives designed as potential modulators of SK3 channel. The toxicity of the newly synthesized compounds and their migration effects were evaluated on the breast cancer cell line MDA-MB-435s. Two molecules (7a and 10c) demonstrated a significant decrease in the SK3 channel-dependent migration as well as the SK3/Orai1-related Ca2+ entry. Current measurements showed that these compounds are more likely SK3-selective. Taken all together these results suggest that such molecules could be considered as promising anti-metastatic drugs in breast cancer.
Collapse
Affiliation(s)
- Sana Kouba
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Julien Braire
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Romain Félix
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Aurélie Chantôme
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Paul-Alain Jaffrès
- University of Brest, CNRS, CEMCA, UMR 6521, 6 Avenue Victor Le Gorgeu, 29238, Brest, France
| | - Jacques Lebreton
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Didier Dubreuil
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Muriel Pipelier
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France
| | - Xuexin Zhang
- Pennylvania State University College of Medicine, Department of Cellular and Molecular Physiology, 700 HMC Crescent Road, Hershey, PA, 17033, USA
| | - Mohamed Trebak
- Pennylvania State University College of Medicine, Department of Cellular and Molecular Physiology, 700 HMC Crescent Road, Hershey, PA, 17033, USA
| | - Christophe Vandier
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France
| | - Monique Mathé-Allainmat
- University of Nantes, CNRS, Faculty of Sciences, Chimie et Interdisciplinarité: Synthèse, Analyse, Modélisation (CEISAM), UMR CNRS 6230, 2, Rue de La Houssinière, 44322, Nantes Cedex 3, France.
| | - Marie Potier-Cartereau
- University of Tours, Inserm U1069 Nutrition, Croissance et Cancer (N2C), Faculty of Medicine, 10 Boulevard Tonnellé, 37032, Tours Cedex, France.
| |
Collapse
|
15
|
Shim H, Brown BM, Singh L, Singh V, Fettinger JC, Yarov-Yarovoy V, Wulff H. The Trials and Tribulations of Structure Assisted Design of K Ca Channel Activators. Front Pharmacol 2019; 10:972. [PMID: 31616290 PMCID: PMC6764326 DOI: 10.3389/fphar.2019.00972] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/29/2019] [Indexed: 11/13/2022] Open
Abstract
Calcium-activated K+ channels constitute attractive targets for the treatment of neurological and cardiovascular diseases. To explain why certain 2-aminobenzothiazole/oxazole-type KCa activators (SKAs) are KCa3.1 selective we previously generated homology models of the C-terminal calmodulin-binding domain (CaM-BD) of KCa3.1 and KCa2.3 in complex with CaM using Rosetta modeling software. We here attempted to employ this atomistic level understanding of KCa activator binding to switch selectivity around and design KCa2.2 selective activators as potential anticonvulsants. In this structure-based drug design approach we used RosettaLigand docking and carefully compared the binding poses of various SKA compounds in the KCa2.2 and KCa3.1 CaM-BD/CaM interface pocket. Based on differences between residues in the KCa2.2 and KCa.3.1 models we virtually designed 168 new SKA compounds. The compounds that were predicted to be both potent and KCa2.2 selective were synthesized, and their activity and selectivity tested by manual or automated electrophysiology. However, we failed to identify any KCa2.2 selective compounds. Based on the full-length KCa3.1 structure it was recently demonstrated that the C-terminal crystal dimer was an artefact and suggested that the "real" binding pocket for the KCa activators is located at the S4-S5 linker. We here confirmed this structural hypothesis through mutagenesis and now offer a new, corrected binding site model for the SKA-type KCa channel activators. SKA-111 (5-methylnaphtho[1,2-d]thiazol-2-amine) is binding in the interface between the CaM N-lobe and the S4-S5 linker where it makes van der Waals contacts with S181 and L185 in the S45A helix of KCa3.1.
Collapse
Affiliation(s)
- Heesung Shim
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States.,Department of Chemistry, University of California, Davis, Davis, CA, United States
| | - Brandon M Brown
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Latika Singh
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Vikrant Singh
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - James C Fettinger
- Department of Chemistry, University of California, Davis, Davis, CA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
16
|
Brown BM, Shim H, Christophersen P, Wulff H. Pharmacology of Small- and Intermediate-Conductance Calcium-Activated Potassium Channels. Annu Rev Pharmacol Toxicol 2019; 60:219-240. [PMID: 31337271 DOI: 10.1146/annurev-pharmtox-010919-023420] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The three small-conductance calcium-activated potassium (KCa2) channels and the related intermediate-conductance KCa3.1 channel are voltage-independent K+ channels that mediate calcium-induced membrane hyperpolarization. When intracellular calcium increases in the channel vicinity, it calcifies the flexible N lobe of the channel-bound calmodulin, which then swings over to the S4-S5 linker and opens the channel. KCa2 and KCa3.1 channels are highly druggable and offer multiple binding sites for venom peptides and small-molecule blockers as well as for positive- and negative-gating modulators. In this review, we briefly summarize the physiological role of KCa channels and then discuss the pharmacophores and the mechanism of action of the most commonly used peptidic and small-molecule KCa2 and KCa3.1 modulators. Finally, we describe the progress that has been made in advancing KCa3.1 blockers and KCa2.2 negative- and positive-gating modulators toward the clinic for neurological and cardiovascular diseases and discuss the remaining challenges.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Heesung Shim
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | | | - Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
17
|
Cancer-Associated Intermediate Conductance Ca 2+-Activated K⁺ Channel K Ca3.1. Cancers (Basel) 2019; 11:cancers11010109. [PMID: 30658505 PMCID: PMC6357066 DOI: 10.3390/cancers11010109] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Several tumor entities have been reported to overexpress KCa3.1 potassium channels due to epigenetic, transcriptional, or post-translational modifications. By modulating membrane potential, cell volume, or Ca2+ signaling, KCa3.1 has been proposed to exert pivotal oncogenic functions in tumorigenesis, malignant progression, metastasis, and therapy resistance. Moreover, KCa3.1 is expressed by tumor-promoting stroma cells such as fibroblasts and the tumor vasculature suggesting a role of KCa3.1 in the adaptation of the tumor microenvironment. Combined, this features KCa3.1 as a candidate target for innovative anti-cancer therapy. However, immune cells also express KCa3.1 thereby contributing to T cell activation. Thus, any strategy targeting KCa3.1 in anti-cancer therapy may also modulate anti-tumor immune activity and/or immunosuppression. The present review article highlights the potential of KCa3.1 as an anti-tumor target providing an overview of the current knowledge on its function in tumor pathogenesis with emphasis on vasculo- and angiogenesis as well as anti-cancer immune responses.
Collapse
|
18
|
Li N, Ding H, Li Z, Liu Y, Wang P. Effect of high-fat diet-induced obesity on the small-conductance Ca 2+-activated K + channel function affecting the contractility of rat detrusor smooth muscle. Int Urol Nephrol 2018; 51:61-72. [PMID: 30361965 DOI: 10.1007/s11255-018-2016-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/19/2018] [Indexed: 12/01/2022]
Abstract
PURPOSE Obesity usually induces overactive bladder (OAB) associated with detrusor overactivity, which is related to increased contractility of the detrusor smooth muscle (DSM). Small-conductance Ca2+-activated K+ (SK) channels play a constitutive role in the regulation of DSM contractility. However, the role of SK channels in the DSM changes in obesity-related OAB is still unknown. Here, we tested the hypothesis that obesity-related OAB is associated with reduced expression and activity of SK channels in DSM and that SK channels activation is a potential treatment for OAB. METHODS Female Sprague-Dawley rats were fed a normal diet (ND) or a high-fat diet (HFD) and weighed after 12 weeks. Urodynamic studies, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and isometric tension recording were performed. RESULTS Increased average body weights and urodynamically demonstrated OAB were observed in HFD rats. qRT-PCR experiments revealed a decrease in the mRNA expression level of SK channel in DSM tissue of the HFD rats. Isometric tension recordings indicated an attenuated relaxation effect of NS309 on the spontaneous phasic and electrical field stimulation-induced contractions that occurred via SK channel activation in HFD DSM strips. CONCLUSIONS Reduced expression and activity of SK channels in the DSM contribute to obesity-related OAB, indicating that SK channels are a potential therapeutic target for OAB.
