1
|
Balkaya M, Dohare P, Chen S, Schober AL, Fidaleo AM, Nalwalk JW, Sah R, Mongin AA. Conditional deletion of LRRC8A in the brain reduces stroke damage independently of swelling-activated glutamate release. iScience 2023; 26:106669. [PMID: 37182109 PMCID: PMC10173736 DOI: 10.1016/j.isci.2023.106669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/03/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
The ubiquitous volume-regulated anion channels (VRACs) facilitate cell volume control and contribute to many other physiological processes. Treatment with non-specific VRAC blockers or brain-specific deletion of the essential VRAC subunit LRRC8A is highly protective in rodent models of stroke. Here, we tested the widely accepted idea that the harmful effects of VRACs are mediated by release of the excitatory neurotransmitter glutamate. We produced conditional LRRC8A knockout either exclusively in astrocytes or in the majority of brain cells. Genetically modified mice were subjected to an experimental stroke (middle cerebral artery occlusion). The astrocytic LRRC8A knockout yielded no protection. Conversely, the brain-wide LRRC8A deletion strongly reduced cerebral infarction in both heterozygous (Het) and full KO mice. Yet, despite identical protection, Het mice had full swelling-activated glutamate release, whereas KO animals showed its virtual absence. These findings suggest that LRRC8A contributes to ischemic brain injury via a mechanism other than VRAC-mediated glutamate release.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Preeti Dohare
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Sophie Chen
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Alexandra L. Schober
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Antonio M. Fidaleo
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Julia W. Nalwalk
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - Rajan Sah
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander A. Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
- Corresponding author
| |
Collapse
|
2
|
Naffaa MM, Al-Ewaidat OA. Ligand modulation of KCNQ-encoded (K V7) potassium channels in the heart and nervous system. Eur J Pharmacol 2021; 906:174278. [PMID: 34174270 DOI: 10.1016/j.ejphar.2021.174278] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/06/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
KCNQ-encoded (KV7) potassium channels are diversely distributed in the human tissues, associated with many physiological processes and pathophysiological conditions. These channels are increasingly used as drug targets for treating diseases. More selective and potent molecules on various types of the KV7 channels are desirable for appropriate therapies. The recent knowledge of the structure and function of human KCNQ-encoded channels makes it more feasible to achieve these goals. This review discusses the role and mechanism of action of many molecules in modulating the function of the KCNQ-encoded potassium channels in the heart and nervous system. The effects of these compounds on KV7 channels help to understand their involvement in many diseases, and to search for more selective and potent ligands to be used in the treatment of many disorders such as various types of cardiac arrhythmias, epilepsy, and pain.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA; Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA.
| | - Ola A Al-Ewaidat
- Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
3
|
Wilson CS, Bach MD, Ashkavand Z, Norman KR, Martino N, Adam AP, Mongin AA. Metabolic constraints of swelling-activated glutamate release in astrocytes and their implication for ischemic tissue damage. J Neurochem 2019; 151:255-272. [PMID: 31032919 DOI: 10.1111/jnc.14711] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/01/2019] [Accepted: 04/10/2019] [Indexed: 12/20/2022]
Abstract
Volume-regulated anion channel (VRAC) is a glutamate-permeable channel that is activated by physiological and pathological cell swelling and promotes ischemic brain damage. However, because VRAC opening requires cytosolic ATP, it is not clear if and how its activity is sustained in the metabolically compromised CNS. In the present study, we used cultured astrocytes - the cell type which shows prominent swelling in stroke - to model how metabolic stress and changes in gene expression may impact VRAC function in the ischemic and post-ischemic brain. The metabolic state of primary rat astrocytes was modified with chemical inhibitors and examined using luciferin-luciferase ATP assays and a Seahorse analyzer. Swelling-activated glutamate release was quantified with the radiotracer D-[3 H]aspartate. The specific contribution of VRAC to swelling-activated glutamate efflux was validated by RNAi knockdown of the essential subunit, leucine-rich repeat-containing 8A (LRRC8A); expression levels of VRAC components were measured with qRT-PCR. Using this methodology, we found that complete metabolic inhibition with the glycolysis blocker 2-deoxy-D-glucose and the mitochondrial poison sodium cyanide reduced astrocytic ATP levels by > 90% and abolished glutamate release from swollen cells (via VRAC). When only mitochondrial respiration was inhibited by cyanide or rotenone, the intracellular ATP levels and VRAC activity were largely preserved. Bypassing glycolysis by providing the mitochondrial substrates pyruvate and/or glutamine led to partial recovery of ATP levels and VRAC activity. Unexpectedly, the metabolic block of VRAC was overridden when ATP-depleted cells were exposed to extreme cell swelling (≥ 50% reduction in medium osmolarity). Twenty-four hour anoxic adaptation caused a moderate reduction in the expression levels of the VRAC component LRRC8A, but no significant changes in VRAC activity. Overall, our findings suggest that (i) astrocytic VRAC activity and metabolism can be sustained by low levels of glucose and (ii) the inhibitory influence of diminishing ATP levels and the stimulatory effect of cellular swelling are the two major factors that govern VRAC activity in the ischemic brain.
Collapse
Affiliation(s)
- Corinne S Wilson
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Martin D Bach
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Zahra Ashkavand
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Kenneth R Norman
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Nina Martino
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
4
|
Liu H, Huang Q, Sun H, Li J, Lin Q, Wu H, Liu C. Effects of separate or combined exposure of nonylphenol and octylphenol on central 5-HT system and related learning and memory in the rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 172:523-529. [PMID: 30743168 DOI: 10.1016/j.ecoenv.2019.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 05/20/2023]
Abstract
This study evaluated toxic effects of nonylphenol (NP) and octylphenol (OP) on central 5-hydroxytryptamine (5-HT) system and related learning and memory in the rats. Male Sprague-Dawley rats were exposed to NP (30, 90, or 270 mg/kg), OP (40, 120, or 360 mg/kg), or a mixture of NP and OP [(mixed with the corresponding NP, OP alone exposed low, medium and high dose according to the natural environment exists NP:OP = 4:1; NOL (24 mg/kg NP+8 mg/kg OP), NOM (72 mg/kg NP+24 mg/kg OP), NOH (216 mg/kg NP+72 mg/kg OP)] by gavage every other day for 30 d. Learning and memory were assessed using a passive-avoidance test. Levels of estrogen receptor β (ERβ), 5-HT, tryptophan hydroxylase 2 (TPH2), monoamine oxidase (MAOA) enzyme, serotonin transporter (SERT), the vesicular monoamine transporter 2 (VMAT2), 5-hydroxytryptamine 1 A (5-HT1A), 5-hydroxytryptamine 3 A (5-HT3A), 5-hydroxytryptamine 3B (5-HT3B), 5-hydroxytryptamine 4 A (5-HT4A) and 5-hydroxytryptamine 6 A (5-HT6A) were measured using ELISA kits. Levels of ERβ, MAOA, SERT, VMAT2, 5-HT1A, 5-HT3A, 5-HT3B, 5-HT4A and 5-HT6A in rat hippocampal reduced by a high dose of NP and/or OP. Levels of TPH2 in rat midbrain and 5-HT in rat hippocampal increased by a high dose of NP and/or OP. In addition, latency was significantly shorter and errors were significantly greater in the high dose NP and NP+OP (NO) groups. Taken together, these results suggest that NP and/or OP may affect learning and memory in rats by inhibiting levels of ERβ, which could then lead to decreases in levels of 5-HT1A, 5-HT3A, 5-HT3B, 5-HT4A, and 5-HT6A in the rat hippocampus. These findings suggested that separate and combined exposure to NP and OP could produce toxic effects on central 5-HT system and related learning and memory in the rats.
