1
|
Lowry AJ, Liang P, Song M, Wan Y, Pei ZM, Yang H, Zhang Y. TMEM16 and OSCA/TMEM63 proteins share a conserved potential to permeate ions and phospholipids. eLife 2024; 13:RP96957. [PMID: 39495104 PMCID: PMC11534332 DOI: 10.7554/elife.96957] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
The calcium-activated TMEM16 proteins and the mechanosensitive/osmolarity-activated OSCA/TMEM63 proteins belong to the Transmembrane Channel/Scramblase (TCS) superfamily. Within the superfamily, OSCA/TMEM63 proteins, as well as TMEM16A and TMEM16B, are thought to function solely as ion channels. However, most TMEM16 members, including TMEM16F, maintain an additional function as scramblases, rapidly exchanging phospholipids between leaflets of the membrane. Although recent studies have advanced our understanding of TCS structure-function relationships, the molecular determinants of TCS ion and lipid permeation remain unclear. Here, we show that single mutations along the transmembrane helix (TM) 4/6 interface allow non-scrambling TCS members to permeate phospholipids. In particular, this study highlights the key role of TM 4 in controlling TCS ion and lipid permeation and offers novel insights into the evolution of the TCS superfamily, suggesting that, like TMEM16s, the OSCA/TMEM63 family maintains a conserved potential to permeate ions and phospholipids.
Collapse
Affiliation(s)
- Augustus J Lowry
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Pengfei Liang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Mo Song
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryGuangdongChina
| | - Yuichun Wan
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
| | - Zhen-Ming Pei
- Department of Biology, Duke UniversityDurhamUnited States
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
- Department of Neurobiology, Duke University School of MedicineDurhamUnited States
| | - Yang Zhang
- Department of Biochemistry, Duke University School of MedicineDurhamUnited States
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryGuangdongChina
| |
Collapse
|
2
|
Ousingsawat J, Schreiber R, Kunzelmann K. Functional Interdependence of Anoctamins May Influence Conclusions from Overexpression Studies. Int J Mol Sci 2024; 25:9998. [PMID: 39337485 PMCID: PMC11432102 DOI: 10.3390/ijms25189998] [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] [Received: 08/05/2024] [Revised: 09/09/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Anoctamin 6 (ANO6, TMEM16F) is a phospholipid (PL) scramblase that moves PLs between both plasma membrane (PM) leaflets and operates as an ion channel. It plays a role in development and is essential for hemostasis, bone mineralization and immune defense. However, ANO6 has also been shown to regulate cellular Ca2+ signaling and PM compartments, thereby controlling the expression of ion channels such as CFTR. Given these pleiotropic effects, we investigated the functional interdependence of the ubiquitous ANO6 with other commonly co-expressed anoctamins. As most expression studies on anoctamins use HEK293 human embryonic kidney cells, we compared ion currents, PL scrambling and Ca2+ signals induced by the overexpression of anoctamins in HEK293 wild-type parental and ANO6-knockout cells. The data suggest that the endogenous expression of ANO6 significantly affects the results obtained from overexpressed anoctamins, particularly after increasing intracellular Ca2+. Thus, a significant interdependence of anoctamins may influence the interpretation of data obtained from the functional analysis of overexpressed anoctamins.
Collapse
Affiliation(s)
| | | | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, University Street 31, D-93053 Regensburg, Germany; (J.O.); (R.S.)
| |
Collapse
|
3
|
Genovese M, Galietta LJV. Anoctamin pharmacology. Cell Calcium 2024; 121:102905. [PMID: 38788257 DOI: 10.1016/j.ceca.2024.102905] [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: 03/21/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024]
Abstract
TMEM16 proteins, also known as anoctamins, are a family of ten membrane proteins with various tissue expression and subcellular localization. TMEM16A (anoctamin 1) is a plasma membrane protein that acts as a calcium-activated chloride channel. It is expressed in many types of epithelial cells, smooth muscle cells and some neurons. In airway epithelial cells, TMEM16A expression is particularly enhanced by inflammatory stimuli that also promote goblet cell metaplasia and mucus hypersecretion. Therefore, pharmacological modulation of TMEM16A could be beneficial to improve mucociliary clearance in chronic obstructive respiratory diseases. However, the correct approach to modulate TMEM16A activity (activation or inhibition) is still debated. Pharmacological inhibitors of TMEM16A could also be useful as anti-hypertensive agents given the TMEM16A role in smooth muscle contraction. In contrast to TMEM16A, TMEM16F (anoctamin 6) behaves as a calcium-activated phospholipid scramblase, responsible for the externalization of phosphatidylserine on cell surface. Inhibitors of TMEM16F could be useful as anti-coagulants and anti-viral agents. The role of other anoctamins as therapeutic targets is still unclear since their physiological role is still to be defined.
Collapse
Affiliation(s)
- Michele Genovese
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy; Department of Translational Medical Sciences (DISMET), University of Naples "Federico II", Italy.
| |
Collapse
|
4
|
Schreiber R, Ousingsawat J, Kunzelmann K. The anoctamins: Structure and function. Cell Calcium 2024; 120:102885. [PMID: 38642428 DOI: 10.1016/j.ceca.2024.102885] [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: 02/21/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/22/2024]
Abstract
When activated by increase in intracellular Ca2+, anoctamins (TMEM16 proteins) operate as phospholipid scramblases and as ion channels. Anoctamin 1 (ANO1) is the Ca2+-activated epithelial anion-selective channel that is coexpressed together with the abundant scramblase ANO6 and additional intracellular anoctamins. In salivary and pancreatic glands, ANO1 is tightly packed in the apical membrane and secretes Cl-. Epithelia of airways and gut use cystic fibrosis transmembrane conductance regulator (CFTR) as an apical Cl- exit pathway while ANO1 supports Cl- secretion mainly by facilitating activation of luminal CFTR and basolateral K+ channels. Under healthy conditions ANO1 modulates intracellular Ca2+ signals by tethering the endoplasmic reticulum, and except of glands its direct secretory contribution as Cl- channel might be small, compared to CFTR. In the kidneys ANO1 supports proximal tubular acid secretion and protein reabsorption and probably helps to excrete HCO3-in the collecting duct epithelium. However, under pathological conditions as in polycystic kidney disease, ANO1 is strongly upregulated and may cause enhanced proliferation and cyst growth. Under pathological condition, ANO1 and ANO6 are upregulated and operate as secretory channel/phospholipid scramblases, partly by supporting Ca2+-dependent processes. Much less is known about the role of other epithelial anoctamins whose potential functions are discussed in this review.
Collapse
Affiliation(s)
- Rainer Schreiber
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany.
| |
Collapse
|
5
|
Kunzelmann K, Ousingsawat J, Schreiber R. VSI: The anoctamins: Structure and function: "Intracellular" anoctamins. Cell Calcium 2024; 120:102888. [PMID: 38657371 DOI: 10.1016/j.ceca.2024.102888] [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: 02/20/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Plasma membrane localized anoctamin 1, 2 and 6 (TMEM16A, B, F) have been examined in great detail with respect to structure and function, but much less is known about the other seven intracellular members of this exciting family of proteins. This is probably due to their limited accessibility in intracellular membranous compartments, such as the endoplasmic reticulum (ER) or endosomes. However, these so-called intracellular anoctamins are also found in the plasma membrane (PM) which adds to the confusion regarding their cellular role. Probably all intracellular anoctamins except of ANO8 operate as intracellular phospholipid (PL) scramblases, allowing for Ca2+-activated, passive transport of phospholipids like phosphatidylserine between both membrane leaflets. Probably all of them also conduct ions, which is probably part of their physiological function. In this brief overview, we summarize key findings on the biological functions of ANO3, 4, 5, 7, 8, 9 and 10 (TMEM16C, D, E, G, H, J, K) that are gradually coming to light. Compartmentalized regulation of intracellular Ca2+ signals, tethering of the ER to specific PM contact sites, and control of intracellular vesicular trafficking appear to be some of the functions of intracellular anoctamins, while loss of function and abnormal expression are the cause for various diseases.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Physiological Institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany.
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany
| |
Collapse
|
6
|
Dibattista M, Pifferi S, Hernandez-Clavijo A, Menini A. The physiological roles of anoctamin2/TMEM16B and anoctamin1/TMEM16A in chemical senses. Cell Calcium 2024; 120:102889. [PMID: 38677213 DOI: 10.1016/j.ceca.2024.102889] [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: 02/29/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Chemical senses allow animals to detect and discriminate a vast array of molecules. The olfactory system is responsible of the detection of small volatile molecules, while water dissolved molecules are detected by taste buds in the oral cavity. Moreover, many animals respond to signaling molecules such as pheromones and other semiochemicals through the vomeronasal organ. The peripheral organs dedicated to chemical detection convert chemical signals into perceivable information through the employment of diverse receptor types and the activation of multiple ion channels. Two ion channels, TMEM16B, also known as anoctamin2 (ANO2) and TMEM16A, or anoctamin1 (ANO1), encoding for Ca2+-activated Cl¯ channels, have been recently described playing critical roles in various cell types. This review aims to discuss the main properties of TMEM16A and TMEM16B-mediated currents and their physiological roles in chemical senses. In olfactory sensory neurons, TMEM16B contributes to amplify the odorant response, to modulate firing, response kinetics and adaptation. TMEM16A and TMEM16B shape the pattern of action potentials in vomeronasal sensory neurons increasing the interspike interval. In type I taste bud cells, TMEM16A is activated during paracrine signaling mediated by ATP. This review aims to shed light on the regulation of diverse signaling mechanisms and neuronal excitability mediated by Ca-activated Cl¯ channels, hinting at potential new roles for TMEM16A and TMEM16B in the chemical senses.
Collapse
Affiliation(s)
- Michele Dibattista
- Department of Translational Biomedicine and Neuroscience, University of Bari A. Moro, 70121 Bari, Italy
| | - Simone Pifferi
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy.
| | - Andres Hernandez-Clavijo
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna Menini
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy.
| |
Collapse
|
7
|
Liang P, Zhang Y, Wan YCS, Ma S, Dong P, Lowry AJ, Francis SJ, Khandelwal S, Delahunty M, Telen MJ, Strouse JJ, Arepally GM, Yang H. Deciphering and disrupting PIEZO1-TMEM16F interplay in hereditary xerocytosis. Blood 2024; 143:357-369. [PMID: 38033286 PMCID: PMC10862370 DOI: 10.1182/blood.2023021465] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/07/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
ABSTRACT Cell-surface exposure of phosphatidylserine (PS) is essential for phagocytic clearance and blood clotting. Although a calcium-activated phospholipid scramblase (CaPLSase) has long been proposed to mediate PS exposure in red blood cells (RBCs), its identity, activation mechanism, and role in RBC biology and disease remain elusive. Here, we demonstrate that TMEM16F, the long-sought-after RBC CaPLSase, is activated by calcium influx through the mechanosensitive channel PIEZO1 in RBCs. PIEZO1-TMEM16F functional coupling is enhanced in RBCs from individuals with hereditary xerocytosis (HX), an RBC disorder caused by PIEZO1 gain-of-function channelopathy. Enhanced PIEZO1-TMEM16F coupling leads to an increased propensity to expose PS, which may serve as a key risk factor for HX clinical manifestations including anemia, splenomegaly, and postsplenectomy thrombosis. Spider toxin GsMTx-4 and antigout medication benzbromarone inhibit PIEZO1, preventing force-induced echinocytosis, hemolysis, and PS exposure in HX RBCs. Our study thus reveals an activation mechanism of TMEM16F CaPLSase and its pathophysiological function in HX, providing insights into potential treatment.