Collapse
Affiliation(s)
- Ning Li
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China
| | - Honglin Ding
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China.,Department of Urology, Affiliated Hospital, Chifeng University, 42 Wangfu Street, Chifeng, Neimeng, China
| | - Zizheng Li
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China
| | - Yili Liu
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China.
| | - Ping Wang
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China
| |
Collapse
|
19
|
Brown BM, Pressley B, Wulff H. KCa3.1 Channel Modulators as Potential Therapeutic Compounds for Glioblastoma. Curr Neuropharmacol 2018; 16:618-626. [PMID: 28676010 PMCID: PMC5997873 DOI: 10.2174/1570159x15666170630164226] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022] Open
Abstract
Background The intermediate-conductance Ca2+-activated K+ channel KCa3.1 is widely expressed in cells of the immune system such as T- and B-lymphocytes, mast cells, macrophages and microglia, but also found in dedifferentiated vascular smooth muscle cells, fibroblasts and many cancer cells including pancreatic, prostate, leukemia and glioblastoma. In all these cell types KCa3.1 plays an important role in cellular activation, migration and proliferation by regulating membrane potential and Ca2+ signaling. Methods and Results KCa3.1 therefore constitutes an attractive therapeutic target for diseases involving excessive proliferation or activation of one more of these cell types and researchers both in academia and in the pharmaceutical industry have developed several potent and selective small molecule inhibitors of KCa3.1. This article will briefly review the available compounds (TRAM-34, senicapoc, NS6180), their binding sites and mechanisms of action, and then discuss the potential usefulness of these compounds for the treatment of brain tumors based on their brain penetration and their efficacy in reducing microglia activation in animal models of ischemic stroke and Alzheimer’s disease. Conclusion Senicapoc, which has previously been in Phase III clinical trials, would be available for repurposing, and could be used to quickly translate findings made with other KCa3.1 blocking tool compounds into clinical trials.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| | - Brandon Pressley
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| |
Collapse
|
20
|
Lively S, Lam D, Wong R, Schlichter LC. Comparing Effects of Transforming Growth Factor β1 on Microglia From Rat and Mouse: Transcriptional Profiles and Potassium Channels. Front Cell Neurosci 2018; 12:115. [PMID: 29780305 PMCID: PMC5946019 DOI: 10.3389/fncel.2018.00115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/11/2018] [Indexed: 12/02/2022] Open
Abstract
The cytokine, transforming growth factor β1 (TGFβ1), is up-regulated after central nervous system (CNS) injuries or diseases involving microglial activation, and it has been proposed as a therapeutic agent for treating neuroinflammation. Microglia can produce and respond to TGFβ1. While rats and mice are commonly used for studying neuroinflammation, very few reports directly compare them. Such studies are important for improving pre-clinical studies and furthering translational progress in developing therapeutic interventions. After intracerebral hemorrhage (ICH) in the rat striatum, the TGFβ1 receptor was highly expressed on microglia/macrophages within the hematoma. We recently found species similarities and differences in response to either a pro-inflammatory (interferon-γ, IFN-γ, +tumor necrosis factor, TNF-α) or anti-inflammatory interleukin-4 (IL-4) stimulus. Here, we assessed whether rat and mouse microglia differ in their responses to TGFβ1. Microglia were isolated from Sprague-Dawley rats and C57BL/6 mice and treated with TGFβ1. We quantified changes in expression of >50 genes, in their morphology, proliferation, apoptosis and in three potassium channels that are considered therapeutic targets. Many inflammatory mediators, immune receptors and modulators showed species similarities, but notable differences included that, for some genes, only one species responded (e.g., Il4r, Il10, Tgfbr2, colony-stimulating factor receptor (Csf1r), Itgam, suppressor of cytokine signaling 1 (Socs1), toll-like receptors 4 (Tlr4), P2rx7, P2ry12), and opposite responses were seen for others (Tgfb1, Myc, Ifngr1). In rat only, TGFβ1 affected microglial morphology and proliferation, but there was no apoptosis in either species. In both species, TGFβ1 dramatically increased Kv1.3 channel expression and current (no effects on Kir2.1). KCa3.1 showed opposite species responses: the current was low in unstimulated rat microglia and greatly increased by TGFβ1 but higher in control mouse cells and decreased by TGFβ1. Finally, we compared TGFβ1 and IL10 (often considered similar anti-inflammatory stimuli) and found many different responses in both species. Overall, the numerous species differences should be considered when characterizing neuroinflammation and microglial activation in vitro and in vivo, and when targeting potassium channels.
Collapse
Affiliation(s)
- Starlee Lively
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada
| | - Doris Lam
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Raymond Wong
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Lyanne C Schlichter
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Chimote AA, Balajthy A, Arnold MJ, Newton HS, Hajdu P, Qualtieri J, Wise-Draper T, Conforti L. A defect in KCa3.1 channel activity limits the ability of CD8 + T cells from cancer patients to infiltrate an adenosine-rich microenvironment. Sci Signal 2018; 11:11/527/eaaq1616. [PMID: 29692361 DOI: 10.1126/scisignal.aaq1616] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The limited ability of cytotoxic T cells to infiltrate solid tumors hampers immune surveillance and the efficacy of immunotherapies in cancer. Adenosine accumulates in solid tumors and inhibits tumor-specific T cells. Adenosine inhibits T cell motility through the A2A receptor (A2AR) and suppression of KCa3.1 channels. We conducted three-dimensional chemotaxis experiments to elucidate the effect of adenosine on the migration of peripheral blood CD8+ T cells from head and neck squamous cell carcinoma (HNSCC) patients. The chemotaxis of HNSCC CD8+ T cells was reduced in the presence of adenosine, and the effect was greater on HNSCC CD8+ T cells than on healthy donor (HD) CD8+ T cells. This response correlated with the inability of CD8+ T cells to infiltrate tumors. The effect of adenosine was mimicked by an A2AR agonist and prevented by an A2AR antagonist. We found no differences in A2AR expression, 3',5'-cyclic adenosine monophosphate abundance, or protein kinase A type 1 activity between HNSCC and HD CD8+ T cells. We instead detected a decrease in KCa3.1 channel activity, but not expression, in HNSCC CD8+ T cells. Activation of KCa3.1 channels by 1-EBIO restored the ability of HNSCC CD8+ T cells to chemotax in the presence of adenosine. Our data highlight the mechanism underlying the increased sensitivity of HNSCC CD8+ T cells to adenosine and the potential therapeutic benefit of KCa3.1 channel activators, which could increase infiltration of these T cells into tumors.