Collapse
Affiliation(s)
- Huan Liu
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Qingyi Huang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Hanzhi Sun
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Jieming Li
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Qianwen Lin
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Haoming Wu
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Chunhong Liu
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
5
|
Okada Y, Okada T, Sato-Numata K, Islam MR, Ando-Akatsuka Y, Numata T, Kubo M, Shimizu T, Kurbannazarova RS, Marunaka Y, Sabirov RZ. Cell Volume-Activated and Volume-Correlated Anion Channels in Mammalian Cells: Their Biophysical, Molecular, and Pharmacological Properties. Pharmacol Rev 2019; 71:49-88. [PMID: 30573636 DOI: 10.1124/pr.118.015917] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
There are a number of mammalian anion channel types associated with cell volume changes. These channel types are classified into two groups: volume-activated anion channels (VAACs) and volume-correlated anion channels (VCACs). VAACs can be directly activated by cell swelling and include the volume-sensitive outwardly rectifying anion channel (VSOR), which is also called the volume-regulated anion channel; the maxi-anion channel (MAC or Maxi-Cl); and the voltage-gated anion channel, chloride channel (ClC)-2. VCACs can be facultatively implicated in, although not directly activated by, cell volume changes and include the cAMP-activated cystic fibrosis transmembrane conductance regulator (CFTR) anion channel, the Ca2+-activated Cl- channel (CaCC), and the acid-sensitive (or acid-stimulated) outwardly rectifying anion channel. This article describes the phenotypical properties and activation mechanisms of both groups of anion channels, including accumulating pieces of information on the basis of recent molecular understanding. To that end, this review also highlights the molecular identities of both anion channel groups; in addition to the molecular identities of ClC-2 and CFTR, those of CaCC, VSOR, and Maxi-Cl were recently identified by applying genome-wide approaches. In the last section of this review, the most up-to-date information on the pharmacological properties of both anion channel groups, especially their half-maximal inhibitory concentrations (IC50 values) and voltage-dependent blocking, is summarized particularly from the standpoint of pharmacological distinctions among them. Future physiologic and pharmacological studies are definitely warranted for therapeutic targeting of dysfunction of VAACs and VCACs.
Collapse
Affiliation(s)
- Yasunobu Okada
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Toshiaki Okada
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Kaori Sato-Numata
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Md Rafiqul Islam
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Yuhko Ando-Akatsuka
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Tomohiro Numata
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Machiko Kubo
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Takahiro Shimizu
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Ranohon S Kurbannazarova
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Yoshinori Marunaka
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Ravshan Z Sabirov
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| |
Collapse
|
6
|
Kow LM, Pfaff DW. Rapid estrogen actions on ion channels: A survey in search for mechanisms. Steroids 2016; 111:46-53. [PMID: 26939826 PMCID: PMC4929851 DOI: 10.1016/j.steroids.2016.02.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 12/31/2022]
Abstract
A survey of nearly two hundred reports shows that rapid estrogenic actions can be detected across a range of kinds of estrogens, a range of doses, on a wide range of tissue, cell and ion channel types. Striking is the fact that preparations of estrogenic agents that do not permeate the cell membrane almost always mimic the actions of the estrogenic agents that do permeate the membrane. All kinds of estrogens, ranging from natural ones, through receptor modulators, endocrine disruptors, phytoestrogens, agonists, and antagonists to novel G-1 and STX, have been reported to be effective. For actions on specific types of ion channels, the possibility of opposing actions, in different cases, is the rule, not the exception. With this variety there is no single, specific action mechanism for estrogens per se, although in some cases estrogens can act directly or via some signaling pathways to affect ion channels. We infer that estrogens can bind a large number of substrates/receptors at the membrane surface. As against the variety of subsequent routes of action, this initial step of the estrogen's binding action is the key.
Collapse
Affiliation(s)
- Lee-Ming Kow
- The Rockefeller University, New York, NY 10065, USA.
| | | |
Collapse
|
7
|
Mongin AA. Volume-regulated anion channel--a frenemy within the brain. Pflugers Arch 2015; 468:421-41. [PMID: 26620797 DOI: 10.1007/s00424-015-1765-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/16/2015] [Accepted: 11/20/2015] [Indexed: 10/22/2022]
Abstract
The volume-regulated anion channel (VRAC) is a ubiquitously expressed yet highly enigmatic member of the superfamily of chloride/anion channels. It is activated by cellular swelling and mediates regulatory cell volume decrease in a majority of vertebrate cells, including those in the central nervous system (CNS). In the brain, besides its crucial role in cellular volume regulation, VRAC is thought to play a part in cell proliferation, apoptosis, migration, and release of physiologically active molecules. Although these roles are not exclusive to the CNS, the relative significance of VRAC in the brain is amplified by several unique aspects of its physiology. One important example is the contribution of VRAC to the release of the excitatory amino acid neurotransmitters glutamate and aspartate. This latter process is thought to have impact on both normal brain functioning (such as astrocyte-neuron signaling) and neuropathology (via promoting the excitotoxic death of neuronal cells in stroke and traumatic brain injury). In spite of much work in the field, the molecular nature of VRAC remained unknown until less than 2 years ago. Two pioneer publications identified VRAC as the heterohexamer formed by the leucine-rich repeat-containing 8 (LRRC8) proteins. These findings galvanized the field and are likely to result in dramatic revisions to our understanding of the place and role of VRAC in the brain, as well as other organs and tissues. The present review briefly recapitulates critical findings in the CNS and focuses on anticipated impact on the LRRC8 discovery on further progress in neuroscience research.
Collapse
Affiliation(s)
- Alexander A Mongin
- Center for Neuropharmacology and Neuroscience, Albany Medical College, 47 New Scotland Ave., Albany, NY, 12208, USA.
| |
Collapse
|
8
|
Maher J, Hunter AC, Mabley JG, Lippiat J, Allen MC. Smooth muscle relaxation and activation of the large conductance Ca(++)-activated K+ (BK(Ca)) channel by novel oestrogens. Br J Pharmacol 2015; 169:1153-65. [PMID: 23586466 DOI: 10.1111/bph.12211] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 02/25/2013] [Accepted: 03/24/2013] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Oestrogens can interact directly with membrane receptors and channels and can activate vascular BK(Ca) channels. We hypothesized that novel oestrogen derivatives could relax smooth muscle by an extracllular effect on the α and β1 subunits of the BK(Ca) channel, rather than at an intracellular site. EXPERIMENTAL APPROACH We studied the effects of novel oestrogens on the tension of pre-contracted isolated rat aortic rings, and on the electrophysiological properties of HEK 293 cells expressing the hSloα or hSloα+β1 subunits. Two of the derivatives incorporated a quaternary ammonium side-chain making them membrane impermeable. KEY RESULTS Oestrone, oestrone oxime and Quat DME-oestradiol relaxed pre-contracted rat aorta, but only Quat DME-oestradiol-induced relaxation was iberiotoxin sensitive. However, only potassium currents recorded in HEK 293 cells over-expressing both hSloα and hSloβ1 were activated by oestrone, oestrone oxime and Quat DME-oestradiol. CONCLUSION AND IMPLICATIONS The novel oestrogens were able to relax smooth muscle, but through different mechanisms. In particular, oestrone oxime required the presence of the endothelium to exert much of its effect, whilst Quat DME-oestradiol depended both on NO and BK(Ca) channel activation. The activation of BK(Ca) currents in HEK 293 cells expressing hSloα+β1 by Quat DME-oestradiol is consistent with an extracellular binding site between the two subunits. The binding site resides between the extracellular N terminal of the α subunit and the extracellular loop between TM1 and 2 of the β1 subunit. Membrane-impermeant Quat DME-oestradiol lacks an exchangeable hydrogen on the A ring obviating antioxidant activity.
