1
|
Arreola J, López-Romero AE, Huerta M, Guzmán-Hernández ML, Pérez-Cornejo P. Insights into the function and regulation of the calcium-activated chloride channel TMEM16A. Cell Calcium 2024; 121:102891. [PMID: 38772195 DOI: 10.1016/j.ceca.2024.102891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
The TMEM16A channel, a member of the TMEM16 protein family comprising chloride (Cl-) channels and lipid scramblases, is activated by the free intracellular Ca2+ increments produced by inositol 1,4,5-trisphosphate (IP3)-induced Ca2+ release after GqPCRs or Ca2+ entry through cationic channels. It is a ubiquitous transmembrane protein that participates in multiple physiological functions essential to mammals' lives. TMEM16A structure contains two identical 10-segment monomers joined at their transmembrane segment 10. Each monomer harbours one independent hourglass-shaped pore gated by Ca2+ ligation to an orthosteric site adjacent to the pore and controlled by two gates. The orthosteric site is created by assembling negatively charged glutamate side chains near the pore´s cytosolic end. When empty, this site generates an electrostatic barrier that controls channel rectification. In addition, an isoleucine-triad forms a hydrophobic gate at the boundary of the cytosolic vestibule and the inner side of the neck. When the cytosolic Ca2+ rises, one or two Ca2+ ions bind to the orthosteric site in a voltage (V)-dependent manner, thus neutralising the electrostatic barrier and triggering an allosteric gating mechanism propagating via transmembrane segment 6 to the hydrophobic gate. These coordinated events lead to pore opening, allowing the Cl- flux to ensure the physiological response. The Ca2+-dependent function of TMEM16A is highly regulated. Anions with higher permeability than Cl- facilitate V dependence by increasing the Ca2+ sensitivity, intracellular protons can replace Ca2+ and induce channel opening, and phosphatidylinositol 4,5-bisphosphate bound to four cytosolic sites likely maintains Ca2+ sensitivity. Additional regulation is afforded by cytosolic proteins, most likely by phosphorylation and protein-protein interaction mechanisms.
Collapse
Affiliation(s)
- Jorge Arreola
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico.
| | - Ana Elena López-Romero
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - Miriam Huerta
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - María Luisa Guzmán-Hernández
- Catedrática CONAHCYT, Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| |
Collapse
|
2
|
Yuan L, Tang Y, Yin L, Lin X, Luo Z, Wang S, Li J, Liang P, Jiang B. The role of Transmembrane Protein 16A (TMEM16A) in pulmonary hypertension. Cardiovasc Pathol 2023; 65:107525. [PMID: 36781068 DOI: 10.1016/j.carpath.2023.107525] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023] Open
Abstract
Transmembrane protein 16A (TMEM16A), a member of the TMEM16 family, is the molecular basis of Ca2+-activated chloride channels (CaCCs) and is involved in a variety of physiological and pathological processes. Previous studies have focused more on respiratory-related diseases and tumors. However, recent studies have identified an important role for TMEM16A in cardiovascular diseases, especially in pulmonary hypertension. TMEM16A is expressed in both pulmonary artery smooth muscle cells and pulmonary artery endothelial cells and is involved in the development of pulmonary hypertension. This paper presents the structure and function of TMEM16A, the pathogenesis of pulmonary hypertension, and highlights the role and mechanism of TMEM16A in pulmonary hypertension, summarizing the controversies in this field and taking into account hypertension and portal hypertension, which have similar pathogenesis. It is hoped that the unique role of TMEM16A in pulmonary hypertension will be illustrated and provide ideas for research in this area.
Collapse
Affiliation(s)
- Ludong Yuan
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China
| | - Yuting Tang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China
| | - Leijing Yin
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China
| | - Xiaofang Lin
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China
| | - Zhengyang Luo
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China
| | - Shuxin Wang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China
| | - Jing Li
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China
| | - Pengfei Liang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bimei Jiang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan China.
| |
Collapse
|
3
|
Rodriguez TC, Zhong L, Simpson H, Gleason E. Reduced Expression of TMEM16A Impairs Nitric Oxide-Dependent Cl− Transport in Retinal Amacrine Cells. Front Cell Neurosci 2022; 16:937060. [PMID: 35966201 PMCID: PMC9363626 DOI: 10.3389/fncel.2022.937060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Postsynaptic cytosolic Cl− concentration determines whether GABAergic and glycinergic synapses are inhibitory or excitatory. We have shown that nitric oxide (NO) initiates the release of Cl− from acidic internal stores into the cytosol of retinal amacrine cells (ACs) thereby elevating cytosolic Cl−. In addition, we found that cystic fibrosis transmembrane conductance regulator (CFTR) expression and Ca2+ elevations are necessary for the transient effects of NO on cytosolic Cl− levels, but the mechanism remains to be elucidated. Here, we investigated the involvement of TMEM16A as a possible link between Ca2+ elevations and cytosolic Cl− release. TMEM16A is a Ca2+-activated Cl− channel that is functionally coupled with CFTR in epithelia. Both proteins are also expressed in neurons. Based on this and its Ca2+ dependence, we test the hypothesis that TMEM16A participates in the NO-dependent elevation in cytosolic Cl− in ACs. Chick retina ACs express TMEM16A as shown by Western blot analysis, single-cell PCR, and immunocytochemistry. Electrophysiology experiments demonstrate that TMEM16A functions in amacrine cells. Pharmacological inhibition of TMEM16A with T16inh-AO1 reduces the NO-dependent Cl− release as indicated by the diminished shift in the reversal potential of GABAA receptor-mediated currents. We confirmed the involvement of TMEM16A in the NO-dependent Cl− release using CRISPR/Cas9 knockdown of TMEM16A. Two different modalities targeting the gene for TMEM16A (ANO1) were tested in retinal amacrine cells: an all-in-one plasmid vector and crRNA/tracrRNA/Cas9 ribonucleoprotein. The all-in-one CRISPR/Cas9 modality did not change the expression of TMEM16A protein and produced no change in the response to NO. However, TMEM16A-specific crRNA/tracrRNA/Cas9 ribonucleoprotein effectively reduces both TMEM16A protein levels and the NO-dependent shift in the reversal potential of GABA-gated currents. These results show that TMEM16A plays a role in the NO-dependent Cl− release from retinal ACs.
