1
|
Hou L, Liu Y, Sun C, Xu R, Cao G, Wang X. Novel Perspective of Cardiovascular Diseases: Volume-Regulatory Anion Channels in the Cell Membrane. MEMBRANES 2022; 12:membranes12070644. [PMID: 35877847 PMCID: PMC9324040 DOI: 10.3390/membranes12070644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/09/2022] [Accepted: 06/18/2022] [Indexed: 11/16/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Although there are established mechanisms and preventions for CVDs, they are not totally elucidative and effective. Emerging evidence suggests that the dysregulation of ion channels in the cell membranes underpins the dysfunction of the cardiovascular system. To date, a variety of cation channels have been widely recognized as important targets for the treatment of CVDs. As a critical component of the anion channels, the volume-regulated anion channel (VRAC) is involved in a series of cell functions by the volume regulation and maintenance of membrane homeostasis. It has been confirmed to play crucial roles in cell action potential generation, cell proliferation, differentiation and apoptosis, and the VRAC appears to be a major participant in metabolic processes during CVDs. This review summarizes the current evidence and progress concerning the VRAC, to determine the future directions and challenges for CVDs for both preventive and therapeutic purposes.
Collapse
|
2
|
Drug development in targeting ion channels for brain edema. Acta Pharmacol Sin 2020; 41:1272-1288. [PMID: 32855530 PMCID: PMC7609292 DOI: 10.1038/s41401-020-00503-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/02/2020] [Indexed: 12/18/2022] Open
Abstract
Cerebral edema is a pathological hallmark of various central nervous system (CNS) insults, including traumatic brain injury (TBI) and excitotoxic injury such as stroke. Due to the rigidity of the skull, edema-induced increase of intracranial fluid significantly complicates severe CNS injuries by raising intracranial pressure and compromising perfusion. Mortality due to cerebral edema is high. With mortality rates up to 80% in severe cases of stroke, it is the leading cause of death within the first week. Similarly, cerebral edema is devastating for patients of TBI, accounting for up to 50% mortality. Currently, the available treatments for cerebral edema include hypothermia, osmotherapy, and surgery. However, these treatments only address the symptoms and often elicit adverse side effects, potentially in part due to non-specificity. There is an urgent need to identify effective pharmacological treatments for cerebral edema. Currently, ion channels represent the third-largest target class for drug development, but their roles in cerebral edema remain ill-defined. The present review aims to provide an overview of the proposed roles of ion channels and transporters (including aquaporins, SUR1-TRPM4, chloride channels, glucose transporters, and proton-sensitive channels) in mediating cerebral edema in acute ischemic stroke and TBI. We also focus on the pharmacological inhibitors for each target and potential therapeutic strategies that may be further pursued for the treatment of cerebral edema.
Collapse
|
3
|
Warcoin E, Clouzeau C, Brignole-Baudouin F, Baudouin C. Hyperosmolarité : effets intracellulaires et implication dans la sécheresse oculaire. J Fr Ophtalmol 2016; 39:641-51. [DOI: 10.1016/j.jfo.2016.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/27/2016] [Accepted: 07/27/2016] [Indexed: 11/26/2022]
|
4
|
Horger KS, Liu H, Rao DK, Shukla S, Sept D, Ambudkar SV, Mayer M. Hydrogel-assisted functional reconstitution of human P-glycoprotein (ABCB1) in giant liposomes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:643-53. [PMID: 25450342 DOI: 10.1016/j.bbamem.2014.10.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 09/17/2014] [Accepted: 10/20/2014] [Indexed: 11/24/2022]
Abstract
This paper describes the formation of giant proteoliposomes containing P-glycoprotein (P-gp) from a solution of small proteoliposomes that had been deposited and partially dried on a film of agarose. This preparation method generated a significant fraction of giant proteoliposomes that were free of internalized vesicles, making it possible to determine the accessible liposome volume. Measuring the intensity of the fluorescent substrate rhodamine 123 (Rho123) inside and outside these giant proteoliposomes determined the concentration of transported substrates of P-gp. Fitting a kinetic model to the fluorescence data revealed the rate of passive diffusion as well as active transport by reconstituted P-gp in the membrane. This approach determined estimates for the membrane permeability coefficient (Ps) of passive diffusion and rate constants of active transport (kT) by P-gp as a result of different experimental conditions. The Ps value for Rho123 was larger in membranes containing P-gp under all assay conditions than in membranes without P-gp indicating increased leakiness in the presence of reconstituted transmembrane proteins. For P-gp liposomes, the kT value was significantly higher in the presence of ATP than in its absence or in the presence of ATP and the competitive inhibitor verapamil. This difference in kT values verified that P-gp was functionally active after reconstitution and quantified the rate of active transport. Lastly, patch clamp experiments on giant proteoliposomes showed ion channel activity consistent with a chloride ion channel protein that co-purified with P-gp. Together, these results demonstrate several advantages of using giant rather than small proteoliposomes to characterize transport properties of transport proteins and ion channels.
Collapse
Affiliation(s)
- Kim S Horger
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI 48109, USA
| | - Haiyan Liu
- Department of Biomedical Engineering, University of Michigan, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Divya K Rao
- Department of Biomedical Engineering, University of Michigan, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Suneet Shukla
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20814, USA
| | - David Sept
- Department of Biomedical Engineering, University of Michigan, 1101 Beal Avenue, Ann Arbor, MI 48109, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20814, USA
| | - Michael Mayer
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, 1101 Beal Avenue, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Swelling-activated Cl− currents and intracellular CLC-3 are involved in proliferation of human pulmonary artery smooth muscle cells. J Hypertens 2014; 32:318-30. [DOI: 10.1097/hjh.0000000000000013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
6
|
Duran C, Chien LT, Hartzell HC. Drosophila bestrophin-1 currents are regulated by phosphorylation via a CaMKII dependent mechanism. PLoS One 2013; 8:e58875. [PMID: 23554946 PMCID: PMC3595206 DOI: 10.1371/journal.pone.0058875] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 02/07/2013] [Indexed: 11/19/2022] Open
Abstract
Cell swelling induced by hypo-osmotic stress results in activation of volume-regulated anion channels (VRAC) that drive a compensatory regulatory volume decrease. We have previously shown that the Best1 gene in Drosophila encodes a VRAC that is also activated by increases in intracellular Ca2+. The role of Best1 as a VRAC has recently been independently confirmed by the Clapham lab in an unbiased RNAi screen. Although dBest1 is clearly a volume-regulated channel, its mechanisms of regulation remain unknown. Here we investigate Drosophila Best1 (dBest1) regulation using the Drosophila S2 cell model system. Because dBest1 activates slowly after establishing whole-cell recording, we tested the hypothesis that the channel is activated by phosphorylation. Two experiments indicate that phosphorylation is required for dBest1 activation: nonspecific protein kinase inhibitors or intracellular perfusion with the non-hydrolyzable ATP analog AMP-PNP dramatically reduce the amplitude of dBest1 currents. Furthermore, intracellular perfusion with ATP-γ-S augments channel activation. The kinase responsible for dBest1 activation is likely Ca2+/calmodulin dependent kinase II (CaMKII), because specific inhibitors of this kinase dramatically inhibit dBest1 current activation. Neither specific PKA inhibitors nor inactive control inhibitors have effects on dBest1currents. Our results demonstrate that dBest1 currents are regulated by phosphorylation via a CaMKII dependent mechanism.