Collapse
Affiliation(s)
- J Maher
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton, UK
| | | | | | | | | |
Collapse
|
9
|
Ferrer T, Aréchiga-Figueroa IA, Shapiro MS, Tristani-Firouzi M, Sanchez-Chapula JA. Tamoxifen inhibition of kv7.2/kv7.3 channels. PLoS One 2013; 8:e76085. [PMID: 24086693 PMCID: PMC3782443 DOI: 10.1371/journal.pone.0076085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 08/21/2013] [Indexed: 11/18/2022] Open
Abstract
KCNQ genes encode five Kv7 K+ channel subunits (Kv7.1–Kv7.5). Four of these (Kv7.2–Kv7.5) are expressed in the nervous system. Kv7.2 and Kv7.3 are the principal molecular components of the slow voltage-gated M-channel, which regulates neuronal excitability. In this study, we demonstrate that tamoxifen, an estrogen receptor antagonist used in the treatment of breast cancer, inhibits Kv7.2/Kv7.3 currents heterologously expressed in human embryonic kidney HEK-293 cells. Current inhibition by tamoxifen was voltage independent but concentration-dependent. The IC50 for current inhibition was 1.68 ± 0.44 µM. The voltage-dependent activation of the channel was not modified. Tamoxifen inhibited Kv7.2 homomeric channels with a higher potency (IC50 = 0.74 ± 0.16 µM). The mutation Kv7.2 R463E increases phosphatidylinositol- 4,5-bisphosphate (PIP2) - channel interaction and diminished dramatically the inhibitory effect of tamoxifen compared with that for wild type Kv7.2. Conversely, the mutation Kv7.2 R463Q, which decreases PIP2 -channel interaction, increased tamoxifen potency. Similar results were obtained on the heteromeric Kv7.2 R463Q/Kv7.3 and Kv7.2 R463E/Kv7.3 channels, compared to Kv7.2/Kv7.3 WT. Overexpression of type 2A PI(4)P5-kinase (PIP5K 2A) significantly reduced tamoxifen inhibition of Kv7.2/Kv7.3 and Kv7.2 R463Q channels. Our results suggest that tamoxifen inhibited Kv7.2/Kv7.3 channels by interfering with PIP2-channel interaction because of its documented interaction with PIP2 and the similar effect of tamoxifen on various PIP2 sensitive channels.
Collapse
Affiliation(s)
- Tania Ferrer
- Unidad de Investigación “Carlos Méndez” del Centro Universitario de Investigaciones Biomédicas de la Universidad de Colima, Colima, Col. México
- * E-mail:
| | - Ivan Arael Aréchiga-Figueroa
- Unidad de Investigación “Carlos Méndez” del Centro Universitario de Investigaciones Biomédicas de la Universidad de Colima, Colima, Col. México
| | - Mark S. Shapiro
- Department of Physiology, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Martin Tristani-Firouzi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Division of Pediatric Cardiology, University of Utah, Salt Lake City, Utah, United States of America
| | - José A. Sanchez-Chapula
- Unidad de Investigación “Carlos Méndez” del Centro Universitario de Investigaciones Biomédicas de la Universidad de Colima, Colima, Col. México
| |
Collapse
|
10
|
Rivera-Guevara C, Pérez-Alvarez V, García-Becerra R, Ordaz-Rosado D, Morales-Ríos MS, Hernández-Gallegos E, Cooney AJ, Bravo-Gómez ME, Larrea F, Camacho J. Genomic action of permanently charged tamoxifen derivatives via estrogen receptor-alpha. Bioorg Med Chem 2010; 18:5593-601. [PMID: 20621492 DOI: 10.1016/j.bmc.2010.06.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/10/2010] [Accepted: 06/14/2010] [Indexed: 11/30/2022]
Abstract
Tamoxifen is a selective estrogen receptor modulator widely used in oncology and reproductive endocrinology. In order to decrease its non-desirable effects and elucidate mechanisms of action, permanently charged tamoxifen derivatives (PCTDs) have been reported. Whether PCTDs have genomic effects remains controversial. Since the clinical relevance of tamoxifen, the necessity to have new anticancer drugs, and in order to gain insights into the mechanisms of action of PCTDs, we obtained six quaternary ammonium salts derived from tamoxifen including three new compounds. We characterized them by nuclear magnetic resonance, X-ray diffraction, electron microscopy, and/or high performance liquid chromatography, and detected them in cell lysates by liquid chromatography coupled to mass spectrometry. We evaluated their binding to estrogen receptor-alpha (ERalpha, their effect on the transcriptional activity mediated by ERalpha (gene reporter assays), and the proliferation of cancer cells (MCF-7 and cells from a cervical cancer primary culture). Structural studies demonstrated the expected identity of the molecules. All PCTDs did bind to ERalpha, one of them induced ERalpha-mediated transcription while two others inhibited such genomic action. Accordingly, PCTDs were detected in cell lysates. PCTDs inhibited cell proliferation, noteworthy, two of them displayed higher inhibition than tamoxifen. Structure-activity analysis suggests that PCTDs permanent positive charge and the length of the aliphatic chain might be associated to the biological responses studied. We suggest genomic effects as a mechanism of action of PCTDs. The experimental approaches here used could lead to a better design of new therapeutic molecules and help to elucidate molecular mechanisms of new anticancer drugs.
Collapse
Affiliation(s)
- Claudia Rivera-Guevara
- Department of Pharmacology, Centro de Investigación y de Estudios Avanzados, Avenida Instituto Politécnico Nacional 2508, México DF 07360, Mexico.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Ponce-Balbuena D, Moreno-Galindo EG, López-Izquierdo A, Ferrer T, Sánchez-Chapula JA. Tamoxifen Inhibits Cardiac ATP-Sensitive and Acetylcholine-Activated K+ Currents in Part by Interfering With Phosphatidylinositol 4,5-Bisphosphate–Channel Interaction. J Pharmacol Sci 2010; 113:66-75. [DOI: 10.1254/jphs.10024fp] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
12
|
Ponce-Balbuena D, López-Izquierdo A, Ferrer T, Rodríguez-Menchaca AA, Aréchiga-Figueroa IA, Sánchez-Chapula JA. Tamoxifen inhibits inward rectifier K+ 2.x family of inward rectifier channels by interfering with phosphatidylinositol 4,5-bisphosphate-channel interactions. J Pharmacol Exp Ther 2009; 331:563-73. [PMID: 19654266 DOI: 10.1124/jpet.109.156075] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tamoxifen, an estrogen receptor antagonist used in the treatment of breast cancer, inhibits the inward rectifier potassium current (I(K1)) in cardiac myocytes by an unknown mechanism. We characterized the inhibitory effects of tamoxifen on Kir2.1, Kir2.2, and Kir2.3 potassium channels that underlie cardiac I(K1). We also studied the effects of 4-hydroxytamoxifen and raloxifene. All three drugs inhibited inward rectifier K(+) 2.x (Kir2.x) family members. The order of inhibition for all three drugs was Kir2.3 > Kir2.1 approximately Kir2.2. The onset of inhibition of Kir2.x current by these compounds was slow (T(1/2) approximately 6 min) and only partially recovered after washout ( approximately 30%). Kir2.x inhibition was concentration-dependent but voltage-independent. The time course and degree of inhibition was independent of external or internal drug application. We tested the hypothesis that tamoxifen interferes with the interaction between the channel and the membrane-delimited channel activator, phosphatidylinositol 4,5-bisphosphate (PIP(2)). Inhibition of Kir2.3 currents was significantly reduced by a single point mutation of I213L, which enhances Kir2.3 interaction with membrane PIP(2). Pretreatment with PIP(2) significantly decreased the inhibition induced by tamoxifen, 4-hydroxytamoxifen, and raloxifene on Kir2.3 channels. Pretreatment with spermine (100 microM) decreased the inhibitory effect of tamoxifen on Kir2.1, probably by strengthening the channel's interaction with PIP(2). In cat atrial and ventricular myocytes, 3 microM tamoxifen inhibited I(K1), but the effect was greater in the former than the latter. The data strongly suggest that tamoxifen, its metabolite, and the estrogen receptor inhibitor raloxifene inhibit Kir2.x channels indirectly by interfering with the interaction between the channel and PIP(2).
