1
|
Park SH, Lee Y, Jeon H, Park J, Kim J, Kang M, Namkung W. Anticancer Effect of Hemin through ANO1 Inhibition in Human Prostate Cancer Cells. Int J Mol Sci 2024; 25:6032. [PMID: 38892219 PMCID: PMC11172662 DOI: 10.3390/ijms25116032] [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: 04/15/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Anoctamin1 (ANO1), a calcium-activated chloride channel, is overexpressed in a variety of cancer cells, including prostate cancer, and is involved in cancer cell proliferation, migration, and invasion. Inhibition of ANO1 in these cancer cells exhibits anticancer effects. In this study, we conducted a screening to identify novel ANO1 inhibitors with anticancer effects using PC-3 human prostate carcinoma cells. Screening of 2978 approved and investigational drugs revealed that hemin is a novel ANO1 inhibitor with an IC50 value of 0.45 μM. Notably, hemin had no significant effect on intracellular calcium signaling and cystic fibrosis transmembrane conductance regulator (CFTR), a cyclic AMP (cAMP)-regulated chloride channel, and it showed a weak inhibitory effect on ANO2 at 3 μM, a concentration that completely inhibits ANO1. Interestingly, hemin also significantly decreased ANO1 protein levels and strongly inhibited the cell proliferation and migration of PC-3 cells in an ANO1-dependent manner. Furthermore, it strongly induced caspase-3 activation, PARP degradation, and apoptosis in PC-3 cells. These findings suggest that hemin possesses anticancer properties via ANO1 inhibition and could be considered for development as a novel treatment for prostate cancer.
Collapse
Affiliation(s)
- So-Hyeon Park
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (S.-H.P.); (Y.L.); (H.J.); (J.P.)
| | - Yechan Lee
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (S.-H.P.); (Y.L.); (H.J.); (J.P.)
| | - Hyejin Jeon
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (S.-H.P.); (Y.L.); (H.J.); (J.P.)
| | - Junghwan Park
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (S.-H.P.); (Y.L.); (H.J.); (J.P.)
| | - Jieun Kim
- Graduate Program of Industrial Pharmaceutical Science, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (J.K.); (M.K.)
| | - Mincheol Kang
- Graduate Program of Industrial Pharmaceutical Science, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (J.K.); (M.K.)
| | - Wan Namkung
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (S.-H.P.); (Y.L.); (H.J.); (J.P.)
- Graduate Program of Industrial Pharmaceutical Science, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; (J.K.); (M.K.)
| |
Collapse
|
2
|
Nguyen DM, Chen TY. Structure and Function of Calcium-Activated Chloride Channels and Phospholipid Scramblases in the TMEM16 Family. Handb Exp Pharmacol 2024; 283:153-180. [PMID: 35792944 DOI: 10.1007/164_2022_595] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The transmembrane protein 16 (TMEM16) family consists of Ca2+-activated chloride channels and phospholipid scramblases. Ten mammalian TMEM16 proteins, TMEM16A-K (with no TMEM16I), and several non-mammalian TMEM16 proteins, such as afTMEM16 and nhTMEM16, have been discovered. All known TMEM16 proteins are homodimeric proteins containing two subunits. Each subunit consists of ten transmembrane helices with Ca2+-binding sites and a single ion-permeation/phospholipid transport pathway. The ion-permeation pathway and the phospholipid transport pathway of TMEM16 proteins have a wide intracellular vestibule, a narrow neck, and a smaller extracellular vestibule. Interestingly, the lining wall of the ion-permeation/phospholipid transport pathway may be formed, at least partially, by membrane phospholipids, though the degree of pore-wall forming by phospholipids likely varies among TMEM16 proteins. Thus, the biophysical properties and activation mechanisms of TMEM16 proteins could differ from each other accordingly. Here we review the current understanding of the structure and function of TMEM16 molecules.
Collapse
Affiliation(s)
- Dung Manh Nguyen
- Center for Neuroscience, University of California, Davis, CA, USA.