Collapse
|
4
|
Klett NJ, Cravetchi O, Allen CN. Long-Term Imaging Reveals a Circadian Rhythm of Intracellular Chloride in Neurons of the Suprachiasmatic Nucleus. J Biol Rhythms 2022; 37:110-123. [PMID: 34994231 PMCID: PMC9203244 DOI: 10.1177/07487304211059770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Both inhibitory and excitatory GABA transmission exist in the mature suprachiasmatic nucleus (SCN), the master pacemaker of circadian physiology. Whether GABA is inhibitory or excitatory depends on the intracellular chloride concentration ([Cl-]i). Here, using the genetically encoded ratiometric probe Cl-Sensor, we investigated [Cl-]i in AVP and VIP-expressing SCN neurons for several days in culture. The chloride ratio (RCl) demonstrated circadian rhythmicity in AVP + neurons and VIP + neurons, but was not detected in GFAP + astrocytes. RCl peaked between ZT 7 and ZT 8 in both AVP + and VIP + neurons. RCl rhythmicity was not dependent on the activity of several transmembrane chloride carriers, action potential generation, or the L-type voltage-gated calcium channels, but was sensitive to GABA antagonists. We conclude that [Cl-]i is under circadian regulation in both AVP + and VIP + neurons.
Collapse
Affiliation(s)
- Nathan J. Klett
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Olga Cravetchi
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Charles N. Allen
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
5
|
Zhong L, Gleason EL. Adenylate Cyclase 1 Links Calcium Signaling to CFTR-Dependent Cytosolic Chloride Elevations in Chick Amacrine Cells. Front Cell Neurosci 2021; 15:726605. [PMID: 34456687 PMCID: PMC8385318 DOI: 10.3389/fncel.2021.726605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/19/2021] [Indexed: 01/03/2023] Open
Abstract
The strength and sign of synapses involving ionotropic GABA and glycine receptors are dependent upon the Cl- gradient. We have shown that nitric oxide (NO) elicits the release of Cl- from internal acidic stores in retinal amacrine cells (ACs); temporarily altering the Cl- gradient and the strength or even sign of incoming GABAergic or glycinergic synapses. The underlying mechanism for this effect of NO requires the cystic fibrosis transmembrane regulator (CFTR) but the link between NO and CFTR activation has not been determined. Here, we test the hypothesis that NO-dependent Ca2+ elevations activate the Ca2+-dependent adenylate cyclase 1 (AdC1) leading to activation of protein kinase A (PKA) whose activity is known to open the CFTR channel. Using the reversal potential of GABA-gated currents to monitor cytosolic Cl-, we established the requirement for Ca2+ elevations. Inhibitors of AdC1 suppressed the NO-dependent increases in cytosolic Cl- whereas inhibitors of other AdC subtypes were ineffective suggesting that AdC1 is involved. Inhibition of PKA also suppressed the action of NO. To address the sufficiency of this pathway in linking NO to elevations in cytosolic Cl-, GABA-gated currents were measured under internal and external zero Cl- conditions to isolate the internal Cl- store. Activators of the cAMP pathway were less effective than NO in producing GABA-gated currents. However, coupling the cAMP pathway activators with the release of Ca2+ from stores produced GABA-gated currents indistinguishable from those stimulated with NO. Together, these results demonstrate that cytosolic Ca2+ links NO to the activation of CFTR and the elevation of cytosolic Cl-.
Collapse
Affiliation(s)
- Li Zhong
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Evanna L Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
6
|
Paik SS, Park YS, Kim IB. Calcium- and Voltage-Dependent Dual Gating ANO1 is an Intrinsic Determinant of Repolarization in Rod Bipolar Cells of the Mouse Retina. Cells 2020; 9:cells9030543. [PMID: 32110998 PMCID: PMC7140511 DOI: 10.3390/cells9030543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 01/09/2023] Open
Abstract
TMEM16A/anoctamin1 (ANO1), a calcium (Ca2+)-activated chloride (Cl-) channel, has many functions in various excitable cells and modulates excitability in both Ca2+- and voltage-gating modes. However, its gating characteristics and role in primary neural cells remain unclear. Here, we characterized its Ca2+- and voltage-dependent components in rod bipolar cells using dissociated and slice preparations of the mouse retina. The I-V curves of Ca2+-dependent ANO1 tail current and voltage-gated Ca2+ channel (VGCC) are similar; as ANO1 is blocked by VGCC inhibitors, ANO1 may be gated by Ca2+ influx through VGCC. The voltage-dependent component of ANO1 has outward rectifying and sustained characteristics and is clearly isolated by the inhibitory effect of Cl- reduction and T16Ainh-A01, a selective ANO1 inhibitor, in high EGTA, a Ca2+ chelator. The voltage-dependent component disappears due to VGCC inhibition, suggesting that Ca2+ is the essential trigger for ANO1. In perforated current-clamping method, the application of T16Ainh-A01 and reduction of Cl- extended excitation periods in rod bipolar cells, revealing that ANO1 induces repolarization during excitation. Overall, ANO1 opens by VGCC activation during physiological excitation of the rod bipolar cell and has a voltage-dependent component. These two gating-modes concurrently provide the intrinsic characteristics of the membrane potential in rod bipolar cells.
Collapse
Affiliation(s)
- Sun-Sook Paik
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 100744, Korea; (S.-S.P.); (Y.S.P.)
| | - Yong Soo Park
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 100744, Korea; (S.-S.P.); (Y.S.P.)
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 100744, Korea; (S.-S.P.); (Y.S.P.)
- Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of Korea, Seoul 100744, Korea
- Correspondence: ; Tel.: +82-2-2258-7263; Fax: +82-2-536-3110
| |
Collapse
|
7
|
Wen X, Thoreson WB. Contributions of glutamate transporters and Ca 2+-activated Cl - currents to feedback from horizontal cells to cone photoreceptors. Exp Eye Res 2019; 189:107847. [PMID: 31628905 DOI: 10.1016/j.exer.2019.107847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/26/2019] [Accepted: 10/15/2019] [Indexed: 02/05/2023]
Abstract
Lateral inhibitory feedback from horizontal cells (HCs) to cones establishes center-surround receptive fields and color opponency in the retina. When HCs hyperpolarize to light, inhibitory feedback to cones increases activation of cone Ca2+ currents (ICa) that can in turn activate additional currents. We recorded simultaneously from cones and HCs to analyze cone currents activated by HC feedback in salamander retina. Depolarization-activated inward tail currents in cones were inhibited by CaCCinh-A01 that inhibits both Ano1 and Ano2 Ca2+-activated Cl- currents (ICl(Ca)). An Ano1-selective inhibitor Ani9 was less effective suggesting that Ano2 is the predominant ICl(Ca) subtype in cones. CaCCinh-A01 inhibited feedback currents more strongly when intracellular Ca2+ in cones was buffered with 0.05 mM EGTA compared to stronger buffering with 5 mM EGTA. By contrast, blocking glutamate transporter anion currents (ICl(Glu)) with TBOA had stronger inhibitory effects on cone feedback currents when Ca2+ buffering was strong. Inward feedback currents ran down at rates intermediate between rundown of glutamate release and ICl(Ca), consistent with contributions to feedback from both ICl(Ca) and ICl(Glu). These results suggest that Cl- channels coupled to glutamate transporters help to speed inward feedback currents initiated by local changes in intracellular [Ca2+] close to synaptic ribbons of cones whereas Ano2 Ca2+-activated Cl- channels contribute to slower components of feedback regulated by spatially extensive changes in intracellular [Ca2+].
Collapse
Affiliation(s)
- Xiangyi Wen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
8
|
Van Hook MJ, Nawy S, Thoreson WB. Voltage- and calcium-gated ion channels of neurons in the vertebrate retina. Prog Retin Eye Res 2019; 72:100760. [PMID: 31078724 PMCID: PMC6739185 DOI: 10.1016/j.preteyeres.2019.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
In this review, we summarize studies investigating the types and distribution of voltage- and calcium-gated ion channels in the different classes of retinal neurons: rods, cones, horizontal cells, bipolar cells, amacrine cells, interplexiform cells, and ganglion cells. We discuss differences among cell subtypes within these major cell classes, as well as differences among species, and consider how different ion channels shape the responses of different neurons. For example, even though second-order bipolar and horizontal cells do not typically generate fast sodium-dependent action potentials, many of these cells nevertheless possess fast sodium currents that can enhance their kinetic response capabilities. Ca2+ channel activity can also shape response kinetics as well as regulating synaptic release. The L-type Ca2+ channel subtype, CaV1.4, expressed in photoreceptor cells exhibits specific properties matching the particular needs of these cells such as limited inactivation which allows sustained channel activity and maintained synaptic release in darkness. The particular properties of K+ and Cl- channels in different retinal neurons shape resting membrane potentials, response kinetics and spiking behavior. A remaining challenge is to characterize the specific distributions of ion channels in the more than 100 individual cell types that have been identified in the retina and to describe how these particular ion channels sculpt neuronal responses to assist in the processing of visual information by the retina.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Scott Nawy
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
9
|
Modes of Accessing Bicarbonate for the Regulation of Membrane Guanylate Cyclase (ROS-GC) in Retinal Rods and Cones. eNeuro 2019; 6:eN-NWR-0393-18. [PMID: 30783616 PMCID: PMC6378327 DOI: 10.1523/eneuro.0393-18.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/20/2018] [Accepted: 01/15/2019] [Indexed: 11/21/2022] Open
Abstract
The membrane guanylate cyclase, ROS-GC, that synthesizes cyclic GMP for use as a second messenger for visual transduction in retinal rods and cones, is stimulated by bicarbonate. Bicarbonate acts directly on ROS-GC1, because it enhanced the enzymatic activity of a purified, recombinant fragment of bovine ROS-GC1 consisting solely of the core catalytic domain. Moreover, recombinant ROS-GC1 proved to be a true sensor of bicarbonate, rather than a sensor for CO2. Access to bicarbonate differed in rods and cones of larval salamander, Ambystoma tigrinum, of unknown sex. In rods, bicarbonate entered at the synapse and diffused to the outer segment, where it was removed by Cl--dependent exchange. In contrast, cones generated bicarbonate internally from endogenous CO2 or from exogenous CO2 that was present in extracellular solutions of bicarbonate. Bicarbonate production from both sources of CO2 was blocked by the carbonic anhydrase inhibitor, acetazolamide. Carbonic anhydrase II expression was verified immunohistochemically in cones but not in rods. In addition, cones acquired bicarbonate at their outer segments as well as at their inner segments. The multiple pathways for access in cones may support greater uptake of bicarbonate than in rods and buffer changes in its intracellular concentration.
Collapse
|
10
|
|
11
|
Benedetto R, Cabrita I, Schreiber R, Kunzelmann K. TMEM16A is indispensable for basal mucus secretion in airways and intestine. FASEB J 2018; 33:4502-4512. [PMID: 30586313 DOI: 10.1096/fj.201801333rrr] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Transmembrane member 16A (TMEM16A) is the Ca2+-activated chloride channel in airways and intestine. It has been associated with goblet cell metaplasia, as expression of TMEM16A is strongly up-regulated in cystic fibrosis and asthma during mucus hypersecretion. However, the possible role of TMEM16A for mucus production or mucus secretion remains obscure, and whether TMEM16A controls the function of intestinal goblet cells is entirely unknown. Basal mucus secretion in lungs occurs through low levels of ATP in the airway surface liquid. Here, we report for the first time that TMEM16A is essential for basal secretion of mucus in airways and intestine. Airway-ciliated and intestinal epithelial-specific knockout of TMEM16A ( TMEM16Aflox/floxFoxJ1, TMEM16Aflox/floxVil1) leads to accumulation of mucus in airway club (Clara) cells and intestinal goblet cells, respectively. Acute ATP-induced mucus secretion by airway club cells is inhibited when TMEM16A is knocked out in ciliated cells, possibly as a result of compromised release of prosecretory cytokines. Knockdown or inhibition of TMEM16A in human Calu3 airway epithelial cells indicates compromised IL-8 release. In intestinal goblet cells lacking expression of TMEM16A, mucus accumulates as a result of compromised ATP-induced secretion. In contrast, cholinergic mucus secretion by compound exocytosis is independent of TMEM16A. The data demonstrate a previously unrecognized role of TMEM16A for membrane exocytosis and describe a novel, ATP-driven pathway for intestinal mucus secretion. We conclude that ATP-dependent mucus secretion in both airways and intestine requires TMEM16A. The present results may form the basis for a novel, therapeutic approach for the treatment of mucus hypersecretion in inflammatory airway and intestinal disease.-Benedetto, R., Cabrita, I., Schreiber, R., Kunzelmann, K. TMEM16A is indispensable for basal mucus secretion in airways and intestine.