Collapse
Affiliation(s)
- Ameet A Chimote
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Andras Balajthy
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Michael J Arnold
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Hannah S Newton
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Peter Hajdu
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Julianne Qualtieri
- Department of Pathology, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Trisha Wise-Draper
- Division of Hematology Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Laura Conforti
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
22
|
Bednenko J, Harriman R, Mariën L, Nguyen HM, Agrawal A, Papoyan A, Bisharyan Y, Cardarelli J, Cassidy-Hanley D, Clark T, Pedersen D, Abdiche Y, Harriman W, van der Woning B, de Haard H, Collarini E, Wulff H, Colussi P. A multiplatform strategy for the discovery of conventional monoclonal antibodies that inhibit the voltage-gated potassium channel Kv1.3. MAbs 2018; 10:636-650. [PMID: 29494279 PMCID: PMC5973702 DOI: 10.1080/19420862.2018.1445451] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Identifying monoclonal antibodies that block human voltage-gated ion channels (VGICs) is a challenging endeavor exacerbated by difficulties in producing recombinant ion channel proteins in amounts that support drug discovery programs. We have developed a general strategy to address this challenge by combining high-level expression of recombinant VGICs in Tetrahymena thermophila with immunization of phylogenetically diverse species and unique screening tools that allow deep-mining for antibodies that could potentially bind functionally important regions of the protein. Using this approach, we targeted human Kv1.3, a voltage-gated potassium channel widely recognized as a therapeutic target for the treatment of a variety of T-cell mediated autoimmune diseases. Recombinant Kv1.3 was used to generate and recover 69 full-length anti-Kv1.3 mAbs from immunized chickens and llamas, of which 10 were able to inhibit Kv1.3 current. Select antibodies were shown to be potent (IC50<10 nM) and specific for Kv1.3 over related Kv1 family members, hERG and hNav1.5.
Collapse
Affiliation(s)
| | - Rian Harriman
- b Department of Immunology , Crystal Bioscience , Emeryville , California , USA
| | | | - Hai M Nguyen
- d Department of Pharmacology , University of California , Davis , California , USA
| | - Alka Agrawal
- a TetraGenetics Inc , Arlington , Massachusetts , USA
| | - Ashot Papoyan
- a TetraGenetics Inc , Arlington , Massachusetts , USA
| | | | | | - Donna Cassidy-Hanley
- e Department of Immunology and Microbiology , Cornell University , Ithaca , New York , USA
| | - Ted Clark
- a TetraGenetics Inc , Arlington , Massachusetts , USA.,e Department of Immunology and Microbiology , Cornell University , Ithaca , New York , USA
| | | | | | | | | | | | | | - Heike Wulff
- d Department of Pharmacology , University of California , Davis , California , USA
| | - Paul Colussi
- a TetraGenetics Inc , Arlington , Massachusetts , USA
| |
Collapse
|
23
|
Mathew John C, Khaddaj Mallat R, George G, Kim T, Mishra RC, Braun AP. Pharmacologic targeting of endothelial Ca 2+-activated K + channels: A strategy to improve cardiovascular function. Channels (Austin) 2018; 12:126-136. [PMID: 29577810 PMCID: PMC5972810 DOI: 10.1080/19336950.2018.1454814] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
Endothelial small and intermediate-conductance, Ca2+-activated K+ channels (KCa2.3 and KCa3.1, respectively) play an important role in the regulation of vascular function and systemic blood pressure. Growing evidence indicates that they are intimately involved in agonist-evoked vasodilation of small resistance arteries throughout the circulation. Small molecule activators of KCa2.x and 3.1 channels, such as SKA-31, can acutely inhibit myogenic tone in isolated resistance arteries, induce effective vasodilation in intact vascular beds, such as the coronary circulation, and acutely decrease systemic blood pressure in vivo. The blood pressure-lowering effect of SKA-31, and early indications of improvement in endothelial dysfunction suggest that endothelial KCa channel activators could eventually be developed into a new class of endothelial targeted agents to combat hypertension or atherosclerosis. This review summarises recent insights into the activation of endothelial Ca2+ activated K+ channels in various vascular beds, and how tools, such as SKA-31, may be beneficial in disease-related conditions.
Collapse
Affiliation(s)
- Cini Mathew John
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rayan Khaddaj Mallat
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Grace George
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Taeyeob Kim
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ramesh C. Mishra
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
24
|
Brown BM, Shim H, Zhang M, Yarov-Yarovoy V, Wulff H. Structural Determinants for the Selectivity of the Positive KCa3.1 Gating Modulator 5-Methylnaphtho[2,1- d]oxazol-2-amine (SKA-121). Mol Pharmacol 2017; 92:469-480. [PMID: 28760780 DOI: 10.1124/mol.117.109421] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/27/2017] [Indexed: 12/20/2022] Open
Abstract
Intermediate-conductance (KCa3.1) and small-conductance (KCa2) calcium-activated K+ channels are gated by calcium binding to calmodulin (CaM) molecules associated with the calmodulin-binding domain (CaM-BD) of these channels. The existing KCa activators, such as naphtho[1,2-d]thiazol-2-ylamine (SKA-31), 6,7-dichloro-1H-indole-2,3-dione 3-oxime (NS309), and 1-ethylbenzimidazolin-2-one (EBIO), activate both channel types with similar potencies. In a previous chemistry effort, we optimized the benzothiazole pharmacophore of SKA-31 toward KCa3.1 selectivity and identified 5-methylnaphtho[2,1-d]oxazol-2-amine (SKA-121), which exhibits 40-fold selectivity for KCa3.1 over KCa2.3. To understand why introduction of a single CH3 group in five-position of the benzothiazole/oxazole system could achieve such a gain in selectivity for KCa3.1 over KCa2.3, we first localized the binding site of the benzothiazoles/oxazoles to the CaM-BD/CaM interface and then used computational modeling software to generate models of the KCa3.1 and KCa2.3 CaM-BD/CaM complexes with SKA-121. Based on a combination of mutagenesis and structural modeling, we suggest that all benzothiazole/oxazole-type KCa activators bind relatively "deep" in the CaM-BD/CaM interface and hydrogen bond with E54 on CaM. In KCa3.1, SKA-121 forms an additional hydrogen bond network with R362. In contrast, NS309 sits more "forward" and directly hydrogen bonds with R362 in KCa3.1. Mutating R362 to serine, the corresponding residue in KCa2.3 reduces the potency of SKA-121 by 7-fold, suggesting that R362 is responsible for the generally greater potency of KCa activators on KCa3.1. The increase in SKA-121's KCa3.1 selectivity compared with its parent, SKA-31, seems to be due to better overall shape complementarity and hydrophobic interactions with S372 and M368 on KCa3.1 and M72 on CaM at the KCa3.1-CaM-BD/CaM interface.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Heesung Shim
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Miao Zhang
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Vladimir Yarov-Yarovoy
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| | - Heike Wulff
- Department of Pharmacology (B.M.B., H.S., H.W.), Department of Physiology and Membrane Biology (V.Y.-Y.), School of Medicine, and Department of Chemistry (H.S.), University of California, Davis, California; and Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (M.Z.)
