1
|
Wu KC, Lee CY, Chan P, Leong IL, Leung YM. Tannic Acid as an Ion Channel Modulator: An Understanding of Its Pharmacological Spectrum. Drug Dev Res 2025; 86:e70098. [PMID: 40342145 DOI: 10.1002/ddr.70098] [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: 04/02/2025] [Revised: 04/02/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025]
Abstract
Tannic acid (TA) is a polyphenol present in many plant foods and beverages such as green tea and wines. As a food additive, it has been recognized by Food and Drug Administration as generally safe. As a candidate drug, its pharmacological effects cover a wide spectrum, ranging from antibacterial, anticancer, cardioprotection to neuroprotection. TA has been shown to modulate a number of ion channels such as Ca2+-activated Cl- channels (CaCC), voltage-gated K+ (Kv) channels and transient receptor potential (TRP) channels, producing effects such as analgesia, antihypertensive effects and reduction of airway hypersensitivity. In this review we focus on how ion channel modulation by TA may account for the pharmacological effects of TA in various cells and organ systems. Further emphasis should be paid to factors, such as dosage and routes of administration, before the pharmacological actions of TA could be translated into therapeutic applications.
Collapse
Affiliation(s)
- King-Chuen Wu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chiu-Yin Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Paul Chan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Iat-Lon Leong
- Division of Cardiology, University Hospital, Macau University of Science and Technology, Macau, China
| | - Yuk-Man Leung
- Department of Physiology, China Medical University, Taichung, Taiwan
| |
Collapse
|
2
|
Alveal M, Méndez A, García A, Henríquez M. Purinergic regulation of pulmonary vascular tone. Purinergic Signal 2024; 20:595-606. [PMID: 38713328 PMCID: PMC11554604 DOI: 10.1007/s11302-024-10010-5] [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: 12/04/2023] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Purinergic signaling is a crucial determinant in the regulation of pulmonary vascular physiology and presents a promising avenue for addressing lung diseases. This intricate signaling system encompasses two primary receptor classes: P1 and P2 receptors. P1 receptors selectively bind adenosine, while P2 receptors exhibit an affinity for ATP, ADP, UTP, and UDP. Functionally, P1 receptors are associated with vasodilation, while P2 receptors mediate vasoconstriction, particularly in basally relaxed vessels, through modulation of intracellular Ca2+ levels. The P2X subtype receptors facilitate extracellular Ca2+ influx, while the P2Y subtype receptors are linked to endoplasmic reticulum Ca2+ release. Notably, the primary receptor responsible for ATP-induced vasoconstriction is P2X1, with α,β-meATP and UDP being identified as potent vasoconstrictor agonists. Interestingly, ATP has been shown to induce endothelium-dependent vasodilation in pre-constricted vessels, associated with nitric oxide (NO) release. In the context of P1 receptors, adenosine stimulation of pulmonary vessels has been unequivocally demonstrated to induce vasodilation, with a clear dependency on the A2B receptor, as evidenced in studies involving guinea pigs and rats. Importantly, evidence strongly suggests that this vasodilation occurs independently of endothelium-mediated mechanisms. Furthermore, studies have revealed variations in the expression of purinergic receptors across different vessel sizes, with reports indicating notably higher expression of P2Y1, P2Y2, and P2Y4 receptors in small pulmonary arteries. While the existing evidence in this area is still emerging, it underscores the urgent need for a comprehensive examination of the specific characteristics of purinergic signaling in the regulation of pulmonary vascular tone, particularly focusing on the disparities observed across different intrapulmonary vessel sizes. Consequently, this review aims to meticulously explore the current evidence regarding the role of purinergic signaling in pulmonary vascular tone regulation, with a specific emphasis on the variations observed in intrapulmonary vessel sizes. This endeavor is critical, as purinergic signaling holds substantial promise in the modulation of vascular tone and in the proactive prevention and treatment of pulmonary vascular diseases.
Collapse
Affiliation(s)
- Marco Alveal
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina Universidad de Chile, Independencia 1027, 7500975, Independencia, Santiago, Chile
| | - Andrea Méndez
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina Universidad de Chile, Independencia 1027, 7500975, Independencia, Santiago, Chile
- Escuela de Kinesiología, Facultad de Salud y Ciencias Sociales, Campus Providencia, Sede Santiago, Universidad de Las Américas, Santiago, Chile
| | - Aline García
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina Universidad de Chile, Independencia 1027, 7500975, Independencia, Santiago, Chile
- Escuela de Graduados, Facultad de Ciencias Veterinarias,, Universidad Austral de Chile, Valdivia, Chile
| | - Mauricio Henríquez
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina Universidad de Chile, Independencia 1027, 7500975, Independencia, Santiago, Chile.
| |
Collapse
|
3
|
Arreola J, López-Romero AE, Huerta M, Guzmán-Hernández ML, Pérez-Cornejo P. Insights into the function and regulation of the calcium-activated chloride channel TMEM16A. Cell Calcium 2024; 121:102891. [PMID: 38772195 DOI: 10.1016/j.ceca.2024.102891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
The TMEM16A channel, a member of the TMEM16 protein family comprising chloride (Cl-) channels and lipid scramblases, is activated by the free intracellular Ca2+ increments produced by inositol 1,4,5-trisphosphate (IP3)-induced Ca2+ release after GqPCRs or Ca2+ entry through cationic channels. It is a ubiquitous transmembrane protein that participates in multiple physiological functions essential to mammals' lives. TMEM16A structure contains two identical 10-segment monomers joined at their transmembrane segment 10. Each monomer harbours one independent hourglass-shaped pore gated by Ca2+ ligation to an orthosteric site adjacent to the pore and controlled by two gates. The orthosteric site is created by assembling negatively charged glutamate side chains near the pore´s cytosolic end. When empty, this site generates an electrostatic barrier that controls channel rectification. In addition, an isoleucine-triad forms a hydrophobic gate at the boundary of the cytosolic vestibule and the inner side of the neck. When the cytosolic Ca2+ rises, one or two Ca2+ ions bind to the orthosteric site in a voltage (V)-dependent manner, thus neutralising the electrostatic barrier and triggering an allosteric gating mechanism propagating via transmembrane segment 6 to the hydrophobic gate. These coordinated events lead to pore opening, allowing the Cl- flux to ensure the physiological response. The Ca2+-dependent function of TMEM16A is highly regulated. Anions with higher permeability than Cl- facilitate V dependence by increasing the Ca2+ sensitivity, intracellular protons can replace Ca2+ and induce channel opening, and phosphatidylinositol 4,5-bisphosphate bound to four cytosolic sites likely maintains Ca2+ sensitivity. Additional regulation is afforded by cytosolic proteins, most likely by phosphorylation and protein-protein interaction mechanisms.
Collapse
Affiliation(s)
- Jorge Arreola
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico.
| | - Ana Elena López-Romero
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - Miriam Huerta
- Jorge Arreola, Physics Institute of Universidad Autónoma de San Luis Potosí. Av. Parque Chapultepec 1570, Privadas del Pedregal, 78295 San Luis Potosí, SLP., Mexico
| | - María Luisa Guzmán-Hernández
- Catedrática CONAHCYT, Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí. Ave. V. Carranza 2905, Los Filtros, San Luis Potosí, SLP 78210, Mexico
| |
Collapse
|
4
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, Chen Z, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Wu C, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024; 57:1243-1259.e8. [PMID: 38744291 PMCID: PMC11168877 DOI: 10.1016/j.immuni.2024.03.023] [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: 04/05/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
Epithelial cells secrete chloride to regulate water release at mucosal barriers, supporting both homeostatic hydration and the "weep" response that is critical for type 2 immune defense against parasitic worms (helminths). Epithelial tuft cells in the small intestine sense helminths and release cytokines and lipids to activate type 2 immune cells, but whether they regulate epithelial secretion is unknown. Here, we found that tuft cell activation rapidly induced epithelial chloride secretion in the small intestine. This response required tuft cell sensory functions and tuft cell-derived acetylcholine (ACh), which acted directly on neighboring epithelial cells to stimulate chloride secretion, independent of neurons. Maximal tuft cell-induced chloride secretion coincided with immune restriction of helminths, and clearance was delayed in mice lacking tuft cell-derived ACh, despite normal type 2 inflammation. Thus, we have uncovered an epithelium-intrinsic response unit that uses ACh to couple tuft cell sensing to the secretory defenses of neighboring epithelial cells.
Collapse
Affiliation(s)
- Tyler E Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Derek B Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew B Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Darshan N Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kaitlyn A Barrow
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucille M Rich
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, CA, USA
| | - Jason S Debley
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Michael R Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
5
|
Akin EJ, Aoun J, Jimenez C, Mayne K, Baeck J, Young MD, Sullivan B, Sanders KM, Ward SM, Bulley S, Jaggar JH, Earley S, Greenwood IA, Leblanc N. ANO1, CaV1.2, and IP3R form a localized unit of EC-coupling in mouse pulmonary arterial smooth muscle. J Gen Physiol 2023; 155:e202213217. [PMID: 37702787 PMCID: PMC10499037 DOI: 10.1085/jgp.202213217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/01/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Pulmonary arterial (PA) smooth muscle cells (PASMC) generate vascular tone in response to agonists coupled to Gq-protein receptor signaling. Such agonists stimulate oscillating calcium waves, the frequency of which drives the strength of contraction. These Ca2+ events are modulated by a variety of ion channels including voltage-gated calcium channels (CaV1.2), the Tmem16a or Anoctamin-1 (ANO1)-encoded calcium-activated chloride (CaCC) channel, and Ca2+ release from the sarcoplasmic reticulum through inositol-trisphosphate receptors (IP3R). Although these calcium events have been characterized, it is unclear how these calcium oscillations underly a sustained contraction in these muscle cells. We used smooth muscle-specific ablation of ANO1 and pharmacological tools to establish the role of ANO1, CaV1.2, and IP3R in the contractile and intracellular Ca2+ signaling properties of mouse PA smooth muscle expressing the Ca2+ biosensor GCaMP3 or GCaMP6. Pharmacological block or genetic ablation of ANO1 or inhibition of CaV1.2 or IP3R, or Ca2+ store depletion equally inhibited 5-HT-induced tone and intracellular Ca2+ waves. Coimmunoprecipitation experiments showed that an anti-ANO1 antibody was able to pull down both CaV1.2 and IP3R. Confocal and superresolution nanomicroscopy showed that ANO1 coassembles with both CaV1.2 and IP3R at or near the plasma membrane of PASMC from wild-type mice. We conclude that the stable 5-HT-induced PA contraction results from the integration of stochastic and localized Ca2+ events supported by a microenvironment comprising ANO1, CaV1.2, and IP3R. In this model, ANO1 and CaV1.2 would indirectly support cyclical Ca2+ release events from IP3R and propagation of intracellular Ca2+ waves.