Collapse
Affiliation(s)
- Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Inês Cabrita
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| |
Collapse
|
12
|
Seo KH, Jin Y, Jung SY, Lee SH. Comprehensive behavioral analyses of anoctamin1/TMEM16A-conditional knockout mice. Life Sci 2018; 207:323-331. [PMID: 29928889 DOI: 10.1016/j.lfs.2018.06.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/07/2018] [Accepted: 06/15/2018] [Indexed: 10/28/2022]
Abstract
AIMS Anoctamin-1 (TMEM16A) is a calcium-activated chloride channel that is involved in numerous physiological conditions. Its role has been identified in electrophysiological and histological studies of genetic knockout animals. Recent cellular localization studies have shown that anoctamin-1 is co-expressed with presynaptic proteins, therefore its role in presynaptic terminals has been suggested. However, behavioral studies are lacking because conventional knockouts of anoctamin-1 are lethal after birth. In this study, we explored the role of anoctamin-1 in presynaptic terminals by analyzing the behavior of mice with conditional knockouts of anoctamin-1 in synapsin1-expressing cells. MAIN METHODS Using a synapsin1-Cre system, we selectively ablated anoctamin-1 in synapsin1 expressing cells. The mice were used in the behavioral experiments when they were between 6 and 9 months of age. KEY FINDINGS The mice with the conditional knockout of anoctamin-1 in synapsin1-expressing cells displayed impaired social behavior. In addition, the mice showed depressive-like behavior and decreased weight. However, these animals displayed normal locomotor activity, cognitive function, and motor coordination. SIGNIFICANCE These results suggested that anoctamin-1 is involved in psychiatric behavior because of its role in the regulation of synaptic transmission in presynaptic terminals.
Collapse
Affiliation(s)
- Kyoung Hee Seo
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeonsun Jin
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sun-Young Jung
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Sung Hoon Lee
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
13
|
Calcium-regulated chloride channel anoctamin-1 is present in the suprachiasmatic nuclei of rats. Neuroreport 2018; 29:334-339. [PMID: 29309309 DOI: 10.1097/wnr.0000000000000967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium-regulated chloride channel (CaCC) anoctamin-1 has been recently identified in neurons. In neurons, which express the Na-K-2Cl cotransporter, activation of CaCCs increases firing frequency, by reversion between the Cl equilibrium potential and the membrane resting potential, leading to membrane depolarization by Cl extrusion from the cell. Although there are no reports of CaCCs present in the suprachiasmatic nuclei (SCN), the fact that Na-K-2Cl cotransporter is present in SCN neurons, where it has been shown to be involved in the excitatory effects of γ-aminobutyric acid, together with the increase of neuronal firing rate induced by release of intracellular Ca after administration of 100 nM ryanodine, leads us to determine whether CaCCs are present in the SCN. Immunohistochemistry and western blots show the expression of the CaCCs anoctamin-1 protein. Quantitative PCR demonstrated the expression of anoctamin-1 mRNA in the SCN. These results clearly indicate the presence of CaCC in SCN of rats.
Collapse
|
14
|
A randomized, placebo-controlled trial of the benzoquinone idebenone in a mouse model of OPA1-related dominant optic atrophy reveals a limited therapeutic effect on retinal ganglion cell dendropathy and visual function. Neuroscience 2016; 319:92-106. [PMID: 26820596 DOI: 10.1016/j.neuroscience.2016.01.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 12/19/2022]
Abstract
Dominant optic atrophy (DOA) arises from mutations in the OPA1 gene that promotes fusion of the inner mitochondrial membrane and plays a role in maintaining ATP levels. Patients display optic disc pallor, retinal ganglion cell (RGC) loss and bilaterally reduced vision. We report a randomized, placebo-controlled trial of idebenone at 2000 mg/kg/day in 56 Opa1 mutant mice (B6;C3-Opa1(Q285STOP)), with RGC dendropathy and visual loss, and 63 wildtype mice. We assessed cellular responses in the retina, brain and liver and RGC morphology, by diolistic labeling, Sholl analysis and quantification of dendritic morphometric features. Vision was assessed by optokinetic responses. ATP levels were raised by 0.57 nmol/mg (97.73%, p=0.035) in brain from idebenone-treated Opa1 mutant mice, but in the liver there was an 80.35% (p=0.011) increase in oxidative damage. NQO1 expression in Opa1 mutant mice was reduced in the brain (to 30.5%, p=0.002) but not in retina, and neither expression level was induced by idebenone. ON-center RGCs failed to show major recovery, other than improvements in secondary dendritic length (by 53.89%, p=0.052) and dendritic territory (by 2.22 × 10(4) μm(2) or 90.24%, p=0.074). An improvement in optokinetic response was observed (by 12.2 ± 3.2s, p=0.003), but this effect was not sustained over time. OFF-center RGCs from idebenone-treated wildtype mice showed shrinkage in total dendritic length by 2.40 mm (48.05%, p=0.025) and a 47.37% diminished Sholl profile (p=0.029). Visual function in wildtype idebenone-treated mice was impaired (2.9 fewer head turns than placebo, p=0.007). Idebenone appears largely ineffective in protecting Opa1 heterozygous RGCs from dendropathy. The detrimental effect of idebenone in wildtype mice has not been previously observed and raises some concerns.