| |
Collapse
|
25
|
Andrei SA, Sijbesma E, Hann M, Davis J, O’Mahony G, Perry MWD, Karawajczyk A, Eickhoff J, Brunsveld L, Doveston RG, Milroy LG, Ottmann C. Stabilization of protein-protein interactions in drug discovery. Expert Opin Drug Discov 2017; 12:925-940. [DOI: 10.1080/17460441.2017.1346608] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Sebastian A. Andrei
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Michael Hann
- Platform Technology and Science, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage, UK
| | - Jeremy Davis
- Department of Chemistry, UCB Celltech, Slough, UK
| | - Gavin O’Mahony
- CVMD Medicinal Chemistry, Innovative Medicines and Early Development, AstraZeneca Gothenburg, Pepparedsleden, Mölndal, Sweden
| | - Matthew W. D. Perry
- RIA Medicinal Chemistry, Innovative Medicines and Early Development, AstraZeneca Gothenburg, Pepparedsleden, Mölndal, Sweden
| | - Anna Karawajczyk
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Dortmund, Germany
| | - Jan Eickhoff
- Assay development & screening, Lead Discovery Center GmbH, Dortmund, Germany
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Richard G. Doveston
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Lech-Gustav Milroy
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Chemistry, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
26
|
Comerma-Steffensen S, Kun A, Hedegaard ER, Mogensen S, Aalkjaer C, Köhler R, Mønster Christensen B, Simonsen U. Down-regulation of K Ca2.3 channels causes erectile dysfunction in mice. Sci Rep 2017. [PMID: 28630432 PMCID: PMC5476588 DOI: 10.1038/s41598-017-04188-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Modulation of endothelial calcium-activated K+ channels has been proposed as an approach to restore arterial endothelial cell function in disease. We hypothesized that small-conductance calcium-activated K+ channels (KCa2.3 or SK3) contributes to erectile function. The research was performed in transgenic mice with overexpression (KCa2.3T/T(−Dox)) or down-regulation (KCa2.3T/T(+Dox)) of the KCa2.3 channels and wild-type C57BL/6-mice (WT). QPCR revealed that KCa2.3 and KCa1.1 channels were the most abundant in mouse corpus cavernosum. KCa2.3 channels were found by immunoreactivity and electron microscopy in the apical-lateral membrane of endothelial cells in the corpus cavernosum. Norepinephrine contraction was enhanced in the corpus cavernosum of KCa2.3T/T(+Dox)versus KCa2.3T/T(−Dox) mice, while acetylcholine relaxation was only reduced at 0.3 µM and relaxations in response to the nitric oxide donor sodium nitroprusside were unaltered. An opener of KCa2 channels, NS309 induced concentration-dependent relaxations of corpus cavernosum. Mean arterial pressure was lower in KCa2.3T/T(−Dox) mice compared with WT and KCa2.3T/T(+Dox) mice. In anesthetized mice, cavernous nerve stimulation augmented in frequency/voltage dependent manner erectile function being lower in KCa2.3T/T(+Dox) mice at low frequencies. Our findings suggest that down-regulation of KCa2.3 channels contributes to erectile dysfunction, and that pharmacological activation of KCa2.3 channels may have the potential to restore erectile function.
Collapse
Affiliation(s)
- Simon Comerma-Steffensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.
| | - Attila Kun
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Elise R Hedegaard
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Susie Mogensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | | | - Ralf Köhler
- Aragon Agency for Investigation and Development (ARAID), Translational Research Unit, Miguel Servet University Hospital, Zaragoza, Spain
| | | | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
27
|
Honrath B, Krabbendam IE, Culmsee C, Dolga AM. Small conductance Ca 2+-activated K + channels in the plasma membrane, mitochondria and the ER: Pharmacology and implications in neuronal diseases. Neurochem Int 2017; 109:13-23. [PMID: 28511953 DOI: 10.1016/j.neuint.2017.05.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/24/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022]
Abstract
Ca2+-activated K+ (KCa) channels regulate after-hyperpolarization in many types of neurons in the central and peripheral nervous system. Small conductance Ca2+-activated K+ (KCa2/SK) channels, a subfamily of KCa channels, are widely expressed in the nervous system, and in the cardiovascular system. Voltage-independent SK channels are activated by alterations in intracellular Ca2+ ([Ca2+]i) which facilitates the opening of these channels through binding of Ca2+ to calmodulin that is constitutively bound to the SK2 C-terminus. In neurons, SK channels regulate synaptic plasticity and [Ca2+]i homeostasis, and a number of recent studies elaborated on the emerging neuroprotective potential of SK channel activation in conditions of excitotoxicity and cerebral ischemia, as well as endoplasmic reticulum (ER) stress and oxidative cell death. Recently, SK channels were discovered in the inner mitochondrial membrane and in the membrane of the endoplasmic reticulum which sheds new light on the underlying molecular mechanisms and pathways involved in SK channel-mediated protective effects. In this review, we will discuss the protective properties of pharmacological SK channel modulation with particular emphasis on intracellularly located SK channels as potential therapeutic targets in paradigms of neuronal dysfunction.
Collapse
Affiliation(s)
- Birgit Honrath
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany; Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Inge E Krabbendam
- Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany
| | - Amalia M Dolga
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany; Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
28
|
Li N, Ding H, He X, Li Z, Liu Y. Expression and function of the small-conductance Ca 2+-activated K + channel is decreased in urinary bladder smooth muscle cells from female guinea pig with partial bladder outlet obstruction. Int Urol Nephrol 2017; 49:1147-1155. [PMID: 28417339 DOI: 10.1007/s11255-017-1592-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/10/2017] [Indexed: 01/10/2023]
Abstract
PURPOSE Overactive bladder (OAB), usually accompanied by partial bladder outlet obstruction (PBOO), is associated with detrusor overactivity (DO) which is related to the increased urinary bladder smooth muscle (UBSM) cells excitability. Small-conductance Ca2+-activated K+ (SK) channels play a constitutive regulatory role of UBSM excitability and contractility. PBOO is associated with the decreased SK channels mRNA expression and the attenuated regulative effect of SK channels on UBSM contractility. However, the regulation of SK channels in PBOO UBSM cell excitability is less clear. Here, we tested the hypothesis that PBOO is associated with decreased expression and function of SK channels in UBSM cells and that SK channels are a potential target for the treatment of OAB. METHODS Cystometry indicated that DO was achieved 2 weeks after PBOO in female guinea pigs. Using this animal model, we conducted single-cell quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and patch-clamp electrophysiology. RESULTS The single-cell qRT-PCR experiments indicated the reduced SK channel mRNA expression in PBOO UBSM cells. Patch-clamp studies revealed that NS309 had a diminished effect on resting membrane potential hyperpolarization via the activation of SK channels in PBOO UBSM cells. Moreover, attenuated whole-cell SK channel currents were demonstrated in PBOO UBSM cells. CONCLUSIONS The attenuated expression and function of SK channels, which results in the increased UBSM cells excitability and contributes to DO, was discovered in PBOO UBSM cells, suggesting that SK channels might be potential therapeutic targets for the control of OAB.
Collapse
Affiliation(s)
- Ning Li
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China
| | - Honglin Ding
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China
| | - Xiaoning He
- Department of Stomatology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China
| | - Zizheng Li
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China
| | - Yili Liu
- Department of Urology, Fourth Affiliated Hospital, China Medical University, 4 Chongshan East Road, Shenyang, Liaoning, China.
| |
Collapse
|
29
|
Simó-Vicens R, Sauter DRP, Grunnet M, Diness JG, Bentzen BH. Effect of antiarrhythmic drugs on small conductance calcium - activated potassium channels. Eur J Pharmacol 2017; 803:118-123. [PMID: 28322838 DOI: 10.1016/j.ejphar.2017.03.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 10/19/2022]
Abstract
Atrial fibrillation (AF) is the most common type of arrhythmia. Current pharmacological treatment for AF is moderately effective and/or increases the risk of serious ventricular adverse effects. To avoid ventricular adverse effects, a new target has been considered, the small conductance calcium-activated K+ channels (KCa2.X, SK channels). In the heart, KCa2.X channels are functionally more important in atria compared to ventricles, and pharmacological inhibition of the channel confers atrial selective prolongation of the cardiac action potential and converts AF to sinus rhythm in animal models of AF. Whether antiarrhythmic drugs (AADs) recommended for treating AF target KCa2.X channels is unknown. To this end, we tested a large number of AADs on the human KCa2.2 and KCa2.3 channels to assess their effect on this new target using automated whole-cell patch clamp. Of the AADs recommended for treatment of AF only dofetilide and propafenone inhibited hKCa2.X channels, with no subtype selectivity. The calculated IC50 were 90±10µmol/l vs 60±10µmol/l for dofetilide and 42±4µmol/l vs 80±20µmol/l for propafenone (hKCa2.3 vs hKCa2.2). Whether this inhibition has clinical importance for their antiarrhythmic effect is unlikely, as the calculated IC50 values are very high compared to the effective free therapeutic plasma concentration of the drugs when used for AF treatment, 40,000-fold for dofetilide and 140-fold higher for propafenone.