Collapse
Affiliation(s)
- Elizabeth J. Akin
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Joydeep Aoun
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Connor Jimenez
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Katie Mayne
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Julius Baeck
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Michael D. Young
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Brennan Sullivan
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Simon Bulley
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jonathan H. Jaggar
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Scott Earley
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| | - Iain A. Greenwood
- Department of Vascular Pharmacology, Molecular and Clinical Science Research Institute, St. George’s University of London, London, UK
| | - Normand Leblanc
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, Reno, NV, USA
| |
Collapse
|
6
|
Varga Á, Madácsy T, Görög M, Kiss A, Susánszki P, Szabó V, Jójárt B, Dudás K, Farkas G, Szederkényi E, Lázár G, Farkas A, Ayaydin F, Pallagi P, Maléth J. Human pancreatic ductal organoids with controlled polarity provide a novel ex vivo tool to study epithelial cell physiology. Cell Mol Life Sci 2023; 80:192. [PMID: 37380797 PMCID: PMC10307727 DOI: 10.1007/s00018-023-04836-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 06/30/2023]
Abstract
Epithelial ion and fluid secretion determine the physiological functions of a broad range of organs, such as the lung, liver, or pancreas. The molecular mechanism of pancreatic ion secretion is challenging to investigate due to the limited access to functional human ductal epithelia. Patient-derived organoids may overcome these limitations, however direct accessibility of the apical membrane is not solved. In addition, due to the vectorial transport of ions and fluid the intraluminal pressure in the organoids is elevated, which may hinder the study of physiological processes. To overcome these, we developed an advanced culturing method for human pancreatic organoids based on the removal of the extracellular matrix that induced an apical-to-basal polarity switch also leading to reversed localization of proteins with polarized expression. The cells in the apical-out organoids had a cuboidal shape, whereas their resting intracellular Ca2+ concentration was more consistent compared to the cells in the apical-in organoids. Using this advanced model, we demonstrated the expression and function of two novel ion channels, the Ca2+ activated Cl- channel Anoctamin 1 (ANO1) and the epithelial Na+ channel (ENaC), which were not considered in ductal cells yet. Finally, we showed that the available functional assays, such as forskolin-induced swelling, or intracellular Cl- measurement have improved dynamic range when performed with apical-out organoids. Taken together our data suggest that polarity-switched human pancreatic ductal organoids are suitable models to expand our toolset in basic and translational research.
Collapse
Affiliation(s)
- Árpád Varga
- Department of Medicine, University of Szeged, Szeged, Hungary
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HCEMM-USZ Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Tamara Madácsy
- Department of Medicine, University of Szeged, Szeged, Hungary
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HCEMM-USZ Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Marietta Görög
- Department of Medicine, University of Szeged, Szeged, Hungary
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary
| | - Aletta Kiss
- Department of Medicine, University of Szeged, Szeged, Hungary
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary
| | - Petra Susánszki
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - Viktória Szabó
- Department of Medicine, University of Szeged, Szeged, Hungary
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HCEMM-USZ Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Boldizsár Jójárt
- Department of Medicine, University of Szeged, Szeged, Hungary
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HCEMM-USZ Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - Krisztina Dudás
- Department of Medicine, University of Szeged, Szeged, Hungary
| | - Gyula Farkas
- Department of Surgery, University of Szeged, Szeged, Hungary
| | | | - György Lázár
- Department of Surgery, University of Szeged, Szeged, Hungary
| | - Attila Farkas
- HCEMM-USZ Functional Cell Biology and Immunology Advanced Core Facility, University of Szeged, Szeged, Hungary
| | - Ferhan Ayaydin
- HCEMM-USZ Functional Cell Biology and Immunology Advanced Core Facility, University of Szeged, Szeged, Hungary
| | - Petra Pallagi
- Department of Medicine, University of Szeged, Szeged, Hungary
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HCEMM-USZ Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary
| | - József Maléth
- Department of Medicine, University of Szeged, Szeged, Hungary.
- ELRN-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, Department of Medicine, University of Szeged, Szeged, 6720, Hungary.
- HCEMM-USZ Molecular Gastroenterology Research Group, University of Szeged, Szeged, Hungary.
| |
Collapse
|
7
|
Zhang L, Wang Y, Yuan W, An C, Tan Q, Ma J. BEST1 Positive Monocytes in Circulation: Visualize Intratumoral Crosstalk between Cancer Cells and Monocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2205915. [PMID: 37088729 DOI: 10.1002/advs.202205915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/29/2023] [Indexed: 05/03/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are characterized by an abundance of monocytes and macrophages recruited from the peripheral blood. However, it has not been determined whether these infiltrated cells can be released back into circulation with a tumor-associated neobiosignature. This study reports that Bestrophin1 (BEST1), a component protein of Ca2+ -activated Cl- channels (CaCCs), is highly expressed on classical monocytes in the peripheral blood of HNSCC patients. This is due to monocyte education by tumor cells, in which tumoral VEGF-A upregulates BEST1 expression on monocytes through the MEK-ERK-ELK1 pathway. This leads to improved secretion of IL-6 and IL-8, which promotes tumor cell proliferation. This work also finds that BEST1 facilitates the motility of monocytes, contributing to the migration of these cells back into circulation. These results suggest that the expression of BEST1 on peripheral monocytes may be a potential tool for monitoring tumor progression, and opens up the possibility of searching for cancer biomarkers on monocytes rather than on the tumor or its products.
Collapse
Affiliation(s)
- Luyao Zhang
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, P. R. China
| | - Yiran Wang
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, P. R. China
| | - Wei Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, P. R. China
| | - Changming An
- Department of Head and Neck Surgery, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, P. R. China
| | - Qin Tan
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, P. R. China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, P. R. China
| |
Collapse
|
8
|
Arreola J, López-Romero AE, Pérez-Cornejo P, Rodríguez-Menchaca AA. Phosphatidylinositol 4,5-Bisphosphate and Cholesterol Regulators of the Calcium-Activated Chloride Channels TMEM16A and TMEM16B. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:279-304. [PMID: 36988885 DOI: 10.1007/978-3-031-21547-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Chloride fluxes through homo-dimeric calcium-activated channels TMEM16A and TMEM16B are critical to blood pressure, gastrointestinal motility, hormone, fluid and electrolyte secretion, pain sensation, sensory transduction, and neuronal and muscle excitability. Their gating depends on the voltage-dependent binding of two intracellular calcium ions to a high-affinity site formed by acidic residues from α-helices 6-8 in each monomer. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), a low-abundant lipid of the inner leaflet, supports TMEM16A function; it allows TMEM16A to evade the down-regulation induced by calcium, poly-L-lysine, or PI(4,5)P2 5-phosphatase. In stark contrast, adding or removing PI(4,5)P2 diminishes or increases TMEM16B function, respectively. PI(4,5)P2-binding sites on TMEM16A, and presumably on TMEM16B, are on the cytosolic side of α-helices 3-5, opposite the calcium-binding sites. This modular structure suggested that PI(4,5)P2 and calcium cooperate to maintain the conductive state in TMEM16A. Cholesterol, the second-largest constituent of the plasma membrane, also regulates TMEM16A though the mechanism, functional outcomes, binding site(s), and effects on TMEM16A and TMEM16B remain unknown.
Collapse
Affiliation(s)
- Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | | | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Aldo A Rodríguez-Menchaca
- Department of Physiology and Biophysics, School of Medicine, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| |
Collapse
|
9
|
Jimenez C, Hawn MB, Akin E, Leblanc N. Translational potential of targeting Anoctamin-1-Encoded Calcium-Activated chloride channels in hypertension. Biochem Pharmacol 2022; 206:115320. [PMID: 36279919 DOI: 10.1016/j.bcp.2022.115320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Calcium-activated chloride channels (CaCC) provide a depolarizing stimulus to a variety of tissues through chloride efflux in response to a rise in internal Ca2+ and voltage. One of these channels, Anoctamin-1 (ANO1 or TMEM16A) is now recognized to play a central role in promoting smooth muscle tone in various types of blood vessels. Its role in hypertension, and thus the therapeutic promise of targeting ANO1, is less straightforward. This review gives an overview of our current knowledge about the potential role ANO1 may play in hypertension within the systemic, portal, and pulmonary vascular systems and the importance of this information when pursuing potential treatment strategies. While the role of ANO1 is well-established in several forms of pulmonary hypertension, its contributions to both the generation of vascular tone and its role in hypertension within the systemic and portal systems are much less clear. This, combined with ANO1's various roles throughout a multitude of tissues throughout the body, command caution when targeting ANO1 as a therapeutic target and may require tissue-selective strategies.
Collapse
Affiliation(s)
- Connor Jimenez
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Matthew B Hawn
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Elizabeth Akin
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Normand Leblanc
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA.
| |
Collapse
|
10
|
Patel AA, Sakurai A, Himmel NJ, Cox DN. Modality specific roles for metabotropic GABAergic signaling and calcium induced calcium release mechanisms in regulating cold nociception. Front Mol Neurosci 2022; 15:942548. [PMID: 36157080 PMCID: PMC9502035 DOI: 10.3389/fnmol.2022.942548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Calcium (Ca2+) plays a pivotal role in modulating neuronal-mediated responses to modality-specific sensory stimuli. Recent studies in Drosophila reveal class III (CIII) multidendritic (md) sensory neurons function as multimodal sensors regulating distinct behavioral responses to innocuous mechanical and nociceptive thermal stimuli. Functional analyses revealed CIII-mediated multimodal behavioral output is dependent upon activation levels with stimulus-evoked Ca2+ displaying relatively low vs. high intracellular levels in response to gentle touch vs. noxious cold, respectively. However, the mechanistic bases underlying modality-specific differential Ca2+ responses in CIII neurons remain incompletely understood. We hypothesized that noxious cold-evoked high intracellular Ca2+ responses in CIII neurons may rely upon Ca2+ induced Ca2+ release (CICR) mechanisms involving transient receptor potential (TRP) channels and/or metabotropic G protein coupled receptor (GPCR) activation to promote cold nociceptive behaviors. Mutant and/or CIII-specific knockdown of GPCR and CICR signaling molecules [GABA B -R2, Gαq, phospholipase C, ryanodine receptor (RyR) and Inositol trisphosphate receptor (IP3R)] led to impaired cold-evoked nociceptive behavior. GPCR mediated signaling, through GABA B -R2 and IP3R, is not required in CIII neurons for innocuous touch evoked behaviors. However, CICR via RyR is required for innocuous touch-evoked behaviors. Disruptions in GABA B -R2, IP3R, and RyR in CIII neurons leads to significantly lower levels of cold-evoked Ca2+ responses indicating GPCR and CICR signaling mechanisms function in regulating Ca2+ release. CIII neurons exhibit bipartite cold-evoked firing patterns, where CIII neurons burst during rapid temperature change and tonically fire during steady state cold temperatures. GABA B -R2 knockdown in CIII neurons resulted in disorganized firing patterns during cold exposure. We further demonstrate that application of GABA or the GABA B specific agonist baclofen potentiates cold-evoked CIII neuron activity. Upon ryanodine application, CIII neurons exhibit increased bursting activity and with CIII-specific RyR knockdown, there is an increase in cold-evoked tonic firing and decrease in bursting. Lastly, our previous studies implicated the TRPP channel Pkd2 in cold nociception, and here, we show that Pkd2 and IP3R genetically interact to specifically regulate cold-evoked behavior, but not innocuous mechanosensation. Collectively, these analyses support novel, modality-specific roles for metabotropic GABAergic signaling and CICR mechanisms in regulating intracellular Ca2+ levels and cold-evoked behavioral output from multimodal CIII neurons.