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Tsung-Yu Chen
- Department of Neurology, Center for Neuroscience, University of California, Davis, CA, USA.
| |
Collapse
|
3
|
Li S, Wang Z, Geng R, Zhang W, Wan H, Kang X, Guo S. TMEM16A ion channel: A novel target for cancer treatment. Life Sci 2023; 331:122034. [PMID: 37611692 DOI: 10.1016/j.lfs.2023.122034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
Cancer draws attention owing to the high morbidity and mortality. It is urgent to develop safe and effective cancer therapeutics. The calcium-activated chloride channel TMEM16A is widely distributed in various tissues and regulates physiological functions. TMEM16A is abnormally expressed in several cancers and associate with tumorigenesis, metastasis, and prognosis. Knockdown or inhibition of TMEM16A in cancer cells significantly inhibits cancer development. Therefore, TMEM16A is considered as a biomarker and therapeutic target for some cancers. This work reviews the cancers associated with TMEM16A. Then, the molecular mechanism of TMEM16A overexpression in cancer was analyzed, and the possible signal transduction mechanism of TMEM16A regulating cancer development was summarized. Finally, TMEM16A inhibitors with anticancer effect and their anticancer mechanism were concluded. We hope to provide new ideas for pharmacological studies on TMEM16A in cancer.
Collapse
Affiliation(s)
- Shuting Li
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Zhichen Wang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Ruili Geng
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Weiwei Zhang
- School of Basic Medical Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Haifu Wan
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China
| | - Xianjiang Kang
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| | - Shuai Guo
- School of Life Sciences, Hebei University, Baoding 071002, Hebei, China; Institute of Life Sciences and Green Development, Hebei University, Baoding 071002, Hebei, China.
| |
Collapse
|
4
|
Wang Y, Hu X, Huang H, Jin Z, Gao J, Guo Y, Zhong Y, Li Z, Zong X, Wang K, Zhang L, Liu Z. Optimization of 4-arylthiophene-3-carboxylic acid derivatives as inhibitors of ANO1: Lead optimization studies toward their analgesic efficacy for inflammatory pain. Eur J Med Chem 2022; 237:114413. [DOI: 10.1016/j.ejmech.2022.114413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/17/2022] [Accepted: 04/21/2022] [Indexed: 11/04/2022]
|
5
|
Ion Channel Involvement in Tumor Drug Resistance. J Pers Med 2022; 12:jpm12020210. [PMID: 35207698 PMCID: PMC8878471 DOI: 10.3390/jpm12020210] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 11/30/2022] Open
Abstract
Over 90% of deaths in cancer patients are attributed to tumor drug resistance. Resistance to therapeutic agents can be due to an innate property of cancer cells or can be acquired during chemotherapy. In recent years, it has become increasingly clear that regulation of membrane ion channels is an important mechanism in the development of chemoresistance. Here, we review the contribution of ion channels in drug resistance of various types of cancers, evaluating their potential in clinical management. Several molecular mechanisms have been proposed, including evasion of apoptosis, cell cycle arrest, decreased drug accumulation in cancer cells, and activation of alternative escape pathways such as autophagy. Each of these mechanisms leads to a reduction of the therapeutic efficacy of administered drugs, causing more difficulty in cancer treatment. Thus, targeting ion channels might represent a good option for adjuvant therapies in order to counteract chemoresistance development.