Collapse
Affiliation(s)
- Rafel Simó-Vicens
- Biomedical Institute, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark; Acesion Pharma, Ole Maaløes Vej 3, DK-2200 Copenhagen, Denmark
| | - Daniel R P Sauter
- Biomedical Institute, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark; Acesion Pharma, Ole Maaløes Vej 3, DK-2200 Copenhagen, Denmark
| | - Morten Grunnet
- Acesion Pharma, Ole Maaløes Vej 3, DK-2200 Copenhagen, Denmark
| | - Jonas G Diness
- Acesion Pharma, Ole Maaløes Vej 3, DK-2200 Copenhagen, Denmark
| | - Bo H Bentzen
- Biomedical Institute, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark; Acesion Pharma, Ole Maaløes Vej 3, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
30
|
Oliván-Viguera A, Lozano-Gerona J, López de Frutos L, Cebolla JJ, Irún P, Abarca-Lachen E, García-Malinis AJ, García-Otín ÁL, Gilaberte Y, Giraldo P, Köhler R. Inhibition of Intermediate-Conductance Calcium-Activated K Channel (KCa3.1) and Fibroblast Mitogenesis by α-Linolenic Acid and Alterations of Channel Expression in the Lysosomal Storage Disorders, Fabry Disease, and Niemann Pick C. Front Physiol 2017; 8:39. [PMID: 28197106 PMCID: PMC5281581 DOI: 10.3389/fphys.2017.00039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/13/2017] [Indexed: 01/02/2023] Open
Abstract
The calcium/calmodulin-gated KCa3.1 channel regulates normal and abnormal mitogenesis by controlling K+-efflux, cell volume, and membrane hyperpolarization-driven calcium-entry. Recent studies suggest modulation of KCa3.1 by omega-3 fatty acids as negative modulators and impaired KCa3.1 functions in the inherited lysosomal storage disorder (LSD), Fabry disease (FD). In the first part of present study, we characterize KCa3.1 in murine and human fibroblasts and test the impact of omega-3 fatty acids on fibroblast proliferation. In the second, we study whether KCa3.1 is altered in the LSDs, FD, and Niemann-Pick disease type C (NPC). Our patch-clamp and mRNA-expression studies on murine and human fibroblasts show functional expression of KCa3.1. KCa currents display the typical pharmacological fingerprint of KCa3.1: Ca2+-activation, potentiation by the positive-gating modulators, SKA-31 and SKA-121, and inhibition by TRAM-34, Senicapoc (ICA-17043), and the negative-gating modulator, 13b. Considering modulation by omega-3 fatty acids we found that α-linolenic acid (α-LA) and docosahexanenoic acid (DHA) inhibit KCa3.1 currents and strongly reduce fibroblast growth. The α-LA-rich linseed oil and γ-LA-rich borage oil at 0.5% produce channel inhibition while α-LA/γ-LA-low oils has no anti-proliferative effect. Concerning KCa3.1 in LSD, mRNA expression studies, and patch-clamp on primary fibroblasts from FD and NPC patients reveal lower KCa3.1-gene expression and membrane expression than in control fibroblasts. In conclusion, the omega-3 fatty acid, α-LA, and α-LA/γ-LA-rich plant oils, inhibit fibroblast KCa3.1 channels and mitogenesis. Reduced fibroblast KCa3.1 functions are a feature and possible biomarker of cell dysfunction in FD and NPC and supports the concept that biased lipid metabolism is capable of negatively modulating KCa3.1 expression.
Collapse
Affiliation(s)
- Aida Oliván-Viguera
- Biomedical Signal Interpretation and Computational Simulation Group, Aragón Institute for Engineering Research (I3A), University of ZaragozaZaragoza, Spain; Instituto de Investigación Sanitaria AragónZaragoza, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and NanomedicineZaragoza, Spain
| | - Javier Lozano-Gerona
- Instituto de Investigación Sanitaria AragónZaragoza, Spain; Aragón Institute of Health SciencesZaragoza, Spain; Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de ZaragozaZaragoza, Spain
| | - Laura López de Frutos
- Instituto de Investigación Sanitaria AragónZaragoza, Spain; Spanish Foundation for the Study and Treatment of Gaucher Disease and Other Lysosomal DisordersZaragoza, Spain
| | - Jorge J Cebolla
- Instituto de Investigación Sanitaria AragónZaragoza, Spain; Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de ZaragozaZaragoza, Spain; Centro de Investigación Biomédica en Red de Enfermedades RarasZaragoza, Spain
| | - Pilar Irún
- Instituto de Investigación Sanitaria AragónZaragoza, Spain; Centro de Investigación Biomédica en Red de Enfermedades RarasZaragoza, Spain
| | - Edgar Abarca-Lachen
- Faculty of Health Sciences, Universidad San Jorge Villanueva de Gállego, Spain
| | | | - Ángel Luis García-Otín
- Instituto de Investigación Sanitaria AragónZaragoza, Spain; Aragón Institute of Health SciencesZaragoza, Spain
| | | | - Pilar Giraldo
- Instituto de Investigación Sanitaria AragónZaragoza, Spain; Aragón Institute of Health SciencesZaragoza, Spain; Spanish Foundation for the Study and Treatment of Gaucher Disease and Other Lysosomal DisordersZaragoza, Spain; Centro de Investigación Biomédica en Red de Enfermedades RarasZaragoza, Spain
| | - Ralf Köhler
- Instituto de Investigación Sanitaria AragónZaragoza, Spain; Aragón Institute of Health SciencesZaragoza, Spain; Centro de Investigación Biomédica en Red de Enfermedades RarasZaragoza, Spain; Aragón Agency for Research and DevelopmentZaragoza, Spain
| |
Collapse
|
31
|
Simonsen U, Wandall-Frostholm C, Oliván-Viguera A, Köhler R. Emerging roles of calcium-activated K channels and TRPV4 channels in lung oedema and pulmonary circulatory collapse. Acta Physiol (Oxf) 2017; 219:176-187. [PMID: 27497091 DOI: 10.1111/apha.12768] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/30/2015] [Accepted: 08/04/2016] [Indexed: 12/12/2022]
Abstract
It has been suggested that the transient receptor potential cation (TRP) channel subfamily V (vanilloid) type 4 (TRPV4) and intermediate conductance calcium-activated potassium (KCa3.1) channels contribute to endothelium-dependent vasodilation. Here, we summarize very recent evidence for a synergistic interplay of TRPV4 and KCa3.1 channels in lung disease. Among the endothelial Ca2+ -permeable TRPs, TRPV4 is best characterized and produces arterial dilation by stimulating Ca2+ -dependent nitric oxide synthesis and endothelium-dependent hyperpolarization. Besides these roles, some TRP channels control endothelial/epithelial barrier functions and vascular integrity, while KCa3.1 channels provide the driving force required for Cl- and water transport in some cells and most secretory epithelia. The three conditions, increased pulmonary venous pressure caused by left heart disease, high inflation pressure and chemically induced lung injury, may lead to activation of TRPV4 channels followed by Ca2+ influx leading to activation of KCa3.1 channels in endothelial cells ultimately leading to acute lung injury. We find that a deficiency in KCa3.1 channels protects against TRPV4-induced pulmonary arterial relaxation, fluid extravasation, haemorrhage, pulmonary circulatory collapse and cardiac arrest in vivo. These data identify KCa3.1 channels as crucial molecular components in downstream TRPV4 signal transduction and as a potential target for the prevention of undesired fluid extravasation, vasodilatation and pulmonary circulatory collapse.