Collapse
|
15
|
Modulating Ca²⁺ signals: a common theme for TMEM16, Ist2, and TMC. Pflugers Arch 2015; 468:475-90. [PMID: 26700940 DOI: 10.1007/s00424-015-1767-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 12/21/2022]
Abstract
Since the discovery of TMEM16A (anoctamin 1, ANO1) as Ca(2+)-activated Cl(-) channel, the protein was found to serve different physiological functions, depending on the type of tissue. Subsequent reports on other members of the anoctamin family demonstrated a broad range of yet poorly understood properties. Compromised anoctamin function is causing a wide range of diseases, such as hearing loss (ANO2), bleeding disorder (ANO6), ataxia and dystonia (ANO3, 10), persistent borrelia and mycobacteria infection (ANO10), skeletal syndromes like gnathodiaphyseal dysplasia and limb girdle muscle dystrophy (ANO5), and cancer (ANO1, 6, 7). Animal models demonstrate CF-like airway disease, asthma, and intestinal hyposecretion (ANO1). Although present data indicate that ANO1 is a Ca(2+)-activated Cl(-) channel, it remains unclear whether all anoctamins form plasma membrane-localized or intracellular chloride channels. We find Ca(2+)-activated Cl(-) currents appearing by expression of most anoctamin paralogs, including the Nectria haematococca homologue nhTMEM16 and the yeast homologue Ist2. As recent studies show a role of anoctamins, Ist2, and the related transmembrane channel-like (TMC) proteins for intracellular Ca(2+) signaling, we will discuss the role of these proteins in generating compartmentalized Ca(2+) signals, which may give a hint as to the broad range of cellular functions of anoctamins.
Collapse
|
16
|
Zhang W, Schmelzeisen S, Parthier D, Frings S, Möhrlen F. Anoctamin Calcium-Activated Chloride Channels May Modulate Inhibitory Transmission in the Cerebellar Cortex. PLoS One 2015; 10:e0142160. [PMID: 26558388 PMCID: PMC4641602 DOI: 10.1371/journal.pone.0142160] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/19/2015] [Indexed: 01/18/2023] Open
Abstract
Calcium-activated chloride channels of the anoctamin (alias TMEM16) protein family fulfill critical functions in epithelial fluid transport, smooth muscle contraction and sensory signal processing. Little is known, however, about their contribution to information processing in the central nervous system. Here we examined the recent finding that a calcium-dependent chloride conductance impacts on GABAergic synaptic inhibition in Purkinje cells of the cerebellum. We asked whether anoctamin channels may underlie this chloride conductance. We identified two anoctamin channel proteins, ANO1 and ANO2, in the cerebellar cortex. ANO1 was expressed in inhibitory interneurons of the molecular layer and the granule cell layer. Both channels were expressed in Purkinje cells but, while ANO1 appeared to be retained in the cell body, ANO2 was targeted to the dendritic tree. Functional studies confirmed that ANO2 was involved in a calcium-dependent mode of ionic plasticity that reduces the efficacy of GABAergic synapses. ANO2 channels attenuated GABAergic transmission by increasing the postsynaptic chloride concentration, hence reducing the driving force for chloride influx. Our data suggest that ANO2 channels are involved in a Ca2+-dependent regulation of synaptic weight in GABAergic inhibition. Thus, in balance with the chloride extrusion mechanism via the co-transporter KCC2, ANO2 appears to regulate ionic plasticity in the cerebellum.
Collapse
Affiliation(s)
- Weiping Zhang
- Department of Animal Molecular Physiology, Centre of Organismal Studies, Im Neuenheimer Feld 504, Heidelberg University, Heidelberg, Germany
| | - Steffen Schmelzeisen
- Department of Animal Molecular Physiology, Centre of Organismal Studies, Im Neuenheimer Feld 504, Heidelberg University, Heidelberg, Germany
| | - Daniel Parthier
- Department of Animal Molecular Physiology, Centre of Organismal Studies, Im Neuenheimer Feld 504, Heidelberg University, Heidelberg, Germany
| | - Stephan Frings
- Department of Animal Molecular Physiology, Centre of Organismal Studies, Im Neuenheimer Feld 504, Heidelberg University, Heidelberg, Germany
| | - Frank Möhrlen
- Department of Animal Molecular Physiology, Centre of Organismal Studies, Im Neuenheimer Feld 504, Heidelberg University, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
17
|
Caputo A, Piano I, Demontis GC, Bacchi N, Casarosa S, Della Santina L, Gargini C. TMEM16A is associated with voltage-gated calcium channels in mouse retina and its function is disrupted upon mutation of the auxiliary α2δ4 subunit. Front Cell Neurosci 2015; 9:422. [PMID: 26557056 PMCID: PMC4617175 DOI: 10.3389/fncel.2015.00422] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/04/2015] [Indexed: 02/02/2023] Open
Abstract
Photoreceptors rely upon highly specialized synapses to efficiently transmit signals to multiple postsynaptic targets. Calcium influx in the presynaptic terminal is mediated by voltage-gated calcium channels (VGCC). This event triggers neurotransmitter release, but also gates calcium-activated chloride channels (TMEM), which in turn regulate VGCC activity. In order to investigate the relationship between VGCC and TMEM channels, we analyzed the retina of wild type (WT) and Cacna2d4 mutant mice, in which the VGCC auxiliary α2δ4 subunit carries a nonsense mutation, disrupting the normal channel function. Synaptic terminals of mutant photoreceptors are disarranged and synaptic proteins as well as TMEM16A channels lose their characteristic localization. In parallel, calcium-activated chloride currents are impaired in rods, despite unaltered TMEM16A protein levels. Co-immunoprecipitation revealed the interaction between VGCC and TMEM16A channels in the retina. Heterologous expression of these channels in tsA-201 cells showed that TMEM16A associates with the CaV1.4 subunit, and the association persists upon expression of the mutant α2δ4 subunit. Collectively, our experiments show association between TMEM16A and the α1 subunit of VGCC. Close proximity of these channels allows optimal function of the photoreceptor synaptic terminal under physiological conditions, but also makes TMEM16A channels susceptible to changes occurring to calcium channels.
Collapse
Affiliation(s)
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa Pisa, Italy
| | | | - Niccolò Bacchi
- Centre for Integrative Biology, University of Trento Trento, Italy
| | - Simona Casarosa
- Centre for Integrative Biology, University of Trento Trento, Italy
| | | | | |
Collapse
|
18
|
Dam VS, Boedtkjer DMB, Aalkjaer C, Matchkov V. The bestrophin- and TMEM16A-associated Ca(2+)- activated Cl(–) channels in vascular smooth muscles. Channels (Austin) 2015; 8:361-9. [PMID: 25478625 PMCID: PMC4203738 DOI: 10.4161/chan.29531] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The presence of Ca2+-activated Cl– currents (ICl(Ca)) in vascular smooth muscle cells (VSMCs) is well established. ICl(Ca) are supposedly important for arterial contraction by linking changes in [Ca2+]i and membrane depolarization. Bestrophins and some members of the TMEM16 protein family were recently associated with ICl(Ca). Two distinct ICl(Ca) are characterized in VSMCs; the cGMP-dependent ICl(Ca) dependent upon bestrophin expression and the ‘classical’ Ca2+-activated Cl– current, which is bestrophin-independent. Interestingly, TMEM16A is essential for both the cGMP-dependent and the classical ICl(Ca). Furthermore, TMEM16A has a role in arterial contraction while bestrophins do not. TMEM16A’s role in the contractile response cannot be explained however only by a simple suppression of the depolarization by Cl– channels. It is suggested that TMEM16A expression modulates voltage-gated Ca2+ influx in a voltage-independent manner and recent studies also demonstrate a complex role of TMEM16A in modulating other membrane proteins.