Collapse
Affiliation(s)
| | | | | | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
11
|
De Jesús-Pérez JJ, López-Romero AE, Posadas O, Segura-Covarrubias G, Aréchiga-Figueroa I, Gutiérrez-Medina B, Pérez-Cornejo P, Arreola J. Gating and anion selectivity are reciprocally regulated in TMEM16A (ANO1). J Gen Physiol 2022; 154:213275. [PMID: 35687042 PMCID: PMC9194859 DOI: 10.1085/jgp.202113027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 05/23/2022] [Indexed: 02/03/2023] Open
Abstract
Numerous essential physiological processes depend on the TMEM16A-mediated Ca2+-activated chloride fluxes. Extensive structure-function studies have helped to elucidate the Ca2+ gating mechanism of TMEM16A, revealing a Ca2+-sensing element close to the anion pore that alters conduction. However, substrate selection and the substrate-gating relationship in TMEM16A remain less explored. Here, we study the gating-permeant anion relationship on mouse TMEM16A expressed in HEK 293 cells using electrophysiological recordings coupled with site-directed mutagenesis. We show that the apparent Ca2+ sensitivity of TMEM16A increased with highly permeant anions and SCN- mole fractions, likely by stabilizing bound Ca2+. Conversely, mutations at crucial gating elements, including the Ca2+-binding site 1, the transmembrane helix 6 (TM6), and the hydrophobic gate, impaired the anion permeability and selectivity of TMEM16A. Finally, we found that, unlike anion-selective wild-type channels, the voltage dependence of unselective TMEM16A mutant channels was less sensitive to SCN-. Therefore, our work identifies structural determinants of selectivity at the Ca2+ site, TM6, and hydrophobic gate and reveals a reciprocal regulation of gating and selectivity. We suggest that this regulation is essential to set ionic selectivity and the Ca2+ and voltage sensitivities in TMEM16A.
Collapse
Affiliation(s)
| | - Ana E. López-Romero
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Odalys Posadas
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | | | - Iván Aréchiga-Figueroa
- Consejo Nacional de Ciencia y Tecnología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Braulio Gutiérrez-Medina
- Advanced Materials Division, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, México
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México,Correspondence to Jorge Arreola:
| |
Collapse
|
12
|
Jaszczak JS, DeVault L, Jan LY, Jan YN. Steroid hormone signaling activates thermal nociception during Drosophila peripheral nervous system development. eLife 2022; 11:e76464. [PMID: 35353036 PMCID: PMC8967384 DOI: 10.7554/elife.76464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 12/27/2022] Open
Abstract
Sensory neurons enable animals to detect environmental changes and avoid harm. An intriguing open question concerns how the various attributes of sensory neurons arise in development. Drosophila melanogaster larvae undergo a behavioral transition by robustly activating a thermal nociceptive escape behavior during the second half of larval development (third instar). The Class IV dendritic arborization (C4da) neurons are multimodal sensors which tile the body wall of Drosophila larvae and detect nociceptive temperature, light, and mechanical force. In contrast to the increase in nociceptive behavior in the third instar, we find that ultraviolet light-induced Ca2+ activity in C4da neurons decreases during the same period of larval development. Loss of ecdysone receptor has previously been shown to reduce nociception in third instar larvae. We find that ligand-dependent activation of ecdysone signaling is sufficient to promote nociceptive responses in second instar larvae and suppress expression of subdued (encoding a TMEM16 channel). Reduction of subdued expression in second instar C4da neurons not only increases thermal nociception but also decreases the response to ultraviolet light. Thus, steroid hormone signaling suppresses subdued expression to facilitate the sensory switch of C4da neurons. This regulation of a developmental sensory switch through steroid hormone regulation of channel expression raises the possibility that ion channel homeostasis is a key target for tuning the development of sensory modalities.
Collapse
Affiliation(s)
- Jacob S Jaszczak
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Laura DeVault
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Department of Developmental Biology, Washington University Medical SchoolSaint LouisUnited States
| | - Lily Yeh Jan
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Yuh Nung Jan
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| |
Collapse
|
13
|
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ signaling pathway that is evolutionarily conserved across eukaryotes. SOCE is triggered physiologically when the endoplasmic reticulum (ER) Ca2+ stores are emptied through activation of inositol 1,4,5-trisphosphate receptors. SOCE is mediated by the Ca2+ release-activated Ca2+ (CRAC) channels, which are highly Ca2+ selective. Upon store depletion, the ER Ca2+-sensing STIM proteins aggregate and gain extended conformations spanning the ER-plasma membrane junctional space to bind and activate Orai, the pore-forming proteins of hexameric CRAC channels. In recent years, studies on STIM and Orai tissue-specific knockout mice and gain- and loss-of-function mutations in humans have shed light on the physiological functions of SOCE in various tissues. Here, we describe recent findings on the composition of native CRAC channels and their physiological functions in immune, muscle, secretory, and neuronal systems to draw lessons from transgenic mice and human diseases caused by altered CRAC channel activity.
Collapse
Affiliation(s)
- Scott M Emrich
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA;
| | - Ryan E Yoast
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA;
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA;
- Department of Pharmacology and Chemical Biology and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Leon-Aparicio D, Sánchez-Solano A, Arreola J, Perez-Cornejo P. Oleic acid blocks the calcium-activated chloride channel TMEM16A/ANO1. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159134. [DOI: 10.1016/j.bbalip.2022.159134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 02/12/2022] [Accepted: 02/17/2022] [Indexed: 11/28/2022]
|
15
|
Klett NJ, Cravetchi O, Allen CN. Long-Term Imaging Reveals a Circadian Rhythm of Intracellular Chloride in Neurons of the Suprachiasmatic Nucleus. J Biol Rhythms 2022; 37:110-123. [PMID: 34994231 PMCID: PMC9203244 DOI: 10.1177/07487304211059770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Both inhibitory and excitatory GABA transmission exist in the mature suprachiasmatic nucleus (SCN), the master pacemaker of circadian physiology. Whether GABA is inhibitory or excitatory depends on the intracellular chloride concentration ([Cl-]i). Here, using the genetically encoded ratiometric probe Cl-Sensor, we investigated [Cl-]i in AVP and VIP-expressing SCN neurons for several days in culture. The chloride ratio (RCl) demonstrated circadian rhythmicity in AVP + neurons and VIP + neurons, but was not detected in GFAP + astrocytes. RCl peaked between ZT 7 and ZT 8 in both AVP + and VIP + neurons. RCl rhythmicity was not dependent on the activity of several transmembrane chloride carriers, action potential generation, or the L-type voltage-gated calcium channels, but was sensitive to GABA antagonists. We conclude that [Cl-]i is under circadian regulation in both AVP + and VIP + neurons.
Collapse
Affiliation(s)
- Nathan J. Klett
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Olga Cravetchi
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
| | - Charles N. Allen
- Oregon Institute for Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Thiruvengadam G, Sreetama SC, Charton K, Hogarth M, Novak JS, Suel-Petat L, Chandra G, Allard B, Richard I, Jaiswal JK. Anoctamin 5 Knockout Mouse Model Recapitulates LGMD2L Muscle Pathology and Offers Insight Into in vivo Functional Deficits. J Neuromuscul Dis 2021; 8:S243-S255. [PMID: 34633328 PMCID: PMC8673513 DOI: 10.3233/jnd-210720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the Anoctamin 5 (Ano5) gene that result in the lack of expression or function of ANO5 protein, cause Limb Girdle Muscular Dystrophy (LGMD) 2L/R12, and Miyoshi Muscular Dystrophy (MMD3). However, the dystrophic phenotype observed in patient muscles is not uniformly recapitulated by ANO5 knockout in animal models of LGMD2L. Here we describe the generation of a mouse model of LGMD2L generated by targeted out-of-frame deletion of the Ano5 gene. This model shows progressive muscle loss, increased muscle weakness, and persistent bouts of myofiber regeneration without chronic muscle inflammation, which recapitulates the mild to moderate skeletal muscle dystrophy reported in the LGMD2L patients. We show that these features of ANO5 deficient muscle are not associated with a change in the calcium-activated sarcolemmal chloride channel activity or compromised in vivo regenerative myogenesis. Use of this mouse model allows conducting in vivo investigations into the functional role of ANO5 in muscle health and for preclinical therapeutic development for LGMD2L.
Collapse
Affiliation(s)
- Girija Thiruvengadam
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Sen Chandra Sreetama
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Karine Charton
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Marshall Hogarth
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - James S Novak
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington DC
| | - Laurence Suel-Petat
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Goutam Chandra
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGene, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Lyon, France
| | - Isabelle Richard
- Généthon INSERM, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Jyoti K Jaiswal
- Center of Genetic Medicine Research, Children's National Health System, MW Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington DC
| |
Collapse
|
19
|
Chandra G, Sreetama SC, Mázala DAG, Charton K, VanderMeulen JH, Richard I, Jaiswal JK. Endoplasmic reticulum maintains ion homeostasis required for plasma membrane repair. J Cell Biol 2021; 220:211873. [PMID: 33688936 PMCID: PMC7953257 DOI: 10.1083/jcb.202006035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 12/11/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022] Open
Abstract
Of the many crucial functions of the ER, homeostasis of physiological calcium increase is critical for signaling. Plasma membrane (PM) injury causes a pathological calcium influx. Here, we show that the ER helps clear this surge in cytoplasmic calcium through an ER-resident calcium pump, SERCA, and a calcium-activated ion channel, Anoctamin 5 (ANO5). SERCA imports calcium into the ER, and ANO5 supports this by maintaining electroneutrality of the ER lumen through anion import. Preventing either of these transporter activities causes cytosolic calcium overload and disrupts PM repair (PMR). ANO5 deficit in limb girdle muscular dystrophy 2L (LGMD2L) patient cells compromises their cytosolic and ER calcium homeostasis. By generating a mouse model of LGMD2L, we find that PM injury causes cytosolic calcium overload and compromises the ability of ANO5-deficient myofibers to repair. Addressing calcium overload in ANO5-deficient myofibers enables them to repair, supporting the requirement of the ER in calcium homeostasis in injured cells and facilitating PMR.