Collapse
Affiliation(s)
- Pengfei Liang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Yang Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Yui Chun S. Wan
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Shang Ma
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX
| | - Ping Dong
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Augustus J. Lowry
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
| | - Samuel J. Francis
- Department of Surgery, Duke University School of Medicine, Durham, NC
| | - Sanjay Khandelwal
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Martha Delahunty
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Marilyn J. Telen
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - John J. Strouse
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | | | - Huanghe Yang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC
- Department of Neurobiology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
8
|
Nguyen DM, Chen TY. Structure and Function of Calcium-Activated Chloride Channels and Phospholipid Scramblases in the TMEM16 Family. Handb Exp Pharmacol 2024; 283:153-180. [PMID: 35792944 DOI: 10.1007/164_2022_595] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The transmembrane protein 16 (TMEM16) family consists of Ca2+-activated chloride channels and phospholipid scramblases. Ten mammalian TMEM16 proteins, TMEM16A-K (with no TMEM16I), and several non-mammalian TMEM16 proteins, such as afTMEM16 and nhTMEM16, have been discovered. All known TMEM16 proteins are homodimeric proteins containing two subunits. Each subunit consists of ten transmembrane helices with Ca2+-binding sites and a single ion-permeation/phospholipid transport pathway. The ion-permeation pathway and the phospholipid transport pathway of TMEM16 proteins have a wide intracellular vestibule, a narrow neck, and a smaller extracellular vestibule. Interestingly, the lining wall of the ion-permeation/phospholipid transport pathway may be formed, at least partially, by membrane phospholipids, though the degree of pore-wall forming by phospholipids likely varies among TMEM16 proteins. Thus, the biophysical properties and activation mechanisms of TMEM16 proteins could differ from each other accordingly. Here we review the current understanding of the structure and function of TMEM16 molecules.
Collapse
Affiliation(s)
- Dung Manh Nguyen
- Center for Neuroscience, University of California, Davis, CA, USA.
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Tsung-Yu Chen
- Department of Neurology, Center for Neuroscience, University of California, Davis, CA, USA.
| |
Collapse
|
9
|
Agbani EO, Chow L, Nicholas J, Skeith L, Schneider P, Gregory A, Mahe E, Yamaura L, Young D, Dufour A, Paul PP, Walker AM, Mukherjee PG, Poole AW, Poon MC, Lee A. Overexpression of facilitative glucose transporter-3 and membrane procoagulation in maternal platelets of preeclamptic pregnancy. J Thromb Haemost 2023; 21:1903-1919. [PMID: 36963633 DOI: 10.1016/j.jtha.2023.03.014] [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: 08/03/2022] [Revised: 01/19/2023] [Accepted: 03/03/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND Preeclampsia (PE) is a hypertensive disorder during pregnancy that results in significant adverse maternal and neonatal outcomes. Platelet activation is present in PE and contributes to the thrombo-hemorrhagic states of the disorder. However, the mechanisms that initiate and/or sustain platelet activation in PE are ill-defined. OBJECTIVES We aimed to characterise this mechanism and the procoagulant potentials of platelets in PE. METHODS In this quantitative observational study, we analyzed platelet procoagulant membrane dynamics in patients with PE (n = 21) compared with age-matched normotensive pregnancies (n = 20), gestational hypertension (n = 10), and non-pregnant female controls (n = 19). We analyzed fluorescently labeled indicators of platelet activation, bioenergetics, and procoagulation (phosphatidylserine exposure and thrombin generation), coupled with high-resolution imaging and thrombelastography. We then validated our findings using flow cytometry, immunoassays, classical pharmacology, and convolutional neural network analysis. RESULTS PE platelets showed significant ultra-structural remodeling, are more extensively preactivated than in healthy pregnancies and can circulate as microaggregates. Preactivated platelets of PE externalized phosphatidylserine and thrombin formed on the platelet membranes. Platelets' expression of facilitative glucose transporter-1 increased in all pregnant groups. However, PE platelets additionally overexpress glucose transporter-3 to enhance glucose uptake and sustain activation and secretion events. Although preeclampsia platelets exposed to subendothelial collagen showed incremental activation, the absolute hemostatic response to collagen was diminished, and likely contributed to greater blood loss perioperatively. CONCLUSIONS We revealed 2 bioenergetic mediators in the mechanism of sustained platelet procoagulation in preeclampsia. Although glucose transporter-1 and glucose transporter-3 remain elusive antiprocoagulant targets, they may be sensitive monitors of PE onset and progression.
Collapse
Affiliation(s)
- Ejaife O Agbani
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Alberta, Canada; Libin Cardiovascular Institute, Calgary, Alberta, Canada.
| | - Lorraine Chow
- Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Joshua Nicholas
- Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Leslie Skeith
- Libin Cardiovascular Institute, Calgary, Alberta, Canada; Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Prism Schneider
- Department of Surgery, Cumming School of Medicine, University of Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Alexander Gregory
- Libin Cardiovascular Institute, Calgary, Alberta, Canada; Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Etienne Mahe
- Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada; Department of Pathology & Laboratory Medicine, University of Calgary, Alberta, Canada
| | - Lisa Yamaura
- Department of Surgery, Cumming School of Medicine, University of Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Daniel Young
- McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Antoine Dufour
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Padma Polash Paul
- Braintoy Inc Calgary and Computational Neuroscience Lab, University of Oxford, England, United Kingdom
| | - Andrew M Walker
- Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | | | - Alastair W Poole
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, England, United Kingdom
| | - Man-Chiu Poon
- Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada; Arnie Charbonneau Cancer Institute, Calgary, Alberta, Canada
| | - Adrienne Lee
- Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada; Division of Hematology, Department of Medicine/Medical Oncology, University of British Columbia, Island Health, Victoria, Canada
| |
Collapse
|
10
|
Tatsumi M, Kishi T, Ishida S, Kawana H, Uwamizu A, Ono Y, Kawakami K, Aoki J, Inoue A. Ectodomain shedding of EGFR ligands serves as an activation readout for TRP channels. PLoS One 2023; 18:e0280448. [PMID: 36668668 PMCID: PMC9858409 DOI: 10.1371/journal.pone.0280448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023] Open
Abstract
Transient receptor potential (TRP) channels are activated by various extracellular and intracellular stimuli and are involved in many physiological events. Because compounds that act on TRP channels are potential candidates for therapeutic agents, a simple method for evaluating TRP channel activation is needed. In this study, we demonstrated that a transforming growth factor alpha (TGFα) shedding assay, previously developed for detecting G-protein-coupled receptor (GPCR) activation, can also detect TRP channel activation. This assay is a low-cost, easily accessible method that requires only an absorbance microplate reader. Mechanistically, TRP-channel-triggered TGFα shedding is achieved by both of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and 17 (ADAM17), whereas the GPCR-induced TGFα shedding response depends solely on ADAM17. This difference may be the result of qualitative or quantitative differences in intracellular Ca2+ kinetics between TRP channels and GPCRs. Use of epidermal growth factor (EGF) and betacellulin (BTC), substrates of ADAM10, improved the specificity of the shedding assay by reducing background responses mediated by endogenously expressed GPCRs. This assay for TRP channel measurement will not only facilitate the high-throughput screening of TRP channel ligands but also contribute to understanding the roles played by TRP channels as regulators of membrane protein ectodomain shedding.
Collapse
Affiliation(s)
- Manae Tatsumi
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takayuki Kishi
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Satoru Ishida
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroki Kawana
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Akiharu Uwamizu
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuki Ono
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kouki Kawakami
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Asuka Inoue
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- * E-mail:
| |
Collapse
|
11
|
Ke K, Li L, Lu C, Zhu Q, Wang Y, Mou Y, Wang H, Jin W. The crosstalk effect between ferrous and other ions metabolism in ferroptosis for therapy of cancer. Front Oncol 2022; 12:916082. [PMID: 36033459 PMCID: PMC9413412 DOI: 10.3389/fonc.2022.916082] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/19/2022] [Indexed: 11/15/2022] Open
Abstract
Ferroptosis is an iron-dependent cell death process characterized by excessive accumulation of reactive oxygen species and lipid peroxidation. The elucidation of ferroptosis pathways may lead to novel cancer therapies. Current evidence suggests that the mechanism of ferroptosis can be summarized as oxidative stress and antioxidant defense mechanisms. During this process, ferrous ions play a crucial role in cellular oxidation, plasma membrane damage, reactive oxygen species removal imbalance and lipid peroxidation. Although, disregulation of intracellular cations (Fe2+, Ca2+, Zn2+, etc.) and anions (Cl-, etc.) have been widely reported to be involved in ferroptosis, their specific regulatory mechanisms have not been established. To further understand the crosstalk effect between ferrous and other ions in ferroptosis, we reviewed the ferroptosis process from the perspective of ions metabolism. In addition, the role of ferrous and other ions in tumor therapy is briefly summarized.
Collapse
Affiliation(s)
- Kun Ke
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Li Li
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Chao Lu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Qicong Zhu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yuanyu Wang
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yiping Mou
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Huiju Wang
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- *Correspondence: Weiwei Jin, ; Huiju Wang,
| | - Weiwei Jin
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- *Correspondence: Weiwei Jin, ; Huiju Wang,
| |
Collapse
|
12
|
Zhang Y, Liang P, Yang L, Shan KZ, Feng L, Chen Y, Liedtke W, Coyne CB, Yang H. Functional coupling between TRPV4 channel and TMEM16F modulates human trophoblast fusion. eLife 2022; 11:e78840. [PMID: 35670667 PMCID: PMC9236608 DOI: 10.7554/elife.78840] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/05/2022] [Indexed: 11/15/2022] Open
Abstract
TMEM16F, a Ca2+-activated phospholipid scramblase (CaPLSase), is critical for placental trophoblast syncytialization, HIV infection, and SARS-CoV2-mediated syncytialization, however, how TMEM16F is activated during cell fusion is unclear. Here, using trophoblasts as a model for cell fusion, we demonstrate that Ca2+ influx through the Ca2+ permeable transient receptor potential vanilloid channel TRPV4 is critical for TMEM16F activation and plays a role in subsequent human trophoblast fusion. GSK1016790A, a TRPV4 specific agonist, robustly activates TMEM16F in trophoblasts. We also show that TRPV4 and TMEM16F are functionally coupled within Ca2+ microdomains in a human trophoblast cell line using patch-clamp electrophysiology. Pharmacological inhibition or gene silencing of TRPV4 hinders TMEM16F activation and subsequent trophoblast syncytialization. Our study uncovers the functional expression of TRPV4 and one of the physiological activation mechanisms of TMEM16F in human trophoblasts, thus providing us with novel strategies to regulate CaPLSase activity as a critical checkpoint of physiologically and disease-relevant cell fusion events.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Pengfei Liang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Liheng Yang
- Department of Molecular Genetics and Microbiology, Duke University Medical CenterDurhamUnited States
| | - Ke Zoe Shan
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University Medical CentreDurhamUnited States
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua HospitalShanghaiChina
| | - Yong Chen
- Department of Neurology, Duke University Medical CenterDurhamUnited States
| | - Wolfgang Liedtke
- Department of Neurology, Duke University Medical CenterDurhamUnited States
- Department of Anesthesiology, Duke University Medical CenterDurhamUnited States
- Department of Neurobiology, Duke University Medical CenterDurhamUnited States
- College of Dentistry, Department of Molecular Pathobiology, NYUNew YorkUnited States
| | - Carolyn B Coyne
- Department of Molecular Genetics and Microbiology, Duke University Medical CenterDurhamUnited States
- Duke Human Vaccine Institute, Duke UniversityDurhamUnited States
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
- Department of Neurobiology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
13
|
Pifferi S, Boccaccio A. Ca 2+-Activated Chloride Channels and Phospholipid Scramblases. Int J Mol Sci 2022; 23:ijms23042158. [PMID: 35216275 PMCID: PMC8875746 DOI: 10.3390/ijms23042158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 01/29/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Simone Pifferi
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
- Correspondence: (S.P.); (A.B.)