Collapse
|
19
|
Maurya DK, Henriques T, Marini M, Pedemonte N, Galietta LJV, Rock JR, Harfe BD, Menini A. Development of the Olfactory Epithelium and Nasal Glands in TMEM16A-/- and TMEM16A+/+ Mice. PLoS One 2015; 10:e0129171. [PMID: 26067252 PMCID: PMC4465891 DOI: 10.1371/journal.pone.0129171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 05/05/2015] [Indexed: 12/16/2022] Open
Abstract
TMEM16A/ANO1 is a calcium-activated chloride channel expressed in several types of epithelia and involved in various physiological processes, including proliferation and development. During mouse embryonic development, the expression of TMEM16A in the olfactory epithelium is dynamic. TMEM16A is expressed at the apical surface of the entire olfactory epithelium at embryonic day E12.5 while from E16.5 its expression is restricted to a region near the transition zone with the respiratory epithelium. To investigate whether TMEM16A plays a role in the development of the mouse olfactory epithelium, we obtained the first immunohistochemistry study comparing the morphological properties of the olfactory epithelium and nasal glands in TMEM16A-/- and TMEM16A+/+ littermate mice. A comparison between the expression of the olfactory marker protein and adenylyl cyclase III shows that genetic ablation of TMEM16A did not seem to affect the maturation of olfactory sensory neurons and their ciliary layer. As TMEM16A is expressed at the apical part of supporting cells and in their microvilli, we used ezrin and cytokeratin 8 as markers of microvilli and cell body of supporting cells, respectively, and found that morphology and development of supporting cells were similar in TMEM16A-/- and TMEM16A+/+ littermate mice. The average number of supporting cells, olfactory sensory neurons, horizontal and globose basal cells were not significantly different in the two types of mice. Moreover, we also observed that the morphology of Bowman’s glands, nasal septal glands and lateral nasal glands did not change in the absence of TMEM16A. Our results indicate that the development of mouse olfactory epithelium and nasal glands does not seem to be affected by the genetic ablation of TMEM16A.
Collapse
Affiliation(s)
- Devendra Kumar Maurya
- Laboratory of Olfactory Transduction, SISSA, International School for Advanced Studies, Trieste, Italy
| | - Tiago Henriques
- Laboratory of Olfactory Transduction, SISSA, International School for Advanced Studies, Trieste, Italy
| | | | | | | | - Jason R. Rock
- Department of Anatomy, UCSF School of Medicine, San Francisco, CA, United States of America
| | - Brian D. Harfe
- Department of Molecular Genetics and Microbiology Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, United States of America
| | - Anna Menini
- Laboratory of Olfactory Transduction, SISSA, International School for Advanced Studies, Trieste, Italy
- * E-mail:
| |
Collapse
|
20
|
Gui D, Li Y, Chen X. Alterations of TMEM16a allostery in human retinal microarterioles in long-standing hypertension. IUBMB Life 2015; 67:348-54. [PMID: 25914185 DOI: 10.1002/iub.1376] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/08/2015] [Indexed: 02/04/2023]
Affiliation(s)
- Dongmei Gui
- Department of Ophthalmology; Sheng Jing Hospital of China Medical University; Shenyang China
| | - Yanfeng Li
- The Second Department of Neurosurgery; The People's Hospital of Liaoning Province; Shenyang China
| | - Xiaolong Chen
- Department of Ophthalmology; Sheng Jing Hospital of China Medical University; Shenyang China
| |
Collapse
|
21
|
Matchkov VV, Boedtkjer DM, Aalkjaer C. The role of Ca2+ activated Cl− channels in blood pressure control. Curr Opin Pharmacol 2015; 21:127-37. [DOI: 10.1016/j.coph.2015.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 12/17/2022]
|
22
|
Four basic residues critical for the ion selectivity and pore blocker sensitivity of TMEM16A calcium-activated chloride channels. Proc Natl Acad Sci U S A 2015; 112:3547-52. [PMID: 25733897 DOI: 10.1073/pnas.1502291112] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
TMEM16A (transmembrane protein 16) (Anoctamin-1) forms a calcium-activated chloride channel (CaCC) that regulates a broad array of physiological properties in response to changes in intracellular calcium concentration. Although known to conduct anions according to the Eisenman type I selectivity sequence, the structural determinants of TMEM16A anion selectivity are not well-understood. Reasoning that the positive charges on basic residues are likely contributors to anion selectivity, we performed whole-cell recordings of mutants with alanine substitution for basic residues within the putative pore region and identified four residues on four different putative transmembrane segments that significantly increased the permeability of the larger halides and thiocyanate relative to that of chloride. Because TMEM16A permeation properties are known to shift with changes in intracellular calcium concentration, we further examined the calcium dependence of anion selectivity. We found that WT TMEM16A but not mutants with alanine substitution at those four basic residues exhibited a clear decline in the preference for larger anions as intracellular calcium was increased. Having implicated these residues as contributing to the TMEM16A pore, we scrutinized candidate small molecules from a high-throughput CaCC inhibitor screen to identify two compounds that act as pore blockers. Mutations of those four putative pore-lining basic residues significantly altered the IC50 of these compounds at positive voltages. These findings contribute to our understanding regarding anion permeation of TMEM16A CaCC and provide valuable pharmacological tools to probe the channel pore.