Collapse
|
6
|
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
|
7
|
Opazo MC, Rivera JC, Gonzalez PA, Bueno SM, Kalergis AM, Riedel CA. Thyroid Gene Mutations in Pregnant and Breastfeeding Women Diagnosed With Transient Congenital Hypothyroidism: Implications for the Offspring's Health. Front Endocrinol (Lausanne) 2021; 12:679002. [PMID: 34721286 PMCID: PMC8551387 DOI: 10.3389/fendo.2021.679002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 09/13/2021] [Indexed: 12/21/2022] Open
Abstract
Fetus and infants require appropriate thyroid hormone levels and iodine during pregnancy and lactation. Nature endorses the mother to supply thyroid hormones to the fetus and iodine to the lactating infant. Genetic variations on thyroid proteins that cause dyshormonogenic congenital hypothyroidism could in pregnant and breastfeeding women impair the delivery of thyroid hormones and iodine to the offspring. The review discusses maternal genetic variations in thyroid proteins that, in the context of pregnancy and/or breastfeeding, could trigger thyroid hormone deficiency or iodide transport defect that will affect the proper development of the offspring.
Collapse
Affiliation(s)
- Maria C. Opazo
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias de la Vida, Departamento de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- Instituto de Ciencias Naturales, Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Santiago, Chile
| | - Juan Carlos Rivera
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias de la Vida, Departamento de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
| | - Pablo A. Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias de la Vida, Departamento de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
8
|
Wang Y, Gao J, Zhao S, Song Y, Huang H, Zhu G, Jiao P, Xu X, Zhang G, Wang K, Zhang L, Liu Z. Discovery of 4-arylthiophene-3-carboxylic acid as inhibitor of ANO1 and its effect as analgesic agent. Acta Pharm Sin B 2021; 11:1947-1964. [PMID: 34386330 PMCID: PMC8343189 DOI: 10.1016/j.apsb.2020.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/26/2020] [Accepted: 11/04/2020] [Indexed: 02/05/2023] Open
Abstract
Anoctamin 1 (ANO1) is a kind of calcium-activated chloride channel involved in nerve depolarization. ANO1 inhibitors display significant analgesic activity by the local peripheral and intrathecal administration. In this study, several thiophenecarboxylic acid and benzoic acid derivatives were identified as novel ANO1 inhibitors through the shape-based virtual screening, among which the 4-arylthiophene-3-carboxylic acid analogues with the best ANO1 inhibitory activity were designed, synthesized and compound 42 (IC50 = 0.79 μmol/L) was finally obtained. Compound 42 selectively inhibited ANO1 without affecting ANO2 and intracellular Ca2+ concentration. Subsequently, the analgesic effect was investigated by intragastric administration in pain models. Compound 42 significantly attenuated allodynia which was induced by formalin and chronic constriction injury. Through homology modeling and molecular dynamics, the binding site was predicted to be located near the calcium-binding region between α6 and α8. Our study validates ANO1 inhibitors having a significant analgesic effect by intragastric administration and also provides selective molecular tools for ANO1-related research.
Collapse
|
9
|
Nowicka-Bauer K, Szymczak-Cendlak M. Structure and Function of Ion Channels Regulating Sperm Motility-An Overview. Int J Mol Sci 2021; 22:ijms22063259. [PMID: 33806823 PMCID: PMC8004680 DOI: 10.3390/ijms22063259] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
Sperm motility is linked to the activation of signaling pathways that trigger movement. These pathways are mainly dependent on Ca2+, which acts as a secondary messenger. The maintenance of adequate Ca2+ concentrations is possible thanks to proper concentrations of other ions, such as K+ and Na+, among others, that modulate plasma membrane potential and the intracellular pH. Like in every cell, ion homeostasis in spermatozoa is ensured by a vast spectrum of ion channels supported by the work of ion pumps and transporters. To achieve success in fertilization, sperm ion channels have to be sensitive to various external and internal factors. This sensitivity is provided by specific channel structures. In addition, novel sperm-specific channels or isoforms have been found with compositions that increase the chance of fertilization. Notably, the most significant sperm ion channel is the cation channel of sperm (CatSper), which is a sperm-specific Ca2+ channel required for the hyperactivation of sperm motility. The role of other ion channels in the spermatozoa, such as voltage-gated Ca2+ channels (VGCCs), Ca2+-activated Cl-channels (CaCCs), SLO K+ channels or voltage-gated H+ channels (VGHCs), is to ensure the activation and modulation of CatSper. As the activation of sperm motility differs among metazoa, different ion channels may participate; however, knowledge regarding these channels is still scarce. In the present review, the roles and structures of the most important known ion channels are described in regard to regulation of sperm motility in animals.