| | - Anna Boccaccio
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy
- Correspondence: (S.P.); (A.B.)
| |
Collapse
|
14
|
Polymodal Control of TMEM16x Channels and Scramblases. Int J Mol Sci 2022; 23:ijms23031580. [PMID: 35163502 PMCID: PMC8835819 DOI: 10.3390/ijms23031580] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
The TMEM16A/anoctamin-1 calcium-activated chloride channel (CaCC) contributes to a range of vital functions, such as the control of vascular tone and epithelial ion transport. The channel is a founding member of a family of 10 proteins (TMEM16x) with varied functions; some members (i.e., TMEM16A and TMEM16B) serve as CaCCs, while others are lipid scramblases, combine channel and scramblase function, or perform additional cellular roles. TMEM16x proteins are typically activated by agonist-induced Ca2+ release evoked by Gq-protein-coupled receptor (GqPCR) activation; thus, TMEM16x proteins link Ca2+-signalling with cell electrical activity and/or lipid transport. Recent studies demonstrate that a range of other cellular factors—including plasmalemmal lipids, pH, hypoxia, ATP and auxiliary proteins—also control the activity of the TMEM16A channel and its paralogues, suggesting that the TMEM16x proteins are effectively polymodal sensors of cellular homeostasis. Here, we review the molecular pathophysiology, structural biology, and mechanisms of regulation of TMEM16x proteins by multiple cellular factors.
Collapse
|
15
|
Wray S, Prendergast C, Arrowsmith S. Calcium-Activated Chloride Channels in Myometrial and Vascular Smooth Muscle. Front Physiol 2021; 12:751008. [PMID: 34867456 PMCID: PMC8637852 DOI: 10.3389/fphys.2021.751008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/24/2021] [Indexed: 11/24/2022] Open
Abstract
In smooth muscle tissues, calcium-activated chloride channels (CaCC) provide the major anionic channel. Opening of these channels leads to chloride efflux and depolarization of the myocyte membrane. In this way, activation of the channels by a rise of intracellular [Ca2+], from a variety of sources, produces increased excitability and can initiate action potentials and contraction or increased tone. We now have a good mechanistic understanding of how the channels are activated and regulated, due to identification of TMEM16A (ANO1) as the molecular entity of the channel, but key questions remain. In reviewing these channels and comparing two distinct smooth muscles, myometrial and vascular, we expose the differences that occur in their activation mechanisms, properties, and control. We find that the myometrium only expresses “classical,” Ca2+-activated, and voltage sensitive channels, whereas both tonic and phasic blood vessels express classical, and non-classical, cGMP-regulated CaCC, which are voltage insensitive. This translates to more complex activation and regulation in vascular smooth muscles, irrespective of whether they are tonic or phasic. We therefore tentatively conclude that although these channels are expressed and functionally important in all smooth muscles, they are probably not part of the mechanisms governing phasic activity. Recent knockdown studies have produced unexpected functional results, e.g. no effects on labour and delivery, and tone increasing in some but decreasing in other vascular beds, strongly suggesting that there is still much to be explored concerning CaCC in smooth muscle.
Collapse
Affiliation(s)
- Susan Wray
- Department of Women and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Clodagh Prendergast
- Department of Women and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Sarah Arrowsmith
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
16
|
Hawn MB, Akin E, Hartzell H, Greenwood IA, Leblanc N. Molecular mechanisms of activation and regulation of ANO1-Encoded Ca 2+-Activated Cl - channels. Channels (Austin) 2021; 15:569-603. [PMID: 34488544 PMCID: PMC8480199 DOI: 10.1080/19336950.2021.1975411] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 08/29/2021] [Indexed: 01/13/2023] Open
Abstract
Ca2+-activated Cl- channels (CaCCs) perform a multitude of functions including the control of cell excitability, regulation of cell volume and ionic homeostasis, exocrine and endocrine secretion, fertilization, amplification of olfactory sensory function, and control of smooth muscle cell contractility. CaCCs are the translated products of two members (ANO1 and ANO2, also known as TMEM16A and TMEM16B) of the Anoctamin family of genes comprising ten paralogs. This review focuses on recent progress in understanding the molecular mechanisms involved in the regulation of ANO1 by cytoplasmic Ca2+, post-translational modifications, and how the channel protein interacts with membrane lipids and protein partners. After first reviewing the basic properties of native CaCCs, we then present a brief historical perspective highlighting controversies about their molecular identity in native cells. This is followed by a summary of the fundamental biophysical and structural properties of ANO1. We specifically address whether the channel is directly activated by internal Ca2+ or indirectly through the intervention of the Ca2+-binding protein Calmodulin (CaM), and the structural domains responsible for Ca2+- and voltage-dependent gating. We then review the regulation of ANO1 by internal ATP, Calmodulin-dependent protein kinase II-(CaMKII)-mediated phosphorylation and phosphatase activity, membrane lipids such as the phospholipid phosphatidyl-(4,5)-bisphosphate (PIP2), free fatty acids and cholesterol, and the cytoskeleton. The article ends with a survey of physical and functional interactions of ANO1 with other membrane proteins such as CLCA1/2, inositol trisphosphate and ryanodine receptors in the endoplasmic reticulum, several members of the TRP channel family, and the ancillary Κ+ channel β subunits KCNE1/5.
Collapse
Affiliation(s)
- M. B. Hawn
- Department of Pharmacology and Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| | - E. Akin
- Department of Pharmacology and Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| | - H.C. Hartzell
- Department of Cell Biology, Emory University School of Medicine, USA
| | - I. A. Greenwood
- Department of Vascular Pharmacology, St. George’s University of London, UK
| | - N. Leblanc
- Department of Pharmacology and Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| |
Collapse
|
17
|
Le SC, Liang P, Lowry AJ, Yang H. Gating and Regulatory Mechanisms of TMEM16 Ion Channels and Scramblases. Front Physiol 2021; 12:787773. [PMID: 34867487 PMCID: PMC8640346 DOI: 10.3389/fphys.2021.787773] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022] Open
Abstract
The transmembrane protein 16 (TMEM16) family consists of Ca2+-activated ion channels and Ca2+-activated phospholipid scramblases (CaPLSases) that passively flip-flop phospholipids between the two leaflets of the membrane bilayer. Owing to their diverse functions, TMEM16 proteins have been implicated in various human diseases, including asthma, cancer, bleeding disorders, muscular dystrophy, arthritis, epilepsy, dystonia, ataxia, and viral infection. To understand TMEM16 proteins in health and disease, it is critical to decipher their molecular mechanisms of activation gating and regulation. Structural, biophysical, and computational characterizations over the past decade have greatly advanced the molecular understanding of TMEM16 proteins. In this review, we summarize major structural features of the TMEM16 proteins with a focus on regulatory mechanisms and gating.
Collapse
Affiliation(s)
- Son C. Le
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
| | - Pengfei Liang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
| | - Augustus J. Lowry
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
- Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
18
|
Anion and Cation Permeability of the Mouse TMEM16F Calcium-Activated Channel. Int J Mol Sci 2021; 22:ijms22168578. [PMID: 34445284 PMCID: PMC8395294 DOI: 10.3390/ijms22168578] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 12/27/2022] Open
Abstract
TMEM16F is involved in several physiological processes, such as blood coagulation, bone development and virus infections. This protein acts both as a Ca2+-dependent phospholipid scramblase and a Ca2+-activated ion channel but several studies have reported conflicting results about the ion selectivity of the TMEM16F-mediated current. Here, we have performed a detailed side-by-side comparison of the ion selectivity of TMEM16F using the whole-cell and inside-out excised patch configurations to directly compare the results. In inside-out configuration, Ca2+-dependent activation was fast and the TMEM16F-mediated current was activated in a few milliseconds, while in whole-cell recordings full activation required several minutes. We determined the relative permeability between Na+ and Cl¯ (PNa/PCl) using the dilution method in both configurations. The TMEM16F-mediated current was highly nonselective, but there were differences depending on the configuration of the recordings. In whole-cell recordings, PNa/PCl was approximately 0.5, indicating a slight preference for Cl¯ permeation. In contrast, in inside-out experiments the TMEM16F channel showed a higher permeability for Na+ with PNa/PCl reaching 3.7. Our results demonstrate that the time dependence of Ca2+ activation and the ion selectivity of TMEM16F depend on the recording configuration.
Collapse
|
19
|
Okada Y, Sato-Numata K, Sabirov RZ, Numata T. Cell Death Induction and Protection by Activation of Ubiquitously Expressed Anion/Cation Channels. Part 2: Functional and Molecular Properties of ASOR/PAC Channels and Their Roles in Cell Volume Dysregulation and Acidotoxic Cell Death. Front Cell Dev Biol 2021; 9:702317. [PMID: 34307382 PMCID: PMC8299559 DOI: 10.3389/fcell.2021.702317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/18/2021] [Indexed: 12/18/2022] Open
Abstract
For survival and functions of animal cells, cell volume regulation (CVR) is essential. Major hallmarks of necrotic and apoptotic cell death are persistent cell swelling and shrinkage, and thus they are termed the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. A number of ubiquitously expressed anion and cation channels play essential roles not only in CVR but also in cell death induction. This series of review articles address the question how cell death is induced or protected with using ubiquitously expressed ion channels such as swelling-activated anion channels, acid-activated anion channels, and several types of TRP cation channels including TRPM2 and TRPM7. In the Part 1, we described the roles of swelling-activated VSOR/VRAC anion channels. Here, the Part 2 focuses on the roles of the acid-sensitive outwardly rectifying (ASOR) anion channel, also called the proton-activated chloride (PAC) anion channel, which is activated by extracellular protons in a manner sharply dependent on ambient temperature. First, we summarize phenotypical properties, the molecular identity, and the three-dimensional structure of ASOR/PAC. Second, we highlight the unique roles of ASOR/PAC in CVR dysfunction and in the induction of or protection from acidotoxic cell death under acidosis and ischemic conditions.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan.,Department of Physiology, School of Medicine, Aichi Medical University, Nagakute, Japan.,Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kaori Sato-Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Ravshan Z Sabirov
- Laboratory of Molecular Physiology, Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Tomohiro Numata
- Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
20
|
Kostritskii AY, Machtens JP. Molecular mechanisms of ion conduction and ion selectivity in TMEM16 lipid scramblases. Nat Commun 2021; 12:2826. [PMID: 33990555 PMCID: PMC8121942 DOI: 10.1038/s41467-021-22724-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/23/2021] [Indexed: 02/03/2023] Open
Abstract
TMEM16 lipid scramblases transport lipids and also operate as ion channels with highly variable ion selectivities and various physiological functions. However, their molecular mechanisms of ion conduction and selectivity remain largely unknown. Using computational electrophysiology simulations at atomistic resolution, we identified the main ion-conductive state of TMEM16 lipid scramblases, in which an ion permeation pathway is lined by lipid headgroups that directly interact with permeating ions in a voltage polarity-dependent manner. We found that lipid headgroups modulate the ion-permeability state and regulate ion selectivity to varying degrees in different scramblase isoforms, depending on the amino-acid composition of the pores. Our work has defined the structural basis of ion conduction and selectivity in TMEM16 lipid scramblases and uncovered the mechanisms responsible for the direct effects of membrane lipids on the conduction properties of ion channels.