Collapse
Affiliation(s)
- Goutam Chandra
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Sen Chandra Sreetama
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Davi A G Mázala
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Karine Charton
- Généthon, Institut National de la Santé et de la Recherche Médicale, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Jack H VanderMeulen
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Isabelle Richard
- Généthon, Institut National de la Santé et de la Recherche Médicale, U951, INTEGRARE Research Unit, University Paris-Saclay, Evry, France
| | - Jyoti K Jaiswal
- Center of Genetic Medicine Research, Children's National Health System, Washington, DC.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
20
|
Metabolic Regulation of Sugar Assimilation for Lipid Production in Aspergillus oryzae BCC7051 through Comparative Transcriptome Perspective. BIOLOGY 2021; 10:biology10090885. [PMID: 34571762 PMCID: PMC8467706 DOI: 10.3390/biology10090885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary Oleaginous fungi are a promising candidate to produce microbial lipids as alternative sources for industrial applications. As lipids are intracellular metabolites with dynamic traits, the fungal ability in utilizing carbon sources for biomass and lipid production is significant in terms of production yield and economic feasibility. This study aimed to explore the metabolic regulation in lipogenesis of oleaginous Aspergillus oryzae BCC7051 at the transcriptional level. Through comparative transcriptome analysis, a set of differentially expressed genes (DEGs) between the xylose- and glucose-grown cultures (C5 and C6 cultures) at fast-growing and lipid-accumulating stages were identified and functionally categorized into transporter proteins and cellular processes. Combining with the growth and lipid phenotypes, the transcriptome results pointed to a crucial link between sugar assimilation, energy, lipid, and other metabolisms in A. oryzae for leveraging the metabolic flux from xylose to fatty acid and lipid biosynthesis in render the oleaginous features. This study provides a remarkable insight in guiding strain optimization and bioprocess development using renewable feedstocks from agroindustrial residues. Abstract Microbial lipid production with cost effectiveness is a prerequisite for the oleochemical sector. In this work, genome-wide transcriptional responses on the utilization of xylose and glucose in oleaginous Aspergillus oryzae were studied with relation to growth and lipid phenotypic traits. Comparative analysis of the active growth (t1) and lipid-accumulating (t2) stages showed that the C5 cultures efficiently consumed carbon sources for biomass and lipid production comparable to the C6 cultures. By pairwise comparison, 599 and 917 differentially expressed genes (DEGs) were identified in the t1 and t2 groups, respectively, in which the consensus DEGs were categorized into polysaccharide-degrading enzymes, membrane transports, and cellular processes. A discrimination in transcriptional responses of DEGs set was also found in various metabolic genes, mostly in carbohydrate, amino acid, lipid, cofactors, and vitamin metabolisms. Although central carbohydrate metabolism was shared among the C5 and C6 cultures, the metabolic functions in acetyl-CoA and NADPH generation, and biosynthesis of terpenoid backbone, fatty acid, sterol, and amino acids were allocated for leveraging biomass and lipid production through at least transcriptional control. This study revealed robust metabolic networks in the oleaginicity of A. oryzae governing glucose/xylose flux toward lipid biosynthesis that provides meaningful hints for further process developments of microbial lipid production using cellulosic sugar feedstocks.
Collapse
|
21
|
Yang Q, Hori M. Characterization of Contractile Machinery of Vascular Smooth Muscles in Hypertension. Life (Basel) 2021; 11:life11070702. [PMID: 34357074 PMCID: PMC8304034 DOI: 10.3390/life11070702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Hypertension is a key risk factor for cardiovascular disease and it is a growing public health problem worldwide. The pathophysiological mechanisms of vascular smooth muscle (VSM) contraction contribute to the development of hypertension. Calcium (Ca2+)-dependent and -independent signaling mechanisms regulate the balance of the myosin light chain kinase and myosin light chain phosphatase to induce myosin phosphorylation, which activates VSM contraction to control blood pressure (BP). Here, we discuss the mechanism of the contractile machinery in VSM, especially RhoA/Rho kinase and PKC/CPI-17 of Ca2+ sensitization pathway in hypertension. The two signaling pathways affect BP in physiological and pathophysiological conditions and are highlighted in pulmonary, pregnancy, and salt-sensitive hypertension.
Collapse
Affiliation(s)
- Qunhui Yang
- Correspondence: ; Tel.: +81-3-5841-7940; Fax: +81-3-5841-8183
| | | |
Collapse
|
22
|
Wang H, Ma D, Zhu X, Liu P, Li S, Yu B, Yang H. Nimodipine inhibits intestinal and aortic smooth muscle contraction by regulating Ca 2+-activated Cl - channels. Toxicol Appl Pharmacol 2021; 421:115543. [PMID: 33872679 DOI: 10.1016/j.taap.2021.115543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/18/2022]
Abstract
Nimodipine is a clinically used dihydropyridine L-type calcium channel antagonist that effectively inhibits transmembrane Ca2+ influx following the depolarization of smooth muscle cells, but the detailed effect on smooth muscle contraction is not fully understood. Ca2+-activated Cl- channels (CaCCs) in vascular smooth muscle cells (VSMCs) may regulate vascular contractility. We found that nimodipine can inhibit transmembrane protein 16A (TMEM16A) activity in a concentration-dependent manner by cell-based fluorescence-quenching assay and short-circuit current analysis, with an IC50 value of ~5 μM. Short-circuit current analysis also showed that nimodipine prevented Ca2+-activated Cl- current in both HT-29 cells and mouse colonic epithelia accompanied by significantly decreased cytoplasmic Ca2+ concentrations. In the absence of extracellular Ca2+, nimodipine still exhibited an inhibitory effect on TMEM16A/CaCCs. Additionally, the application of nimodipine to CFTR-expressing FRT cells and mouse colonic mucosa resulted in mild activation of CFTR-mediated Cl- currents. Nimodipine inhibited basolateral CCh-activated K+ channel activity with no effect on Na+/K+-ATPase activity. Evaluation of intestinal smooth muscle contraction showed that nimodipine inhibits intestinal smooth muscle contractility and frequency, with an activity pattern that was similar to that of non-specific inhibitors of CaCCs. In aortic smooth muscle, the expression of TMEM16A in thoracic aorta is higher than that in abdominal aorta, corresponding to stronger maximum contractility in thoracic aorta smooth muscle stimulated by phenylephrine (PE) and Eact. Nimodipine completely inhibited the contraction of aortic smooth muscle stimulated by Eact, and partially inhibited the contraction stimulated by PE. In summary, the results indicate that nimodipine effectively inhibits TMEM16A/CaCCs by reduction transmembrane Ca2+ influx and directly interacting with TMEM16A, explaining the mechanisms of nimodipine relaxation of intestinal and aortic smooth muscle contraction and providing new targets for pharmacological applications.
Collapse
MESH Headings
- Animals
- Anoctamin-1/antagonists & inhibitors
- Anoctamin-1/metabolism
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Calcium Channel Blockers/toxicity
- Calcium Signaling/drug effects
- HT29 Cells
- Humans
- Ileum/drug effects
- Ileum/metabolism
- In Vitro Techniques
- Male
- Mice, Inbred C57BL
- Muscle Contraction/drug effects
- Muscle, Smooth/drug effects
- Muscle, Smooth/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Nimodipine/toxicity
- Rats
- Rats, Sprague-Dawley
- Vasoconstriction/drug effects
- Mice
Collapse
Affiliation(s)
- Hao Wang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian 116000, PR China; Laboratory medical college, Jilin Medical University, Jilin 132013, PR China
| | - Di Ma
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian 116000, PR China
| | - Xiaojuan Zhu
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian 116000, PR China
| | - Panyue Liu
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian 116000, PR China
| | - Shuai Li
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian 116000, PR China
| | - Bo Yu
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian 116000, PR China.
| | - Hong Yang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University, Dalian 116000, PR China.
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Shan W, Hu Y, Ding J, Yang X, Lou J, Du Q, Liao Q, Luo L, Xu J, Xie R. Advances in Ca 2+ modulation of gastrointestinal anion secretion and its dysregulation in digestive disorders (Review). Exp Ther Med 2020; 20:8. [PMID: 32934673 PMCID: PMC7471861 DOI: 10.3892/etm.2020.9136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/22/2020] [Indexed: 11/29/2022] Open
Abstract
Intracellular calcium (Ca2+) is a critical cell signaling component in gastrointestinal (GI) physiology. Cytosolic calcium ([Ca2+]cyt), as a secondary messenger, controls GI epithelial fluid and ion transport, mucus and neuropeptide secretion, as well as synaptic transmission and motility. The key roles of Ca2+ signaling in other types of secretory cell (including those in the airways and salivary glands) are well known. However, its action in GI epithelial secretion and the underlying molecular mechanisms have remained to be fully elucidated. The present review focused on the role of [Ca2+]cyt in GI epithelial anion secretion. Ca2+ signaling regulates the activities of ion channels and transporters involved in GI epithelial ion and fluid transport, including Cl- channels, Ca2+-activated K+ channels, cystic fibrosis (CF) transmembrane conductance regulator and anion/HCO3- exchangers. Previous studies by the current researchers have focused on this field over several years, providing solid evidence that Ca2+ signaling has an important role in the regulation of GI epithelial anion secretion and uncovering underlying molecular mechanisms. The present review is largely based on previous studies by the current researchers and provides an overview of the currently known molecular mechanisms of GI epithelial anion secretion with an emphasis on Ca2+-mediated ion secretion and its dysregulation in GI disorders. In addition, previous studies by the current researchers demonstrated that different regulatory mechanisms are in place for GI epithelial HCO3- and Cl- secretion. An increased understanding of the roles of Ca2+ signaling and its targets in GI anion secretion may lead to the development of novel strategies to inhibit GI diseases, including the enhancement of fluid secretion in CF and protection of the GI mucosa in ulcer diseases.
Collapse
Affiliation(s)
- Weixi Shan
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yanxia Hu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jianhong Ding
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xiaoxu Yang
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jun Lou
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qian Du
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qiushi Liao
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Lihong Luo
- Department of Oncology and Geriatrics, Traditional Chinese Medicine Hospital of Chishui City, Guizhou 564700, P.R. China
| | - Jingyu Xu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Rui Xie
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
26
|
Skitchenko RK, Usoltsev D, Uspenskaya M, Kajava AV, Guskov A. Census of halide-binding sites in protein structures. Bioinformatics 2020; 36:3064-3071. [PMID: 32022861 PMCID: PMC7214031 DOI: 10.1093/bioinformatics/btaa079] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/02/2022] Open
Abstract
Motivation Halides are negatively charged ions of halogens, forming fluorides (F−), chlorides (Cl−), bromides (Br−) and iodides (I−). These anions are quite reactive and interact both specifically and non-specifically with proteins. Despite their ubiquitous presence and important roles in protein function, little is known about the preferences of halides binding to proteins. To address this problem, we performed the analysis of halide–protein interactions, based on the entries in the Protein Data Bank. Results We have compiled a pipeline for the quick analysis of halide-binding sites in proteins using the available software. Our analysis revealed that all of halides are strongly attracted by the guanidinium moiety of arginine side chains, however, there are also certain preferences among halides for other partners. Furthermore, there is a certain preference for coordination numbers in the binding sites, with a correlation between coordination numbers and amino acid composition. This pipeline can be used as a tool for the analysis of specific halide–protein interactions and assist phasing experiments relying on halides as anomalous scatters. Availability and implementation All data described in this article can be reproduced via complied pipeline published at https://github.com/rostkick/Halide_sites/blob/master/README.md. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Dmitrii Usoltsev
- Institute BioEngineering, ITMO University, Saint-Petersburg 197101, Russia
| | - Mayya Uspenskaya
- Institute BioEngineering, ITMO University, Saint-Petersburg 197101, Russia
| | - Andrey V Kajava
- Institute BioEngineering, ITMO University, Saint-Petersburg 197101, Russia.,Centre de Recherche en Biologie cellulaire de Montpellier (CRBM), UMR 5237 CNRS, Universite Montpellier, Montpellier 34293, France
| | - Albert Guskov
- Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Groningen 9747 AG, the Netherlands
| |
Collapse
|
27
|
|
28
|
Abstract
Cl- is the major extracellular (Cl-out) and intracellular (Cl-in) anion whose concentration is actively regulated by multiple transporters. These transporters generate Cl- gradients across the plasma membrane and between the cytoplasm and intracellular organelles. [Cl-]in changes rapidly in response to cell stimulation and influences many physiological functions, as well as cellular and systemic homeostasis. However, less appreciated is the signaling function of Cl-. Cl- interacts with multiple proteins to directly modify their activity. This review highlights the signaling function of Cl- and argues that Cl- is a bona fide signaling ion, a function deserving extensive exploration.