Collapse
Affiliation(s)
- Daniela Ponce-Balbuena
- Unidad de Investigación Carlos Méndez del Centro Universitario de Investigaciones Biomédicas de la Universidad de Colima, Colima, Colima, México
| | | | | | | | | | | |
Collapse
|
13
|
Beca S, Pavlov E, Kargacin ME, Aschar-Sobbi R, French RJ, Kargacin GJ. Inhibition of a cardiac sarcoplasmic reticulum chloride channel by tamoxifen. Pflugers Arch 2008; 457:121-35. [PMID: 18458943 DOI: 10.1007/s00424-008-0510-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 03/21/2008] [Accepted: 03/25/2008] [Indexed: 11/25/2022]
Abstract
Anion and cation channels present in the sarcoplasmic reticulum (SR) are believed to be necessary to maintain the electroneutrality of SR membrane during Ca(2+) uptake by the SR Ca(2+) pump (SERCA). Here we incorporated canine cardiac SR ion channels into lipid bilayers and studied the effects of tamoxifen and other antiestrogens on these channels. A Cl(-) channel was identified exhibiting multiple subconductance levels which could be divided into two primary conductance bands. Tamoxifen decreases the time the channel spends in its higher, voltage-sensitive band and the mean channel current. The lower, voltage-insensitive, conductance band is not affected by tamoxifen, nor is a K(+) channel present in the cardiac SR preparation. By examining SR Ca(2+) uptake, SERCA ATPase activity, and SR ion channels in the same preparation, we also estimated SERCA transport current, SR Cl(-) and K(+) currents, and the density of SERCA, Cl(-), and K(+) channels in cardiac SR membranes.
Collapse
Affiliation(s)
- Sanja Beca
- Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | | | | | | | | | | |
Collapse
|
14
|
Mongin AA. Disruption of ionic and cell volume homeostasis in cerebral ischemia: The perfect storm. ACTA ACUST UNITED AC 2007; 14:183-93. [PMID: 17961999 DOI: 10.1016/j.pathophys.2007.09.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The mechanisms of brain tissue damage in stroke are strongly linked to the phenomenon of excitotoxicity, which is defined as damage or death of neural cells due to excessive activation of receptors for the excitatory neurotransmitters glutamate and aspartate. Under physiological conditions, ionotropic glutamate receptors mediate the processes of excitatory neurotransmission and synaptic plasticity. In ischemia, sustained pathological release of glutamate from neurons and glial cells causes prolonged activation of these receptors, resulting in massive depolarization and cytoplasmic Ca(2+) overload. High cytoplasmic levels of Ca(2+) activate many degradative processes that, depending on the metabolic status, cause immediate or delayed death of neural cells. This traditional view has been expanded by a number of observations that implicate Cl(-) channels and several types of non-channel transporter proteins, such as the Na(+),K(+),2Cl(-) cotransporter, Na(+)/H(+) exchanger, and Na(+)/Ca(2+) exchanger, in the development of glutamate toxicity. Some of these ion transporters increase tissue damage by promoting pathological cell swelling and necrotic cell death, while others contribute to a long-term accumulation of cytoplasmic Ca(2+). This brief review is aimed at illustrating how the dysregulation of various ion transport processes combine in a 'perfect storm' that disrupts neural ionic homeostasis and culminates in the irreversible damage and death of neural cells. The clinical relevance of individual transporters as targets for therapeutic intervention in stroke is also briefly discussed.
Collapse
Affiliation(s)
- Alexander A Mongin
- Center for Neuropharmacology and Neuroscience, Albany Medical College, 47 New Scotland Avenue (MC-136), Albany, NY 12208, USA
| |
Collapse
|
15
|
Marrero-Alonso J, García Marrero B, Gómez T, Díaz M. Functional inhibition of intestinal and uterine muscles by non-permeant triphenylethylene derivatives. Eur J Pharmacol 2006; 532:115-27. [PMID: 16466652 DOI: 10.1016/j.ejphar.2005.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Revised: 11/07/2005] [Accepted: 11/14/2005] [Indexed: 10/25/2022]
Abstract
We have previously shown that the triphenylethylene antiestrogen tamoxifen reversibly inhibited spontaneous contractile activity in isolated duodenal muscle. Now, we have synthesized different quaternary ammonium salts of tamoxifen by changing the substituents on the nitrogen of the alkylaminoethoxy side-chain, to obtain plasma membrane impermeable compounds. Synthesized molecules were N-desmethyl-tamoxifen-hydrochloride, ethylbromide-tamoxifen and butylbromide-tamoxifen, which differed in the size of their ionic side-chain. All compounds rapidly and reversibly inhibited spontaneous and CaCl(2)-induced contractions in mouse duodenum and uterus. Dose-response analyses revealed a structure-activity relationship where the larger the side-chain the higher the inhibitory potency. Fourier analyses on triphenylethylene-relaxed duodenal tissues showed that harmonic components of contractile activity were readily recovered upon exposure to the L-type calcium channel agonist 1,4-dihydro-2,6-dimethyl-5-nitro-4-[2-(trifluoromethyl)phenyl]-pyridine-3-carboxilic acid methyl ester (BAY-K644). Likewise, BAY-K644 completely reversed triphenylethylene-induced effects on uterine tonic tension. Our experiments suggest that impermeant tamoxifen derivatives relax visceral smooth muscle through a membrane-mediated non-genomic mechanism that involves inhibition of L-type calcium channels.
Collapse
Affiliation(s)
- Jorge Marrero-Alonso
- Laboratorio de Fisiología Animal, Departamento de Biología Animal, Universidad de La Laguna, 38206 Tenerife, Spain
| | | | | | | |
Collapse
|
16
|
Willett MC, Dluzen DE. Tamoxifen increases methamphetamine-evoked dopamine output from superfused striatal tissue fragments of male mice. Brain Res 2004; 1029:186-94. [PMID: 15542073 DOI: 10.1016/j.brainres.2004.09.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2004] [Indexed: 11/16/2022]
Abstract
The antiestrogen, tamoxifen (TMX), has been shown to function as a neuroprotectant against the nigrostriatal dopaminergic (NSDA) neurotoxin, methamphetamine (MA), within male mice. In the present report, we examined the effects of a combined infusion of TMX and MA within superfused striatal tissue fragments of male mice as an approach to understand some of the bases for TMX to function as a NSDA neuroprotectant within male mice. In Experiment 1, a coinfusion of TMX at 1, 10, or 100 pg/ml were all equally effective in increasing MA-evoked dopamine (DA) output as compared with a 0 pg/ml (control) dose. In Experiment 2, we tested whether this effect of TMX was specific for MA-evoked DA output by coinfusing TMX with a depolarizing concentration of potassium chloride (K+ -30 mM). No statistically significant differences were obtained between superfusions of striatal tissue fragments stimulated with K+ in the presence or absence of TMX (100 pg/ml). In Experiment 3, we assessed whether these effects of TMX may be exerted upon the dopamine transporter (DAT) by coinfusing DA (1 microM) in the presence or absence of TMX (100 pg/ml). No differences in DA recovery rates were obtained between superfusions performed in the presence or absence of TMX. Taken together, these results show that the striatum of male mice is very sensitive to the modulatory effects of TMX upon MA-evoked DA output. These effects of TMX appear to be relatively specific for MA-evoked DA output, as K+ -stimulated DA was not altered by TMX, and do not appear to exert these effects by altering dopamine transporter function.