Collapse
Affiliation(s)
- Andrei Y. Kostritskii
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, and JARA-HPC, Forschungszentrum Jülich, Jülich, Germany ,grid.1957.a0000 0001 0728 696XInstitute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany ,grid.1957.a0000 0001 0728 696XDepartment of Physics, RWTH Aachen University, Aachen, Germany
| | - Jan-Philipp Machtens
- grid.8385.60000 0001 2297 375XInstitute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, and JARA-HPC, Forschungszentrum Jülich, Jülich, Germany ,grid.1957.a0000 0001 0728 696XInstitute of Clinical Pharmacology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
21
|
Kolesnikov D, Perevoznikova A, Gusev K, Glushankova L, Kaznacheyeva E, Shalygin A. Electrophysiological Properties of Endogenous Single Ca 2+ Activated Cl - Channels Induced by Local Ca 2+ Entry in HEK293. Int J Mol Sci 2021; 22:4767. [PMID: 33946319 PMCID: PMC8124839 DOI: 10.3390/ijms22094767] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
Microdomains formed by proteins of endoplasmic reticulum and plasma membrane play a key role in store-operated Ca2+ entry (SOCE). Ca2+ release through inositol 1,4,5-trisphosphate receptor (IP3R) and subsequent Ca2+ store depletion activate STIM (stromal interaction molecules) proteins, sensors of intraluminal Ca2+, which, in turn, open the Orai channels in plasma membrane. Downstream to this process could be activated TRPC (transient receptor potential-canonical) calcium permeable channels. Using single channel patch-clamp technique we found that a local Ca2+ entry through TRPC1 channels activated endogenous Ca2+-activated chloride channels (CaCCs) with properties similar to Anoctamin6 (TMEM16F). Our data suggest that their outward rectification is based on the dependence from membrane potential of both the channel conductance and the channel activity: (1) The conductance of active CaCCs highly depends on the transmembrane potential (from 3 pS at negative potentials till 60 pS at positive potentials); (2) their activity (NPo) is enhanced with increasing Ca2+ concentration and/or transmembrane potential, conversely lowering of intracellular Ca2+ concentration reduced the open state dwell time; (3) CaCC amplitude is only slightly increased by intracellular Ca2+ concentration. Experiments with Ca2+ buffering by EGTA or BAPTA suggest close local arrangement of functional CaCCs and TRPC1 channels. It is supposed that Ca2+-activated chloride channels are involved in Ca2+ entry microdomains.
Collapse
Affiliation(s)
| | | | | | | | - Elena Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, 194064 St. Petersburg, Russia; (D.K.); (A.P.); (K.G.); (L.G.)
| | - Alexey Shalygin
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Avenue, 194064 St. Petersburg, Russia; (D.K.); (A.P.); (K.G.); (L.G.)
| |
Collapse
|
22
|
ANO7: Insights into topology, function, and potential applications as a biomarker and immunotherapy target. Tissue Cell 2021; 72:101546. [PMID: 33940566 DOI: 10.1016/j.tice.2021.101546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/21/2021] [Accepted: 04/11/2021] [Indexed: 01/01/2023]
Abstract
Anoctamin 7 (ANO7) is a member of the transmembrane protein TMEM16 family. It has a conservative topology similar to other members in this family, such as the typical eight-transmembrane domain, but it also has unique features. Although the ion channel role of ANO7 has been well accepted, evolutionary analyses and relevant studies suggest that ANO7 may be a multi-facet protein in function. Studies have shown that ANO7 may also function as a scramblase. ANO7 is highly expressed in prostate cancer as well as normal prostate tissues. A considerable amount of evidence has confirmed that ANO7 is associated with human physiology and pathology, particularly with the development of prostate cancer, which makes ANO7 a good candidate as a diagnostic and prognostic biomarker. In addition, ANO7 may be a potential target for prostate cancer immunotherapy. Antibody-based or T cell-mediated immunotherapies against prostate cancer by targeting ANO7 have been highly anticipated. ANO7 may also correlate with several other types of cancers or diseases, where further studies are warranted.
Collapse
|
23
|
Connolly A, Panes R, Tual M, Lafortune R, Bellemare-Pelletier A, Gagnon E. TMEM16F mediates bystander TCR-CD3 membrane dissociation at the immunological synapse and potentiates T cell activation. Sci Signal 2021; 14:eabb5146. [PMID: 33758060 DOI: 10.1126/scisignal.abb5146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Electrostatic interactions regulate many aspects of T cell receptor (TCR) activity, including enabling the dynamic binding of the TCR-associated CD3ε and CD3ζ chains to anionic lipids in the plasma membrane to prevent spontaneous phosphorylation. Substantial changes in the electrostatic potential of the plasma membrane occur at the immunological synapse, the interface between a T cell and an antigen-presenting cell. Here, we investigated how the electrostatic interactions that promote dynamic membrane binding of the TCR-CD3 cytoplasmic domains are modulated during signaling and affect T cell activation. We found that Ca2+-dependent activation of the phosphatidylserine scramblase TMEM16F, which was previously implicated in T cell activation, reduced the electrostatic potential of the plasma membrane during immunological synapse formation by locally redistributing phosphatidylserine. This, in turn, increased the dissociation of bystander TCR-CD3 cytoplasmic domains from the plasma membrane and enhanced TCR-dependent signaling and consequently T cell activation. This study establishes the molecular basis for the role of TMEM16F in bystander TCR-induced signal amplification and identifies enhancement of TMEM16F function as a potential therapeutic strategy for promoting T cell activation.
Collapse
Affiliation(s)
- Audrey Connolly
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Rébecca Panes
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Margaux Tual
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Raphaël Lafortune
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| | - Angélique Bellemare-Pelletier
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada
| | - Etienne Gagnon
- Institut de Recherche en Immunologie et Cancérologie, 2950 Chemin de la Polytechnique, Montréal, Québec H3T1J4, Canada.
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, 2900 Édouard-Montpetit, Montréal, Québec H3T1J4, Canada
| |
Collapse
|
24
|
Roh JW, Hwang GE, Kim WK, Nam JH. Ca 2+ Sensitivity of Anoctamin 6/TMEM16F Is Regulated by the Putative Ca 2+-Binding Reservoir at the N-Terminal Domain. Mol Cells 2021; 44:88-100. [PMID: 33658434 PMCID: PMC7941003 DOI: 10.14348/molcells.2021.2203] [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: 10/14/2020] [Revised: 01/05/2021] [Accepted: 01/26/2021] [Indexed: 11/27/2022] Open
Abstract
Anoctamin 6/TMEM16F (ANO6) is a dual-function protein with Ca2+-activated ion channel and Ca2+-activated phospholipid scramblase activities, requiring a high intracellular Ca2+ concentration (e.g., half-maximal effective Ca2+ concentration [EC50] of [Ca2+]i > 10 μM), and strong and sustained depolarization above 0 mV. Structural comparison with Anoctamin 1/TMEM16A (ANO1), a canonical Ca2+- activated chloride channel exhibiting higher Ca2+ sensitivity (EC50 of 1 μM) than ANO6, suggested that a homologous Ca2+-transferring site in the N-terminal domain (Nt) might be responsible for the differential Ca2+ sensitivity and kinetics of activation between ANO6 and ANO1. To elucidate the role of the putative Ca2+-transferring reservoir in the Nt (Nt-CaRes), we constructed an ANO6-1-6 chimera in which Nt-CaRes was replaced with the corresponding domain of ANO1. ANO6- 1-6 showed higher sensitivity to Ca2+ than ANO6. However, neither the speed of activation nor the voltage-dependence differed between ANO6 and ANO6-1-6. Molecular dynamics simulation revealed a reduced Ca2+ interaction with Nt- CaRes in ANO6 than ANO6-1-6. Moreover, mutations on potentially Ca2+-interacting acidic amino acids in ANO6 Nt- CaRes resulted in reduced Ca2+ sensitivity, implying direct interactions of Ca2+ with these residues. Based on these results, we cautiously suggest that the net charge of Nt- CaRes is responsible for the difference in Ca2+ sensitivity between ANO1 and ANO6.
Collapse
Affiliation(s)
- Jae Won Roh
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
| | - Ga Eun Hwang
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
| | - Woo Kyung Kim
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Korea
| |
Collapse
|
25
|
Divalent Cation Modulation of Ion Permeation in TMEM16 Proteins. Int J Mol Sci 2021; 22:ijms22042209. [PMID: 33672260 PMCID: PMC7926781 DOI: 10.3390/ijms22042209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 01/01/2023] Open
Abstract
Intracellular divalent cations control the molecular function of transmembrane protein 16 (TMEM16) family members. Both anion channels (such as TMEM16A) and phospholipid scramblases (such as TMEM16F) in this family are activated by intracellular Ca2+ in the low µM range. In addition, intracellular Ca2+ or Co2+ at mM concentrations have been shown to further potentiate the saturated Ca2+-activated current of TMEM16A. In this study, we found that all alkaline earth divalent cations in mM concentrations can generate similar potentiation effects in TMEM16A when applied intracellularly, and that manipulations thought to deplete membrane phospholipids weaken the effect. In comparison, mM concentrations of divalent cations minimally potentiate the current of TMEM16F but significantly change its cation/anion selectivity. We suggest that divalent cations may increase local concentrations of permeant ions via a change in pore electrostatic potential, possibly acting through phospholipid head groups in or near the pore. Monovalent cations appear to exert a similar effect, although with a much lower affinity. Our findings resolve controversies regarding the ion selectivity of TMEM16 proteins. The physiological role of this mechanism, however, remains elusive because of the nearly constant high cation concentrations in cytosols.
Collapse
|
26
|
Liang P, Yang H. Molecular underpinning of intracellular pH regulation on TMEM16F. J Gen Physiol 2021; 153:e202012704. [PMID: 33346788 PMCID: PMC7754671 DOI: 10.1085/jgp.202012704] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/29/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
TMEM16F, a dual-function phospholipid scramblase and ion channel, is important in blood coagulation, skeleton development, HIV infection, and cell fusion. Despite advances in understanding its structure and activation mechanism, how TMEM16F is regulated by intracellular factors remains largely elusive. Here we report that TMEM16F lipid scrambling and ion channel activities are strongly influenced by intracellular pH (pHi). We found that low pHi attenuates, whereas high pHi potentiates, TMEM16F channel and scramblase activation under physiological concentrations of intracellular Ca2+ ([Ca2+]i). We further demonstrate that TMEM16F pHi sensitivity depends on [Ca2+]i and exhibits a bell-shaped relationship with [Ca2+]i: TMEM16F channel activation becomes increasingly pHi sensitive from resting [Ca2+]i to micromolar [Ca2+]i, but when [Ca2+]i increases beyond 15 µM, pHi sensitivity gradually diminishes. The mutation of a Ca2+-binding residue that markedly reduces TMEM16F Ca2+ sensitivity (E667Q) maintains the bell-shaped relationship between pHi sensitivity and Ca2+ but causes a dramatic shift of the peak [Ca2+]i from 15 µM to 3 mM. Our biophysical characterizations thus pinpoint that the pHi regulatory effects on TMEM16F stem from the competition between Ca2+ and protons for the primary Ca2+-binding residues in the pore. Within the physiological [Ca2+]i range, the protonation state of the primary Ca2+-binding sites influences Ca2+ binding and regulates TMEM16F activation. Our findings thus uncover a regulatory mechanism of TMEM16F by pHi and shine light on our understanding of the pathophysiological roles of TMEM16F in diseases with dysregulated pHi, including cancer.