Collapse
Affiliation(s)
- U. Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology; Aarhus University; Aarhus C Denmark
| | - C. Wandall-Frostholm
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology; Aarhus University; Aarhus C Denmark
| | - A. Oliván-Viguera
- Translational Research Unit; University Hospital Miguel Servet and IACS/IIS; Aragonese Agency for Investigation and Development (ARAID); Zaragoza Spain
| | - R. Köhler
- Translational Research Unit; University Hospital Miguel Servet and IACS/IIS; Aragonese Agency for Investigation and Development (ARAID); Zaragoza Spain
| |
Collapse
|
32
|
Nguyen HM, Grössinger EM, Horiuchi M, Davis KW, Jin LW, Maezawa I, Wulff H. Differential Kv1.3, KCa3.1, and Kir2.1 expression in "classically" and "alternatively" activated microglia. Glia 2016; 65:106-121. [PMID: 27696527 PMCID: PMC5113690 DOI: 10.1002/glia.23078] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/15/2016] [Indexed: 11/10/2022]
Abstract
Microglia are highly plastic cells that can assume different phenotypes in response to microenvironmental signals. Lipopolysaccharide (LPS) and interferon-γ (IFN-γ) promote differentiation into classically activated M1-like microglia, which produce high levels of pro-inflammatory cytokines and nitric oxide and are thought to contribute to neurological damage in ischemic stroke and Alzheimer's disease. IL-4 in contrast induces a phenotype associated with anti-inflammatory effects and tissue repair. We here investigated whether these microglia subsets vary in their K+ channel expression by differentiating neonatal mouse microglia into M(LPS) and M(IL-4) microglia and studying their K+ channel expression by whole-cell patch-clamp, quantitative PCR and immunohistochemistry. We identified three major types of K+ channels based on their biophysical and pharmacological fingerprints: a use-dependent, outwardly rectifying current sensitive to the KV 1.3 blockers PAP-1 and ShK-186, an inwardly rectifying Ba2+ -sensitive Kir 2.1 current, and a Ca2+ -activated, TRAM-34-sensitive KCa 3.1 current. Both KV 1.3 and KCa 3.1 blockers inhibited pro-inflammatory cytokine production and iNOS and COX2 expression demonstrating that KV 1.3 and KCa 3.1 play important roles in microglia activation. Following differentiation with LPS or a combination of LPS and IFN-γ microglia exhibited high KV 1.3 current densities (∼50 pA/pF at 40 mV) and virtually no KCa 3.1 and Kir currents, while microglia differentiated with IL-4 exhibited large Kir 2.1 currents (∼ 10 pA/pF at -120 mV). KCa 3.1 currents were generally low but moderately increased following stimulation with IFN-γ or ATP (∼10 pS/pF). This differential K+ channel expression pattern suggests that KV 1.3 and KCa 3.1 inhibitors could be used to inhibit detrimental neuroinflammatory microglia functions. GLIA 2016;65:106-121.
Collapse
Affiliation(s)
- Hai M Nguyen
- Department of Pharmacology, University of California, Davis, California
| | - Eva M Grössinger
- Department of Pharmacology, University of California, Davis, California
| | - Makoto Horiuchi
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California.,M.I.N.D. Institute, University of California Davis Medical Center, Davis, Sacramento, California
| | - Kyle W Davis
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California.,M.I.N.D. Institute, University of California Davis Medical Center, Davis, Sacramento, California
| | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California.,M.I.N.D. Institute, University of California Davis Medical Center, Davis, Sacramento, California
| | - Heike Wulff
- Department of Pharmacology, University of California, Davis, California
| |
Collapse
|
33
|
Choi S, Kim JA, Li H, Shin K, Oh GT, Lee Y, Oh S, Pewzner‐Jung Y, Futerman AH, Suh SH. KCa 3.1 upregulation preserves endothelium-dependent vasorelaxation during aging and oxidative stress. Aging Cell 2016; 15:801-10. [PMID: 27363720 PMCID: PMC5013018 DOI: 10.1111/acel.12502] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2016] [Indexed: 01/24/2023] Open
Abstract
Endothelial oxidative stress develops with aging and reactive oxygen species impair endothelium-dependent relaxation (EDR) by decreasing nitric oxide (NO) availability. Endothelial KCa 3.1, which contributes to EDR, is upregulated by H2 O2 . We investigated whether KCa 3.1 upregulation compensates for diminished EDR to NO during aging-related oxidative stress. Previous studies identified that the levels of ceramide synthase 5 (CerS5), sphingosine, and sphingosine 1-phosphate were increased in aged wild-type and CerS2 mice. In primary mouse aortic endothelial cells (MAECs) from aged wild-type and CerS2 null mice, superoxide dismutase (SOD) was upregulated, and catalase and glutathione peroxidase 1 (GPX1) were downregulated, when compared to MAECs from young and age-matched wild-type mice. Increased H2 O2 levels induced Fyn and extracellular signal-regulated kinases (ERKs) phosphorylation and KCa 3.1 upregulation. Catalase/GPX1 double knockout (catalase(-/-) /GPX1(-/-) ) upregulated KCa 3.1 in MAECs. NO production was decreased in aged wild-type, CerS2 null, and catalase(-/-) /GPX1(-/-) MAECs. However, KCa 3.1 activation-induced, N(G) -nitro-l-arginine-, and indomethacin-resistant EDR was increased without a change in acetylcholine-induced EDR in aortic rings from aged wild-type, CerS2 null, and catalase(-/-) /GPX1(-/-) mice. CerS5 transfection or exogenous application of sphingosine or sphingosine 1-phosphate induced similar changes in levels of the antioxidant enzymes and upregulated KCa 3.1. Our findings suggest that, during aging-related oxidative stress, SOD upregulation and downregulation of catalase and GPX1, which occur upon altering the sphingolipid composition or acyl chain length, generate H2 O2 and thereby upregulate KCa 3.1 expression and function via a H2 O2 /Fyn-mediated pathway. Altogether, enhanced KCa 3.1 activity may compensate for decreased NO signaling during vascular aging.
Collapse
Affiliation(s)
- Shinkyu Choi
- Department of Physiology Medical School Ewha Womans University Seoul South Korea
| | - Ji Aee Kim
- Department of Physiology Medical School Ewha Womans University Seoul South Korea
| | - Hai‐yan Li
- Department of Physiology Medical School Ewha Womans University Seoul South Korea
| | - Kyong‐Oh Shin
- College of Pharmacy and MRC Chungbuk National University Chongju South Korea
| | - Goo Taeg Oh
- Department of Life Sciences Ewha Womans University Seoul South Korea
| | - Yong‐Moon Lee
- College of Pharmacy and MRC Chungbuk National University Chongju South Korea
| | - Seikwan Oh
- Department of Molecular Medicine Medical School Ewha Womans University Seoul South Korea
| | - Yael Pewzner‐Jung
- Department of Biological Chemistry Weizmann Institute of Science Rehovot Israel
| | - Anthony H. Futerman
- Department of Biological Chemistry Weizmann Institute of Science Rehovot Israel
| | - Suk Hyo Suh
- Department of Physiology Medical School Ewha Womans University Seoul South Korea
| |
Collapse
|
34
|
Abstract
Small-conductance Ca2+-activated potassium (SK) channels are relative newcomers within the field of cardiac electrophysiology. In recent years, an increased focus has been given to these channels because they might constitute a relatively atrial-selective target. This review will give a general introduction to SK channels followed by their proposed function in the heart under normal and pathophysiological conditions. It is revealed how antiarrhythmic effects can be obtained by SK channel inhibition in a number of species in situations of atrial fibrillation. On the contrary, the beneficial effects of SK channel inhibition in situations of heart failure are questionable and still needs investigation. The understanding of cardiac SK channels is rapidly increasing these years, and it is hoped that this will clarify whether SK channel inhibition has potential as a new anti–atrial fibrillation principle.