Collapse
Affiliation(s)
- Benjamin P Lüscher
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Laura Vachel
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Ehud Ohana
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
29
|
A network of phosphatidylinositol 4,5-bisphosphate binding sites regulates gating of the Ca 2+-activated Cl - channel ANO1 (TMEM16A). Proc Natl Acad Sci U S A 2019; 116:19952-19962. [PMID: 31515451 DOI: 10.1073/pnas.1904012116] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ANO1 (TMEM16A) is a Ca2+-activated Cl- channel that regulates diverse cellular functions including fluid secretion, neuronal excitability, and smooth muscle contraction. ANO1 is activated by elevation of cytosolic Ca2+ and modulated by phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. Here, we describe a closely concerted experimental and computational study, including electrophysiology, mutagenesis, functional assays, and extended sampling of lipid-protein interactions with molecular dynamics (MD) to characterize PI(4,5)P2 binding modes and sites on ANO1. ANO1 currents in excised inside-out patches activated by 270 nM Ca2+ at +100 mV are increased by exogenous PI(4,5)P2 with an EC50 = 1.24 µM. The effect of PI(4,5)P2 is dependent on membrane voltage and Ca2+ and is explained by a stabilization of the ANO1 Ca2+-bound open state. Unbiased atomistic MD simulations with 1.4 mol% PI(4,5)P2 in a phosphatidylcholine bilayer identified 8 binding sites with significant probability of binding PI(4,5)P2 Three of these sites captured 85% of all ANO1-PI(4,5)P2 interactions. Mutagenesis of basic amino acids near the membrane-cytosol interface found 3 regions of ANO1 critical for PI(4,5)P2 regulation that correspond to the same 3 sites identified by MD. PI(4,5)P2 is stabilized by hydrogen bonding between amino acid side chains and phosphate/hydroxyl groups on PI(4,5)P2 Binding of PI(4,5)P2 alters the position of the cytoplasmic extension of TM6, which plays a crucial role in ANO1 channel gating, and increases the accessibility of the inner vestibule to Cl- ions. We propose a model consisting of a network of 3 PI(4,5)P2 binding sites at the cytoplasmic face of the membrane allosterically regulating ANO1 channel gating.
Collapse
|
30
|
Dayal A, Ng SFJ, Grabner M. Ca 2+-activated Cl - channel TMEM16A/ANO1 identified in zebrafish skeletal muscle is crucial for action potential acceleration. Nat Commun 2019; 10:115. [PMID: 30631052 PMCID: PMC6328546 DOI: 10.1038/s41467-018-07918-z] [Citation(s) in RCA: 24] [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: 02/07/2018] [Accepted: 12/03/2018] [Indexed: 01/16/2023] Open
Abstract
The Ca2+-activated Cl- channel (CaCC) TMEM16A/Anoctamin 1 (ANO1) is expressed in gastrointestinal epithelia and smooth muscle cells where it mediates secretion and intestinal motility. However, ANO1 Cl- conductance has never been reported to play a role in skeletal muscle. Here we show that ANO1 is robustly expressed in the highly evolved skeletal musculature of the euteleost species zebrafish. We characterised ANO1 as bonafide CaCC which is activated close to maximum by Ca2+ ions released from the SR during excitation-contraction (EC) coupling. Consequently, our study addressed the question about the physiological advantage of implementation of ANO1 into the euteleost skeletal-muscle EC coupling machinery. Our results reveal that Cl- influx through ANO1 plays an essential role in restricting the width of skeletal-muscle action potentials (APs) by accelerating the repolarisation phase. Resulting slimmer APs enable higher AP-frequencies and apparently tighter controlled, faster and stronger muscle contractions, crucial for high speed movements.
Collapse
Affiliation(s)
- Anamika Dayal
- Department of Medical Genetics, Molecular and Clinical Pharmacology, Division of Biochemical Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1, A-6020, Innsbruck, Austria.
| | - Shu Fun J Ng
- Department of Medical Genetics, Molecular and Clinical Pharmacology, Division of Biochemical Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1, A-6020, Innsbruck, Austria
| | - Manfred Grabner
- Department of Medical Genetics, Molecular and Clinical Pharmacology, Division of Biochemical Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1, A-6020, Innsbruck, Austria.
| |
Collapse
|
31
|
Lee YH, Yi GS. Prediction of Novel Anoctamin1 (ANO1) Inhibitors Using 3D-QSAR Pharmacophore Modeling and Molecular Docking. Int J Mol Sci 2018; 19:ijms19103204. [PMID: 30336555 PMCID: PMC6214110 DOI: 10.3390/ijms19103204] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 11/24/2022] Open
Abstract
Recently, anoctamin1 (ANO1), a calcium-activated chloride channel, has been considered an important drug target, due to its involvement in various physiological functions, as well as its possibility for treatment of cancer, pain, diarrhea, hypertension, and asthma. Although several ANO1 inhibitors have been discovered by high-throughput screening, a discovery of new ANO1 inhibitors is still in the early phase, in terms of their potency and specificity. Moreover, there is no computational model to be able to identify a novel lead candidate of ANO1 inhibitor. Therefore, three-dimensional quantitative structure-activity relationship (3D-QSAR) pharmacophore modeling approach was employed for identifying the essential chemical features to be required in the inhibition of ANO1. The pharmacophore hypothesis 2 (Hypo2) was selected as the best model based on the highest correlation coefficient of prediction on the test set (0.909). Hypo2 comprised a hydrogen bond acceptor, a hydrogen bond donor, a hydrophobic, and a ring aromatic feature with good statistics of the total cost (73.604), the correlation coefficient of the training set (0.969), and the root-mean-square deviation (RMSD) value (0.946). Hypo2 was well assessed by the test set, Fischer randomization, and leave-one-out methods. Virtual screening of the ZINC database with Hypo2 retrieved the 580 drug-like candidates with good potency and ADMET properties. Finally, two compounds were selected as novel lead candidates of ANO1 inhibitor, based on the molecular docking score and the interaction analysis. In this study, the best pharmacophore model, Hypo2, with notable predictive ability was successfully generated, and two potential leads of ANO1 inhibitors were identified. We believe that these compounds and the 3D-QSAR pharmacophore model could contribute to discovering novel and potent ANO1 inhibitors in the future.
Collapse
Affiliation(s)
- Yoon Hyeok Lee
- Department of Bio and Brain engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea.
| | - Gwan-Su Yi
- Department of Bio and Brain engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea.
| |
Collapse
|
32
|
Gomes Castro AJ, Cazarolli LH, Bretanha LC, Sulis PM, Rey Padilla DP, Aragón Novoa DM, Dambrós BF, Pizzolatti MG, Mena Barreto Silva FR. The potent insulin secretagogue effect of betulinic acid is mediated by potassium and chloride channels. Arch Biochem Biophys 2018; 648:20-26. [PMID: 29704483 DOI: 10.1016/j.abb.2018.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 12/13/2022]
Abstract
Betulinic acid (BA) has been described as an insulin secretagogue which may explain its potent antihyperglycemic effect; however, the exact role of BA as an insulinogenic agent is not clear. The aim of this study was to investigate the mechanism of BA on calcium influx and static insulin secretion in pancreatic islets isolated from euglycemic rats. We found that BA triggers calcium influx by a mechanism dependent on ATP-dependent potassium channels and L-type voltage-dependent calcium channels. Additionally, the voltage-dependent and calcium-dependent chloride channels are also involved in the mechanism of BA, probably due to an indirect stimulation of calcium entry and increased intracellular calcium. Additionally, the downstream activation of PKC, which is necessary for the effect of BA on calcium influx, is involved in the full stimulatory response of the triterpene. BA stimulated the static secretion of insulin in pancreatic islets, indicating that the abrupt calcium influx may be a key step in its secretagogue effect. As such, BA stimulates insulin secretion through the activation of electrophysiological mechanisms, such as the closure of potassium channels and opening of calcium and chloride channels, inducing cellular depolarization associated with metabolic-biochemical effects, in turn activating PKC and ensuring the secretion of insulin.
Collapse
Affiliation(s)
- Allisson Jhonatan Gomes Castro
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Luisa Helena Cazarolli
- Universidade Federal da Fronteira Sul, Campus Universitário Laranjeiras do Sul, Laranjeiras do Sul, PR, Brazil
| | - Lizandra C Bretanha
- Departamento de Química, Centro de Ciências Físicas e Matemáticas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Paola Miranda Sulis
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Diana Patricia Rey Padilla
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Universidad Nacional de Colombia, Departamento de Farmácia, Facultad de Ciencias, Bogotá, Colombia
| | | | - Betina Fernanda Dambrós
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Moacir G Pizzolatti
- Departamento de Química, Centro de Ciências Físicas e Matemáticas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | |
Collapse
|
33
|
Calcium-regulated chloride channel anoctamin-1 is present in the suprachiasmatic nuclei of rats. Neuroreport 2018; 29:334-339. [PMID: 29309309 DOI: 10.1097/wnr.0000000000000967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium-regulated chloride channel (CaCC) anoctamin-1 has been recently identified in neurons. In neurons, which express the Na-K-2Cl cotransporter, activation of CaCCs increases firing frequency, by reversion between the Cl equilibrium potential and the membrane resting potential, leading to membrane depolarization by Cl extrusion from the cell. Although there are no reports of CaCCs present in the suprachiasmatic nuclei (SCN), the fact that Na-K-2Cl cotransporter is present in SCN neurons, where it has been shown to be involved in the excitatory effects of γ-aminobutyric acid, together with the increase of neuronal firing rate induced by release of intracellular Ca after administration of 100 nM ryanodine, leads us to determine whether CaCCs are present in the SCN. Immunohistochemistry and western blots show the expression of the CaCCs anoctamin-1 protein. Quantitative PCR demonstrated the expression of anoctamin-1 mRNA in the SCN. These results clearly indicate the presence of CaCC in SCN of rats.