Collapse
Affiliation(s)
- Karolina Nowicka-Bauer
- Department of Chemical Physics, Faculty of Chemistry, Adam Mickiewicz University in Poznań, 61-614 Poznan, Poland
- Correspondence:
| | - Monika Szymczak-Cendlak
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University in Poznań, 61-614 Poznan, Poland;
| |
Collapse
|
10
|
McMahon DB, Carey RM, Kohanski MA, Adappa ND, Palmer JN, Lee RJ. PAR-2-activated secretion by airway gland serous cells: role for CFTR and inhibition by Pseudomonas aeruginosa. Am J Physiol Lung Cell Mol Physiol 2021; 320:L845-L879. [PMID: 33655758 DOI: 10.1152/ajplung.00411.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Airway submucosal gland serous cells are important sites of fluid secretion in conducting airways. Serous cells also express the cystic fibrosis (CF) transmembrane conductance regulator (CFTR). Protease-activated receptor 2 (PAR-2) is a G protein-coupled receptor that activates secretion from intact airway glands. We tested if and how human nasal serous cells secrete fluid in response to PAR-2 stimulation using Ca2+ imaging and simultaneous differential interference contrast imaging to track isosmotic cell shrinking and swelling reflecting activation of solute efflux and influx pathways, respectively. During stimulation of PAR-2, serous cells exhibited dose-dependent increases in intracellular Ca2+. At stimulation levels >EC50 for Ca2+, serous cells simultaneously shrank ∼20% over ∼90 s due to KCl efflux reflecting Ca2+-activated Cl- channel (CaCC, likely TMEM16A)-dependent secretion. At lower levels of PAR-2 stimulation (<EC50 for Ca2+), shrinkage was not evident due to failure to activate CaCC. Low levels of cAMP-elevating VIP receptor (VIPR) stimulation, also insufficient to activate secretion alone, synergized with low-level PAR-2 stimulation to elicit fluid secretion dependent on both cAMP and Ca2+ to activate CFTR and K+ channels, respectively. Polarized cultures of primary serous cells also exhibited synergistic fluid secretion. Pre-exposure to Pseudomonas aeruginosa conditioned media inhibited PAR-2 activation by proteases but not peptide agonists in primary nasal serous cells, Calu-3 bronchial cells, and primary nasal ciliated cells. Disruption of synergistic CFTR-dependent PAR-2/VIPR secretion may contribute to reduced airway surface liquid in CF. Further disruption of the CFTR-independent component of PAR-2-activated secretion by P. aeruginosa may also be important to CF pathophysiology.
Collapse
Affiliation(s)
- Derek B McMahon
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ryan M Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Michael A Kohanski
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nithin D Adappa
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - James N Palmer
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Robert J Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.,Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Grigoriev VV. [Calcium-activated chloride channels: structure, properties, role in physiological and pathological processes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:17-33. [PMID: 33645519 DOI: 10.18097/pbmc20216701017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Ca2+-activated chloride channels (CaCC) are a class of intracellular calcium activated chloride channels that mediate numerous physiological functions. In 2008, the molecular structure of CaCC was determined. CaCC are formed by the protein known as anoctamine 1 (ANO1 or TMEM16A). CaCC mediates the secretion of Cl- in secretory epithelia, such as the airways, salivary glands, intestines, renal tubules, and sweat glands. The presence of CaCC has also been recognized in the vascular muscles, smooth muscles of the respiratory tract, which control vascular tone and hypersensitivity of the respiratory tract. TMEM16A is activated in many cancers; it is believed that TMEM16A is involved in carcinogenesis. TMEM16A is also involved in cancer cells proliferation. The role of TMEM16A in the mechanisms of hypertension, asthma, cystic fibrosis, nociception, and dysfunction of the gastrointestinal tract has been determined. In addition to TMEM16A, its isoforms are involved in other physiological and pathophysiological processes. TMEM16B (or ANO2) is involved in the sense of smell, while ANO6 works like scramblase, and its mutation causes a rare bleeding disorder, known as Scott syndrome. ANO5 is associated with muscle and bone diseases. TMEM16A interacts with various cellular signaling pathways including: epidermal growth factor receptor (EGFR), mitogen-activated protein kinases (MAPK), calmodulin (CaM) kinases, transforming growth factor TGF-β. The review summarizes existing information on known natural and synthetic compounds that can block/modulate CaCC currents and their effect on some pathologies in which CaCC is involved.