Collapse
Affiliation(s)
- Charity Duran
- Department of Cell Biology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Li-Ting Chien
- Department of Cell Biology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - H. Criss Hartzell
- Department of Cell Biology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
7
|
Anion-sensitive fluorophore identifies the Drosophila swell-activated chloride channel in a genome-wide RNA interference screen. PLoS One 2012; 7:e46865. [PMID: 23056495 PMCID: PMC3464265 DOI: 10.1371/journal.pone.0046865] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 09/06/2012] [Indexed: 12/21/2022] Open
Abstract
When cells swell in hypo-osmotic solutions, chloride-selective ion channels (Cl(swell)) activate to reduce intracellular osmolality and prevent catastrophic cell rupture. Despite intensive efforts to assign a molecular identity to the mammalian Cl(swell) channel, it remains unknown. In an unbiased genome-wide RNA interference (RNAi) screen of Drosophila cells stably expressing an anion-sensitive fluorescent indicator, we identify Bestrophin 1 (dBest1) as the Drosophila Cl(swell) channel. Of the 23 screen hits with mammalian homologs and predicted transmembrane domains, only RNAi specifically targeting dBest1 eliminated the Cl(swell) current (I(Clswell)). We further demonstrate the essential contribution of dBest1 to Drosophila I(Clswell) with the introduction of a human Bestrophin disease-associated mutation (W94C). Overexpression of the W94C construct in Drosophila cells significantly reduced the endogenous I(Clswell). We confirm that exogenous expression of dBest1 alone in human embryonic kidney (HEK293) cells creates a clearly identifiable Drosophila-like I(Clswell). In contrast, activation of mouse Bestrophin 2 (mBest2), the closest mammalian ortholog of dBest1, is swell-insensitive. The first 64 residues of dBest1 conferred swell activation to mBest2. The chimera, however, maintains mBest2-like pore properties, strongly indicating that the Bestrophin protein forms the Cl(swell) channel itself rather than functioning as an essential auxiliary subunit. dBest1 is an anion channel clearly responsive to swell; this activation depends upon its N-terminus.
Collapse
|
8
|
Mcferrin MB, Sontheimer H. A role for ion channels in glioma cell invasion. NEURON GLIA BIOLOGY 2012; 2:39-49. [PMID: 16520829 PMCID: PMC1389710 DOI: 10.1017/s17440925x06000044] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Many cells, including neuronal and glial progenitor cells, stem cells and microglial cells, have the capacity to move through the extracellular spaces of the developing and mature brain. This is particularly pronounced in astrocyte-derived tumors, gliomas, which diffusely infiltrate the normal brain. Although a significant body of literature exists regarding signals that are involved in the guidance of cells and their processes, little attention has been paid to cell-shape and cell-volume changes of migratory cells. However, extracellular spaces in the brain are very narrow and represent a major obstacle that requires cells to dynamically regulate their volume. Recent studies in glioma cells show that this involves the secretion of Cl(-) and K(+) with water. Pharmacological inhibition of Cl(-) channels impairs their ability to migrate and limits tumor progression in experimental tumor models. One Cl(-)-channel inhibitor, chlorotoxin, is currently in Phase II clinical trials to treat malignant glioma. This article reviews our current knowledge of cell-volume changes and the role of ion channels during the migration of glioma cells. It also discusses evidence that supports the importance of channel-mediated cell-volume changes in the migration of immature neurons and progenitor cells during development. New unpublished data is presented, which demonstrates that Cl(-) and K(+) channels involved in cell shrinkage localize to lipid-raft domains on the invadipodia of glioma cells and that their presence might be regulated by trafficking of these proteins in and out of lipid rafts.
Collapse
Affiliation(s)
| | - Harald Sontheimer
- Correspondence should be addressed to: Harald Sontheimer, Ph.D. Department of Neurobiology, The University of Alabama at Birmingham 1119 6th Ave S. CIRC 545 USA phone: +1 205 975 5805 fax: +1 205 975 5518
| |
Collapse
|
9
|
Fuchigami T, Matsuzaki T, Ihara S. Possible Roles of ENaC and Cl-Channel in Wound Closure inXenopus laevisEmbryos. Zoolog Sci 2011; 28:703-11. [DOI: 10.2108/zsj.28.703] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
10
|
Mao J, Xu B, Li H, Chen L, Jin X, Zhu J, Wang W, Zhu L, Zuo W, Chen W, Wang L. Lack of association between stretch-activated and volume-activated Cl− currents in hepatocellular carcinoma cells. J Cell Physiol 2011; 226:1176-85. [DOI: 10.1002/jcp.22443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
11
|
Kolbaev SN, Luhmann HJ, Kilb W. Activity-dependent scaling of GABAergic excitation by dynamic Cl- changes in Cajal-Retzius cells. Pflugers Arch 2011; 461:557-65. [PMID: 21336585 DOI: 10.1007/s00424-011-0935-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/04/2011] [Accepted: 02/04/2011] [Indexed: 11/29/2022]
Abstract
To unravel the functional implications of activity-dependent Cl- changes during early stages of neuronal development, we determined which changes in the GABA reversal potential (E (GABA)) and GABAergic rheobase shifts were induced by episodes of GABA(A) receptor activation using gramicidin-perforated patch-clamp recordings from Cajal-Retzius cells in tangential cortical slices of newborn mice. Under this condition, focal application of the GABA(A) agonist muscimol (10 μM) depolarized the membrane by 15 ± 0.8 mV (n = 35). Such subthreshold GABAergic depolarizations considerably reduced the rheobase, corresponding to an excitatory action. After repetitive focal muscimol applications (50 pulses at 0.5 Hz) a significant reduction of E (GABA) and an attenuation of the excitatory GABAergic rheobase shift were observed, while the GABAergic membrane conductance and the absolute value of the rheobase were unaltered after the muscimol pulses. Bath application of 100 μM carbachol induced bursts of spontaneous GABAergic postsynaptic potentials. Both, E (GABA) and the excitatory GABAergic rheobase shift was significantly reduced after such barrage of carbachol-induced GABAergic postsynaptic potentials, while neither the GABAergic membrane conductance nor the absolute value of the rheobase was affected under this condition. Both results indicate that GABAergic activity itself can limit the excitatory effects of GABA(A) receptor activation, which supports the hypothesis that the low capacity of the Cl- homeostasis in immature neurons could be a substrate for synaptic scaling and homeostatic plasticity.
Collapse
Affiliation(s)
- Sergey N Kolbaev
- Institute of Physiology and Pathophysiology, University Medical Center Mainz, Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany
| | | | | |
Collapse
|
12
|
Du RH, Tang YB, Zhou JG, Guan YY. Effects of Tween 80 on volume-regulated chloride channel and cell proliferation in rat basilar artery smooth muscle cell. J Pharm Pharmacol 2011; 63:253-60. [DOI: 10.1111/j.2042-7158.2010.01209.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Abstract
Objectives
We have previously found that volume-regulated chloride current (VRCC) is involved in cell cycle progression and cell proliferation. This study was to examine the effect of Tween 80, a nonionic surfactant, on VRCC and cell proliferation in rat basilar artery smooth muscle cells (BASMCs).
Methods
VRCC was recorded using a whole-cell patch clamp. Cell proliferation and cell cycle were determined by CCK-8, cell count and flow cytometry.
Key findings
The results showed that endothelin-1 promotes cell cycle transition from the G0/G1 phase to the S phase and significantly increases VRCC in BASMCs. The effect of Tween 80 on VRCC is reversible and concentration dependent. However, this chemical has no effect on the calcium-activated chloride channel. Tween 80 also concentration-dependently inhibits BASMCs proliferation and arrests cells in the G1/S checkpoint. The antiproliferative effect is paralleled with the inhibitory effect on VRCC.
Conclutision
Our study demonstrates that the inhibitory effect of Tween 80 on VRCC contributes importantly to arrest of the cell cycle and prevention of cell proliferation.