Collapse
|
34
|
Ma MM, Gao M, Guo KM, Wang M, Li XY, Zeng XL, Sun L, Lv XF, Du YH, Wang GL, Zhou JG, Guan YY. TMEM16A Contributes to Endothelial Dysfunction by Facilitating Nox2 NADPH Oxidase-Derived Reactive Oxygen Species Generation in Hypertension. Hypertension 2017; 69:892-901. [PMID: 28320851 DOI: 10.1161/hypertensionaha.116.08874] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 12/28/2016] [Accepted: 02/16/2017] [Indexed: 01/09/2023]
Abstract
Ca2+-activated Cl- channels play a crucial role in various physiological processes. However, the role of TMEM16A in vascular endothelial dysfunction during hypertension is unclear. In this study, we investigated the specific involvement of TMEM16A in regulating endothelial function and blood pressure and the underlying mechanism. Reverse transcription-polymerase chain reaction, Western blotting, coimmunoprecipitation, confocal imaging, patch-clamp recordings, and TMEM16A endothelial-specific transgenic and knockout mice were used. We found that TMEM16A was expressed abundantly and functioned as a Ca2+-activated Cl- channel in endothelial cells. Angiotensin II induced endothelial dysfunction with an increase in TMEM16A expression. The knockout of endothelial-specific TMEM16A significantly lowered the blood pressure and ameliorated endothelial dysfunction in angiotensin II-induced hypertension, whereas the overexpression of endothelial-specific TMEM16A resulted in the opposite effects. These results were related to the increased reactive oxygen species production, Nox2-containing NADPH oxidase activation, and Nox2 and p22phox protein expression that were facilitated by TMEM16A on angiotensin II-induced hypertensive challenge. Moreover, TMEM16A directly bound with Nox2 and reduced the degradation of Nox2 through the proteasome-dependent degradation pathway. Therefore, TMEM16A is a positive regulator of endothelial reactive oxygen species generation via Nox2-containing NADPH oxidase, which induces endothelial dysfunction and hypertension. Modification of TMEM16A may be a novel therapeutic strategy for endothelial dysfunction-associated diseases.
Collapse
Affiliation(s)
- Ming-Ming Ma
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.).
| | - Min Gao
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Kai-Min Guo
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Mi Wang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Xiang-Yu Li
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Xue-Lin Zeng
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Lu Sun
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Xiao-Fei Lv
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Yan-Hua Du
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Guan-Lei Wang
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Jia-Guo Zhou
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| | - Yong-Yuan Guan
- From the Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (M.-M.M., X.-Y.L., X.-L.Z., L.S., X.-F.L., Y.-H.D., G.-L.W., J.-G.Z., Y.-Y.G.); Department of Pharmacy, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (M.G.); Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China (K.-M.G.); and Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (M.W.)
| |
Collapse
|
35
|
M De la Fuente I, Malaina I, Pérez-Samartín A, Boyano MD, Pérez-Yarza G, Bringas C, Villarroel Á, Fedetz M, Arellano R, Cortes JM, Martínez L. Dynamic properties of calcium-activated chloride currents in Xenopus laevis oocytes. Sci Rep 2017; 7:41791. [PMID: 28198817 PMCID: PMC5304176 DOI: 10.1038/srep41791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/30/2016] [Indexed: 11/18/2022] Open
Abstract
Chloride is the most abundant permeable anion in the cell, and numerous studies in the last two decades highlight the great importance and broad physiological role of chloride currents mediated anion transport. They participate in a multiplicity of key processes, as for instance, the regulation of electrical excitability, apoptosis, cell cycle, epithelial secretion and neuronal excitability. In addition, dysfunction of Cl− channels is involved in a variety of human diseases such as epilepsy, osteoporosis and different cancer types. Historically, chloride channels have been of less interest than the cation channels. In fact, there seems to be practically no quantitative studies of the dynamics of chloride currents. Here, for the first time, we have quantitatively studied experimental calcium-activated chloride fluxes belonging to Xenopus laevis oocytes, and the main results show that the experimental Cl− currents present an informational structure characterized by highly organized data sequences, long-term memory properties and inherent “crossover” dynamics in which persistent correlations arise at short time intervals, while anti-persistent behaviors become dominant in long time intervals. Our work sheds some light on the understanding of the informational properties of ion currents, a key element to elucidate the physiological functional coupling with the integrative dynamics of metabolic processes.
Collapse
Affiliation(s)
- Ildefonso M De la Fuente
- Department of Nutrition, CEBAS-CSIC Institute, Espinardo University Campus, Murcia, Spain.,Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Iker Malaina
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Alberto Pérez-Samartín
- Department of Neurosciences, Faculty of Medicine and Dentistry, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - María Dolores Boyano
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Gorka Pérez-Yarza
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Carlos Bringas
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Álvaro Villarroel
- Biophysics Unit, CSIC, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - María Fedetz
- Department of Biochemistry and Pharmacology, Institute of Parasitology and Biomedicine "López-Neyra", CSIC, Granada, Spain
| | - Rogelio Arellano
- Laboratory of Cellular Neurophysiology, Neurobiology Institute, UNAM, Querétaro, México
| | - Jesus M Cortes
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country, UPV/EHU, Leioa, Spain.,BioCruces Health Research Institute, Cruces University Hospital, Barakaldo, Spain.,IKERBASQUE: The Basque Foundation for Science, Bilbao, Spain
| | - Luis Martínez
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa, Spain
| |
Collapse
|
36
|
Fujii S, Suzuki K, Kawamoto A, Ishibashi F, Nakata T, Murano T, Ito G, Shimizu H, Mizutani T, Oshima S, Tsuchiya K, Nakamura T, Araki A, Ohtsuka K, Okamoto R, Watanabe M. PGE 2 is a direct and robust mediator of anion/fluid secretion by human intestinal epithelial cells. Sci Rep 2016; 6:36795. [PMID: 27827428 PMCID: PMC5101536 DOI: 10.1038/srep36795] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 10/21/2016] [Indexed: 12/13/2022] Open
Abstract
Intestinal epithelial cells (IECs) play an indispensable role in maintaining body fluid balance partly through their ability to regulate anion/fluid secretion. Yet in various inflammatory gastrointestinal diseases, over-secretion of anions results in symptoms such as severe diarrhoea. Endogenous mediators, such as vasoactive intestinal peptide or prostaglandin E2 (PGE2), regulate intestinal anion/fluid secretion, but their direct effect on purified human IECs has never been described in detail. Based on a previously described intestinal organoid swelling model, we established a 3D-scanner-assisted quantification method to evaluate the anion/fluid secretory response of cultured human IECs. Among various endogenous secretagogues, we found that PGE2 had the lowest EC50 value with regard to the induction of swelling of the jejunal and colonic organoids. This PGE2-mediated swelling response was dependent on environmental Cl- concentrations as well as on several channels and transporters as shown by a series of chemical inhibitor studies. The concomitant presence of various inflammatory cytokines with PGE2 failed to modulate the PGE2-mediated organoid swelling response. Therefore, the present study features PGE2 as a direct and robust mediator of anion/fluid secretion by IECs in the human intestine.
Collapse
Affiliation(s)
- Satoru Fujii
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohei Suzuki
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ami Kawamoto
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fumiaki Ishibashi
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toru Nakata
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsuro Murano
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Go Ito
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Hiromichi Shimizu
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeru Oshima
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kiichiro Tsuchiya
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsuya Nakamura
- Department of Advanced Therapeutics in GI Diseases, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiro Araki
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuo Ohtsuka
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,Center for Stem Cell and Regenerative Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
37
|
Senatore A, Raiss H, Le P. Physiology and Evolution of Voltage-Gated Calcium Channels in Early Diverging Animal Phyla: Cnidaria, Placozoa, Porifera and Ctenophora. Front Physiol 2016; 7:481. [PMID: 27867359 PMCID: PMC5095125 DOI: 10.3389/fphys.2016.00481] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 10/07/2016] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium (Cav) channels serve dual roles in the cell, where they can both depolarize the membrane potential for electrical excitability, and activate transient cytoplasmic Ca2+ signals. In animals, Cav channels play crucial roles including driving muscle contraction (excitation-contraction coupling), gene expression (excitation-transcription coupling), pre-synaptic and neuroendocrine exocytosis (excitation-secretion coupling), regulation of flagellar/ciliary beating, and regulation of cellular excitability, either directly or through modulation of other Ca2+-sensitive ion channels. In recent years, genome sequencing has provided significant insights into the molecular evolution of Cav channels. Furthermore, expanded gene datasets have permitted improved inference of the species phylogeny at the base of Metazoa, providing clearer insights into the evolution of complex animal traits which involve Cav channels, including the nervous system. For the various types of metazoan Cav channels, key properties that determine their cellular contribution include: Ion selectivity, pore gating, and, importantly, cytoplasmic protein-protein interactions that direct sub-cellular localization and functional complexing. It is unclear when these defining features, many of which are essential for nervous system function, evolved. In this review, we highlight some experimental observations that implicate Cav channels in the physiology and behavior of the most early-diverging animals from the phyla Cnidaria, Placozoa, Porifera, and Ctenophora. Given our limited understanding of the molecular biology of Cav channels in these basal animal lineages, we infer insights from better-studied vertebrate and invertebrate animals. We also highlight some apparently conserved cellular functions of Cav channels, which might have emerged very early on during metazoan evolution, or perhaps predated it.
Collapse
Affiliation(s)
- Adriano Senatore
- Department of Biology, University of Toronto Mississauga Mississauga, ON, Canada
| | - Hamad Raiss
- Department of Biology, University of Toronto Mississauga Mississauga, ON, Canada
| | - Phuong Le
- Department of Biology, University of Toronto Mississauga Mississauga, ON, Canada
| |
Collapse
|
38
|
Abstract
Fluid shear stress is an important environmental cue that governs vascular physiology and pathology, but the molecular mechanisms that mediate endothelial responses to flow are only partially understood. Gating of ion channels by flow is one mechanism that may underlie many of the known responses. Here, we review the literature on endothelial ion channels whose activity is modulated by flow with an eye toward identifying important questions for future research.
Collapse
Affiliation(s)
- Kristin A Gerhold
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and
| | - Martin A Schwartz
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, Connecticut
| |
Collapse
|
39
|
Seo Y, Lee HK, Park J, Jeon DK, Jo S, Jo M, Namkung W. Ani9, A Novel Potent Small-Molecule ANO1 Inhibitor with Negligible Effect on ANO2. PLoS One 2016; 11:e0155771. [PMID: 27219012 PMCID: PMC4878759 DOI: 10.1371/journal.pone.0155771] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/03/2016] [Indexed: 12/13/2022] Open
Abstract
Anoctamin1 (ANO1)/transmembrane protein 16A (TMEM16A), a calcium-activated chloride channel (CaCC), is involved in many physiological functions such as fluid secretion, smooth muscle contraction, nociception and cancer progression. To date, only a few ANO1 inhibitors have been described, and these have low potency and selectivity for ANO1. Here, we performed a high-throughput screening to identify highly potent and selective small molecule inhibitors of ANO1. Three novel ANO1 inhibitors were discovered from screening of 54,400 synthetic small molecules, and they were found to fully block ANO1 channel activity with an IC50 < 3 μM. Electrophysiological analysis revealed that the most potent inhibitor, 2-(4-chloro-2-methylphenoxy)-N-[(2-methoxyphenyl)methylideneamino]-acetamide (Ani9), completely inhibited ANO1 chloride current with submicromolar potency. Notably, unlike previous small-molecule ANO1 inhibitors identified to date, Ani9 displayed high selectivity for ANO1 as compared to ANO2, which shares a high amino acid homology to ANO1. In addition, Ani9 did not affect the intracellular calcium signaling and CFTR chloride channel activity. Our results suggest that Ani9 may be a useful pharmacological tool for studying ANO1 and a potential development candidate for drug therapy of cancer, hypertension, pain, diarrhea and asthma.