Collapse
Affiliation(s)
- Matthew C Willett
- Department of Anatomy, Northeastern Ohio Universities College of Medicine (NEOUCOM), 4209 State Route 44, PO Box 95, Rootstown, OH 44272-0095, USA
| | | |
Collapse
|
17
|
Coelho RR, Souza EP, Soares PMG, Meireles AVP, Santos GCM, Scarparo HC, Assreuy AMS, Criddle DN. Effects of chloride channel blockers on hypotonicity-induced contractions of the rat trachea. Br J Pharmacol 2003; 141:367-73. [PMID: 14691057 PMCID: PMC1574202 DOI: 10.1038/sj.bjp.0705615] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. We have investigated the inhibitory effects of blockers of volume-activated (Cl(vol)) and calcium-activated (Cl(Ca)) chloride channels on hypotonic solution (HS)-induced contractions of rat trachea, comparing their effects with those of the voltage-dependent calcium channel (VDCC) blocker nifedpine. 2. HS elicited large, stable contractions that were partially dependent on the cellular chloride gradient; a reduction to 41.45+/-7.71% of the control response was obtained when extracellular chloride was removed. In addition, HS-induced responses were reduced to 26.8+/-5.6% of the control by 1 microm nifedipine, and abolished under calcium-free conditions, indicating a substantial requirement for extracellular calcium entry, principally via VDCCs. 3. The established Cl(vol) blockers tamoxifen (</=10 microm) and 4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid (1-100 microm), at concentrations previously reported to inhibit Cl(vol) in smooth muscle, did not significantly inhibit HS-induced contractions. 4. In contrast, the recognized Cl(Ca) blocker niflumic acid (NFA; 1-100 microm) produced a reversible, concentration-dependent inhibition of HS responses, with a reduction to 36.6+/-6.4% of control contractions at the highest concentration. The mixed Cl(vol) and Cl(Ca) blocker, 5-nitro 2-(3-phenylpropylamine) benzoic acid (NPPB; 10-100 microm) also elicited concentration-related inhibition of HS-induced contractions, producing a decrease to 35.9+/-11.3% of the control at 100 microm. 5. Our results show that HS induces reversible, chloride-dependent contractions of rat isolated trachea that were inhibited by NFA and NPPB, while exhibiting little sensitivity to recognized blockers of Cl(vol). The data support the possibility that opening of calcium-activated chloride channels under hypotonic conditions in respiratory smooth muscle may ultimately lead to VDCC-mediated calcium entry and contraction.
Collapse
Affiliation(s)
- Roberta R Coelho
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
| | - Emmanuel P Souza
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
| | - Pedro M G Soares
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
| | - Ana Vaneska P Meireles
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
| | - Geam C M Santos
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
| | - Henrique C Scarparo
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
- Departamento de Clínica Odontológica da Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará, R. Monsenhor Furtado w/n, Fortaleza CE 60441-750, Brazil
| | - Ana Maria S Assreuy
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
| | - David N Criddle
- Laboratório de Farmacologia dos Canais Iônicos – LAFACI, Departamento de Ciências Fisiológicas, CCS, Universidade Estadual do Ceará, Av. Paranjana 1700, Fortaleza CE 60740-000, Brazil
- MRC Secretory Control Research Group, The Physiological Laboratory, University of Liverpool, Crown Street, Liverpool L69 3BX
- Author for correspondence:
| |
Collapse
|
18
|
He J, Kargacin ME, Kargacin GJ, Ward CA. Tamoxifen inhibits Na+ and K+ currents in rat ventricular myocytes. Am J Physiol Heart Circ Physiol 2003; 285:H661-8. [PMID: 12702490 DOI: 10.1152/ajpheart.00686.2002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tamoxifen is an estrogen receptor antagonist used in the treatment of breast cancer. However, tamoxifen has been shown to induce QT prolongation of the electrocardiogram, thereby potentially causing life-threatening polymorphic ventricular arrhythmias. The purpose of the present study was to elucidate the electrophysiological mechanism(s) that underlie the arrhythmogenic effects of tamoxifen. We used standard ruptured whole cell and perforated patch-clamping techniques on rat ventricular myocytes to investigate the effects of tamoxifen on cardiac action potential (AP) waveforms and the underlying K+ currents. Tamoxifen (3 micromol/l) markedly prolonged AP duration, decreased maximal rate of depolarization, and decreased resting membrane potential. At this concentration, tamoxifen significantly depressed the Ca2+-independent transient outward K+ current (Ito), sustained outward delayed rectifier K+ current (Isus), inward rectifier K+ current (IK1), and Na+ current (INa) in the myocytes. Lower concentrations of tamoxifen (1 micromol/l) also decreased the resting membrane potential and significantly depressed IK1 to 79 +/- 5% (n = 5; at -120 mV) of pretreatment values. The results of this study indicate that inhibition of Ito, Isus, and IK1 by tamoxifen may underlie AP prolongation in cardiac myocytes and thereby contribute to prolonged QT interval observed in patients.
Collapse
Affiliation(s)
- Jianying He
- Department of Physiology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | | | | | |
Collapse
|
19
|
Arias HR, Kem WR, Trudell JR, Blanton MP. Unique general anesthetic binding sites within distinct conformational states of the nicotinic acetylcholine receptor. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 54:1-50. [PMID: 12785284 DOI: 10.1016/s0074-7742(03)54002-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
General anesthesia is a complex behavioral state provoked by the pharmacological action of a broad range of structurally different hydrophobic molecules called general anesthetics (GAs) on receptor members of the genetically linked ligand-gated ion channel (LGIC) superfamily. This superfamily includes nicotinic acetylcholine (AChRs), type A and C gamma-aminobutyric acid (GABAAR and GABACR), glycine (GlyR), and type 3 5-hydroxytryptamine (5-HT3R) receptors. This review focuses on recent advances in the localization of GA binding sites on conformationally and compositionally distinct AChRs. The experimental evidence outlined in this review suggests that: 1. Several neuronal-type AChRs might be targets for the pharmacological action of distinct GAs. 2. The molecular components of a specific GA binding site on a certain receptor subtype are different from the structural determinants of the locus for the same GA on a different receptor subtype. 3. There are unique binding sites for distinct GAs in the same receptor protein. 4. A GA can activate, potentiate, or inhibit an ion channel, indicating the existence of more than one binding site for the same GA. 5. The affinity of a specific GA depends on the conformational state of the receptor. 6. GAs inhibition channels by at least two mechanisms, an open-channel-blocking and/or an allosteric mechanism. 7. Certain GAs may inhibit AChR function by competing for the agonist binding sites or by augmenting the desensitization rate.
Collapse
Affiliation(s)
- Hugo R Arias
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California 91766, USA
| | | | | | | |
Collapse
|
20
|
Baumgarten CM, Clemo HF. Swelling-activated chloride channels in cardiac physiology and pathophysiology. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2003; 82:25-42. [PMID: 12732266 DOI: 10.1016/s0079-6107(03)00003-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Characteristics and functions of the cardiac swelling-activated Cl current (I(Cl,swell)) are considered in physiologic and pathophysiologic settings. I(Cl,swell) is broadly distributed throughout the heart and is stimulated not only by osmotic and hydrostatic increases in cell volume, but also by agents that alter membrane tension and direct mechanical stretch. The current is outwardly rectifying, reverses between the plateau and resting potentials (E(m)), and is time-independent over the physiologic voltage range. Consequently, I(Cl,swell) shortens action potential duration, depolarizes E(m), and acts to decrease cell volume. Because it is activated by stimuli that also activate cation stretch-activated channels, I(Cl,swell) should be considered as a potential effector of mechanoelectrical feedback. I(Cl,swell) is activated in ischemic and non-ischemic dilated cardiomyopathies and perhaps during ischemia and reperfusion. I(Cl,swell) plays a role in arrhythmogenesis, myocardial injury, preconditioning, and apoptosis of myocytes. As a result, I(Cl,swell) potentially is a novel therapeutic target.
Collapse
Affiliation(s)
- Clive M Baumgarten
- Department of Physiology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298-0551, USA. clive.baumgarten.vcu.edu
| | | |
Collapse
|
21
|
Rybalchenko V, Santos-Sacchi J. Cl- flux through a non-selective, stretch-sensitive conductance influences the outer hair cell motor of the guinea-pig. J Physiol 2003; 547:873-91. [PMID: 12562920 PMCID: PMC2342734 DOI: 10.1113/jphysiol.2002.036434] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Outer hair cells underlie high frequency cochlear amplification in mammals. Fast somatic motility can be driven by voltage-dependent conformational changes in the motor protein, prestin, which resides exclusively within lateral plasma membrane of the cell. Yet, how a voltage-driven motor could contribute to high frequency amplification, despite the low-pass membrane filter of the cell, remains an enigma. The recent identification of prestin's Cl- sensitivity revealed an alternative mechanism in which intracellular Cl- fluctuations near prestin could influence the motor. We report the existence of a stretch-sensitive conductance within the lateral membrane that passes anions and cations and is gated at acoustic rates. The resultant intracellular Cl- oscillations near prestin may drive motor protein transitions, as evidenced by pronounced shifts in prestin's state-probability function along the voltage axis. The sensitivity of prestin's state probability to intracellular Cl- levels betokens a more complicated role for Cl- than a simple extrinsic voltage sensor. Instead, we suggest an allosteric modulation of prestin by Cl- and other anions. Finally, we hypothesize that prestin sensitivity to anion flux through the mechanically activated lateral membrane can provide a driving force that circumvents the membrane's low-pass filter, thus permitting amplification at high acoustic frequencies.