Collapse
Affiliation(s)
- V V Grigoriev
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
12
|
Chen W, Gu M, Gao C, Chen B, Yang J, Xie X, Wang X, Sun J, Wang J. The Prognostic Value and Mechanisms of TMEM16A in Human Cancer. Front Mol Biosci 2021; 8:542156. [PMID: 33681289 PMCID: PMC7930745 DOI: 10.3389/fmolb.2021.542156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
As a calcium ion-dependent chloride channel transmembrane protein 16A (TMEM16A) locates on the cell membrane. Numerous research results have shown that TMEM16A is abnormally expressed in many cancers. Mechanically, TMEM16A participates in cancer proliferation and migration by affecting the MAPK and CAMK signaling pathways. Additionally, it is well documented that TMEM16A exerts a regulative impact on the hyperplasia of cancer cells by interacting with EGFR in head and neck squamous cell carcinoma (HNSCC), an epithelial growth factor receptor in head and neck squamous cell carcinoma respectively. Meanwhile, as an EGFR activator, TMEM16A is considered as an oncogene or a tumor-promoting factor. More and more experimental data showed that down-regulation of TMEM16A or gene targeted therapy may be an effective treatment for cancer. This review summarized its role in various cancers and research advances related to its clinical application included treatment and diagnosis.
Collapse
Affiliation(s)
- Wenjian Chen
- Anhui Province Children's Hospital Affiliated to Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China
| | - Meng Gu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Chaobing Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of AnHui Medical University, Hefei, China
| | - Bangjie Chen
- First Clinical Medical College of Anhui Medical University, Hefei, China
| | - Junfa Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Xiaoli Xie
- Anhui Medicine Centralized Procurement Service Center, Hefei, China
| | - Xinyi Wang
- First Clinical Medical College of Anhui Medical University, Hefei, China
| | - Jun Sun
- Anhui Province Children's Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Jinian Wang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
TMEM106C contributes to the malignant characteristics and poor prognosis of hepatocellular carcinoma. Aging (Albany NY) 2021; 13:5585-5606. [PMID: 33591950 PMCID: PMC7950261 DOI: 10.18632/aging.202487] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Transmembrane protein (TMEM) is a kind of integral membrane protein that spans biological membranes. The functions of most members of the TMEM family are unknown. Here, we conducted bioinformatic analysis and biological validation to investigate the role of TMEM106C in HCC. First, GEPIA and OncomineTM were used to analyze TMEM106C expression, which was verified by real-time PCR and western blot analyses. Then, the biological functions of TMEM106C were explored by CCK8 and transwell assays. The prognostic value of TMEM106C was analyzed by UALCAN. LinkedOmics was used to analyze TMEM106C pathways generated by Gene Ontology. A protein-protein interaction network (PPI) was constructed by GeneMANIA. We demonstrated that TMEM106C was overexpressed in HCC and that inhibition of TMEM106C significantly suppressed the proliferation and metastasis of HCC through targeting CENPM and DLC-1. Upregulation of TMEM106C was closely correlated with sex, tumor stage, tumor grade and prognosis. Overexpression of TMEM106C was linked to functional networks involving organelle fission and cell cycle signaling pathways through the regulation of CDK kinases, E2F1 transcription factors and miRNAs. Our data demonstrated that TMEM106C contributes to malignant characteristics and poor prognosis in HCC, which may serve as a prognostic biomarker and potential therapeutic target.