Collapse
Affiliation(s)
- Ren-Hong Du
- Department of Pharmacology, Zhongshan School of Medicine, and Vascular Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Yong-Bo Tang
- Department of Pharmacology, Zhongshan School of Medicine, and Vascular Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Jia-Guo Zhou
- Department of Pharmacology, Zhongshan School of Medicine, and Vascular Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Yong-Yuan Guan
- Department of Pharmacology, Zhongshan School of Medicine, and Vascular Research Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
13
|
Terhag J, Cavara NA, Hollmann M. Cave Canalem: How endogenous ion channels may interfere with heterologous expression in Xenopus oocytes. Methods 2010; 51:66-74. [DOI: 10.1016/j.ymeth.2010.01.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 01/26/2010] [Accepted: 01/27/2010] [Indexed: 10/19/2022] Open
|
14
|
Inoue H, Takahashi N, Okada Y, Konishi M. Volume-sensitive outwardly rectifying chloride channel in white adipocytes from normal and diabetic mice. Am J Physiol Cell Physiol 2010; 298:C900-9. [PMID: 20107039 DOI: 10.1152/ajpcell.00450.2009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The volume-sensitive outwardly rectifying (VSOR) chloride channel is ubiquitously expressed and involved in cell volume regulation after osmotic swelling, called regulatory volume decrease (RVD), in various cell types. In adipocytes, the expression of the VSOR channel has not been explored to date. Here, by employing the whole-cell patch-clamp technique, we examined whether or not the VSOR channel is expressed in white adipocytes freshly isolated from epididymal fat pads of normal (C57BL/6 or KK) and diabetic (KKA(y)) mice. Whole cell voltage-clamp recordings revealed that Cl(-) currents were gradually activated upon cell swelling induced by application of a hypotonic solution, both in normal and diabetic adipocytes. Although both the mean cell size (or cell capacitance) and the current magnitude in KKA(y) adipocytes were larger than those in C57BL/6 cells, the current density was significantly lower in KKA(y) adipocytes (23.32 +/- 1.94 pA in C57BL/6 adipocytes vs. 13.04 +/- 2.41 pA in KKA(y) adipocytes at +100 mV). Similarly, the current density in diabetic KKA(y) adipocytes was lower than that in adipocytes from KK mice (a parental strain of KKA(y) mice), which do not present diabetes until an older age. The current was inhibited by Cl(-) channel blockers, 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB) and glibenclamide, or hypertonic solution, and showed outward rectification and inactivation kinetics at large positive potentials. These electrophysiological and pharmacological properties are consistent with those of the VSOR channel in other cell types. Moreover, adipocytes showed RVD, which was inhibited by NPPB. In KKA(y) adipocytes, RVD was significantly slower (tau; 8.42 min in C57BL/6 adipocytes vs. 11.97 min in KKA(y) adipocytes) and incomplete during the recording period (25 min). It is concluded that the VSOR channel is functionally expressed and involved in volume regulation in white adipocytes. RVD is largely impaired in adipocytes from diabetic mice, presumably as a consequence of the lower density of the functional VSOR channel in the plasma membrane.
Collapse
Affiliation(s)
- Hana Inoue
- Dept. of Physiology, Tokyo Medical Univ., 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan.
| | | | | | | |
Collapse
|
15
|
Abstract
In mammalian brain, neurons and astrocytes are reported to express various chloride and anion channels, but the evidence for functional expression of Ca(2+)-activated anion channel (CAAC) and its molecular identity have been lacking. Here we report electrophysiological evidence for the CAAC expression and its molecular identity by mouse Bestrophin 1 (mBest1) in astrocytes of the mouse brain. Using Ca(2+) imaging and perforated-patch-clamp analysis, we demonstrate that astrocytes displayed an inward current at holding potential of -70 mV that was dependent on an increase in intracellular Ca(2+) after G(alphaq)-coupled receptor activation. This current was mediated mostly by anions and was sensitive to well known anion channel blockers such as niflumic acid, 5-nitro-2(3-phenylpropylamino)-benzoic acid, and flufenamic acid. To find the molecular identity of the anion channel responsible for the CAAC current, we analyzed the expression of candidate genes and found that the mRNA for mouse mBest1 is predominantly expressed in acutely dissociated or cultured astrocytes. Whole-cell patch-clamp analysis using HEK293T cells heterologously expressing full-length mBest1 showed a Ca(2+)-dependent current mediated by mBest1, with a complete impairment of the current by a putative pore mutation, W93C. Furthermore, mBest1-mediated CAAC from cultured astrocytes was significantly reduced by expression of mBest1-specific short hairpin RNA (shRNA), suggesting that the CAAC is mediated by a channel encoded by mBest1. Finally, hippocampal CA1 astrocytes in hippocampal slice also showed mBest1-mediated CAAC because it was inhibited by mBest1-specific shRNA. Collectively, these data provide molecular evidence that the mBest1 channel is responsible for CAAC function in astrocytes.
Collapse
|
16
|
Li Y, O'Neill C, Day ML. Activation of a Chloride Channel by a Trophic Ligand Is Required for Development of the Mouse Preimplantation Embryo In Vitro1. Biol Reprod 2009; 81:759-67. [DOI: 10.1095/biolreprod.108.074567] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
17
|
Okada Y, Sato K, Numata T. Pathophysiology and puzzles of the volume-sensitive outwardly rectifying anion channel. J Physiol 2009; 587:2141-9. [PMID: 19171657 PMCID: PMC2697288 DOI: 10.1113/jphysiol.2008.165076] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Accepted: 01/22/2009] [Indexed: 11/08/2022] Open
Abstract
Cell swelling activates or upregulates a number of anion channels. Of the volume-activated or -regulated anion channels (VAACs or VRACs), the volume-sensitive outwardly rectifying anion channel (VSOR) is most prominently activated and ubiquitously expressed. This channel is known to be involved in a variety of physiological processes including cell volume regulation, cell proliferation, differentiation and cell migration as well as cell turnover involving apoptosis. Recent studies have shown that VSOR activity is also involved in a number of pathophysiological processes including the acquisition of cisplatin resistance by cancer cells, ischaemia-reperfusion-induced death of cardiomyocytes and hippocampal neurons, glial necrosis under lactacidosis as well as neuronal necrosis under excitotoxicity. Moreover, VSOR serves as the pathway for glutamate release from astrocytes under ischaemic conditions and when stimulated by bradykinin, an initial mediator of inflammation. So far, many signalling molecules including the EGF receptor, PI3K, Src, PLCgamma and Rho/Rho kinase have been implicated in the regulation of VSOR activity. However, our pharmacological studies suggest that these signals are not essential components of the swelling-induced VSOR activation mechanism even though some of these signals may play permissive or modulatory roles. Molecular identification of VSOR is required to address the question of how cells sense volume expansion and activate VSOR.
Collapse
Affiliation(s)
- Yasunobu Okada
- Department of Cell Physiology, National Institute for Physiological Sciences, Myodaiji-cho, Okazaki 444-8585, Japan.
| | | | | |
Collapse
|
18
|
Salmon MD, Ahluwalia J. Swell activated chloride channel function in human neutrophils. Biochem Biophys Res Commun 2009; 381:462-5. [DOI: 10.1016/j.bbrc.2009.02.147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 02/26/2009] [Indexed: 11/24/2022]
|
19
|
Wertheimer EV, Salicioni AM, Liu W, Trevino CL, Chavez J, Hernández-González EO, Darszon A, Visconti PE. Chloride Is essential for capacitation and for the capacitation-associated increase in tyrosine phosphorylation. J Biol Chem 2008; 283:35539-50. [PMID: 18957426 PMCID: PMC2602906 DOI: 10.1074/jbc.m804586200] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 09/19/2008] [Indexed: 01/03/2023] Open
Abstract
After epididymal maturation, sperm capacitation, which encompasses a complex series of molecular events, endows the sperm with the ability to fertilize an egg. This process can be mimicked in vitro in defined media, the composition of which is based on the electrolyte concentration of the oviductal fluid. It is well established that capacitation requires Na(+), HCO(3)(-), Ca(2+), and a cholesterol acceptor; however, little is known about the function of Cl(-) during this important process. To determine whether Cl(-), in addition to maintaining osmolarity, actively participates in signaling pathways that regulate capacitation, Cl(-) was replaced by either methanesulfonate or gluconate two nonpermeable anions. The absence of Cl(-) did not affect sperm viability, but capacitation-associated processes such as the increase in tyrosine phosphorylation, the increase in cAMP levels, hyperactivation, the zona pellucidae-induced acrosome reaction, and most importantly, fertilization were abolished or significantly reduced. Interestingly, the addition of cyclic AMP agonists to sperm incubated in Cl(-)-free medium rescued the increase in tyrosine phosphorylation and hyperactivation suggesting that Cl(-) acts upstream of the cAMP/protein kinase A signaling pathway. To investigate Cl(-) transport, sperm incubated in complete capacitation medium were exposed to a battery of anion transport inhibitors. Among them, bumetanide and furosemide, two blockers of Na(+)/K(+)/Cl(-) cotransporters (NKCC), inhibited all capacitation-associated events, suggesting that these transporters may mediate Cl(-) movements in sperm. Consistent with these results, Western blots using anti-NKCC1 antibodies showed the presence of this cotransporter in mature sperm.