Collapse
|
23
|
Chae YJ, Lee HJ, Jeon JH, Kim IB, Choi JS, Sung KW, Hahn SJ. Effects of donepezil on hERG potassium channels. Brain Res 2014; 1597:77-85. [PMID: 25498859 DOI: 10.1016/j.brainres.2014.11.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/04/2014] [Accepted: 11/27/2014] [Indexed: 11/26/2022]
Abstract
Donepezil is a potent, selective inhibitor of acetylcholinesterase, which is used for the treatment of Alzheimer's disease. Whole-cell patch-clamp technique and Western blot analyses were used to study the effects of donepezil on the human ether-a-go-go-related gene (hERG) channel. Donepezil inhibited the tail current of the hERG in a concentration-dependent manner with an IC50 of 1.3 μM. The metabolites of donepezil, 6-ODD and 5-ODD, inhibited the hERG currents in a similar concentration-dependent manner; the IC50 values were 1.0 and 1.5 μM, respectively. A fast drug perfusion system demonstrated that donepezil interacted with both the open and inactivated states of the hERG. A fast application of donepezil during the tail currents inhibited the open state of the hERG in a concentration-dependent manner with an IC50 of 2.7 μM. Kinetic analysis of donepezil in an open state of the hERG yielded blocking and unblocking rate constants of 0.54 µM(-1)s(-1) and 1.82 s(-1), respectively. The block of the hERG by donepezil was voltage-dependent with a steep increase across the voltage range of channel activation. Donepezil caused a reduction in the hERG channel protein trafficking to the plasma membrane at low concentration, but decreased the channel protein expression at higher concentrations. These results suggest that donepezil inhibited the hERG at a supratherapeutic concentration, and that it did so by preferentially binding to the activated (open and/or inactivated) states of the channels and by inhibiting the trafficking and expression of the hERG channel protein in the plasma membrane.
Collapse
Affiliation(s)
- Yun Ju Chae
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, 222 Banpo-daero, Seocho-gu, Korea
| | - Hong Joon Lee
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Ji Hyun Jeon
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, 43-1 Yeokgok 2-dong, Wonmi-gu, Bucheon, Gyeonggi-do, Korea
| | - Ki-Wug Sung
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Sang June Hahn
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, 222 Banpo-daero, Seocho-gu, Korea.
| |
Collapse
|
24
|
Zahratka JA, Williams PDE, Summers PJ, Komuniecki RW, Bamber BA. Serotonin differentially modulates Ca2+ transients and depolarization in a C. elegans nociceptor. J Neurophysiol 2014; 113:1041-50. [PMID: 25411461 DOI: 10.1152/jn.00665.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Monoamines and neuropeptides modulate neuronal excitability and synaptic strengths, shaping circuit activity to optimize behavioral output. In C. elegans, a pair of bipolar polymodal nociceptors, the ASHs, sense 1-octanol to initiate escape responses. In the present study, 1-octanol stimulated large increases in ASH Ca(2+), mediated by L-type voltage-gated Ca(2+) channels (VGCCs) in the cell soma and L-plus P/Q-type VGCCs in the axon, which were further amplified by Ca(2+) released from intracellular stores. Importantly, 1-octanol-dependent aversive responses were not inhibited by reducing ASH L-VGCC activity genetically or pharmacologically. Serotonin, an enhancer of 1-octanol avoidance, potentiated 1-octanol-dependent ASH depolarization measured electrophysiologically, but surprisingly, decreased the ASH somal Ca(2+) transients. These results suggest that ASH somal Ca(2+) transient amplitudes may not always be predictive of neuronal depolarization and synaptic output. Therefore, although increases in steady-state Ca(2+) can reliably indicate when neurons become active, quantitative relationships between Ca(2+) transient amplitudes and neuronal activity may not be as straightforward as previously anticipated.
Collapse
Affiliation(s)
- Jeffrey A Zahratka
- Department of Biological Sciences, The University of Toledo, Toledo, Ohio
| | - Paul D E Williams
- Department of Biological Sciences, The University of Toledo, Toledo, Ohio
| | - Philip J Summers
- Department of Biological Sciences, The University of Toledo, Toledo, Ohio
| | | | - Bruce A Bamber
- Department of Biological Sciences, The University of Toledo, Toledo, Ohio
| |
Collapse
|
25
|
Liu Y, Zhang H, Huang D, Qi J, Xu J, Gao H, Du X, Gamper N, Zhang H. Characterization of the effects of Cl⁻ channel modulators on TMEM16A and bestrophin-1 Ca²⁺ activated Cl⁻ channels. Pflugers Arch 2014; 467:1417-1430. [PMID: 25078708 DOI: 10.1007/s00424-014-1572-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/13/2014] [Accepted: 06/30/2014] [Indexed: 11/30/2022]
Abstract
The Ca(2+) activated Cl(-) channels (CaCCs) play a multitude of important physiological functions. A number of candidate proteins have been proposed to form CaCC, but only two families, the bestrophins and the TMEM16 proteins, recapitulate the properties of native CaCC in expression systems. Studies of endogenous CaCCs are hindered by the lack of specific pharmacology as most Cl(-) channel modulators lack selectivity and a systematic comparison of the effects of these modulators on TMEM16A and bestrophin is missing. In the present study, we studied seven Cl(-) channel inhibitors: niflumic acid (NFA), NPPB, flufenamic acid (FFA), DIDS, tannic acid, CaCCinh-A01 and T16Ainh-A01 for their effects on TMEM16A and bestrophin-1 (Best1) stably expressed in CHO (Chinese hamster ovary) cells using patch clamp technique. Among seven inhibitors studied, NFA showed highest selectivity for TMEM16A (IC50 of 7.40 ± 0.95 μM) over Best1 (IC50 of 102.19 ± 15.05 μM). In contrast, DIDS displayed a reverse selectivity inhibiting Best1 with IC50 of 3.93 ± 0.73 μM and TMEM16A with IC50 of 548.86 ± 25.57 μM. CaCCinh-A01 was the most efficacious blocker for both TMEM16A and Best1 channels. T16Ainh-A01 partially inhibited TMEM16A currents but had no effect on Best1 currents. Tannic acid, NPPB and FFA had variable intermediate effects. Potentiation of channel activity by some of these modulators and the effects on TMEM16A deactivation kinetics were also described. Characterization of Cl(-) channel modulators for their effects on TMEM16A and Best1 will facilitate future studies of native CaCCs.