Collapse
|
14
|
Feng S, Dang S, Han TW, Ye W, Jin P, Cheng T, Li J, Jan YN, Jan LY, Cheng Y. Cryo-EM Studies of TMEM16F Calcium-Activated Ion Channel Suggest Features Important for Lipid Scrambling. Cell Rep 2020; 28:567-579.e4. [PMID: 31291589 PMCID: PMC6684876 DOI: 10.1016/j.celrep.2019.06.023] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/03/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022] Open
Abstract
As a Ca2+-activated lipid scramblase and ion channel that mediates Ca2+ influx, TMEM16F relies on both functions to facilitate extracellular vesicle generation, blood coagulation, and bone formation. How a bona fide ion channel scrambles lipids remains elusive. Our structural analyses revealed the coexistence of an intact channel pore and PIP2-dependent protein conformation changes leading to membrane distortion. Correlated to the extent of membrane distortion, many tightly bound lipids are slanted. Structure-based mutagenesis studies further reveal that neutralization of some lipid-binding residues or those near membrane distortion specifically alters the onset of lipid scrambling, but not Ca2+ influx, thus identifying features outside of channel pore that are important for lipid scrambling. Together, our studies demonstrate that membrane distortion does not require open hydrophilic grooves facing the membrane interior and provide further evidence to suggest separate pathways for lipid scrambling and ion permeation. TMEM16F is a calcium-activated ion channel and lipid scramblase linked to the bleeding disorder Scott syndrome. Feng et al. examine cryo-EM structures of TMEM16F with or without Ca2+ ions and PIP2 nanodisc supplementation and identify structural features for lipid binding and membrane distortion critical for lipid scrambling activity.
Collapse
Affiliation(s)
- Shengjie Feng
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shangyu Dang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tina Wei Han
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Wenlei Ye
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Peng Jin
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tong Cheng
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Junrui Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh Nung Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lily Yeh Jan
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
15
|
Ji Q, Shi S, Guo S, Zhan Y, Zhang H, Chen Y, An H. Activation of TMEM16A by natural product canthaxanthin promotes gastrointestinal contraction. FASEB J 2020; 34:13430-13444. [PMID: 32812278 DOI: 10.1096/fj.202000443rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 11/11/2022]
Abstract
Transmembrane 16A (TMEM16A), also known as anoctamin 1, is the molecular basis of the calcium-activated chloride channels. TMEM16A is present in interstitial cells of Cajal, which are the pacemaker cells that control smooth muscle contraction. TMEM16A is implicated in gastrointestinal disorders. Activation of TMEM16A is believed to promote the gastrointestinal muscle contraction. Here, we report a highly efficient, nontoxic, and selective activator of TMEM16A, canthaxanthin (CX). The study using molecular docking and site-directed mutation revealed that CX-specific binging site in TMEM16A is K769. CX was also found to promote the contraction of smooth muscle cells in gastrointestinal tract through activation of TMEM16A channels, which provides an excellent basis for development of CX as a chemical tool and potential therapeutic for gastrointestinal dysfunction.