Collapse
Affiliation(s)
- Eva V Wertheimer
- Department of Veterinary and Animal Science, Paige Laboratories, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Tamoxifen does not inhibit the swell activated chloride channel in human neutrophils during the respiratory burst. Biochem Biophys Res Commun 2008; 375:596-601. [DOI: 10.1016/j.bbrc.2008.08.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Accepted: 08/13/2008] [Indexed: 11/18/2022]
|
21
|
Hartzell HC, Qu Z, Yu K, Xiao Q, Chien LT. Molecular physiology of bestrophins: multifunctional membrane proteins linked to best disease and other retinopathies. Physiol Rev 2008; 88:639-72. [PMID: 18391176 DOI: 10.1152/physrev.00022.2007] [Citation(s) in RCA: 257] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This article reviews the current state of knowledge about the bestrophins, a newly identified family of proteins that can function both as Cl(-) channels and as regulators of voltage-gated Ca(2+) channels. The founding member, human bestrophin-1 (hBest1), was identified as the gene responsible for a dominantly inherited, juvenile-onset form of macular degeneration called Best vitelliform macular dystrophy. Mutations in hBest1 have also been associated with a small fraction of adult-onset macular dystrophies. It is proposed that dysfunction of bestrophin results in abnormal fluid and ion transport by the retinal pigment epithelium, resulting in a weakened interface between the retinal pigment epithelium and photoreceptors. There is compelling evidence that bestrophins are Cl(-) channels, but bestrophins remain enigmatic because it is not clear that the Cl(-) channel function can explain Best disease. In addition to functioning as a Cl(-) channel, hBest1 also is able to regulate voltage-gated Ca(2+) channels. Some bestrophins are activated by increases in intracellular Ca(2+) concentration, but whether bestrophins are the molecular counterpart of Ca(2+)-activated Cl(-) channels remains in doubt. Bestrophins are also regulated by cell volume and may be a member of the volume-regulated anion channel family.
Collapse
Affiliation(s)
- H Criss Hartzell
- Department of Cell Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | |
Collapse
|
22
|
Franco R, Panayiotidis MI, de la Paz LDO. Autocrine signaling involved in cell volume regulation: the role of released transmitters and plasma membrane receptors. J Cell Physiol 2008; 216:14-28. [PMID: 18300263 DOI: 10.1002/jcp.21406] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cell volume regulation is a basic homeostatic mechanism transcendental for the normal physiology and function of cells. It is mediated principally by the activation of osmolyte transport pathways that result in net changes in solute concentration that counteract cell volume challenges in its constancy. This process has been described to be regulated by a complex assortment of intracellular signal transduction cascades. Recently, several studies have demonstrated that alterations in cell volume induce the release of a wide variety of transmitters including hormones, ATP and neurotransmitters, which have been proposed to act as extracellular signals that regulate the activation of cell volume regulatory mechanisms. In addition, changes in cell volume have also been reported to activate plasma membrane receptors (including tyrosine kinase receptors, G-protein coupled receptors and integrins) that have been demonstrated to participate in the regulatory process of cell volume. In this review, we summarize recent studies about the role of changes in cell volume in the regulation of transmitter release as well as in the activation of plasma membrane receptors and their further implications in the regulation of the signaling machinery that regulates the activation of osmolyte flux pathways. We propose that the autocrine regulation of Ca2+-dependent and tyrosine phosphorylation-dependent signaling pathways by the activation of plasma membrane receptors and swelling-induced transmitter release is necessary for the activation/regulation of osmolyte efflux pathways and cell volume recovery. Furthermore, we emphasize the importance of studying these extrinsic signals because of their significance in the understanding of the physiology of cell volume regulation and its role in cell biology in vivo, where the constraint of the extracellular space might enhance the autocrine or even paracrine signaling induced by these released transmitters.
Collapse
Affiliation(s)
- Rodrigo Franco
- Laboratory of Cell Biology and Signal Transduction, Biomedical Research Unit, FES-Iztacala, UNAM, Mexico.
| | | | | |
Collapse
|
23
|
d'Anglemont de Tassigny A, Berdeaux A, Souktani R, Henry P, Ghaleh B. The volume-sensitive chloride channel inhibitors prevent both contractile dysfunction and apoptosis induced by doxorubicin through PI3kinase, Akt and Erk 1/2. Eur J Heart Fail 2008; 10:39-46. [PMID: 18164246 DOI: 10.1016/j.ejheart.2007.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Revised: 10/17/2007] [Accepted: 11/12/2007] [Indexed: 01/17/2023] Open
Abstract
Contractile dysfunction and cardiomyopathies secondary to apoptotic cell death are limiting factors for treating cancer with doxorubicin. Inhibition of volume-sensitive chloride currents (I(Cl,vol)) has been reported to blunt doxorubicin-induced apoptosis in cardiomyocytes. To investigate cellular contractility during acute induction of apoptosis by doxorubicin and to determine whether I(Cl,vol) inhibitors are able to prevent the subsequent contractile dysfunction, electrically paced ventricular myocytes freshly isolated from adult rabbits were acutely exposed to doxorubicin in the presence and absence of I(Cl,vol) inhibitors IAA-94 or DIDS. Doxorubicin induced increases in both annexin V labelling and caspase-3 activity and decreases in cell volume. Alteration in cardiac contractility was observed after doxorubicin exposure. Both IAA-94 and DIDS abolished the doxorubicin-induced decreases in peak shortening and cell volume as well as the increases in caspase-3 activity and annexin V labelling. These protective effects of I(Cl,vol) inhibitors were abolished by previous inhibition of PI(3)kinase, Akt and Erk 1/2. Thus, I(Cl,vol) inhibitors prevent doxorubicin-induced apoptosis and subsequent contractile dysfunction through PI(3)kinase/Akt and Erk 1/2. Inhibition of I(Cl,vol) may represent a new pharmacological strategy for developing cytoprotective drugs against apoptotic cell death and contractile dysfunction.
Collapse
|
24
|
Chien LT, Hartzell HC. Drosophila bestrophin-1 chloride current is dually regulated by calcium and cell volume. ACTA ACUST UNITED AC 2008; 130:513-24. [PMID: 17968025 PMCID: PMC2151665 DOI: 10.1085/jgp.200709795] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mutations in the human bestrophin-1 (hBest1) gene are responsible for Best vitelliform macular dystrophy, however the mechanisms leading to retinal degeneration have not yet been determined because the function of the bestrophin protein is not fully understood. Bestrophins have been proposed to comprise a new family of Cl(-) channels that are activated by Ca(2+). While the regulation of bestrophin currents has focused on intracellular Ca(2+), little is known about other pathways/mechanisms that may also regulate bestrophin currents. Here we show that Cl(-) currents in Drosophila S2 cells, that we have previously shown are mediated by bestrophins, are dually regulated by Ca(2+) and cell volume. The bestrophin Cl(-) currents were activated in a dose-dependent manner by osmotic pressure differences between the internal and external solutions. The increase in the current was accompanied by cell swelling. The volume-regulated Cl(-) current was abolished by treating cells with each of four different RNAi constructs that reduced dBest1 expression. The volume-regulated current was rescued by transfecting with dBest1. Furthermore, cells not expressing dBest1 were severely depressed in their ability to regulate their cell volume. Volume regulation and Ca(2+) regulation can occur independently of one another: the volume-regulated current was activated in the complete absence of Ca(2+) and the Ca(2+)-activated current was activated independently of alterations in cell volume. These two pathways of bestrophin channel activation can interact; intracellular Ca(2+) potentiates the magnitude of the current activated by changes in cell volume. We conclude that in addition to being regulated by intracellular Ca(2+), Drosophila bestrophins are also novel members of the volume-regulated anion channel (VRAC) family that are necessary for cell volume homeostasis.