Collapse
Affiliation(s)
- Pengfei Liang
- Department of Biochemistry, Duke University Medical Center, Durham, NC
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC
- Department of Neurobiology, Duke University Medical Center, Durham, NC
| |
Collapse
|
27
|
Le SC, Yang H. Structure-Function of TMEM16 Ion Channels and Lipid Scramblases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:87-109. [PMID: 35138612 PMCID: PMC11020148 DOI: 10.1007/978-981-16-4254-8_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The TMEM16 protein family comprises two novel classes of structurally conserved but functionally distinct membrane transporters that function as Ca2+-dependent Cl- channels (CaCCs) or dual functional Ca2+-dependent ion channels and phospholipid scramblases. Extensive functional and structural studies have advanced our understanding of TMEM16 molecular mechanisms and physiological functions. TMEM16A and TMEM16B CaCCs control transepithelial fluid transport, smooth muscle contraction, and neuronal excitability, whereas TMEM16 phospholipid scramblases mediate the flip-flop of phospholipids across the membrane to allow phosphatidylserine externalization, which is essential in a plethora of important processes such as blood coagulation, bone development, and viral and cell fusion. In this chapter, we summarize the major methods in studying TMEM16 ion channels and scramblases and then focus on the current mechanistic understanding of TMEM16 Ca2+- and voltage-dependent channel gating as well as their ion and phospholipid permeation.
Collapse
Affiliation(s)
- Son C Le
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
28
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
29
|
Feng S, Dang S, Han TW, Ye W, Jin P, Cheng T, Li J, Jan YN, Jan LY, Cheng Y. Cryo-EM Studies of TMEM16F Calcium-Activated Ion Channel Suggest Features Important for Lipid Scrambling. Cell Rep 2020; 28:567-579.e4. [PMID: 31291589 PMCID: PMC6684876 DOI: 10.1016/j.celrep.2019.06.023] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/03/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022] Open
Abstract
As a Ca2+-activated lipid scramblase and ion channel that mediates Ca2+ influx, TMEM16F relies on both functions to facilitate extracellular vesicle generation, blood coagulation, and bone formation. How a bona fide ion channel scrambles lipids remains elusive. Our structural analyses revealed the coexistence of an intact channel pore and PIP2-dependent protein conformation changes leading to membrane distortion. Correlated to the extent of membrane distortion, many tightly bound lipids are slanted. Structure-based mutagenesis studies further reveal that neutralization of some lipid-binding residues or those near membrane distortion specifically alters the onset of lipid scrambling, but not Ca2+ influx, thus identifying features outside of channel pore that are important for lipid scrambling. Together, our studies demonstrate that membrane distortion does not require open hydrophilic grooves facing the membrane interior and provide further evidence to suggest separate pathways for lipid scrambling and ion permeation. TMEM16F is a calcium-activated ion channel and lipid scramblase linked to the bleeding disorder Scott syndrome. Feng et al. examine cryo-EM structures of TMEM16F with or without Ca2+ ions and PIP2 nanodisc supplementation and identify structural features for lipid binding and membrane distortion critical for lipid scrambling activity.
Collapse
Affiliation(s)
- Shengjie Feng
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shangyu Dang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tina Wei Han
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Wenlei Ye
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Peng Jin
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tong Cheng
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Junrui Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh Nung Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lily Yeh Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
30
|
Öhlinger T, Müllner EW, Fritz M, Sauer T, Werning M, Baron DM, Salzer U. Lysophosphatidic acid-induced pro-thrombotic phosphatidylserine exposure and ionophore-induced microvesiculation is mediated by the scramblase TMEM16F in erythrocytes. Blood Cells Mol Dis 2020; 83:102426. [PMID: 32222693 DOI: 10.1016/j.bcmd.2020.102426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 12/31/2022]
Abstract
Recent studies indicate that erythrocytes actively modulate blood clotting and thrombus formation. The lipid mediator lysophosphatidic acid (LPA) is produced by activated platelets, and triggers a signaling process in erythrocytes. This results in cellular calcium uptake and exposure of phosphatidylserine (PS) at the cell surface, thereby generating activated membrane binding sites for factors of the clotting cascade. Moreover, erythrocytes of patients with a bleeding disorder and mutations in the scramblase TMEM16F show impaired PS exposure and microvesiculation upon treatment with calcium ionophore. We report that TMEM16F inhibitors tannic acid (TA) and epigallocatechin-3-gallate (EGCG) inhibit LPA-induced PS exposure and calcium uptake at low micromolar concentrations; fluoxetine, an antidepressant and a known activator of TMEM16F, enhances these processes. These effectors likewise modulate erythrocyte PS exposure and microvesicle shedding induced by calcium ionophore treatment. Further, LPA-treated erythrocytes triggered thrombin generation in platelet-free plasma which was partially impaired in the presence of TA and EGCG. Thus, this study suggests that LPA activates the scramblase TMEM16F in erythrocytes, thereby possibly mediating a pro-thrombotic function in these cells. EGCG as well as fluoxetine, substances with potentially high plasma concentrations due to alimentation or medical treatment, should be considered as potential effectors of systemic hemostatic regulation.
Collapse
Affiliation(s)
- Thomas Öhlinger
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria; Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Ernst W Müllner
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Magdalena Fritz
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Thomas Sauer
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Maike Werning
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - David M Baron
- Department of Anaesthesia, Intensive Care Medicine and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Ulrich Salzer
- Center for Medical Biochemistry, Max Perutz Laboratories, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
31
|
Munemasa T, Gao X, Melvin JE, Mukaibo T. Ano6 disruption impairs acinar cell regulatory volume decrease and protein secretion in murine submandibular salivary glands. J Cell Physiol 2020; 235:8533-8545. [PMID: 32329061 DOI: 10.1002/jcp.29697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/24/2020] [Accepted: 03/29/2020] [Indexed: 11/09/2022]
Abstract
The widely expressed Anoctamin 6 (Ano6) supports different Ca2+ -dependent functions, but little is known about its role in salivary glands. Mouse submandibular gland (SMG) acinar cells exhibited a robust regulatory volume decrease (RVD) following cell swelling that was reduced approximately 70% in Ano6-/- mice. Ca2+ -free conditions nearly eliminated the RVD response suggesting that Ano6 is an obligatory component of the cell volume-activated, Ca2+ -dependent RVD pathway in salivary gland acinar cells. Ex vivo agonist-stimulated secretion of water and ions was unaffected by Ano6 disruption under both isotonic and hypotonic conditions suggesting that Ano6 does not play a major role in fluid and electrolyte secretion. In contrast, the total amount of β-adrenergic-dependent protein secretion by the SMG was significantly reduced in Ano6-/- mice. Closer inspection of these latter results revealed that protein secretion was affected only in the female SMG by Ano6 disruption. These results indicate that Ano6 modulates the RVD response and protein secretion by salivary gland acinar cells.
Collapse
Affiliation(s)
- Takashi Munemasa
- Secretory Mechanisms and Dysfunctions Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland.,Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Xin Gao
- Secretory Mechanisms and Dysfunctions Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - James E Melvin
- Secretory Mechanisms and Dysfunctions Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Taro Mukaibo
- Secretory Mechanisms and Dysfunctions Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland.,Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
32
|
Wilke BU, Kummer KK, Leitner MG, Kress M. Chloride - The Underrated Ion in Nociceptors. Front Neurosci 2020; 14:287. [PMID: 32322187 PMCID: PMC7158864 DOI: 10.3389/fnins.2020.00287] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/12/2020] [Indexed: 01/06/2023] Open
Abstract
In contrast to pain processing neurons in the spinal cord, where the importance of chloride conductances is already well established, chloride homeostasis in primary afferent neurons has received less attention. Sensory neurons maintain high intracellular chloride concentrations through balanced activity of Na+-K+-2Cl- cotransporter 1 (NKCC1) and K+-Cl- cotransporter 2 (KCC2). Whereas in other cell types activation of chloride conductances causes hyperpolarization, activation of the same conductances in primary afferent neurons may lead to inhibitory or excitatory depolarization depending on the actual chloride reversal potential and the total amount of chloride efflux during channel or transporter activation. Dorsal root ganglion (DRG) neurons express a multitude of chloride channel types belonging to different channel families, such as ligand-gated, ionotropic γ-aminobutyric acid (GABA) or glycine receptors, Ca2+-activated chloride channels of the anoctamin/TMEM16, bestrophin or tweety-homolog family, CLC chloride channels and transporters, cystic fibrosis transmembrane conductance regulator (CFTR) as well as volume-regulated anion channels (VRACs). Specific chloride conductances are involved in signal transduction and amplification at the peripheral nerve terminal, contribute to excitability and action potential generation of sensory neurons, or crucially shape synaptic transmission in the spinal dorsal horn. In addition, chloride channels can be modified by a plethora of inflammatory mediators affecting them directly, via protein-protein interaction, or through signaling cascades. Since chloride channels as well as mediators that modulate chloride fluxes are regulated in pain disorders and contribute to nociceptor excitation and sensitization it is timely and important to emphasize their critical role in nociceptive primary afferents in this review.
Collapse
Affiliation(s)
| | | | | | - Michaela Kress
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
33
|
Centeio R, Cabrita I, Benedetto R, Talbi K, Ousingsawat J, Schreiber R, Sullivan JK, Kunzelmann K. Pharmacological Inhibition and Activation of the Ca 2+ Activated Cl - Channel TMEM16A. Int J Mol Sci 2020; 21:ijms21072557. [PMID: 32272686 PMCID: PMC7177308 DOI: 10.3390/ijms21072557] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/26/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
TMEM16A is a Ca2+ activated Cl− channel with important functions in airways, intestine, and other epithelial organs. Activation of TMEM16A is proposed as a therapy in cystic fibrosis (CF) to reinstall airway Cl− secretion and to enhance airway surface liquid (ASL). This CFTR-agnostic approach is thought to improve mucociliary clearance and lung function in CF. This could indeed improve ASL, however, mucus release and airway contraction may also be induced by activators of TMEM16A, particularly in inflamed airways of patients with asthma, COPD, or CF. Currently, both activators and inhibitors of TMEM16A are developed and examined in different types of tissues. Here we compare activation and inhibition of endogenous and overexpressed TMEM16A and analyze potential off-target effects. The three well-known blockers benzbromarone, niclosamide, and Ani9 inhibited both TMEM16A and ATP-induced Ca2+ increase by variable degrees, depending on the cell type. Niclosamide, while blocking Ca2+ activated TMEM16A, also induced a subtle but significant Ca2+ store release and inhibited store-operated Ca2+ influx. Niclosamide, benzbromarone and Ani9 also affected TMEM16F whole cell currents, indicating limited specificity for these inhibitors. The compounds Eact, cinnamaldehyde, and melittin, as well as the phosphatidylinositol diC8-PIP2 are the reported activators of TMEM16A. However, the compounds were unable to activate endogenous TMEM16A in HT29 colonic epithelial cells. In contrast, TMEM16A overexpressed in HEK293 cells was potently stimulated by these activators. We speculate that overexpressed TMEM16A might have a better accessibility to intracellular Ca2+, which causes spontaneous activity even at basal intracellular Ca2+ concentrations. Small molecules may therefore potentiate pre-stimulated TMEM16A currents, but may otherwise fail to activate silent endogenous TMEM16A.