Collapse
Affiliation(s)
- Volodymyr Rybalchenko
- Department of Surgery (Otolaryngology), Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | | |
Collapse
|
22
|
Chesnoy-Marchais D. Potentiation of glycine responses by dideoxyforskolin and tamoxifen in rat spinal neurons. Eur J Neurosci 2003; 17:681-91. [PMID: 12603258 DOI: 10.1046/j.1460-9568.2003.02481.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dideoxyforskolin, a forskolin analogue unable to stimulate adenylate cyclase, and tamoxifen, an antioestrogen widely used against breast cancer, are both known to block some Cl- channels. Their effects on Cl- responses to glycine or GABA have been tested here by using whole-cell recording from cultured spinal neurons. Dideoxyforskolin (4 or 16 microm) and tamoxifen (0.2-5 microm) both potentiate responses to low glycine concentrations. They also induce blocking effects, predominant at high glycine concentrations. At 5 microm, tamoxifen increased responses to 15 microm glycine by a factor >4.5, reaching 20 in some neurons. Potentiation by extracellular dideoxyforskolin or tamoxifen persisted after intracellular application of the modulator and was not due to Zn2+ contamination. Potentiation by tamoxifen also persisted in a Ca2+-free extracellular solution, after intracellular Ca2+ buffering and protein kinase C blockade. Thus, the critical sites of action are not intracellular. The EC50 for glycine was lowered 6.6-fold by 5 microm tamoxifen. The kinetics and voltage-dependence of the effects of tamoxifen on glycine responses support the idea that this hydrophobic drug may act from a site located within the membrane. Tamoxifen (5 micro m) also increased responses to 2 micro m GABA by a factor of 3.5, but barely affected peak responses to 20 microm GABA. The demonstration that tamoxifen affects some of the main inhibitory receptors should be useful for better evaluating its neurological effects. Furthermore, the results identify a new class of molecules that potentiate glycine receptor function.
Collapse
Affiliation(s)
- Dominique Chesnoy-Marchais
- Laboratoire de Neurobiologie Moléculaire et Cellulaire, CNRS UMR-8544, Ecole Normale Supérieure, 46 rue d'Ulm, 75005, Paris, France.
| |
Collapse
|
23
|
Díaz M. Triphenylethylene antiestrogen-induced acute relaxation of mouse duodenal muscle. Possible involvement of Ca2+ channels. Eur J Pharmacol 2002; 445:257-66. [PMID: 12079691 DOI: 10.1016/s0014-2999(02)01649-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The nonsteroidal antiestrogens tamoxifen, 4-OH-tamoxifen and toremifene rapidly inhibited spontaneous contractile activity and reduced basal tone in isolated mouse duodenal muscle. Inhibition was rapid in onset ( approximately 2 min) and was not mimicked by the pure steroidal antiestrogen 7alpha-[9-[(4,4,5,5,5-pentafluoropentyl)sulfinyl]nonyl]-estra-1,3,5(10)-triene-3,17beta-diol (ICI182,780) indicating the involvement of non-genomic mechanisms. Inhibition by tamoxifen and 4-OH-tamoxifen were observed at concentrations comparable to those reached in antiestrogen adjuvant therapy. Antiestrogen-relaxed tissues showed no response to KCl depolarisation or K(+) channel blockade but displayed clear transient responses to acethylcholine or to the muscarinic receptor agonist carbachol. Frequency analysis showed that spontaneous activity could be readily restored in antiestrogen-relaxed tissues by the exposure to the L-type Ca(2+) channel agonist 1,4-dihydro-2,6-dimethyl-5-nitro-4-[2-(trifluoromethyl)phenyl]-pyridine-3-carboxilic acid methyl ester (BAY K8644). Our experiments suggest that triphenylethylene antiestrogens relax duodenal intestinal muscle via a mechanism that involves inhibition of L-type Ca(2+) channels but not activation of K(+) channels.
Collapse
Affiliation(s)
- Mario Díaz
- Laboratorio de Fisiología Animal, Departamento de Biología Animal, Universidad de La Laguna, 38206 Tenerife, Spain.
| |
Collapse
|
24
|
Chen D, Wu CF, Shi B, Xu YM. Tamoxifen and toremifene impair retrieval, but not acquisition, of spatial information processing in mice. Pharmacol Biochem Behav 2002; 72:417-21. [PMID: 11900814 DOI: 10.1016/s0091-3057(01)00782-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present study examines the effects of tamoxifen (TAM) or toremifene (TOR), two triphenylethylene antiestrogen agents, on spatial information in mice by using Morris water maze. In a 30-s free swim trial, the TAM- or TOR-treated mice (intraperitoneally, 30 min before test) spent shorter time than the blank control mice in target quadrant. Compared to saline control group, animals exposed to TAM (1-10 mg/kg i.p., once a day for 5 days) or TOR (3-30 mg/kg i.p., once a day for 5 days) did not show significant difference on the acquisition of place task in Morris water maze. These results suggest that TAM, at the doses of 1-10 mg/kg, and TOR, at the doses of 3-30 mg/kg, impair the retrieval, but not the acquisition, of spatial information task in Morris water maze. It seems, however, that TOR is more potent than TAM on impairing memory retrieval.
Collapse
Affiliation(s)
- Duo Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110015, China
| | | | | | | |
Collapse
|
25
|
Dick GM, Hunter AC, Sanders KM. Ethylbromide tamoxifen, a membrane-impermeant antiestrogen, activates smooth muscle calcium-activated large-conductance potassium channels from the extracellular side. Mol Pharmacol 2002; 61:1105-13. [PMID: 11961128 DOI: 10.1124/mol.61.5.1105] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Smooth-muscle calcium-activated large-conductance potassium channels (BK channels) are activated by tamoxifen and 17-beta-estradiol. This increase in NP(o), the number of channels, N, multiplied by open probability, depends on the presence of the regulatory beta1-subunit. Furthermore, a previous study indicated that 17-beta-estradiol might bind an extracellular site on the beta1-subunit. Because tamoxifen and 17-beta-estradiol may share a common binding site, we hypothesized that tamoxifen activates BK channels through a site on the extracellular surface of the membrane. A membrane-impermeant analog of tamoxifen, ethylbromide tamoxifen, was synthesized and used to test this hypothesis in whole-cell, outside-out, cell-attached, and inside-out patches from canine colonic smooth muscle cells. Ethylbromide tamoxifen is positively charged and is therefore membrane-impermeant. In whole-cell experiments, ethylbromide tamoxifen increased K(+) current at potentials positive to +40 mV, which has previously been attributed to BK channels. Unlike tamoxifen, ethylbromide tamoxifen did not inhibit delayed rectifier current. In outside-out patches, ethylbromide tamoxifen increased BK channel NP(o) with an EC(50) value of 1 microM. Ethylbromide tamoxifen did not increase BK channel NP(o) in cell-attached or inside-out patches; however, subsequent addition of equimolar tamoxifen did. Both drugs diminished BK channel unitary conductance to a degree that paralleled the effect on NP(o), suggesting an additional interaction with the pore-forming alpha-subunit. An interaction of tamoxifen with the pore was supported by a right shift in the concentration-response curve for tetraethylammonium; similar results were evident with iberiotoxin and charybdotoxin block. Our data suggest that ethylbromide tamoxifen does not easily traverse the plasma membrane and that tamoxifen binding responsible for activation of BK channels is at an extracellular site. The tamoxifen binding site may be within the extracellular loop of the BK channel beta1-subunit or, alternatively, on an as-yet-unidentified mediator that has an extracellular binding site.