Collapse
Affiliation(s)
- Li-Ting Chien
- Department of Cell Biology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | |
Collapse
|
25
|
Ducharme G, Newell EW, Pinto C, Schlichter LC. Small-conductance Cl- channels contribute to volume regulation and phagocytosis in microglia. Eur J Neurosci 2007; 26:2119-30. [PMID: 17927776 DOI: 10.1111/j.1460-9568.2007.05802.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The shape and volume of microglia (brain immune cells) change when they activate during brain inflammation and become migratory and phagocytic. Swollen rat microglia express a large Cl(-) current (I(Clswell)), whose biophysical properties and functional roles are poorly understood and whose molecular identity is unknown. We constructed a fingerprint of useful biophysical properties for comparison with I(Clswell) in other cell types and with cloned Cl(-) channels. The microglial I(Clswell) was rapidly activated by cell swelling but not by voltage, and showed no time-dependence during voltage-clamp steps. Like I(Clswell) in many cell types, the halide selectivity sequence was I(-) > Br(-) > Cl(-) > F(-). However, it differed in lacking inactivation, even at +100 mV with high extracellular Mg(2+), and in having a much lower single-channel conductance: 1-3 pS. Based on these fundamental differences, the microglia channel is apparently a different gene product than the more common intermediate-conductance I(Clswell). Microglia express several candidate genes, with relative mRNA expression levels of: CLIC1 > ClC3 > I(Cln) > or = ClC2 > Best2 > Best1 > or = Best3 > Best4. Using a pharmacological toolbox, we show that all drugs that reduced the microglia current (NPPB, IAA-94, flufenamic acid and DIOA) increased the resting cell volume in isotonic solution and inhibited the regulatory volume decrease that followed cell swelling in hypotonic solution. Both channel blockers tested (NPPB and flufenamic acid) dose-dependently inhibited microglia phagocytosis of E. coli bacteria. Because I(Clswell) is involved in microglia functions that involve shape and volume changes, it is potentially important for controlling their ability to migrate to damage sites and phagocytose dead cells and debris.
Collapse
Affiliation(s)
- Guillaume Ducharme
- Toronto Western Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada M5T 2S8
| | | | | | | |
Collapse
|
26
|
Hoffmann EK, Schettino T, Marshall WS. The role of volume-sensitive ion transport systems in regulation of epithelial transport. Comp Biochem Physiol A Mol Integr Physiol 2007; 148:29-43. [PMID: 17289411 DOI: 10.1016/j.cbpa.2006.11.023] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 11/08/2006] [Accepted: 11/23/2006] [Indexed: 11/25/2022]
Abstract
This review focuses on using the knowledge on volume-sensitive transport systems in Ehrlich ascites tumour cells and NIH-3T3 cells to elucidate osmotic regulation of salt transport in epithelia. Using the intestine of the European eel (Anguilla anguilla) (an absorptive epithelium of the type described in the renal cortex thick ascending limb (cTAL)) we have focused on the role of swelling-activated K+- and anion-conductive pathways in response to hypotonicity, and on the role of the apical (luminal) Na+-K+-2Cl- cotransporter (NKCC2) in the response to hypertonicity. The shrinkage-induced activation of NKCC2 involves an interaction between the cytoskeleton and protein phosphorylation events via PKC and myosin light chain kinase (MLCK). Killifish (Fundulus heteroclitus) opercular epithelium is a Cl(-)-secreting epithelium of the type described in exocrine glands, having a CFTR channel on the apical side and the Na+/K+ ATPase, NKCC1 and a K+ channel on the basolateral side. Osmotic control of Cl- secretion across the operculum epithelium includes: (i) hyperosmotic shrinkage activation of NKCC1 via PKC, MLCK, p38, OSR1 and SPAK; (ii) deactivation of NKCC by hypotonic cell swelling and a protein phosphatase, and (iii) a protein tyrosine kinase acting on the focal adhesion kinase (FAK) to set levels of NKCC activity.
Collapse
Affiliation(s)
- E K Hoffmann
- Department of Molecular Biology, The August Krogh Building, University of Copenhagen, Denmark.
| | | | | |
Collapse
|
27
|
Suh KS, Mutoh M, Mutoh T, Li L, Ryscavage A, Crutchley JM, Dumont RA, Cheng C, Yuspa SH. CLIC4 mediates and is required for Ca2+-induced keratinocyte differentiation. J Cell Sci 2007; 120:2631-40. [PMID: 17636002 DOI: 10.1242/jcs.002741] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Keratinocyte differentiation requires integrating signaling among intracellular ionic changes, kinase cascades, sequential gene expression, cell cycle arrest, and programmed cell death. We now show that Cl(-) intracellular channel 4 (CLIC4) expression is increased in both mouse and human keratinocytes undergoing differentiation induced by Ca(2+), serum and the protein kinase C (PKC)-activator, 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Elevation of CLIC4 is associated with signaling by PKCdelta, and knockdown of CLIC4 protein by antisense or shRNA prevents Ca(2+)-induced keratin 1, keratin 10 and filaggrin expression and cell cycle arrest in differentiating keratinocytes. CLIC4 is cytoplasmic in actively proliferating keratinocytes in vitro, but the cytoplasmic CLIC4 translocates to the nucleus in keratinocytes undergoing growth arrest by differentiation, senescence or transforming growth factor beta (TGFbeta) treatment. Targeting CLIC4 to the nucleus of keratinocytes via adenoviral transduction increases nuclear Cl(-) content and enhances expression of differentiation markers in the absence of elevated Ca(2+). In vivo, CLIC4 is localized to the epidermis in mouse and human skin, where it is predominantly nuclear in quiescent cells. These results suggest that CLIC4 participates in epidermal homeostasis through both alterations in the level of expression and subcellular localization. Nuclear CLIC4, possibly by altering the Cl(-) and pH of the nucleus, contributes to cell cycle arrest and the specific gene expression program associated with keratinocyte terminal differentiation.
Collapse
Affiliation(s)
- Kwang S Suh
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Guan YY, Wang GL, Zhou JG. The ClC-3 Cl− channel in cell volume regulation, proliferation and apoptosis in vascular smooth muscle cells. Trends Pharmacol Sci 2006; 27:290-6. [PMID: 16697056 DOI: 10.1016/j.tips.2006.04.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Revised: 03/09/2006] [Accepted: 04/19/2006] [Indexed: 01/23/2023]
Abstract
The volume-regulated Cl(-) current (I(Cl.vol)) is responsible for the transmembrane Cl(-) transport that is involved in cell volume regulatory mechanisms. Although the regulation of cell volume is a fundamental function of healthy cells for maintaining constant size, the molecular genetic identification of I(Cl.vol) is still being debated. Recent studies in vascular smooth muscle support the idea that ClC-3, a member of the voltage-gated ClC Cl(-) channel family, is the molecular component involved in the activation or regulation of I(Cl.vol). Moreover, gene-targeting studies in vascular smooth muscle cells (VSMCs) and other cell types indicate emerging roles of ClC-3 in cell proliferation and apoptosis. These findings indicate that ClC-3 might be involved in modulating vascular remodeling in hypertension and arteriosclerosis.
Collapse
Affiliation(s)
- Yong-Yuan Guan
- Department of Pharmacology, Zhongshan Medical College, Sun Yat-sen University, 74 Zhongshan 2 Road, Guangzhou 510089, China.
| | | | | |
Collapse
|
29
|
Abstract
Taurine, an important mediator of cellular volume regulation in the central nervous system, is accumulated into neurons and glia by means of a highly specific sodium-dependent membrane transporter. During hyperosmotic cell shrinkage, net cellular taurine content increases as taurine transporter activity is enhanced via elevated gene expression of the transporter protein. In hypo-osmotic conditions, taurine is rapidly lost from cells by means of taurine-conducting membrane channels. We reasoned that changes in taurine transporter activity also might accompany cell swelling to minimize re-accumulation of taurine from the extracellular space. Thus, we determined the kinetic and pharmacological characteristics of neuronal taurine transport and the response to osmotic swelling. Accumulation of radioactive taurine is strongly temperature dependent and occurs via saturable and non-saturable pathways. At concentrations of taurine expected in extracellular fluid in vivo, 98% of taurine accumulation would occur via the saturable pathway. This pathway obeys Michaelis-Menten kinetics with a Km of 30.0 +/- 8.8 microm (mean +/- SE) and Jmax of 2.1 +/- 0.2 nmol/mg protein min. The saturable pathway is dependent on extracellular sodium with an effective binding constant of 80.0 +/- 3.1 mm and a Hill coefficient of 2.1 +/- 0.1. This pathway is inhibited by structural analogues of taurine and by the anion channel inhibitors, 4,4'-diisothiocyanostilbene-2, 2'-disulfonic acid (DIDS) and 5-nitro-2-(3 phenylpropylamino) benzoic acid (NPPB). NPPB, but not DIDS, also reduces the ATP content of the cell cultures. Osmotic swelling at constant extracellular sodium concentration reduces the Jmax of the saturable transport pathway by approximately 48%, increases Kdiff for the non-saturable pathway by 77%, but has no effect on cellular ATP content. These changes in taurine transport occurring in swollen neurons in vivo would contribute to net reduction of taurine content and resulting volume regulation.