Collapse
|
35
|
|
36
|
Oliván-Viguera A, Valero MS, Pinilla E, Amor S, García-Villalón ÁL, Coleman N, Laría C, Calvín-Tienza V, García-Otín ÁL, Fernández-Fernández JM, Murillo MD, Gálvez JA, Díaz-de-Villegas MD, Badorrey R, Simonsen U, Rivera L, Wulff H, Köhler R. Vascular Reactivity Profile of Novel KCa 3.1-Selective Positive-Gating Modulators in the Coronary Vascular Bed. Basic Clin Pharmacol Toxicol 2016; 119:184-92. [PMID: 26821335 DOI: 10.1111/bcpt.12560] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/17/2016] [Indexed: 12/12/2022]
Abstract
Opening of intermediate-conductance calcium-activated potassium channels (KC a 3.1) produces membrane hyperpolarization in the vascular endothelium. Here, we studied the ability of two new KC a 3.1-selective positive-gating modulators, SKA-111 and SKA-121, to (1) evoke porcine endothelial cell KC a 3.1 membrane hyperpolarization, (2) induce endothelium-dependent and, particularly, endothelium-derived hyperpolarization (EDH)-type relaxation in porcine coronary arteries (PCA) and (3) influence coronary artery tone in isolated rat hearts. In whole-cell patch-clamp experiments on endothelial cells of PCA (PCAEC), KC a currents evoked by bradykinin (BK) were potentiated ≈7-fold by either SKA-111 or SKA-121 (both at 1 μM) and were blocked by a KC a 3.1 blocker, TRAM-34. In membrane potential measurements, SKA-111 and SKA-121 augmented bradykinin-induced hyperpolarization. Isometric tension measurements in large- and small-calibre PCA showed that SKA-111 and SKA-121 potentiated endothelium-dependent relaxation with intact NO synthesis and EDH-type relaxation to BK by ≈2-fold. Potentiation of the BK response was prevented by KC a 3.1 inhibition. In Langendorff-perfused rat hearts, SKA-111 potentiated coronary vasodilation elicited by BK. In conclusion, our data show that positive-gating modulation of KC a 3.1 channels improves BK-induced membrane hyperpolarization and endothelium-dependent relaxation in small and large PCA as well as in the coronary circulation of rats. Positive-gating modulators of KC a 3.1 could be therapeutically useful to improve coronary blood flow and counteract impaired coronary endothelial dysfunction in cardiovascular disease.
Collapse
Affiliation(s)
| | - Marta Sofía Valero
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Sara Amor
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Nichole Coleman
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Celia Laría
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain
| | - Víctor Calvín-Tienza
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain
| | - Ángel-Luis García-Otín
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain
| | - José M Fernández-Fernández
- Laboratori de Fisiologia Molecular i Canalopaties, Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - M Divina Murillo
- Department of Pharmacology and Physiology, Veterinary Faculty, University of Zaragoza, Zaragoza, Spain
| | - José A Gálvez
- Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC - Universidad de Zaragoza, Zaragoza, Spain
| | - María D Díaz-de-Villegas
- Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC - Universidad de Zaragoza, Zaragoza, Spain
| | - Ramón Badorrey
- Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC - Universidad de Zaragoza, Zaragoza, Spain
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Luis Rivera
- Department of Physiology, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Heike Wulff
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Ralf Köhler
- Aragon Institute of Health Sciences & IIS, Zaragoza, Spain.,Aragon Agency for Research and Development (ARAID), Zaragoza, Spain
| |
Collapse
|
37
|
Köhler R, Oliván-Viguera A, Wulff H. Endothelial Small- and Intermediate-Conductance K Channels and Endothelium-Dependent Hyperpolarization as Drug Targets in Cardiovascular Disease. ADVANCES IN PHARMACOLOGY 2016; 77:65-104. [DOI: 10.1016/bs.apha.2016.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Christophersen P, Wulff H. Pharmacological gating modulation of small- and intermediate-conductance Ca(2+)-activated K(+) channels (KCa2.x and KCa3.1). Channels (Austin) 2015. [PMID: 26217968 DOI: 10.1080/19336950.2015.1071748] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
This short review discusses pharmacological modulation of the opening/closing properties (gating) of small- and intermediate-conductance Ca(2+)-activated K(+) channels (KCa2 and KCa3.1) with special focus on mechanisms-of-action, selectivity, binding sites, and therapeutic potentials. Despite KCa channel gating-modulation being a relatively novel field in drug discovery, efforts in this area have already revealed a surprising plethora of pharmacological sites-of-actions and channel subtype selectivity exerted by different chemical classes. The currently published positive modulators show that such molecules are potentially useful for the treatment of various neurodegenerative disorders such as ataxia, alcohol dependence, and epilepsy as well as hypertension. The negative KCa2 modulators are very effective agents for atrial fibrillation. The prediction is that further unraveling of the molecular details of gating pharmacology will allow for the design of even more potent and subtype selective KCa modulators entering into drug development for these indications.
Collapse
Affiliation(s)
| | - Heike Wulff
- b Department of Pharmacology ; University of California, Davis ; Davis , CA USA
| |
Collapse
|
39
|
Chen YJ, Wallace BK, Yuen N, Jenkins DP, Wulff H, O'Donnell ME. Blood-brain barrier KCa3.1 channels: evidence for a role in brain Na uptake and edema in ischemic stroke. Stroke 2014; 46:237-44. [PMID: 25477223 DOI: 10.1161/strokeaha.114.007445] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE KCa3.1, a calcium-activated potassium channel, regulates ion and fluid secretion in the lung and gastrointestinal tract. It is also expressed on vascular endothelium where it participates in blood pressure regulation. However, the expression and physiological role of KCa3.1 in blood-brain barrier (BBB) endothelium has not been investigated. BBB endothelial cells transport Na(+) and Cl(-) from the blood into the brain transcellularly through the co-operation of multiple cotransporters, exchangers, pumps, and channels. In the early stages of cerebral ischemia, when the BBB is intact, edema formation occurs by processes involving increased BBB transcellular Na(+) transport. This study evaluated whether KCa3.1 is expressed on and participates in BBB ion transport. METHODS The expression of KCa3.1 on cultured cerebral microvascular endothelial cells, isolated microvessels, and brain sections was evaluated by Western blot and immunohistochemistry. Activity of KCa3.1 on cerebral microvascular endothelial cells was examined by K(+) flux assays and patch-clamp. Magnetic resonance spectroscopy and MRI were used to measure brain Na(+) uptake and edema formation in rats with focal ischemic stroke after TRAM-34 treatment. RESULTS KCa3.1 current and channel protein were identified on bovine cerebral microvascular endothelial cells and freshly isolated rat microvessels. In situ KCa3.1 expression on BBB endothelium was confirmed in rat and human brain sections. TRAM-34 treatment significantly reduced Na(+) uptake, and cytotoxic edema in the ischemic brain. CONCLUSIONS BBB endothelial cells exhibit KCa3.1 protein and activity and pharmacological blockade of KCa3.1 seems to provide an effective therapeutic approach for reducing cerebral edema formation in the first 3 hours of ischemic stroke.