Collapse
Affiliation(s)
- Yohan Seo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
| | - Ho K. Lee
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
| | - Jinhong Park
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
| | - Dong-kyu Jeon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
| | - Sungwoo Jo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
| | - Minjae Jo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
| | - Wan Namkung
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
- * E-mail:
| |
Collapse
|
40
|
Ohya S, Kito H, Hatano N, Muraki K. Recent advances in therapeutic strategies that focus on the regulation of ion channel expression. Pharmacol Ther 2016; 160:11-43. [PMID: 26896566 DOI: 10.1016/j.pharmthera.2016.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A number of different ion channel types are involved in cell signaling networks, and homeostatic regulatory mechanisms contribute to the control of ion channel expression. Profiling of global gene expression using microarray technology has recently provided novel insights into the molecular mechanisms underlying the homeostatic and pathological control of ion channel expression. It has demonstrated that the dysregulation of ion channel expression is associated with the pathogenesis of neural, cardiovascular, and immune diseases as well as cancers. In addition to the transcriptional, translational, and post-translational regulation of ion channels, potentially important evidence on the mechanisms controlling ion channel expression has recently been accumulated. The regulation of alternative pre-mRNA splicing is therefore a novel therapeutic strategy for the treatment of dominant-negative splicing disorders. Epigenetic modification plays a key role in various pathological conditions through the regulation of pluripotency genes. Inhibitors of pre-mRNA splicing and histone deacetyalase/methyltransferase have potential as potent therapeutic drugs for cancers and autoimmune and inflammatory diseases. Moreover, membrane-anchoring proteins, lysosomal and proteasomal degradation-related molecules, auxiliary subunits, and pharmacological agents alter the protein folding, membrane trafficking, and post-translational modifications of ion channels, and are linked to expression-defect channelopathies. In this review, we focused on recent insights into the transcriptional, spliceosomal, epigenetic, and proteasomal regulation of ion channel expression: Ca(2+) channels (TRPC/TRPV/TRPM/TRPA/Orai), K(+) channels (voltage-gated, KV/Ca(2+)-activated, KCa/two-pore domain, K2P/inward-rectifier, Kir), and Ca(2+)-activated Cl(-) channels (TMEM16A/TMEM16B). Furthermore, this review highlights expression of these ion channels in expression-defect channelopathies.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya 464-8650, Japan.
| |
Collapse
|
41
|
Yang T, Colecraft HM. Calmodulin regulation of TMEM16A and 16B Ca(2+)-activated chloride channels. Channels (Austin) 2015; 10:38-44. [PMID: 26083059 DOI: 10.1080/19336950.2015.1058455] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ca(2+)-activated chloride channels encoded by TMEM16A and 16B are important for regulating epithelial mucus secretion, cardiac and neuronal excitability, smooth muscle contraction, olfactory transduction, and cell proliferation. Whether and how the ubiquitous Ca(2+) sensor calmodulin (CaM) regulates the activity of TMEM16A and 16B channels has been controversial and the subject of an ongoing debate. Recently, using a bioengineering approach termed ChIMP (Channel Inactivation induced by Membrane-tethering of an associated Protein) we argued that Ca(2+)-free CaM (apoCaM) is pre-associated with functioning TMEM16A and 16B channel complexes in live cells. Further, the pre-associated apoCaM mediates Ca(2+)-dependent sensitization of activation (CDSA) and Ca(2+)-dependent inactivation (CDI) of some TMEM16A splice variants. In this review, we discuss these findings in the context of previous and recent results relating to Ca(2+)-dependent regulation of TMEM16A/16B channels and the putative role of CaM. We further discuss potential future directions for these nascent ideas on apoCaM regulation of TMEM16A/16B channels, noting that such future efforts will benefit greatly from the pioneering work of Dr. David T. Yue and colleagues on CaM regulation of voltage-dependent calcium channels.
Collapse
Affiliation(s)
- Tingting Yang
- a Department of Physiology and Cellular Biophysics ; Columbia University; College of Physicians and Surgeons ; New York , NY USA
| | - Henry M Colecraft
- a Department of Physiology and Cellular Biophysics ; Columbia University; College of Physicians and Surgeons ; New York , NY USA
| |
Collapse
|
42
|
Leblanc N, Forrest AS, Ayon RJ, Wiwchar M, Angermann JE, Pritchard HAT, Singer CA, Valencik ML, Britton F, Greenwood IA. Molecular and functional significance of Ca(2+)-activated Cl(-) channels in pulmonary arterial smooth muscle. Pulm Circ 2015; 5:244-68. [PMID: 26064450 DOI: 10.1086/680189] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022] Open
Abstract
Increased peripheral resistance of small distal pulmonary arteries is a hallmark signature of pulmonary hypertension (PH) and is believed to be the consequence of enhanced vasoconstriction to agonists, thickening of the arterial wall due to remodeling, and increased thrombosis. The elevation in arterial tone in PH is attributable, at least in part, to smooth muscle cells of PH patients being more depolarized and displaying higher intracellular Ca(2+) levels than cells from normal subjects. It is now clear that downregulation of voltage-dependent K(+) channels (e.g., Kv1.5) and increased expression and activity of voltage-dependent (Cav1.2) and voltage-independent (e.g., canonical and vanilloid transient receptor potential [TRPC and TRPV]) Ca(2+) channels play an important role in the functional remodeling of pulmonary arteries in PH. This review focuses on an anion-permeable channel that is now considered a novel excitatory mechanism in the systemic and pulmonary circulations. It is permeable to Cl(-) and is activated by a rise in intracellular Ca(2+) concentration (Ca(2+)-activated Cl(-) channel, or CaCC). The first section outlines the biophysical and pharmacological properties of the channel and ends with a description of the molecular candidate genes postulated to encode for CaCCs, with particular emphasis on the bestrophin and the newly discovered TMEM16 and anoctamin families of genes. The second section provides a review of the various sources of Ca(2+) activating CaCCs, which include stimulation by mobilization from intracellular Ca(2+) stores and Ca(2+) entry through voltage-dependent and voltage-independent Ca(2+) channels. The third and final section summarizes recent findings that suggest a potentially important role for CaCCs and the gene TMEM16A in PH.
Collapse
Affiliation(s)
- Normand Leblanc
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Abigail S Forrest
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Ramon J Ayon
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Michael Wiwchar
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Jeff E Angermann
- School of Community Health Sciences, University of Nevada, Reno, Nevada, USA
| | - Harry A T Pritchard
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| | - Cherie A Singer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Maria L Valencik
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Fiona Britton
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Iain A Greenwood
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| |
Collapse
|
43
|
Li Q, Cai H. ANO1 taking center stage: blood pressure regulation in SHRs. J Mol Cell Cardiol 2015; 82:216-7. [PMID: 25817885 DOI: 10.1016/j.yjmcc.2015.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 03/18/2015] [Indexed: 11/26/2022]
Affiliation(s)
- Qiang Li
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA 90095, USA
| | - Hua Cai
- Divisions of Molecular Medicine and Cardiology, Departments of Anesthesiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Cherian OL, Menini A, Boccaccio A. Multiple effects of anthracene-9-carboxylic acid on the TMEM16B/anoctamin2 calcium-activated chloride channel. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1005-13. [PMID: 25620774 DOI: 10.1016/j.bbamem.2015.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/04/2015] [Accepted: 01/13/2015] [Indexed: 10/24/2022]
Abstract
Ca(2+)-activated Cl(-) currents (CaCCs) play important roles in many physiological processes. Recent studies have shown that TMEM16A/anoctamin1 and TMEM16B/anoctamin2 constitute CaCCs in several cell types. Here we have investigated for the first time the extracellular effects of the Cl(-) channel blocker anthracene-9-carboxylic acid (A9C) and of its non-charged analogue anthracene-9-methanol (A9M) on TMEM16B expressed in HEK 293T cells, using the whole-cell patch-clamp technique. A9C caused a voltage-dependent block of outward currents and inhibited a larger fraction of the current as depolarization increased, whereas the non-charged A9M produced a small, not voltage dependent block of outward currents. A similar voltage-dependent block by A9C was measured both when TMEM16B was activated by 1.5 and 13μM Ca(2+). However, in the presence of 1.5μM Ca(2+) (but not in 13μM Ca(2+)), A9C also induced a strong potentiation of tail currents measured at -100mV after depolarizing voltages, as well as a prolongation of the deactivation kinetics. On the contrary, A9M did not produce potentiation of tail currents, showing that the negative charge is required for potentiation. Our results provide the first evidence that A9C has multiple effects on TMEM16B and that the negative charge of A9C is necessary both for voltage-dependent block and for potentiation. Future studies are required to identify the molecular mechanisms underlying these complex effects of A9C on TMEM16B. Understanding these mechanisms will contribute to the elucidation of the structure and functional properties of TMEM16B channels.
Collapse
Affiliation(s)
- O Lijo Cherian
- Neurobiology Group, SISSA, International School for Advanced Studies, Via Bonomea 265, 34136 Trieste, Italy
| | - Anna Menini
- Neurobiology Group, SISSA, International School for Advanced Studies, Via Bonomea 265, 34136 Trieste, Italy
| | - Anna Boccaccio
- Istituto di Biofisica, CNR, Via De Marini 6, 16149 Genova, Italy.
| |
Collapse
|
45
|
Wang K, Chen C, Ma J, Lao J, Pang Y. Contribution of calcium-activated chloride channel to elevated pulmonary artery pressure in pulmonary arterial hypertension induced by high pulmonary blood flow. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:146-154. [PMID: 25755701 PMCID: PMC4348827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 12/22/2014] [Indexed: 06/04/2023]
Abstract
The correlation between calcium-activated chloride channel (CaCC) and pulmonary arterial hypertension (PAH) induced by high pulmonary blood flow remains uncertain. In this study, we investigated the possible role and effects of CaCC in this disease. Sixty rats were randomly assigned to normal, sham, and shunt groups. Rats in the shunt group underwent abdominal aorta and inferior vena cava shunt surgery. The pulmonary artery pressure was measured by catheterization. Pathological changes, right ventricle hypertrophy index (RVHI), arterial wall area/vessel area (W/V), and arterial wall thickness/vessel external diameter (T/D) were analyzed by optical microscopy. Electrophysiological characteristics of pulmonary arterial smooth muscle cells (PASMCs) were investigated using patch clamp technology. After 11 weeks of shunting, PAH and pulmonary vascular structural remodeling (PVSR) developed, accompanied by increased pulmonary pressure and pathological interstitial pulmonary changes. Compared with normal and sham groups, pulmonary artery pressure, RVHI, W/V, and T/D of the shunt group rats increased significantly. Electrophysiological results showed primary CaCC characteristics. Compared with normal and sham groups, membrane capacitance and current density of PASMCs in the shunt group increased significantly, which were subsequently attenuated following chloride channel blocker niflumic acid (NFA) treatment. To conclude, CaCC contributed to PAH induced by high pulmonary blood flow and may represent a potential target for treatment of PAH.