Collapse
Affiliation(s)
- James E Olson
- Department of Emergency Medicine, Wright State University School of Medicine, Cox Institute, Kettering, Ohio 45429, USA.
| | | |
Collapse
|
30
|
Stutzin A, Hoffmann EK. Swelling-activated ion channels: functional regulation in cell-swelling, proliferation and apoptosis. Acta Physiol (Oxf) 2006; 187:27-42. [PMID: 16734740 DOI: 10.1111/j.1748-1716.2006.01537.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cell volume regulation is one of the most fundamental homeostatic mechanisms and essential for normal cellular function. At the same time, however, many physiological mechanisms are associated with regulatory changes in cell size meaning that the set point for cell volume regulation is under physiological control. Thus, cell volume is under a tight and dynamic control and abnormal cell volume regulation will ultimately lead to severe cellular dysfunction, including alterations in cell proliferation and cell death. This review describes the different swelling-activated ion channels that participate as key players in the maintenance of normal steady-state cell volume, with particular emphasis on the intracellular signalling pathways responsible for their regulation during hypotonic stress, cell proliferation and apoptosis.
Collapse
Affiliation(s)
- A Stutzin
- Centro de Estudios Moleculares de la Célula and Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | | |
Collapse
|
31
|
Moreland JG, Davis AP, Bailey G, Nauseef WM, Lamb FS. Anion channels, including ClC-3, are required for normal neutrophil oxidative function, phagocytosis, and transendothelial migration. J Biol Chem 2006; 281:12277-88. [PMID: 16522634 DOI: 10.1074/jbc.m511030200] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NADPH oxidase activity, phagocytosis, and cell migration are essential functions of polymorphonuclear leukocytes (PMNs) in host defense. The cytoskeletal reorganization necessary to perform these functions has been extensively studied, but the role of cell volume regulation, which is likely dependent upon anion channels, has not been defined. Mice lacking the anion channel ClC-3 (Clcn3(-/-)) died from presumed sepsis following intravascular catheter placement, whereas Clcn3(+/+) littermates survived. We hypothesized that ClC-3 has a critical role in host defense and reasoned that PMN function would be compromised in these mice. Clcn3(-/-) PMNs displayed markedly reduced NADPH oxidase activity in response to opsonized zymosan and modestly reduced activity after phorbol 12-myristate 13-acetate. Human PMNs treated with the anion channel inhibitors niflumic acid or 5-nitro-2-(3-phenylpropylamino)benzoic acid had a very similar defect. ClC-3 protein was detected in the secretory vesicles and secondary granules of resting PMNs and was up-regulated to the phagosomal membrane. Clcn3(-/-) PMNs and human PMNs lacking normal anion channel function both exhibited reduced uptake of opsonized zymosan at 1, 5, and 10 min in a synchronized phagocytosis assay. Niflumic acid-treated PMNs also had impaired transendothelial migration in vitro, whereas migration in vivo was not altered in Clcn3(-/-) PMNs. Selective inhibition of the swelling-activated chloride channel with tamoxifen profoundly reduced PMN migration but had no effect on NADPH oxidase activity. In summary, PMNs lacking normal anion channel function exhibited reduced NADPH oxidase activity, diminished phagocytosis, and impaired migration. ClC-3 was specifically involved in the respiratory burst and phagocytosis.
Collapse
Affiliation(s)
- Jessica G Moreland
- Division of Critical Care, Department of Pediatrics,The University of Iowa, Iowa City 52242, USA.
| | | | | | | | | |
Collapse
|
32
|
Klausen TK, Bergdahl A, Hougaard C, Christophersen P, Pedersen SF, Hoffmann EK. Cell cycle-dependent activity of the volume- and Ca2+-activated anion currents in Ehrlich lettre ascites cells. J Cell Physiol 2006; 210:831-42. [PMID: 17111356 DOI: 10.1002/jcp.20918] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recent evidence implicates the volume-regulated anion current (VRAC) and other anion currents in control or modulation of cell cycle progression; however, the precise involvement of anion channels in this process is unclear. Here, Cl- currents in Ehrlich Lettre Ascites (ELA) cells were monitored during cell cycle progression, under three conditions: (i) after osmotic swelling (i.e., VRAC), (ii) after an increase in the free intracellular Ca2+ concentration (i.e., the Ca2+-activated Cl- current, CaCC), and (iii) under steady-state isotonic conditions. The maximal swelling-activated VRAC current decreased in G1 and increased in early S phase, compared to that in G0. The isotonic steady-state current, which seems to be predominantly VRAC, also decreased in G1, and increased again in early S phase, to a level similar to that in G0. In contrast, the maximal CaCC current (500 nM free Ca2+ in the pipette), was unaltered from G0 to G1, but decreased in early S phase. A novel high-affinity anion channel inhibitor, the acidic di-aryl-urea NS3728, which inhibited both VRAC and CaCC, attenuated ELA cell growth, suggesting a possible mechanistic link between cell cycle progression and cell cycle-dependent changes in the capacity for conductive Cl- transport. It is suggested that in ELA cells, entrance into the S phase requires an increase in VRAC activity and/or an increased potential for regulatory volume decrease (RVD), and at the same time a decrease in CaCC magnitude.
Collapse
Affiliation(s)
- Thomas Kjaer Klausen
- Department of Biochemistry, Institute for Molecular Biology and Physiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
33
|
Dronne MA, Boissel JP, Grenier E. A mathematical model of ion movements in grey matter during a stroke. J Theor Biol 2005; 240:599-615. [PMID: 16368113 DOI: 10.1016/j.jtbi.2005.10.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Revised: 10/24/2005] [Accepted: 10/26/2005] [Indexed: 10/25/2022]
Abstract
The development of cytotoxic oedema during a stroke consists in cell swelling and shrinking of the extracellular space. This phenomenon is triggered by ion movements through voltage-gated channels, exchangers and pumps. During ischaemia, sodium, calcium and chloride enter the neurons whereas potassium and glutamate are expelled out of the cells. A mathematical model is proposed to represent the long-term dynamics of membrane potentials, cell volumes and ionic concentrations in intracellular and extracellular spaces during a stroke and to study the influence of each ionic current on cell swelling. The model relies on electrophysiological mechanisms and takes into account the behaviour of two types of cells: neurons and also astrocytes known to play a key role in the excitotoxic process in grey matter. The results obtained when a severe or a moderate ischaemia is simulated are consistent with those observed in the in vitro and in vivo experiments. As this model appears to be robust, it is used to perform illustrative simulations aimed at studying the effect of some channel blockers on cell swelling. This approach may help to explore new therapeutic strategies in order to reduce stroke damage.
Collapse
Affiliation(s)
- Marie-Aimée Dronne
- Service de Pharmacologie Clinique, EA 3736, Faculté de Médecine Laennec, Rue Guillaume Paradin, BP 8071, 69376 Lyon, cedex 08, France.
| | | | | |
Collapse
|
34
|
Abstract
Many cells, including neuronal and glial progenitor cells, stem cells and microglial cells, have the capacity to move through the extracellular spaces of the developing and mature brain. This is particularly pronounced in astrocyte-derived tumors, gliomas, which diffusely infiltrate the normal brain. Although a significant body of literature exists regarding signals that are involved in the guidance of cells and their processes, little attention has been paid to cell-shape and cell-volume changes of migratory cells. However, extracellular spaces in the brain are very narrow and represent a major obstacle that requires cells to dynamically regulate their volume. Recent studies in glioma cells show that this involves the secretion of Cl− and K+ with water. Pharmacological inhibition of Cl− channels impairs their ability to migrate and limits tumor progression in experimental tumor models. One Cl−-channel inhibitor, chlorotoxin, is currently in Phase II clinical trials to treat malignant glioma. This article reviews our current knowledge of cell-volume changes and the role of ion channels during the migration of glioma cells. It also discusses evidence that supports the importance of channel-mediated cell-volume changes in the migration of immature neurons and progenitor cells during development. New unpublished data is presented, which demonstrates that Cl− and K+ channels involved in cell shrinkage localize to lipid-raft domains on the invadipodia of glioma cells and that their presence might be regulated by trafficking of these proteins in and out of lipid rafts.