Collapse
Affiliation(s)
- Yi-Je Chen
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis.
| | - Breanna K Wallace
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - Natalie Yuen
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - David P Jenkins
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - Heike Wulff
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| | - Martha E O'Donnell
- From the Department of Pharmacology (Y.-J.C., D.P.J., H.W.) and Department of Physiology and Membrane Biology (B.K.W., N.Y., M.E.O.), University of California, Davis
| |
Collapse
|
40
|
Oliván-Viguera A, Valero MS, Coleman N, Brown BM, Laría C, Murillo MD, Gálvez JA, Díaz-de-Villegas MD, Wulff H, Badorrey R, Köhler R. A novel pan-negative-gating modulator of KCa2/3 channels, fluoro-di-benzoate, RA-2, inhibits endothelium-derived hyperpolarization-type relaxation in coronary artery and produces bradycardia in vivo. Mol Pharmacol 2014; 87:338-48. [PMID: 25468883 DOI: 10.1124/mol.114.095745] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Small/intermediate conductance KCa channels (KCa2/3) are Ca(2+)/calmodulin regulated K(+) channels that produce membrane hyperpolarization and shape neurologic, epithelial, cardiovascular, and immunologic functions. Moreover, they emerged as therapeutic targets to treat cardiovascular disease, chronic inflammation, and some cancers. Here, we aimed to generate a new pharmacophore for negative-gating modulation of KCa2/3 channels. We synthesized a series of mono- and dibenzoates and identified three dibenzoates [1,3-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate) (RA-2), 1,2-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate), and 1,4-phenylenebis(methylene) bis(3-fluoro-4-hydroxybenzoate)] with inhibitory efficacy as determined by patch clamp. Among them, RA-2 was the most drug-like and inhibited human KCa3.1 with an IC50 of 17 nM and all three human KCa2 subtypes with similar potencies. RA-2 at 100 nM right-shifted the KCa3.1 concentration-response curve for Ca(2+) activation. The positive-gating modulator naphtho[1,2-d]thiazol-2-ylamine (SKA-31) reversed channel inhibition at nanomolar RA-2 concentrations. RA-2 had no considerable blocking effects on distantly related large-conductance KCa1.1, Kv1.2/1.3, Kv7.4, hERG, or inwardly rectifying K(+) channels. In isometric myography on porcine coronary arteries, RA-2 inhibited bradykinin-induced endothelium-derived hyperpolarization (EDH)-type relaxation in U46619-precontracted rings. Blood pressure telemetry in mice showed that intraperitoneal application of RA-2 (≤100 mg/kg) did not increase blood pressure or cause gross behavioral deficits. However, RA-2 decreased heart rate by ≈145 beats per minute, which was not seen in KCa3.1(-/-) mice. In conclusion, we identified the KCa2/3-negative-gating modulator, RA-2, as a new pharmacophore with nanomolar potency. RA-2 may be of use to generate structurally new types of negative-gating modulators that could help to define the physiologic and pathomechanistic roles of KCa2/3 in the vasculature, central nervous system, and during inflammation in vivo.
Collapse
Affiliation(s)
- Aida Oliván-Viguera
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Marta Sofía Valero
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Nicole Coleman
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Brandon M Brown
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Celia Laría
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - María Divina Murillo
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - José A Gálvez
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - María D Díaz-de-Villegas
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Heike Wulff
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Ramón Badorrey
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.)
| | - Ralf Köhler
- Aragon Institute of Health Sciences, Zaragoza, Spain (A.O.-V., R.K.); GIMACES, Facultad de Ciencias de la Salud, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V., C.L.); Department of Pharmacology, School of Medicine, University of California Davis, Davis, California (N.C., B.M.B, H.W.); Departamento de Farmacología y Fisiología, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain (M.D.M.); Departamento de Catálisis y Procesos Catalíticos, Instituto de Síntesis Química y Catálisis Homogénea, Consejo Superior de Investigaciones Científicas-Universidad de Zaragoza, Zaragoza, Spain (M.D.D.-V., J.A.G., R.B.); and Fundación Agencia Aragonesa para la Investigación y Desarrollo (R.K.).
| |
Collapse
|
41
|
Cui M, Qin G, Yu K, Bowers MS, Zhang M. Targeting the Small- and Intermediate-Conductance Ca-Activated Potassium Channels: The Drug-Binding Pocket at the Channel/Calmodulin Interface. Neurosignals 2014; 22:65-78. [PMID: 25300231 DOI: 10.1159/000367896] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
The small- and intermediate-conductance Ca(2+)-activated potassium (SK/IK) channels play important roles in the regulation of excitable cells in both the central nervous and cardiovascular systems. Evidence from animal models has implicated SK/IK channels in neurological conditions such as ataxia and alcohol use disorders. Further, genome-wide association studies have suggested that cardiovascular abnormalities such as arrhythmias and hypertension are associated with single nucleotide polymorphisms that occur within the genes encoding the SK/IK channels. The Ca(2+) sensitivity of the SK/IK channels stems from a constitutively bound Ca(2+)-binding protein: calmodulin. Small-molecule positive modulators of SK/IK channels have been developed over the past decade, and recent structural studies have revealed that the binding pocket of these positive modulators is located at the interface between the channel and calmodulin. SK/IK channel positive modulators can potentiate channel activity by enhancing the coupling between Ca(2+) sensing via calmodulin and mechanical opening of the channel. Here, we review binding pocket studies that have provided structural insight into the mechanism of action for SK/IK channel positive modulators. These studies lay the foundation for structure-based drug discovery efforts that can identify novel SK/IK channel positive modulators.
Collapse
Affiliation(s)
- Meng Cui
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Va., USA
| | | | | | | | | |
Collapse
|
42
|
Coleman N, Brown BM, Oliván-Viguera A, Singh V, Olmstead MM, Valero MS, Köhler R, Wulff H. New positive Ca2+-activated K+ channel gating modulators with selectivity for KCa3.1. Mol Pharmacol 2014; 86:342-57. [PMID: 24958817 DOI: 10.1124/mol.114.093286] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Small-conductance (KCa2) and intermediate-conductance (KCa3.1) calcium-activated K(+) channels are voltage-independent and share a common calcium/calmodulin-mediated gating mechanism. Existing positive gating modulators like EBIO, NS309, or SKA-31 activate both KCa2 and KCa3.1 channels with similar potency or, as in the case of CyPPA and NS13001, selectively activate KCa2.2 and KCa2.3 channels. We performed a structure-activity relationship (SAR) study with the aim of optimizing the benzothiazole pharmacophore of SKA-31 toward KCa3.1 selectivity. We identified SKA-111 (5-methylnaphtho[1,2-d]thiazol-2-amine), which displays 123-fold selectivity for KCa3.1 (EC50 111 ± 27 nM) over KCa2.3 (EC50 13.7 ± 6.9 μM), and SKA-121 (5-methylnaphtho[2,1-d]oxazol-2-amine), which displays 41-fold selectivity for KCa3.1 (EC50 109 nM ± 14 nM) over KCa2.3 (EC50 4.4 ± 1.6 μM). Both compounds are 200- to 400-fold selective over representative KV (KV1.3, KV2.1, KV3.1, and KV11.1), NaV (NaV1.2, NaV1.4, NaV1.5, and NaV1.7), as well as CaV1.2 channels. SKA-121 is a typical positive-gating modulator and shifts the calcium-concentration response curve of KCa3.1 to the left. In blood pressure telemetry experiments, SKA-121 (100 mg/kg i.p.) significantly lowered mean arterial blood pressure in normotensive and hypertensive wild-type but not in KCa3.1(-/-) mice. SKA-111, which was found in pharmacokinetic experiments to have a much longer half-life and to be much more brain penetrant than SKA-121, not only lowered blood pressure but also drastically reduced heart rate, presumably through cardiac and neuronal KCa2 activation when dosed at 100 mg/kg. In conclusion, with SKA-121, we generated a KCa3.1-specific positive gating modulator suitable for further exploring the therapeutical potential of KCa3.1 activation.
Collapse
Affiliation(s)
- Nichole Coleman
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Brandon M Brown
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Aida Oliván-Viguera
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Vikrant Singh
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Marilyn M Olmstead
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Marta Sofia Valero
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Ralf Köhler
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| | - Heike Wulff
- Department of Pharmacology (N.C., B.M.B., V.S., H.W.), School of Medicine, and Department of Chemistry (M.M.O.), University of California, Davis, California; Aragon Institute of Health Sciences, Instituto de Investigación Sanitaria, Fundación Agencia Aragonesa para la Investigación y el Desarrollo, Zaragoza, Spain (A.O.-V., R.K.); and Grupo de Investigación del Medio Ambiente del Centro de Estudios Superiores, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Spain (M.S.V.)
| |
Collapse
|