Collapse
Affiliation(s)
- Yani Liu
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China
| | - Huiran Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China
| | - Dongyang Huang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China
| | - Jinlong Qi
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China
| | - Jiaxi Xu
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China
| | - Haixia Gao
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China
| | - Xiaona Du
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China
| | - Nikita Gamper
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Hailin Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Department of Pharmacology, Hebei Medical University, Shijizhuang, Heibei, China.
| |
Collapse
|
26
|
Vocke K, Dauner K, Hahn A, Ulbrich A, Broecker J, Keller S, Frings S, Möhrlen F. Calmodulin-dependent activation and inactivation of anoctamin calcium-gated chloride channels. ACTA ACUST UNITED AC 2014; 142:381-404. [PMID: 24081981 PMCID: PMC3787769 DOI: 10.1085/jgp.201311015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Calcium-dependent chloride channels serve critical functions in diverse biological systems. Driven by cellular calcium signals, the channels codetermine excitatory processes and promote solute transport. The anoctamin (ANO) family of membrane proteins encodes three calcium-activated chloride channels, named ANO 1 (also TMEM16A), ANO 2 (also TMEM16B), and ANO 6 (also TMEM16F). Here we examined how ANO 1 and ANO 2 interact with Ca2+/calmodulin using nonstationary current analysis during channel activation. We identified a putative calmodulin-binding domain in the N-terminal region of the channel proteins that is involved in channel activation. Binding studies with peptides indicated that this domain, a regulatory calmodulin-binding motif (RCBM), provides two distinct modes of interaction with Ca2+/calmodulin, one at submicromolar Ca2+ concentrations and one in the micromolar Ca2+ range. Functional, structural, and pharmacological data support the concept that calmodulin serves as a calcium sensor that is stably associated with the RCBM domain and regulates the activation of ANO 1 and ANO 2 channels. Moreover, the predominant splice variant of ANO 2 in the brain exhibits Ca2+/calmodulin-dependent inactivation, a loss of channel activity within 30 s. This property may curtail ANO 2 activity during persistent Ca2+ signals in neurons. Mutagenesis data indicated that the RCBM domain is also involved in ANO 2 inactivation, and that inactivation is suppressed in the retinal ANO 2 splice variant. These results advance the understanding of Ca2+ regulation in anoctamin Cl− channels and its significance for the physiological function that anoctamin channels subserve in neurons and other cell types.
Collapse
Affiliation(s)
- Kerstin Vocke
- Department of Molecular Physiology, Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Cho SJ, Jeon JH, Chun DI, Yeo SW, Kim IB. Anoctamin 1 expression in the mouse auditory brainstem. Cell Tissue Res 2014; 357:563-9. [PMID: 24853671 DOI: 10.1007/s00441-014-1897-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/15/2014] [Indexed: 01/07/2023]
Abstract
Calcium-activated chloride channels (CaCCs) are involved in numerous physiological functions, including the epithelial movement of fluid. Anoctamin 1 (ANO1) has recently been cloned and characterized as a CaCC and is known to be expressed in various secretory epithelia and in nervous tissues such as the dorsal root ganglia and retina. However, data regarding the expression, function, and cellular and subcellular localization of CaCCs in the brain are still limited. We investigated the distribution and expression patterns of ANO1 in adult mouse brain. Reverse transcriptase plus the polymerase chain reaction, Western blot, and immunohistochemical analyses demonstrated that ANO1 was widely distributed throughout the brain. Furthermore, ANO1 was strongly expressed in two auditory brainstem nuclei: the medial nucleus of the trapezoid body (MNTB) and the anteroventral cochlear nucleus (AVCN). Double-labeling experiments revealed that this ANO1 expression was exclusive to the presynaptic endings of both the MNTB and AVCN. ANO1 is thus mainly localized at presynaptic terminals in various brain regions, specifically in two auditory brainstem nuclei, the MNTB and AVCN, and might therefore contribute to the high-frequency synaptic transmission of auditory signals.
Collapse
Affiliation(s)
- Sang Jae Cho
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea
| | | | | | | | | |
Collapse
|
28
|
Yi E, Lee J, Lee CJ. Developmental Role of Anoctamin-1/TMEM16A in Ca(2+)-Dependent Volume Change in Supporting Cells of the Mouse Cochlea. Exp Neurobiol 2013; 22:322-9. [PMID: 24465148 PMCID: PMC3897694 DOI: 10.5607/en.2013.22.4.322] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 12/14/2013] [Accepted: 12/14/2013] [Indexed: 11/19/2022] Open
Abstract
Mammalian cochlea undergoes morphological and functional changes during the postnatal period, around the hearing onset. Major changes during the initial 2 postnatal weeks of mouse include maturation of sensory hair cells and supporting cells, and acquisition of afferent and efferent innervations. During this period, supporting cells in the greater epithelial ridge (GER) of the cochlea exhibit spontaneous and periodic activities which involves ATP, increase in intracellular Ca(2+), and cell volume change. This Ca(2+)-dependent volume change has been proposed to involve chloride channels or transporters. We found that the spontaneous volume changes were eliminated by anion channel blocker, 100 µM NPPB. Among candidates, expression of Anoctamin-1 (Ano1 or TMEM16A), bestriphin-1 and NKCC1 were investigated in whole-mount cochlea of P9-10 mice. Immunolabeling indicated high level of Ano1 expression in the GER, but not of betrophin-1 or NKCC1. Double-labeling with calretinin and confocal image analysis further elucidated the cellular localization of Ano1 immunoreactivity in supporting cells. It was tested if the Ano1 expression exhibits similar time course to the spontaneous activities in postnatal cochlear supporting cells. Cochlear preparations from P2-3, P5-6, P9-10, P15-16 mice were subjected to immunolabeling. High level of Ano1 immunoreactivity was observed in the GER of P2-3, P5-6, P9-10 cochleae, but not of P15-17 cochleae. Taken together, the localization and time course in Ano1 expression pattern correlates with the spontaneous, periodic volume changes recorded in postnatal cochlear supporting cells. From these results we propose that Ano1 is the pacemaker of spontaneous activities in postnatal cochlea.
Collapse
Affiliation(s)
- Eunyoung Yi
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan 534-729, Korea
| | - Jaekwang Lee
- WCI Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791, Korea
| | - C Justin Lee
- WCI Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791, Korea. ; KU-KIST Graduate School of Converging Science & Techonology, Korea University, Seoul 136-790, Korea
| |
Collapse
|