Collapse
|
35
|
Yeung CH, Barfield JP, Cooper TG. Chloride Channels in Physiological Volume Regulation of Human Spermatozoa1. Biol Reprod 2005; 73:1057-63. [PMID: 16033995 DOI: 10.1095/biolreprod.105.044123] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
As with other mammalian species, human spermatozoa experience a decrease in extracellular osmolarity in cervical mucus upon ejaculation, which requires the efflux of osmolytes and water to counteract swelling that hinders mucus penetration. Recent evidence for the operation of K+ channels in the process of volume regulation suggests parallel involvement of Cl-/anion channels for electro-neutrality as in somatic cells. This was studied using ejaculated spermatozoa washed at seminal osmolality and incubated for 30 min in a medium of mucus osmolality in the presence of Cl- channel blockers. Increases in cell size measured as laser forward-scatter by flow cytometry were detected in the presence of 100 microM 5-nitro-2(3-phenylpropylamino) benzoic acid, 400 microM diisothiocyanato-stilbene-2,2'-disulphonic acid, and 20 microM tamoxifen. No volume changes were found with 400 microM 4-acetamido-4'-isothiocyanato-stilbene-2,2'-disulphonic acid, 200 microM verapamil, or niflumic acid, whereas 1 mM niflumic acid induced shrinkage. Among the candidate channel proteins, Western blotting revealed the presence of ClC-3 (CLCN3) at 87 kDa, but the absence of ClC-2 (CLCN2) from sperm proteins in all samples tested. ICln (CLNS1A) was found in only one of eight samples. Immunocytochemistry localized CLCN3 to the sperm tail. To confirm molecular identities, sperm mRNA was extracted and checked for quality by the presence of protamine 2 transcripts and the absence of sperm DNA and leukocyte mRNA using reverse transcription-polymerase chain reaction. Transcripts of Clcn3 were found in all samples and that of Clns1a in some but not all samples. Clcn3 was therefore considered the most likely candidate of Cl- channel involved in volume regulation of human sperm.
Collapse
Affiliation(s)
- C H Yeung
- Institute of Reproductive Medicine of the University, D-48129 Münster, Germany.
| | | | | |
Collapse
|
36
|
Yeung CH, Barfield JP, Cooper TG. The role of anion channels and Ca2+ in addition to K+ channels in the physiological volume regulation of murine spermatozoa. Mol Reprod Dev 2005; 71:368-79. [PMID: 15803461 DOI: 10.1002/mrd.20261] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Studies in the human, transgenic mice, and cattle indicate that sperm cell volume regulation plays an important role in male fertility as spermatozoa encounter a hypo-osmotic challenge upon ejaculation into the female tract. Physiological regulatory volume decrease (RVD) was examined using flow cytometry in murine sperm released into incubation medium mimicking uterine osmolality and including putative channel inhibitors. The involvement of K+ channels was indicated by the recovery of volume regulation by the K+ ionophore valinomycin in defective sperm from infertile transgenic mice, and from blockage of RVD by quinine in normal sperm. However, in neither case was the recovery complete. The involvement of volume-sensitive osmolyte and anion channels (VSOAC) were investigated using blockers effective in other cell types. NPPB (5-nitro-2(3-phenylpropylamino) benzoic acid) and tamoxifen inhibited RVD but SITS (4-acetamido-4'-isothiocyanato-stilbene-2,2'-disulphonic acid) at 0.4 and 1 mM had no effect whereas DIDS (di-isothiocyanato-stilbene-2,2'-disulphonic acid) at 1 mM enhanced RVD. Verapamil, but not another P-glycoprotein antagonist cyclosporin, caused sperm swelling which persisted in the presence of valinomycin, in Ca2+-free medium and in the presence of thapsigargin, but swelling was abolished by the Ca2+ ionophore A23187. Nifedipine was slightly effective in blocking RVD. Analysis by Western blotting failed to reveal ClC-2 and ClC-3 members of the chloride channel family in murine or rat sperm proteins despite signal bands in positive tissue controls. These findings implicate the involvement of some unidentified VSOAC in sperm volume regulation, which is probably Ca+-dependent.
Collapse
Affiliation(s)
- Ching-Hei Yeung
- Institute of Reproductive Medicine of the University Clinic, Münster, Germany.
| | | | | |
Collapse
|
37
|
Martin RE, Henry RI, Abbey JL, Clements JD, Kirk K. The 'permeome' of the malaria parasite: an overview of the membrane transport proteins of Plasmodium falciparum. Genome Biol 2005; 6:R26. [PMID: 15774027 PMCID: PMC1088945 DOI: 10.1186/gb-2005-6-3-r26] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Revised: 12/31/2004] [Accepted: 01/28/2005] [Indexed: 11/24/2022] Open
Abstract
Bioinformatic and expression analyses attribute putative functions to transporters and channels encoded by the Plasmodium falciparum genome. The malaria parasite has substantially more membrane transport proteins than previously thought. Background The uptake of nutrients, expulsion of metabolic wastes and maintenance of ion homeostasis by the intraerythrocytic malaria parasite is mediated by membrane transport proteins. Proteins of this type are also implicated in the phenomenon of antimalarial drug resistance. However, the initial annotation of the genome of the human malaria parasite Plasmodium falciparum identified only a limited number of transporters, and no channels. In this study we have used a combination of bioinformatic approaches to identify and attribute putative functions to transporters and channels encoded by the malaria parasite, as well as comparing expression patterns for a subset of these. Results A computer program that searches a genome database on the basis of the hydropathy plots of the corresponding proteins was used to identify more than 100 transport proteins encoded by P. falciparum. These include all the transporters previously annotated as such, as well as a similar number of candidate transport proteins that had escaped detection. Detailed sequence analysis enabled the assignment of putative substrate specificities and/or transport mechanisms to all those putative transport proteins previously without. The newly-identified transport proteins include candidate transporters for a range of organic and inorganic nutrients (including sugars, amino acids, nucleosides and vitamins), and several putative ion channels. The stage-dependent expression of RNAs for 34 candidate transport proteins of particular interest are compared. Conclusion The malaria parasite possesses substantially more membrane transport proteins than was originally thought, and the analyses presented here provide a range of novel insights into the physiology of this important human pathogen.
Collapse
Affiliation(s)
- Rowena E Martin
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia
| | - Roselani I Henry
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia
| | - Janice L Abbey
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia
| | - John D Clements
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia
- Division of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia
| | - Kiaran Kirk
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia
| |
Collapse
|
38
|
d'Anglemont de Tassigny A, Souktani R, Henry P, Ghaleh B, Berdeaux A. Volume-sensitive chloride channels (ICl,vol) mediate doxorubicin-induced apoptosis through apoptotic volume decrease in cardiomyocytes. Fundam Clin Pharmacol 2005; 18:531-8. [PMID: 15482374 DOI: 10.1111/j.1472-8206.2004.00273.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Apoptosis is associated with early changes in cell volume through a mechanism called apoptotic volume decrease (AVD). As volume-sensitive chloride channels (I(Cl,vol)) are known to play a key role in the regulation of cell volume, this study investigated the role of I(Cl,vol) and AVD in doxorubicin-induced apoptotic cell death in adult rabbit ventricular cardiomyocytes. Exposure of cardiomyocytes to 1 microm doxorubicin induced a rapid and significant reduction in cell volume of cardiomyocytes (average of 15%), i.e. AVD as well as increases in the early markers of apoptosis, annexin V labeling and caspase-3 activity. Doxorubicin also induced the activation of a current characterized as I(Cl,vol) on the basis of the external chloride sensitivity and pharmacological properties with the patch clamp technique. Doxorubicin-induced AVD and apoptosis were both abolished when cardiomyocytes were exposed to the I(Cl,vol) inhibitors 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB) (0.1 mM) or indanyloxyacetic acid 94 (IAA-94) (10 microM). The crucial role of I(Cl,vol) during AVD and apoptosis was confirmed using C(2)-ceramide, another pro-apoptotic compound. These results demonstrate that activation of I(Cl,vol) plays a major role in the mechanism leading to cell shrinkage and apoptosis-induced AVD by agents such as doxorubicin or C(2)-ceramide in adult cardiomyocytes.