Collapse
Affiliation(s)
- Raquel Centeio
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Inês Cabrita
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Roberta Benedetto
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Khaoula Talbi
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | | | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
- * Correspondence: ; Tel.: +49-(0)941-943-4302; Fax: +49-(0)941-943-4315
| |
Collapse
|
34
|
Ye W, Han TW, He M, Jan YN, Jan LY. Dynamic change of electrostatic field in TMEM16F permeation pathway shifts its ion selectivity. eLife 2019; 8:e45187. [PMID: 31318330 PMCID: PMC6690719 DOI: 10.7554/elife.45187] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/17/2019] [Indexed: 12/11/2022] Open
Abstract
TMEM16F is activated by elevated intracellular Ca2+, and functions as a small-conductance ion channel and as a phospholipid scramblase. In contrast to its paralogs, the TMEM16A/B calcium-activated chloride channels, mouse TMEM16F has been reported as a cation-, anion-, or non-selective ion channel, without a definite conclusion. Starting with the Q559K mutant that shows no current rundown and less outward rectification in excised patch, we found that the channel shifted its ion selectivity in response to the change of intracellular Ca2+ concentration, with an increased permeability ratio of Cl- to Na+ (PCl-/PNa+) at a higher Ca2+ level. The gradual shift of relative ion permeability did not correlate with the channel activation state. Instead, it was indicative of an alteration of electrostatic field in the permeation pathway. The dynamic change of ion selectivity suggests a charge-screening mechanism for TMEM16F ion conduction, and it provides hints to further studies of TMEM16F physiological functions.
Collapse
Affiliation(s)
- Wenlei Ye
- Department of PhysiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Tina W Han
- Department of PhysiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Mu He
- Department of PhysiologyUniversity of California, San FranciscoSan FranciscoUnited States
| | - Yuh Nung Jan
- Department of PhysiologyUniversity of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteSan FranciscoUnited States
- Department of Biochemistry and BiophysicsUniversity of California, San FranciscoSan FranciscoUnited States
| | - Lily Yeh Jan
- Department of PhysiologyUniversity of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteSan FranciscoUnited States
- Department of Biochemistry and BiophysicsUniversity of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
35
|
Ousingsawat J, Schreiber R, Kunzelmann K. TMEM16F/Anoctamin 6 in Ferroptotic Cell Death. Cancers (Basel) 2019; 11:E625. [PMID: 31060306 PMCID: PMC6562394 DOI: 10.3390/cancers11050625] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 12/21/2022] Open
Abstract
Ca2+ activated Cl- channels (TMEM16A; ANO1) support cell proliferation and cancer growth. Expression of TMEM16A is strongly enhanced in different types of malignomas. In contrast, TMEM16F (ANO6) operates as a Ca2+ activated chloride/nonselective ion channel and scrambles membrane phospholipids to expose phosphatidylserine at the cell surface. Both phospholipid scrambling and cell swelling induced through activation of nonselective ion currents appear to destabilize the plasma membrane thereby causing cell death. There is growing evidence that activation of TMEM16F contributes to various forms of regulated cell death. In the present study, we demonstrate that ferroptotic cell death, occurring during peroxidation of plasma membrane phospholipids activates TMEM16F. Ferroptosis was induced by erastin, an inhibitor of the cystine-glutamate antiporter and RSL3, an inhibitor of glutathione peroxidase 4 (GPX4). Cell death was largely reduced in the intestinal epithelium, and in peritoneal macrophages isolated from mice with tissue-specific knockout of TMEM16F. We show that TMEM16F is activated during erastin and RSL3-induced ferroptosis. In contrast, inhibition of ferroptosis by ferrostatin-1 and by inhibitors of TMEM16F block TMEM16F currents and inhibit cell death. We conclude that activation of TMEM16F is a crucial component during ferroptotic cell death, a finding that may be useful to induce cell death in cancer cells.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| |
Collapse
|
36
|
Lin H, Jun I, Woo JH, Lee MG, Kim SJ, Nam JH. Temperature-dependent increase in the calcium sensitivity and acceleration of activation of ANO6 chloride channel variants. Sci Rep 2019; 9:6706. [PMID: 31040335 PMCID: PMC6491614 DOI: 10.1038/s41598-019-43162-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/12/2019] [Indexed: 02/08/2023] Open
Abstract
Anoctamin-6 (ANO6) belongs to a family of calcium (Ca2+)-activated chloride channels (CaCCs), with three splicing variants (V1, V2, and V5) showing plasma membrane expression. Unlike other CaCCs, ANO6 requires a non-physiological intracellular free calcium concentration ([Ca2+]i > 1 μM) and several minutes for full activation under a whole-cell patch clamp. Therefore, its physiological role as an ion channel is uncertain and it is more commonly considered a Ca2+-dependent phospholipid scramblase. Here, we demonstrate that physiological temperature (37 °C) increases ANO6 Ca2+ sensitivity under a whole-cell patch clamp; V1 was activated by 1 μM [Ca2+]i, whereas V2 and V5 were activated by 300 nM [Ca2+]i. Increasing the temperature to 42 °C led to activation of all ANO6 variants by 100 nM [Ca2+]i. The delay time for activation of the three variants was significantly shortened at 37 °C. Notably, the temperature-dependent Ca2+-sensitisation of ANO6 became insignificant under inside-out patch clamp, suggesting critical roles of unknown cytosolic factors. Unlike channel activity, 27 °C but not 37 °C (physiological temperature) induced the scramblase activity of ANO6 at submicromolar [Ca2+]i (300 nM), irrespective of variant type. Our results reveal a physiological ion conducting property of ANO6 at 37 °C and suggest that ANO6 channel function acts separately from its scramblase activity.
Collapse
Affiliation(s)
- Haiyue Lin
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ikhyun Jun
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Joo Han Woo
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea.
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
37
|
Taylor KA, Mahaut-Smith MP. A major interspecies difference in the ionic selectivity of megakaryocyte Ca 2+-activated channels sensitive to the TMEM16F inhibitor CaCCinh-A01. Platelets 2019; 30:962-966. [PMID: 31008669 PMCID: PMC6816474 DOI: 10.1080/09537104.2019.1595560] [Citation(s) in RCA: 5] [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: 11/16/2018] [Revised: 02/12/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
TMEM16F is a surface membrane protein critical for platelet procoagulant activity, which exhibits both phospholipid scramblase and ion channel activities following sustained elevation of cytosolic Ca2+. The extent to which the ionic permeability of TMEM16F is important for platelet scramblase responses remains controversial. To date, only one study has reported the electrophysiological properties of TMEM16F in cells of platelet/megakaryocyte lineage, which observed cation-selectivity within excised patch recordings from murine marrow-derived megakaryocytes. This contrasts with reports using whole-cell recordings that describe this channel as displaying either selectivity for anions or being relatively non-selective amongst the major physiological monovalent ions. We have studied TMEM16F expression and channel activity in primary rat and mouse megakaryocytes and the human erythroleukemic (HEL) cell line that exhibits megakaryocytic surface markers. Immunocytochemical analysis was consistent with surface TMEM16F expression in cells from all three species. Whole-cell recordings in the absence of K+-selective currents revealed an outwardly rectifying conductance activated by a high intracellular Ca2+ concentration in all three species. These currents appeared after 5-6 minutes and were blocked by CaCCinh-A01, properties typical of TMEM16F. Ion substitution experiments showed that the underlying conductance was predominantly Cl--permeable in rat megakaryocytes and HEL cells, yet non-selective between monovalent anions and cations in mouse megakaryocytes. In conclusion, the present study further highlights the difference in ionic selectivity of TMEM16F in platelet lineage cells of the mouse compared to other mammalian species. This provides additional support for the ionic "leak" hypothesis that the scramblase activity of TMEM16F does not rely upon its ability to conduct ions of a specific type.