Collapse
Affiliation(s)
- Gregory M Dick
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA.
| | | | | |
Collapse
|
26
|
Chen D, Wu CF, Shi B, Xu YM. Tamoxifen and toremifene cause impairment of learning and memory function in mice. Pharmacol Biochem Behav 2002; 71:269-76. [PMID: 11812532 DOI: 10.1016/s0091-3057(01)00656-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Tamoxifen (TAM) and toremifene (TOR) are two antiestrogen agents frequently used in the treatment of breast cancer. They are currently being assessed as the prophylactic for patients at high risk of developing tumors. However, the side effects of these drugs on memory function have drawn attention in clinical usage. In the present study, it is demonstrated in mice that TAM and TOR significantly shortened the escaping latency or increased the number of errors, respectively, by using the step-down and step-through passive avoidance tests. By using an appetitively motivated task in T-maze, it is demonstrated that TAM and TOR significantly delayed the latency of finding food in well-trained mice. TAM appeared to impair memory consolidation and retrieval processes, rather than acquisition of memory, whereas TOR appeared to impair acquisition, consolidation, and retrieval processes. These results provide experimental support for the clinical findings that have shown that these drugs impaired memory function in patients routinely taking the drugs and suggest that caution should be taken for using these drugs as the prophylactics for those at risk of developing tumors.
Collapse
Affiliation(s)
- Duo Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110015, China
| | | | | | | |
Collapse
|
27
|
Best L. Inhibition of glucose-induced electrical activity by 4-hydroxytamoxifen in rat pancreatic beta-cells. Cell Signal 2002; 14:69-73. [PMID: 11747991 DOI: 10.1016/s0898-6568(01)00223-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The antioestrogen 4-hydroxytamoxifen (10 or 2 microM) abolished the generation of action potentials and repolarized the membrane potential in rat pancreatic beta-cells stimulated by 16 mM glucose. This effect was slowly reversible upon withdrawal of the drug. In cells stimulated by tolbutamide (100 microM), application of 4-hydroxytamoxifen again inhibited action-potential generation but failed to repolarize the membrane potential. 4-Hydroxytamoxifen inhibited voltage-sensitive calcium currents and activity of the volume-sensitive anion channel. The drug had no effect on net K(+) conductance of the cell. Insulin release stimulated by either glucose or tolbutamide was inhibited by 4-hydroxytamoxifen. It is concluded that 4-hydroxytamoxifen impairs beta-cell electrical and secretory activity by inhibiting calcium and anion channel currents. This effect could contribute towards hyperglycaemia during therapy with tamoxifen, of which 4-hydroxytamoxifen is the major metabolite. This study also reveals differences between the depolarizing actions of glucose and tolbutamide in the beta-cell.
Collapse
Affiliation(s)
- Leonard Best
- Department of Medicine, University of Manchester, Oxford Road, Manchester M13 9WL, UK.
| |
Collapse
|
28
|
Dick GM, Sanders KM. (Xeno)estrogen sensitivity of smooth muscle BK channels conferred by the regulatory beta1 subunit: a study of beta1 knockout mice. J Biol Chem 2001; 276:44835-40. [PMID: 11590153 DOI: 10.1074/jbc.m106851200] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Estrogen and xenoestrogens (i.e. agents that are not steroids but possess estrogenic activity) increase the open probability (P(o)) of large conductance Ca(2+)-activated K(+) (BK) channels in smooth muscle. The mechanism of action may involve the regulatory beta1 subunit. We used beta1 subunit knockout (beta1-/-) mice to test the hypothesis that the regulatory beta1 subunit is essential for the activation of BK channels by tamoxifen, 4-OH tamoxifen (a major biologically active metabolite), and 17beta-estradiol in native myocytes. Patch clamp recordings demonstrate BK channels from beta1-/- mice were similar to wild type with the exception of markedly reduced Ca(2+)/voltage sensitivity and faster activation kinetics. In wild type myocytes, (xeno)estrogens increased NP(o) (P(o) x the number of channels, N), shifted the voltage of half-activation (V(12)) to more negative potentials, and decreased unitary conductance. These effects were non-genomic and direct, because they were rapid, reversible, and observed in cell-free patches. None of the (xeno)estrogens increased the NP(o) of BK channels from beta1-/- mice, but all three agents decreased single channel conductance. Thus, (xeno)estrogens increase BK NP(o) through a mechanism involving the beta1 subunit. The decrease in conductance did not require the beta1 subunit and probably reflects an interaction with the pore-forming alpha subunit. We demonstrate regulation of smooth muscle BK channels by physiological (steroid hormones) and pharmacological (chemotherapeutic) agents and reveal the critical role of the beta1 subunit in these responses in native myocytes.
Collapse
Affiliation(s)
- G M Dick
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA.
| | | |
Collapse
|
29
|
Nakazawa K, Ohno Y. Modulation by estrogens and xenoestrogens of recombinant human neuronal nicotinic receptors. Eur J Pharmacol 2001; 430:175-83. [PMID: 11711029 DOI: 10.1016/s0014-2999(01)01389-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The effects of estrogens and xenoestrogens on human neuronal nicotinic acetylcholine receptor/channels were examined by expressing recombinant channels in Xenopus oocytes. When functional channels were expressed with alpha3 and beta4 subunits, estrogens (17beta-estradiol, 17alpha-estradiol, 17alpha-ethynylestradiol and diethylstilbestrol) and xenoestrogens (bisphenol A, p-nonylphenol and p-octylphenol) inhibited an ionic current activated by acetylcholine at concentrations up to 100 microM. When the subunit combination was changed to alpha4beta2, diethystilbestrol and the xenoestrogens inhibited the acetylcholine-activated current, but 17beta-estradiol or 17alpha-estradiol did not. For 17alpha-ethynylestradiol, the current through the alpha4beta2 receptor/channel was inhibited at 1 microM, but it was markedly enhanced at 10 and 100 microM. Tamoxifen (10 microM), an antiestrogen, itself inhibited the acetylcholine-activated current but did not antagonize the current modulations induced by the estrogens and the xenoestrogens. These and additional results suggest that human neuronal nicotinic acetylcholine receptors are the targets of non-genomic actions of estrogens and xenoestrogens.
Collapse
Affiliation(s)
- K Nakazawa
- Division of Pharmacology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya, 158-8501, Tokyo, Japan.
| | | |
Collapse
|
30
|
Dick GM, Rossow CF, Smirnov S, Horowitz B, Sanders KM. Tamoxifen activates smooth muscle BK channels through the regulatory beta 1 subunit. J Biol Chem 2001; 276:34594-9. [PMID: 11454866 DOI: 10.1074/jbc.m104689200] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Estrogen (17beta-estradiol; 17betaE) and xenoestrogens, estrogenic compounds that are not steroid hormones, have non-genomic actions at plasma membrane receptors unrelated to the nuclear estrogen receptor. The open probability (P(o)) of large conductance Ca(2+)/voltage-sensitive k(+)(BK) channels is increased by 17betaE through the regulatory beta1 subunit. The pharmacological nature of the putative membrane binding site is unclear. We probed the site by determining whether tamoxifen ((Z)-1-(p-dimethylaminoethoxy-phenyl)-1,2-diphenyl-1-butene; Tx), a chemotherapeutic xenoestrogen, increased P(o) in clinically relevant concentrations (0.1-10 microm). In whole cell patch clamp recordings on canine colonic myocytes, which express the beta1 subunit, Tx activated charybdotoxin-sensitive K(+) current. In single channel experiments, Tx increased the NP(o) (P(o) x number channels; N) and decreased the unitary conductance (gamma) of BK channels. Tx increased NP(o) (EC(50) = 0.65 microm) in excised membrane patches independent of Ca(2+) changes. The Tx mechanism of action requires the beta1 subunit, as Tx increased the NP(o) of Slo alpha expressed in human embryonic kidney cells only in the presence of the beta1 subunit. Tx decreased gamma of the alpha subunit expressed alone, without effect on NP(o). Our data indicate that Tx increases BK channel activity in therapeutic concentrations and reveal novel pharmacological properties attributable to the alpha and beta1 subunits. These data shed light on BK channel structure and function, non-genomic mechanisms of regulation, and physiologically and therapeutically relevant effects of xenoestrogens.