Collapse
Affiliation(s)
- Alexandra d'Anglemont de Tassigny
- Laboratoire de Pharmacologie, Faculté de Médecine de Créteil, Université Paris XII, France and Laboratoire de Pharmacologie, INSERM E00.01, Faculté de Médecine Paris-Sud, 94270 Le Kremlin-Bicêtre, France
| | | | | | | | | |
Collapse
|
39
|
d'Anglemont de Tassigny A, Ghaleh B, Souktani R, Henry P, Berdeaux A. Hypo-osmotic stress inhibits doxorubicin-induced apoptosis via a protein kinase A-dependent mechanism in cardiomyocytes. Clin Exp Pharmacol Physiol 2005; 31:438-43. [PMID: 15236631 DOI: 10.1111/j.1440-1681.2004.04025.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The clinical use of doxorubicin is limited by the development of severe cardiomyopathies linked, at least in part, to an abnormal increase in the rate of apoptotic cell death. Because cell shrinkage is considered to be a crucial step at the onset of apoptosis, the aim of the present study was to investigate whether a brief hypo-osmotic stress, which leads to an increase in cell volume, could interfere with the induction of apoptosis by doxorubicin in adult cardiomyocytes. 2. Cell volume expansion results in intracellular accumulation of cAMP, so we secondarily tested whether the protective effect of hypo-osmotic stress could be related to the cAMP pathway. Accordingly, apoptosis was induced by doxorubicin (1 micromol/L) in cardiomyocytes freshly isolated from New Zealand adult rabbit hearts. 3. Exposure to doxorubicin in an iso-osmotic medium (290 mOsmol/kg H2O) induced a rapid decrease in cell volume, as well as increases in annexin V labelling and caspase-3 activity, two biological markers of apoptosis. These effects of doxorubicin were abolished by 15 min pretreatment with hypo-osmotic stress at 220 mOsmol/kgH2O (HS 220). 4. This cytoprotective effect of HS 220 was still observed when doxorubicin was added to the medium 60 min later, but it was abolished when the pretreatment by HS 220 was associated with the protein kinase A inhibitor KT 5720 (200 nmol/L). 5. Conversely, 15 min pretreatment with either the cAMP analogue 8-bromo-cAMP (0.5 mmol/L) or the adenylate cyclase activator forskolin (10 micromol/L) inhibited apoptosis induced by doxorubicin. 6. In conclusion, these results demonstrate that: (i) apoptosis induced by doxorubicin can be counteracted by a hypo-osmotic stress in adult cardiomyocytes; and (ii) activation of the protein kinase A-dependent pathway plays a major role in the mechanism leading to the cytoprotective effect induced by a hypo-osmotic stress.
Collapse
|
40
|
Ernest NJ, Weaver AK, Van Duyn LB, Sontheimer HW. Relative contribution of chloride channels and transporters to regulatory volume decrease in human glioma cells. Am J Physiol Cell Physiol 2005; 288:C1451-60. [PMID: 15659714 PMCID: PMC2548409 DOI: 10.1152/ajpcell.00503.2004] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Primary brain tumors (gliomas) often present with peritumoral edema. Their ability to thrive in this osmotically altered environment prompted us to examine volume regulation in human glioma cells, specifically the relative contribution of Cl(-) channels and transporters to this process. After a hyposmotic challenge, cultured astrocytes, D54-MG glioma cells, and glioma cells from human patient biopsies exhibited a regulatory volume decrease (RVD). Although astrocytes were not able to completely reestablish their original prechallenge volumes, glioma cells exhibited complete volume recovery, sometimes recovering to a volume smaller than their original volumes (V(Post-RVD) < V(baseline)). In glioma cells, RVD was largely inhibited by treatment with a combination of Cl(-) channel inhibitors, 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB) and Cd(2+) (V(Post-RVD) > 1.4*V(baseline)). Volume regulation was also attenuated to a lesser degree by the addition of R-(+)-[(2-n-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-1H-inden-5-yl)oxy]acetic acid (DIOA), a known K(+)-Cl(-) cotransporter (KCC) inhibitor. To dissect the relative contribution of channels vs. transporters in RVD, we took advantage of the comparatively high temperature dependence of transport processes vs. channel-mediated diffusion. Cooling D54-MG glioma cells to 15 degrees C resulted in a loss of DIOA-sensitive volume regulation. Moreover, at 15 degrees C, the channel blockers NPPB + Cd(2+) completely inhibited RVD and cells behaved like perfect osmometers. The calculated osmolyte flux during RVD under these experimental conditions suggests that the relative contribution of Cl(-) channels vs. transporters to this process is approximately 60-70% and approximately 30-40%, respectively. Finally, we identified several candidate proteins that may be involved in RVD, including the Cl(-) channels ClC-2, ClC-3, ClC-5, ClC-6, and ClC-7 and the transporters KCC1 and KCC3a.
Collapse
Affiliation(s)
- Nola Jean Ernest
- Department of Neurobiology, University of Alabama at Birmingham, USA
| | | | | | | |
Collapse
|
41
|
Shibata H, Satoh TO, Ugawa T, Masuda N, Yanai-Inamura H, Abe A, Kondo Y, Kuramochi T, Akamatsu S, Uchida W. Characterization of the Pharmacology of YM-198313 on Volume-Regulated Anion Channels. Biol Pharm Bull 2005; 28:1187-91. [PMID: 15997095 DOI: 10.1248/bpb.28.1187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of the volume-regulated anion channels (VRAC) is considered to be involved in arrhythmia, but it has not yet been fully elucidated because of the lack of its high affinitive and selective compounds. A newly synthesized compound, YM-198313 (sodium 4-({[2-(methylthio)benzyl]amino}-5-[(1-phenylethyl)thio]isothiazol-3-olate), strongly inhibited VRAC in HeLa cells with an IC50 of 3.03+/-0.05 microM. However, YM-198313 weakly affected both the Ca2+-activated Cl- channels in HTC cells and the cAMP-activated Cl- channels in T84 cells, demonstrating that this compound is selective for VRAC among Cl- channels. At 10 microM, YM-198313 almost completely (100+/-7.8%) inhibited the VRAC current in guinea pig atrial myocytes. However, at the same concentration, YM-198313 showed little inhibitory effect on the cardiac cation currents in ventricular myocytes. We believe that YM-198313 is a potent and selective VRAC inhibitor, therefore, it should be use to clarify the role VRAC plays in arrhythmia.
Collapse
Affiliation(s)
- Hiroshi Shibata
- Institute for Drug Discovery Research, Yamanouchi Pharmaceutical Co., Ltd., Ibaraki, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Larkin D, Murphy D, Reilly DF, Cahill M, Sattler E, Harriott P, Cahill DJ, Moran N. ICln, a Novel Integrin αIIbβ3-Associated Protein, Functionally Regulates Platelet Activation. J Biol Chem 2004; 279:27286-93. [PMID: 15075326 DOI: 10.1074/jbc.m402159200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A critical role for the conserved alpha-integrin cytoplasmic motif, KVGFFKR, is recognized in the regulation of activation of the platelet integrin alpha(IIb)beta(3). To understand the molecular mechanisms of this regulation, we sought to determine the nature of the protein interactions with this cytoplasmic motif. We used a tagged synthetic peptide, biotin-KVGFFKR, to probe a high density protein expression array (37,200 recombinant human proteins) for high affinity interactions. A number of potential integrin-binding proteins were identified. One such protein, a chloride channel regulatory protein, ICln, was characterized further because its affinity for the integrin peptide was highest as was its expression in platelets. We verified the presence of ICln in human platelets by PCR, Western blots, immunohistochemistry, and its co-association with alpha(IIb)beta(3) by surface plasmon resonance. The affinity of this interaction was 82.2 +/- 24.4 nm in a cell free assay. ICln co-immunoprecipitates with alpha(IIb)beta(3) in platelet lysates demonstrating that this interaction is physiologically relevant. Furthermore, immobilized KVGFFKR peptides, but not control KAAAAAR peptides, specifically extract ICln from platelet lysates. Acyclovir (100 microm to 5 mm), a pharmacological inhibitor of the ICln chloride channel, specifically inhibits integrin activation (PAC-1 expression) and platelet aggregation without affecting CD62 P expression confirming a specific role for ICln in integrin activation. In parallel, a cell-permeable peptide corresponding to the potential integrin-recognition domain on ICln (AKFEEE, 10-100 microm) also inhibits platelet function. Thus, we have identified, verified, and characterized a novel functional interaction between the platelet integrin and ICln, in the platelet membrane.
Collapse
Affiliation(s)
- Deirdre Larkin
- Department of Clinical Pharmacology, Royal College of Surgeons in Ireland, Dublin
| | | | | | | | | | | | | | | |
Collapse
|