Collapse
Affiliation(s)
- Kirk A. Taylor
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- National Heart and Lung Institute, Cardio-respiratory Section, Imperial College London, London, UK
| | | |
Collapse
|
38
|
Kunzelmann K, Ousingsawat J, Benedetto R, Cabrita I, Schreiber R. Contribution of Anoctamins to Cell Survival and Cell Death. Cancers (Basel) 2019; 11:E382. [PMID: 30893776 PMCID: PMC6468699 DOI: 10.3390/cancers11030382] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 02/07/2023] Open
Abstract
Before anoctamins (TMEM16 proteins) were identified as a family of Ca2+-activated chloride channels and phospholipid scramblases, the founding member anoctamin 1 (ANO1, TMEM16A) was known as DOG1, a marker protein for gastrointestinal stromal tumors (GIST). Meanwhile, ANO1 has been examined in more detail, and the role of ANO1 in cell proliferation and the development of different types of malignomas is now well established. While ANO5, ANO7, and ANO9 may also be relevant for growth of cancers, evidence has been provided for a role of ANO6 (TMEM16F) in regulated cell death. The cellular mechanisms by which anoctamins control cell proliferation and cell death, respectively, are just emerging; however, the pronounced effects of anoctamins on intracellular Ca2+ levels are likely to play a significant role. Recent results suggest that some anoctamins control membrane exocytosis by setting Ca2+i levels near the plasma membrane, and/or by controlling the intracellular Cl- concentration. Exocytosis and increased membrane trafficking induced by ANO1 and ANO6 may enhance membrane expression of other chloride channels, such as CFTR and volume activated chloride channels (VRAC). Notably, ANO6-induced phospholipid scrambling with exposure of phosphatidylserine is pivotal for the sheddase function of disintegrin and metalloproteinase (ADAM). This may support cell death and tumorigenic activity of IL-6 by inducing IL-6 trans-signaling. The reported anticancer effects of the anthelminthic drug niclosamide are probably related to the potent inhibitory effect on ANO1, apart from inducing cell cycle arrest through the Let-7d/CDC34 axis. On the contrary, pronounced activation of ANO6 due to a large increase in intracellular calcium, activation of phospholipase A2 or lipid peroxidation, can lead to ferroptotic death of cancer cells. It therefore appears reasonable to search for both inhibitors and potent activators of TMEM16 in order to interfere with cancer growth and metastasis.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Ines Cabrita
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| |
Collapse
|
39
|
Kunzelmann K, Ousingsawat J, Cabrita I, Doušová T, Bähr A, Janda M, Schreiber R, Benedetto R. TMEM16A in Cystic Fibrosis: Activating or Inhibiting? Front Pharmacol 2019; 10:3. [PMID: 30761000 PMCID: PMC6362895 DOI: 10.3389/fphar.2019.00003] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
The inflammatory airway disease cystic fibrosis (CF) is characterized by airway obstruction due to mucus hypersecretion, airway plugging, and bronchoconstriction. The cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel is dysfunctional in CF, leading to defects in epithelial transport. Although CF pathogenesis is still disputed, activation of alternative Cl- channels is assumed to improve lung function in CF. Two suitable non-CFTR Cl- channels are present in the airway epithelium, the Ca2+ activated channel TMEM16A and SLC26A9. Activation of these channels is thought to be feasible to improve hydration of the airway mucus and to increase mucociliary clearance. Interestingly, both channels are upregulated during inflammatory lung disease. They are assumed to support fluid secretion, necessary to hydrate excess mucus and to maintain mucus clearance. During inflammation, however, TMEM16A is upregulated particularly in mucus producing cells, with only little expression in ciliated cells. Recently it was shown that knockout of TMEM16A in ciliated cells strongly compromises Cl- conductance and attenuated mucus secretion, but does not lead to a CF-like lung disease and airway plugging. Along this line, activation of TMEM16A by denufosol, a stable purinergic ligand, failed to demonstrate any benefit to CF patients in earlier studies. It rather induced adverse effects such as cough. A number of studies suggest that TMEM16A is essential for mucus secretion and possibly also for mucus production. Evidence is now provided for a crucial role of TMEM16A in fusion of mucus-filled granules with the apical plasma membrane and cellular exocytosis. This is probably due to local Ca2+ signals facilitated by TMEM16A. Taken together, TMEM16A supports fluid secretion by ciliated airway epithelial cells, but also maintains excessive mucus secretion during inflammatory airway disease. Because TMEM16A also supports airway smooth muscle contraction, inhibition rather than activation of TMEM16A might be the appropriate treatment for CF lung disease, asthma and COPD. As a number of FDA-approved and well-tolerated drugs have been shown to inhibit TMEM16A, evaluation in clinical trials appears timely.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | | | - Inês Cabrita
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Tereza Doušová
- Department of Pediatrics, Second Faculty of Medicine, University Hospital Motol, Charles University in Prague, Prague, Czechia
| | - Andrea Bähr
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Munich, Germany
- Innere Medizin I, Klinikum Rechts der Isar der TU München, München, Germany
| | - Melanie Janda
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| |
Collapse
|
40
|
Nguyen DM, Chen LS, Yu WP, Chen TY. Comparison of ion transport determinants between a TMEM16 chloride channel and phospholipid scramblase. J Gen Physiol 2019; 151:518-531. [PMID: 30670476 PMCID: PMC6445582 DOI: 10.1085/jgp.201812270] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/02/2019] [Indexed: 01/03/2023] Open
Abstract
The I-V relation of the TMEM16A channel is linear, whereas that of the TMEM16F scramblase is outwardly rectifying. Nguyen et al. show that rectification of TMEM16A is regulated by the charge of residue 584 but that rectification of TMEM16F is affected by aromatic residues at the equivalent position. Two TMEM16 family members, TMEM16A and TMEM16F, have different ion transport properties. Upon activation by intracellular Ca2+, TMEM16A—a Ca2+-activated Cl− channel—is more selective for anions than cations, whereas TMEM16F—a phospholipid scramblase—appears to transport both cations and anions. Under saturating Ca2+ conditions, the current–voltage (I-V) relationships of these two proteins also differ; the I-V curve of TMEM16A is linear, while that of TMEM16F is outwardly rectifying. We previously found that mutating a positively charged lysine residue (K584) in the ion transport pathway to glutamine converted the linear I-V curve of TMEM16A to an outwardly rectifying curve. Interestingly, the corresponding residue in the outwardly rectifying TMEM16F is also a glutamine (Q559). Here, we examine the ion transport functions of TMEM16 molecules and compare the roles of K584 of TMEM16A and Q559 of TMEM16F in controlling the rectification of their respective I-V curves. We find that rectification of TMEM16A is regulated electrostatically by the side-chain charge on the residue at position 584, whereas the charge on residue 559 in TMEM16F has little effect. Unexpectedly, mutation of Q559 to aromatic amino acid residues significantly alters outward rectification in TMEM16F. These same mutants show reduced Ca2+-induced current rundown (or desensitization) compared with wild-type TMEM16F. A mutant that removes the rundown of TMEM16F could facilitate the study of ion transport mechanisms in this phospholipid scramblase in the same way that a CLC-0 mutant in which inactivation (or closure of the slow gate) is suppressed was used in our previous studies.
Collapse
Affiliation(s)
- Dung M Nguyen
- Graduate Group of Pharmacology and Toxicology, University of California, Davis, Davis, CA
| | - Louisa S Chen
- Center for Neuroscience, University of California, Davis, Davis, CA
| | - Wei-Ping Yu
- Center for Neuroscience, University of California, Davis, Davis, CA
| | - Tsung-Yu Chen
- Center for Neuroscience, University of California, Davis, Davis, CA .,Department of Neurology, University of California, Davis, Davis, CA
| |
Collapse
|
41
|
Okada Y, Okada T, Sato-Numata K, Islam MR, Ando-Akatsuka Y, Numata T, Kubo M, Shimizu T, Kurbannazarova RS, Marunaka Y, Sabirov RZ. Cell Volume-Activated and Volume-Correlated Anion Channels in Mammalian Cells: Their Biophysical, Molecular, and Pharmacological Properties. Pharmacol Rev 2019; 71:49-88. [PMID: 30573636 DOI: 10.1124/pr.118.015917] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
There are a number of mammalian anion channel types associated with cell volume changes. These channel types are classified into two groups: volume-activated anion channels (VAACs) and volume-correlated anion channels (VCACs). VAACs can be directly activated by cell swelling and include the volume-sensitive outwardly rectifying anion channel (VSOR), which is also called the volume-regulated anion channel; the maxi-anion channel (MAC or Maxi-Cl); and the voltage-gated anion channel, chloride channel (ClC)-2. VCACs can be facultatively implicated in, although not directly activated by, cell volume changes and include the cAMP-activated cystic fibrosis transmembrane conductance regulator (CFTR) anion channel, the Ca2+-activated Cl- channel (CaCC), and the acid-sensitive (or acid-stimulated) outwardly rectifying anion channel. This article describes the phenotypical properties and activation mechanisms of both groups of anion channels, including accumulating pieces of information on the basis of recent molecular understanding. To that end, this review also highlights the molecular identities of both anion channel groups; in addition to the molecular identities of ClC-2 and CFTR, those of CaCC, VSOR, and Maxi-Cl were recently identified by applying genome-wide approaches. In the last section of this review, the most up-to-date information on the pharmacological properties of both anion channel groups, especially their half-maximal inhibitory concentrations (IC50 values) and voltage-dependent blocking, is summarized particularly from the standpoint of pharmacological distinctions among them. Future physiologic and pharmacological studies are definitely warranted for therapeutic targeting of dysfunction of VAACs and VCACs.
Collapse
Affiliation(s)
- Yasunobu Okada
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Toshiaki Okada
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Kaori Sato-Numata
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Md Rafiqul Islam
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Yuhko Ando-Akatsuka
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Tomohiro Numata
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Machiko Kubo
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Takahiro Shimizu
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Ranohon S Kurbannazarova
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Yoshinori Marunaka
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| | - Ravshan Z Sabirov
- Departments of Physiology and Systems Bioscience (Y.O.) and Molecular Cell Physiology (Y.M.), Kyoto Prefectural University of Medicine, Kyoto, Japan; Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Japan (Y.O., T.O., M.R.I., M.K., R.Z.S.); Department of Physiology, School of Medicine, Fukuoka University, Fukuoka, Japan (K.S.-N., T.N.); Department of Cell Physiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan (Y.A.-A.); Department of Pharmaceutical Physiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (T.S.); Laboratory of Molecular Physiology, Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, Tashkent, Uzbekistan (R.S.K., R.Z.S.); and Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan (Y.M.)
| |
Collapse
|
42
|
Lin H, Roh J, Woo JH, Kim SJ, Nam JH. TMEM16F/ANO6, a Ca 2+-activated anion channel, is negatively regulated by the actin cytoskeleton and intracellular MgATP. Biochem Biophys Res Commun 2018; 503:2348-2354. [PMID: 29964013 DOI: 10.1016/j.bbrc.2018.06.160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/28/2018] [Indexed: 01/08/2023]
Abstract
Anoctamin 6 (ANO6/TMEM16F) is a recently identified membrane protein that has both phospholipid scramblase activity and anion channel function activated by relatively high [Ca2+]i. In addition to the low sensitivity to Ca2+, the activation of ANO6 Cl- conductance is very slow (>3-5 min to reach peak level at 10 μM [Ca2+]i), with subsequent inactivation. In a whole-cell patch clamp recording of ANO6 current (IANO6,w-c), disruption of the actin cytoskeleton with cytochalasin-D (cytoD) significantly accelerated the activation kinetics, while actin filament-stabilizing agents (phalloidin and jasplakinolide) commonly inhibited IANO6,w-c. Inside-out patch clamp recording of ANO6 (IANO6,i-o) showed immediate activation by raising [Ca2+]i. We also found that intracellular ATP (3 mM MgATP in pipette solution) decelerated the activation of IANO6,w-c, and also prevented the inactivation of IANO6,w-c. However, the addition of cytoD still accelerated both activation and inactivation of IANO6,w-c. We conclude that the actin cytoskeleton and intracellular ATP play major roles in the Ca2+-dependent activation and inactivation of IANO6,w-c, respectively.
Collapse
Affiliation(s)
- Haiyue Lin
- Department of Physiology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jaewon Roh
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea
| | - Joo Han Woo
- Department of Physiology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
43
|
Molecular Identities and ATP Release Activities of Two Types of Volume-Regulatory Anion Channels, VSOR and Maxi-Cl. CURRENT TOPICS IN MEMBRANES 2018; 81:125-176. [PMID: 30243431 DOI: 10.1016/bs.ctm.2018.07.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
An elaborate volume regulation system based on interplay of ion channels and transporters was evolved to cope with constant osmotic challenges caused by intensive metabolism, transport and other physiological/pathophysiological events. In animal cells, two types of anion channels are directly activated by cell swelling and involved in the regulatory volume decrease (RVD): volume-sensitive outwardly rectifying anion channel (VSOR), also called volume-regulated anion channel (VRAC), and Maxi-Cl which is the most major type of maxi-anion channel (MAC). These two channels have very different biophysical profiles and exhibit opposite dependence on intracellular ATP. After several decades of verifying many false-positive candidates for VSOR and Maxi-Cl, LRRC8 family proteins emerged as major VSOR components, and SLCO2A1 protein as a core of Maxi-Cl. Still, neither of these proteins alone can fully reproduce the native channel phenotypes suggesting existence of missing components. Although both VSOR and Maxi-Cl have pores wide enough to accommodate bulky ATP4- and MgATP2- anions, evidence accumulated hitherto, based on pharmacological and gene silencing experiments, suggests that Maxi-Cl, but not VSOR, serves as one of the major pathways for the release of ATP from swollen and ischemic/hypoxic cells. Relations of VSOR and Maxi-Cl with diseases and their selective pharmacology are the topics promoted by recent advance in molecular identification of the two volume-activated, volume-regulatory anion channels.