Collapse
Affiliation(s)
- G M Dick
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA.
| | | | | | | | | |
Collapse
|
31
|
Vitális B, Sebestyén L, Sike M, Sólyom S, Hársing LG. Binding characteristics of GYKI-46 903, a non-competitive ligand at 5-HT3 receptors. Pharmacol Res 2001; 43:291-9. [PMID: 11401422 DOI: 10.1006/phrs.2000.0774] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
GYKI-46903 [(+)-(5S,6R)-4-(4-fluorophenyl)-6-propionyloxy-1-aza-bicyclo[3.3.1]-non-3-ene-hydrochloride], a cognition enhancer identified as a non-competitive antagonist of 5-HT3receptors in isolated guinea-pig ileum, was investigated for allosteric action at 5-HT3 receptors in rat cortical membranes by using [3H]granisetron. Equilibrium and kinetic protocols were applied and the competitive antagonist granisetron was included as a negative control. In competition studies, both granisetron and GYKI-46 903 displaced the radioligand with K(i) values of 0.20 +/- 0.02 and 79.84 +/- 0.28 nM, respectively. The inhibition curve for GYKI-46 903 resulted in a Hill slope significantly greater than unity ( 1.37 +/- 0.11), whereas the slope for granisetron was 0.88 +/- 0.08, not different from unity. These results indicate non-competitive and competitive interactions, respectively. Scatchard analysis yielded a linear plot, suggesting a single population of binding sites with a Kd of 0.13 +/- 0.01 nM and a Bmax) of 13.15 +/- 0.34 fmol per mg of protein. Scatchard plots obtained in the absence and presence of granisetron (0.1-3 nM) or GYKI-46 903 (30-1000 nM) revealed a concentration-dependent increase in Kd values by either of these compounds. Granisetron left the Bmax unchanged, but there was a significant increase in the Bmax by GYKI-46 903, which could point to an atypical allosteric interaction. The Schild plot derived from the Kd shifts induced by granisetron was linear with a slope of 1.02, not different from unity, as expected from a competitive interaction. The Schild regression for GYKI-46 903 was linear with a slope of 1.20, deviating significantly from unity, which may also indicate an allosteric interaction. Both the association and dissociation curves of [3H]granisetron were monoexponential. The dissociation rate constant (K(-1)) and the association rate constant (K(+1)) were 0.32 +/- 0.01 min(-1) and 1.15 min(-1) x nM(-1), respectively. The dissociation driven by an excess concentration of ondansetron ( 1 microM) in the absence and presence of granisetron (0.1-3 nM) or GYKI-46 903 (30-10 000 nM) was not influenced by the compounds under study, as compared with the control, indicating the lack of an allosteric effect on the dissociation. Summing up, the binding profile of GYKI-46 903 may reflect a mixed type of action, including a negative allosteric interaction.
Collapse
Affiliation(s)
- B Vitális
- Institute for Drug Research Ltd., Budapest, Hungary
| | | | | | | | | |
Collapse
|
32
|
Kairies P. Tamoxifen and risk of endometrial cancer. Lancet 2001; 357:68. [PMID: 11197381 DOI: 10.1016/s0140-6736(05)71566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
33
|
Allen MC, Gale PA, Hunter AC, Lloyd A, Hardy SP. Membrane impermeant antioestrogens discriminate between ligand- and voltage-gated cation channels in NG108-15 cells. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1509:229-36. [PMID: 11118534 DOI: 10.1016/s0005-2736(00)00297-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Native 5-HT(3) and AChR ligand-gated cation channels can be inhibited (blocked) by the non-steroidal antioestrogen tamoxifen. However, the exact site and mechanism of inhibition by tamoxifen on these channels remain unclear. We have investigated the action of the membrane impermeant quaternary derivative, ethylbromide tamoxifen (EBT), on native ligand-gated 5-HT(3) receptor channels and voltage-gated K(+) channels in NG108-15 cells using whole cell patch clamp. Extracellular EBT inhibited whole cell cationic currents of 5-HT(3) receptors with IC(50) of 0.22+/-0.4 microM (n(H)=1.05+/-0.2). The channel block was characterised by voltage independent and use independent behaviour (similar to that of tamoxifen). EBT was unable to inhibit voltage-gated K(+) currents in NG108-15 cells. This was in contrast to the inhibition by tamoxifen which, at similar concentrations, accelerated the apparent inactivation of these outward K(+) currents. The inhibition of 5-HT(3) receptors by a membrane impermeant derivative of tamoxifen supports the view that the binding site for antioestrogens is extracellular and the inhibition is not mediated through genomic/transcriptional activity.
Collapse
Affiliation(s)
- M C Allen
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Lewes Road, BN2 4GJ, Brighton, UK
| | | | | | | | | |
Collapse
|
34
|
Welsh DG, Nelson MT, Eckman DM, Brayden JE. Swelling-activated cation channels mediate depolarization of rat cerebrovascular smooth muscle by hyposmolarity and intravascular pressure. J Physiol 2000; 527 Pt 1:139-48. [PMID: 10944177 PMCID: PMC2270055 DOI: 10.1111/j.1469-7793.2000.t01-1-00139.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
1. Increases in intravascular pressure depolarize vascular smooth muscle cells. Based on the attenuating effects of Cl- channel antagonists, it has been suggested that swelling-activated Cl- channels may be integral to this response. Consequently, this study tested for the presence of a swelling-activated Cl- conductance in both intact rat cerebral arteries and isolated rat smooth muscle cells. 2. A 50 mosmol l-1 hyposmotic challenge (300 to 250 mosmol l-1) constricted rat cerebral arteries. This constriction contained all the salient features of a pressure-induced response including smooth muscle cell depolarization and a rise in intracellular Ca2+ that was blocked by voltage-operated Ca2+ channel antagonists. The hyposmotically induced depolarization was attenuated by DIDS (300 microM) and tamoxifen (1 microM), a response consistent with the presence of a swelling-activated Cl- conductance. 3. A swelling-activated current was identified in cerebral vascular smooth muscle cells. This current was sensitive to Cl- channel antagonists including DIDS (300 microM), tamoxifen (1 microM) and IAA-94 (100 microM). However, contrary to expectations, the reversal potential of this swelling-activated current shifted with the Na+ equilibrium potential and not the Cl- equilibrium potential, indicating that the swelling-activated current was carried by cations and not anions. The swelling-activated cation current was blocked by Gd3+, a cation channel antagonist. 4. Gd3+ also blocked both swelling- and pressure-induced depolarization of smooth muscle cells in intact cerebral arteries. 5. These findings suggest that swelling- and pressure-induced depolarization arise from the activation of a cation conductance. This current is inhibited by DIDS, tamoxifen, IAA-94 and gadolinium.
Collapse
Affiliation(s)
- D G Welsh
- Department of Pharmacology, University of Vermont, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
35
|
Allen MC. Agonist and antagonist effects of histamine H3 receptor ligands on 5-HT3 receptor-mediated ion currents in NG108-15 cells. Eur J Pharmacol 1998; 361:261-8. [PMID: 9865516 DOI: 10.1016/s0014-2999(98)00719-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The ability of histamine H3 receptor ligands to interact with 5-HT3 receptors in NG108-15 cells was studied using the whole cell patch clamp recording technique. Imetit, a histamine H3 receptor agonist, generated inward currents and exhibited weak partial agonist activity at the 5-HT3 receptor (EC50 = 11.8 microM). Imetit-induced currents were slow to desensitize and at a high concentration reduced in size. The histamine H3 receptor antagonists iodophenpropit and thioperamide did not generate inward currents but were able to inhibit 5-hydroxytryptamine (5-HT) responses with an IC50 of 1.57+/-0.3 microM and 13.7+/-3.5 microM, respectively. Thioperamide is probably a non-competitive antagonist which may have more than one binding site on the receptor.
Collapse
Affiliation(s)
- M C Allen
- Department of Pharmacy, University of Brighton, UK
| |
Collapse
|