Collapse
Affiliation(s)
- Kai Wang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University Nanning, China
| | - Chuansi Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University Nanning, China
| | - Jianfa Ma
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University Nanning, China
| | - Jinquan Lao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University Nanning, China
| | - Yusheng Pang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University Nanning, China
| |
Collapse
|
46
|
Preassociated apocalmodulin mediates Ca2+-dependent sensitization of activation and inactivation of TMEM16A/16B Ca2+-gated Cl- channels. Proc Natl Acad Sci U S A 2014; 111:18213-8. [PMID: 25489088 DOI: 10.1073/pnas.1420984111] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ca(2+)-activated chloride currents carried via transmembrane proteins TMEM16A and TMEM16B regulate diverse processes including mucus secretion, neuronal excitability, smooth muscle contraction, olfactory signal transduction, and cell proliferation. Understanding how TMEM16A/16B are regulated by Ca(2+) is critical for defining their (patho)/physiological roles and for rationally targeting them therapeutically. Here, using a bioengineering approach--channel inactivation induced by membrane-tethering of an associated protein (ChIMP)--we discovered that Ca(2+)-free calmodulin (apoCaM) is preassociated with TMEM16A/16B channel complexes. The resident apoCaM mediates two distinct Ca(2+)-dependent effects on TMEM16A, as revealed by expression of dominant-negative CaM1234. These effects are Ca(2+)-dependent sensitization of activation (CDSA) and Ca(2+)-dependent inactivation (CDI). CDI and CDSA are independently mediated by the N and C lobes of CaM, respectively. TMEM16A alternative splicing provides a mechanism for tuning apoCaM effects. Channels lacking splice segment b selectively lost CDI, and segment a is necessary for apoCaM preassociation with TMEM16A. The results reveal multidimensional regulation of TMEM16A/16B by preassociated apoCaM and introduce ChIMP as a versatile tool to probe the macromolecular complex and function of Ca(2+)-activated chloride channels.
Collapse
|
47
|
Salzer I, Gafar H, Gindl V, Mahlknecht P, Drobny H, Boehm S. Excitation of rat sympathetic neurons via M1 muscarinic receptors independently of Kv7 channels. Pflugers Arch 2014; 466:2289-303. [PMID: 24668449 PMCID: PMC4233321 DOI: 10.1007/s00424-014-1487-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 01/14/2023]
Abstract
The slow cholinergic transmission in autonomic ganglia is known to be mediated by an inhibition of Kv7 channels via M1 muscarinic acetylcholine receptors. However, in the present experiments using primary cultures of rat superior cervical ganglion neurons, the extent of depolarisation caused by the M1 receptor agonist oxotremorine M did not correlate with the extent of Kv7 channel inhibition in the very same neuron. This observation triggered a search for additional mechanisms. As the activation of M1 receptors leads to a boost in protein kinase C (PKC) activity in sympathetic neurons, various PKC enzymes were inhibited by different means. Interference with classical PKC isoforms led to reductions in depolarisations and in noradrenaline release elicited by oxotremorine M, but left the Kv7 channel inhibition by the muscarinic agonist unchanged. M1 receptor-induced depolarisations were also altered when extra- or intracellular Cl(-) concentrations were changed, as were depolarising responses to γ-aminobutyric acid. Depolarisations and noradrenaline release triggered by oxotremorine M were reduced by the non-selective Cl(-) channel blockers 4-acetamido-4'-isothiocyanato-stilbene-2,2'-disulfonic acid and niflumic acid. Oxotremorine M induced slowly rising inward currents at negative membrane potentials that were blocked by inhibitors of Ca(2+)-activated Cl(-) and TMEM16A channels and attenuated by PKC inhibitors. These channel blockers also reduced oxotremorine M-evoked noradrenaline release. Together, these results reveal that slow cholinergic excitation of sympathetic neurons involves the activation of classical PKCs and of Ca(2+)-activated Cl(-) channels in addition to the well-known inhibition of Kv7 channels.
Collapse
Affiliation(s)
- Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria
| | - Hend Gafar
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria
| | - Viola Gindl
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria
| | - Peter Mahlknecht
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria
| | - Helmut Drobny
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria
| | - Stefan Boehm
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1090 Vienna, Austria
| |
Collapse
|
48
|
Betto G, Cherian OL, Pifferi S, Cenedese V, Boccaccio A, Menini A. Interactions between permeation and gating in the TMEM16B/anoctamin2 calcium-activated chloride channel. ACTA ACUST UNITED AC 2014; 143:703-18. [PMID: 24863931 PMCID: PMC4035747 DOI: 10.1085/jgp.201411182] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Extracellular anions more permeant than Cl− modulate TMEM16B gating to promote channel opening, whereas less permeant anions favor channel closure. At least two members of the TMEM16/anoctamin family, TMEM16A (also known as anoctamin1) and TMEM16B (also known as anoctamin2), encode Ca2+-activated Cl− channels (CaCCs), which are found in various cell types and mediate numerous physiological functions. Here, we used whole-cell and excised inside-out patch-clamp to investigate the relationship between anion permeation and gating, two processes typically viewed as independent, in TMEM16B expressed in HEK 293T cells. The permeability ratio sequence determined by substituting Cl− with other anions (PX/PCl) was SCN− > I− > NO3− > Br− > Cl− > F− > gluconate. When external Cl− was substituted with other anions, TMEM16B activation and deactivation kinetics at 0.5 µM Ca2+ were modified according to the sequence of permeability ratios, with anions more permeant than Cl− slowing both activation and deactivation and anions less permeant than Cl− accelerating them. Moreover, replacement of external Cl− with gluconate, or sucrose, shifted the voltage dependence of steady-state activation (G-V relation) to more positive potentials, whereas substitution of extracellular or intracellular Cl− with SCN− shifted G-V to more negative potentials. Dose–response relationships for Ca2+ in the presence of different extracellular anions indicated that the apparent affinity for Ca2+ at +100 mV increased with increasing permeability ratio. The apparent affinity for Ca2+ in the presence of intracellular SCN− also increased compared with that in Cl−. Our results provide the first evidence that TMEM16B gating is modulated by permeant anions and provide the basis for future studies aimed at identifying the molecular determinants of TMEM16B ion selectivity and gating.
Collapse
Affiliation(s)
- Giulia Betto
- Neurobiology Group, International School for Advanced Studies (SISSA), 34136 Trieste, Italy
| | - O Lijo Cherian
- Neurobiology Group, International School for Advanced Studies (SISSA), 34136 Trieste, Italy
| | - Simone Pifferi
- Neurobiology Group, International School for Advanced Studies (SISSA), 34136 Trieste, Italy
| | - Valentina Cenedese
- Neurobiology Group, International School for Advanced Studies (SISSA), 34136 Trieste, Italy
| | - Anna Boccaccio
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy
| | - Anna Menini
- Neurobiology Group, International School for Advanced Studies (SISSA), 34136 Trieste, Italy
| |
Collapse
|
49
|
Bijos DA, Drake MJ, Vahabi B. Anoctamin-1 in the juvenile rat urinary bladder. PLoS One 2014; 9:e106190. [PMID: 25181534 PMCID: PMC4152174 DOI: 10.1371/journal.pone.0106190] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/01/2014] [Indexed: 01/21/2023] Open
Abstract
Purpose To investigate presence, location and functional role of calcium-activated chloride channel (CaCC) Anoctamin-1 (Ano1) in rat urinary bladder. Materials and Methods Bladders from 3 week old Wistar rats were studied. End-point PCR on total mRNA was used to assess the expression of Ano1. Immunofluorescent labelling of whole mount bladder tissue imaged with confocal microscope allowed localization of Ano1 and vimentin immunopositive cells. The effects of CaCC blockers: niflumic acid (NFA) (3,10,30 µM) and 5-Nitro-2-(3-phenylpropylamino)benzoic acid (NPPB) (10, 30 µM) on spontaneous phasic contractile activity of intact (with mucosa) and denuded (without mucosa) detrusor strips were measured under isometric tension in organ baths (n = 141, N = 60). Results Ano1 expression was found at mRNA level in mucosa and detrusor layers. Confocal microscopy revealed presence of Ano1 immunopositive cells in mucosa and in detrusor layers; a subpopulation of vimentin positive cells expressed Ano1. Both chloride channel blockers reduced the amplitude and frequency of phasic contractions in denuded and intact strips. Conclusions Ano1 is expressed in rat urinary bladder and is present in cells sharing markers with interstitial cells. CaCC blockers reduced phasic activity of the bladder tissue. Ano1 is expressed in the bladder and plays a role in its spontaneous phasic contractile activity.
Collapse
Affiliation(s)
- Dominika A. Bijos
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Urological Institute, North Bristol NHS Trust, Bristol, United Kingdom
| | - Marcus J. Drake
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Urological Institute, North Bristol NHS Trust, Bristol, United Kingdom
- * E-mail:
| | - Bahareh Vahabi
- Bristol Urological Institute, North Bristol NHS Trust, Bristol, United Kingdom
- Department of Biological, Biomedical and Analytical Sciences, University of the West of England, Bristol, United Kingdom
| |
Collapse
|
50
|
Singh RD, Gibbons SJ, Saravanaperumal SA, Du P, Hennig GW, Eisenman ST, Mazzone A, Hayashi Y, Cao C, Stoltz GJ, Ordog T, Rock JR, Harfe BD, Szurszewski JH, Farrugia G. Ano1, a Ca2+-activated Cl- channel, coordinates contractility in mouse intestine by Ca2+ transient coordination between interstitial cells of Cajal. J Physiol 2014; 592:4051-68. [PMID: 25063822 DOI: 10.1113/jphysiol.2014.277152] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Interstitial cells of Cajal (ICC) are pacemaker cells that generate electrical activity to drive contractility in the gastrointestinal tract via ion channels. Ano1 (Tmem16a), a Ca(2+)-activated Cl(-) channel, is an ion channel expressed in ICC. Genetic deletion of Ano1 in mice resulted in loss of slow waves in smooth muscle of small intestine. In this study, we show that Ano1 is required to maintain coordinated Ca(2+) transients between myenteric ICC (ICC-MY) of small intestine. First, we found spontaneous Ca(2+) transients in ICC-MY in both Ano1 WT and knockout (KO) mice. However, Ca(2+) transients within the ICC-MY network in Ano1 KO mice were uncoordinated, while ICC-MY Ca(2+) transients in Ano1 WT mice were rhythmic and coordinated. To confirm the role of Ano1 in the loss of Ca(2+) transient coordination, we used pharmacological inhibitors of Ano1 activity and shRNA-mediated knock down of Ano1 expression in organotypic cultures of Ano1 WT small intestine. Coordinated Ca(2+) transients became uncoordinated using both these approaches, supporting the conclusion that Ano1 is required to maintain coordination/rhythmicity of Ca(2+) transients. We next determined the effect on smooth muscle contractility using spatiotemporal maps of contractile activity in Ano1 KO and WT tissues. Significantly decreased contractility that appeared to be non-rhythmic and uncoordinated was observed in Ano1 KO jejunum. In conclusion, Ano1 has a previously unidentified role in the regulation of coordinated gastrointestinal smooth muscle function through coordination of Ca(2+) transients in ICC-MY.
Collapse
Affiliation(s)
- Raman Deep Singh
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Simon J Gibbons
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | | | - Peng Du
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Grant W Hennig
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Seth T Eisenman
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Amelia Mazzone
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Yujiro Hayashi
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Chike Cao
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Gary J Stoltz
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Tamas Ordog
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Jason R Rock
- Department of Anatomy, UCSF School of Medicine, San Francisco, CA, USA
| | - Brian D Harfe
- Department of Molecular Genetics and Microbiology Genetics Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | - Joseph H Szurszewski
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| | - Gianrico Farrugia
- Department of Physiology and Biomedical Engineering Enteric NeuroScience Program, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|