Collapse
|
44
|
Falzone ME, Malvezzi M, Lee BC, Accardi A. Known structures and unknown mechanisms of TMEM16 scramblases and channels. J Gen Physiol 2018; 150:933-947. [PMID: 29915161 PMCID: PMC6028493 DOI: 10.1085/jgp.201711957] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
Falzone et al. interpret the mechanisms underlying the activity of TMEM16 family members from recent structural and functional work. The TMEM16 family of membrane proteins is composed of both Ca2+-gated Cl− channels and Ca2+-dependent phospholipid scramblases. The functional diversity of TMEM16s underlies their involvement in numerous signal transduction pathways that connect changes in cytosolic Ca2+ levels to cellular signaling networks. Indeed, defects in the function of several TMEM16s cause a variety of genetic disorders, highlighting their fundamental pathophysiological importance. Here, we review how our mechanistic understanding of TMEM16 function has been shaped by recent functional and structural work. Remarkably, the recent determination of near-atomic-resolution structures of TMEM16 proteins of both functional persuasions has revealed how relatively minimal rearrangements in the substrate translocation pathway are sufficient to precipitate the dramatic functional differences that characterize the family. These structures, when interpreted in the light of extensive functional analysis, point to an unusual mechanism for Ca2+-dependent activation of TMEM16 proteins in which substrate permeation is regulated by a combination of conformational rearrangements and electrostatics. These breakthroughs pave the way to elucidate the mechanistic bases of ion and lipid transport by the TMEM16 proteins and unravel the molecular links between these transport activities and their function in human pathophysiology.
Collapse
Affiliation(s)
- Maria E Falzone
- Department of Biochemistry, Weill Cornell Medical School, New York, NY
| | - Mattia Malvezzi
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Byoung-Cheol Lee
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Alessio Accardi
- Department of Biochemistry, Weill Cornell Medical School, New York, NY .,Department of Anesthesiology, Weill Cornell Medical School, New York, NY.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medical School, New York, NY
| |
Collapse
|
45
|
Di Zanni E, Gradogna A, Scholz-Starke J, Boccaccio A. Gain of function of TMEM16E/ANO5 scrambling activity caused by a mutation associated with gnathodiaphyseal dysplasia. Cell Mol Life Sci 2018; 75:1657-1670. [PMID: 29124309 PMCID: PMC5897490 DOI: 10.1007/s00018-017-2704-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/10/2017] [Accepted: 10/30/2017] [Indexed: 12/31/2022]
Abstract
Mutations in the human TMEM16E (ANO5) gene are associated both with the bone disease gnathodiaphyseal dysplasia (GDD; OMIM: 166260) and muscle dystrophies (OMIM: 611307, 613319). However, the physiological function of TMEM16E has remained unclear. We show here that human TMEM16E, when overexpressed in mammalian cell lines, displayed partial plasma membrane localization and gave rise to phospholipid scrambling (PLS) as well as non-selective ionic currents with slow time-dependent activation at highly depolarized membrane potentials. While the activity of wild-type TMEM16E depended on elevated cytosolic Ca2+ levels, a mutant form carrying the GDD-causing T513I substitution showed PLS and large time-dependent ion currents even at low cytosolic Ca2+ concentrations. Contrarily, mutation of the homologous position in the Ca2+-activated Cl- channel TMEM16B paralog hardly affected its function. In summary, these data provide the first direct demonstration of Ca2+-dependent PLS activity for TMEM16E and suggest a gain-of-function phenotype related to a GDD mutation.
Collapse
Affiliation(s)
- Eleonora Di Zanni
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy
| | - Antonella Gradogna
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy
| | - Joachim Scholz-Starke
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy.
| | - Anna Boccaccio
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Via de Marini 6, 16149, Genova, Italy.
| |
Collapse
|
46
|
Rahmati N, Hoebeek FE, Peter S, De Zeeuw CI. Chloride Homeostasis in Neurons With Special Emphasis on the Olivocerebellar System: Differential Roles for Transporters and Channels. Front Cell Neurosci 2018; 12:101. [PMID: 29765304 PMCID: PMC5938380 DOI: 10.3389/fncel.2018.00101] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
The intraneuronal ionic composition is an important determinant of brain functioning. There is growing evidence that aberrant homeostasis of the intracellular concentration of Cl- ([Cl-]i) evokes, in addition to that of Na+ and Ca2+, robust impairments of neuronal excitability and neurotransmission and thereby neurological conditions. More specifically, understanding the mechanisms underlying regulation of [Cl-]i is crucial for deciphering the variability in GABAergic and glycinergic signaling of neurons, in both health and disease. The homeostatic level of [Cl-]i is determined by various regulatory mechanisms, including those mediated by plasma membrane Cl- channels and transporters. This review focuses on the latest advances in identification, regulation and characterization of Cl- channels and transporters that modulate neuronal excitability and cell volume. By putting special emphasis on neurons of the olivocerebellar system, we establish that Cl- channels and transporters play an indispensable role in determining their [Cl-]i and thereby their function in sensorimotor coordination.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Freek E. Hoebeek
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- NIDOD Institute, Wilhelmina Children's Hospital, University Medical Center Utrecht and Brain Center Rudolf Magnus, Utrecht, Netherlands
| | - Saša Peter
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam, Netherlands
| |
Collapse
|
47
|
Muratori C, Pakhomov AG, Gianulis E, Meads J, Casciola M, Mollica PA, Pakhomova ON. Activation of the phospholipid scramblase TMEM16F by nanosecond pulsed electric fields (nsPEF) facilitates its diverse cytophysiological effects. J Biol Chem 2017; 292:19381-19391. [PMID: 28982976 PMCID: PMC5702676 DOI: 10.1074/jbc.m117.803049] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/20/2017] [Indexed: 12/17/2022] Open
Abstract
Nanosecond pulsed electric fields (nsPEF) are emerging as a novel modality for cell stimulation and tissue ablation. However, the downstream protein effectors responsible for nsPEF bioeffects remain to be established. Here we demonstrate that nsPEF activate TMEM16F (or Anoctamin 6), a protein functioning as a Ca2+-dependent phospholipid scramblase and Ca2+-activated chloride channel. Using confocal microscopy and patch clamp recordings, we investigated the relevance of TMEM16F activation for several bioeffects triggered by nsPEF, including phosphatidylserine (PS) externalization, nanopore-conducted currents, membrane blebbing, and cell death. In HEK 293 cells treated with a single 300-ns pulse of 25.5 kV/cm, Tmem16f expression knockdown and TMEM16F-specific inhibition decreased nsPEF-induced PS exposure by 49 and 42%, respectively. Moreover, the Tmem16f silencing significantly decreased Ca2+-dependent chloride channel currents activated in response to the nanoporation. Tmem16f expression also affected nsPEF-induced cell blebbing, with only 20% of the silenced cells developing blebs compared with 53% of the control cells. This inhibition of cellular blebbing correlated with a 25% decrease in cytosolic free Ca2+ transient at 30 s after nanoporation. Finally, in TMEM16F-overexpressing cells, a train of 120 pulses (300 ns, 20 Hz, 6 kV/cm) decreased cell survival to 34% compared with 51% in control cells (*, p < 0.01). Taken together, these results indicate that TMEM16F activation by nanoporation mediates and enhances the diverse cellular effects of nsPEF.
Collapse
Affiliation(s)
| | | | - Elena Gianulis
- From the Frank Reidy Research Center for Bioelectrics, and
| | - Jade Meads
- From the Frank Reidy Research Center for Bioelectrics, and
| | - Maura Casciola
- From the Frank Reidy Research Center for Bioelectrics, and
| | - Peter A Mollica
- the Department of Medical Diagnostics and Translational Sciences, Old Dominion University, Norfolk, Virginia 23508
| | | |
Collapse
|
48
|
Procoagulant platelets: generation, function, and therapeutic targeting in thrombosis. Blood 2017; 130:2171-2179. [DOI: 10.1182/blood-2017-05-787259] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/12/2017] [Indexed: 11/20/2022] Open
Abstract
Abstract
Current understanding of how platelets localize coagulation to wound sites has come mainly from studies of a subpopulation of activated platelets. In this review, we summarize data from the last 4 decades that have described these platelets with a range of descriptive titles and attributes. We identify striking overlaps in the reported characteristics of these platelets, which imply a single subpopulation of versatile platelets and thus suggest that their commonality requires unification of their description. We therefore propose the term procoagulant platelet as the unifying terminology. We discuss the agonist requirements and molecular drivers for the dramatic morphological transformation platelets undergo when becoming procoagulant. Finally, we provide perspectives on the biomarker potential of procoagulant platelets for thrombotic events as well as on the possible clinical benefits of inhibitors of carbonic anhydrase enzymes and the water channel Aquaporin-1 for targeting this subpopulation of platelets as antiprocoagulant antithrombotics.
Collapse
|
49
|
Jiang T, Yu K, Hartzell HC, Tajkhorshid E. Lipids and ions traverse the membrane by the same physical pathway in the nhTMEM16 scramblase. eLife 2017; 6:28671. [PMID: 28917060 PMCID: PMC5628016 DOI: 10.7554/elife.28671] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022] Open
Abstract
From bacteria to mammals, different phospholipid species are segregated between the inner and outer leaflets of the plasma membrane by ATP-dependent lipid transporters. Disruption of this asymmetry by ATP-independent phospholipid scrambling is important in cellular signaling, but its mechanism remains incompletely understood. Using MD simulations coupled with experimental assays, we show that the surface hydrophilic transmembrane cavity exposed to the lipid bilayer on the fungal scramblase nhTMEM16 serves as the pathway for both lipid translocation and ion conduction across the membrane. Ca2+ binding stimulates its open conformation by altering the structure of transmembrane helices that line the cavity. We have identified key amino acids necessary for phospholipid scrambling and validated the idea that ions permeate TMEM16 Cl- channels via a structurally homologous pathway by showing that mutation of two residues in the pore region of the TMEM16A Ca2+-activated Cl- channel convert it into a robust scramblase.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Biochemistry, Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, United States.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Kuai Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, United States
| | - Emad Tajkhorshid
- Department of Biochemistry, Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, United States.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, United States
| |
Collapse
|
50
|
Wang H, Zou L, Ma K, Yu J, Wu H, Wei M, Xiao Q. Cell-specific mechanisms of TMEM16A Ca 2+-activated chloride channel in cancer. Mol Cancer 2017; 16:152. [PMID: 28893247 PMCID: PMC5594453 DOI: 10.1186/s12943-017-0720-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/01/2017] [Indexed: 02/08/2023] Open
Abstract
TMEM16A (known as anoctamin 1) Ca2+-activated chloride channel is overexpressed in many tumors. TMEM16A overexpression can be caused by gene amplification in many tumors harboring 11q13 amplification. TMEM16A expression is also controlled in many cancer cells via transcriptional regulation, epigenetic regulation and microRNAs. In addition, TMEM16A activates different signaling pathways in different cancers, e.g. the EGFR and CAMKII signaling in breast cancer, the p38 and ERK1/2 signaling in hepatoma, the Ras-Raf-MEK-ERK1/2 signaling in head and neck squamous cell carcinoma and bladder cancer, and the NFκB signaling in glioma. Furthermore, TMEM16A overexpression has been reported to promote, inhibit, or produce no effects on cell proliferation and migration in different cancer cells. Since TMEM16A exerts different roles in different cancer cells via activation of distinct signaling pathways, we try to develop the idea that TMEM16A regulates cancer cell proliferation and migration in a cell-dependent mechanism. The cell-specific role of TMEM16A may depend on the cellular environment that is predetermined by TMEM16A overexpression mechanisms specific for a particular cancer type. TMEM16A may exert its cell-specific role via its associated protein networks, phosphorylation by different kinases, and involvement of different signaling pathways. In addition, we discuss the role of TMEM16A channel activity in cancer, and its clinical use as a prognostic and predictive marker in different cancers. This review highlights the cell-type specific mechanisms of TMEM16A in cancer, and envisions the promising use of TMEM16A inhibitors as a potential treatment for TMEM16A-overexpressing cancers.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Liang Zou
- Department of Anesthesiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 China
| | - Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Jiankun Yu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| |
Collapse
|