Collapse
Affiliation(s)
- Qiushuang Ji
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China.,School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Sai Shi
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China.,School of Electrical Engineering, Hebei University of Technology, Tianjin, China
| | - Shuai Guo
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China.,School of Electrical Engineering, Hebei University of Technology, Tianjin, China
| | - Yong Zhan
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China.,School of Electrical Engineering, Hebei University of Technology, Tianjin, China
| | - Hailin Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Pharmacology and Toxicology for New Drug, Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China.,School of Electrical Engineering, Hebei University of Technology, Tianjin, China
| |
Collapse
|
16
|
Hunziker M, O'Donnell AM, Gosemann J, Alvarez LA, Puri P. Altered anoctamin-1 and tyrosine phosphorylation in congenital ureteropelvic junction obstruction. J Pediatr Surg 2020; 55:1621-1625. [PMID: 32087933 DOI: 10.1016/j.jpedsurg.2020.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/26/2019] [Accepted: 02/05/2020] [Indexed: 01/30/2023]
Abstract
PURPOSE Ureteropelvic junction (UPJ) obstruction is the most common cause of congenital hydronephrosis in children. The pathophysiology of UPJ obstruction and the exact mechanism of pelviureteral peristalsis are poorly understood. Anoctamin-1 (ANO1), a Ca2+-activated chloride channel, has been shown to play a key role in muscle wall contractions in the gastrointestinal tract. We designed this study to investigate the hypothesis that ANO1 is expressed in smooth muscle cells (SMCs) of the human UPJ and that tyrosine phosphorylation is altered in UPJ obstruction. MATERIALS AND METHODS Fresh frozen specimens of UPJ obstruction (n = 28) and control specimens from patients who underwent Wilms' tumor nephrectomy (n = 20) were prepared. Western blot (WB) was performed to evaluate levels of ANO1 protein expression and changes in tyrosine phosphorylation. In addition analysis of ANO1 and phalloidin using confocal-immunofluoresence-double staining and 3D reconstruction were carried out. RESULTS Our WB results revealed increased tyrosine phosphorylation in UPJ obstruction samples compared to controls, and decreased ANO1 expression in UPJ obstruction. Confocal microscopy showed that ANO1 immunoreactivity was decreased in SMCs of UPJ obstruction compared to controls. CONCLUSIONS We provide evidence, for the first time, of the presence of ANO1 expression in the human UPJ. We speculate that altered tyrosine phosphorylation, observed in UPJ obstruction, may lead to a failure of transmission of peristaltic waves in UPJ obstruction by inhibiting Ca2+-activated chloride channels in SMCs.
Collapse
Affiliation(s)
- Manuela Hunziker
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; School of Medicine and Medical Science and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Anne-Marie O'Donnell
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Jan Gosemann
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Luis A Alvarez
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Prem Puri
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; School of Medicine and Medical Science and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland.
| |
Collapse
|
17
|
Satou R, Shibukawa Y, Kimura M, Sugihara N. Light conditions affect rhythmic expression of aquaporin 5 and anoctamin 1 in rat submandibular glands. Heliyon 2019; 5:e02792. [PMID: 31844723 PMCID: PMC6895735 DOI: 10.1016/j.heliyon.2019.e02792] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 09/20/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022] Open
Abstract
Circadian rhythms regulate various physiological functions and are, therefore, essential for health. Light helps regulate the master and peripheral clocks. The secretion rates of saliva and electrolytes follow a circadian rhythm as well. However, the relationship between the molecular mechanism of saliva water secretion and the peripheral circadian rhythm in salivary glands is not yet clear. The transmembrane proteins aquaporin5 (Aqp5) and anoctamin1 (Ano1) are essential for water transport in the submandibular glands (SGs). The purpose of this study was to reveal the effect of light conditioning on the peripheral clock in SGs. We examined temporal expression patterns among clock genes, Aqp5 and Ano1, in rat SGs under light/dark (LD) and dark/dark (DD) conditions. We observed circadian rhythmic expression of Bmal1, Per2, Cry1, Aqp5, and Ano1 mRNAs under both LD and DD conditions. The expression levels of Aqp5 and Ano1 peaked 6 h earlier under the DD condition than under the LD condition. Maintenance of the circadian rhythm of Aqp5 and Ano1 expression even under the DD condition indicates that Aqp5 and Ano1 may be controlled by clock genes; such genes are called clock-controlled genes (CCGs). Western blot analysis revealed the circadian oscillation and peak shift of AQP5 and ANO1expression under DD conditions. Clock genes may regulate the rhythmic expression of Ano1 and Aqp5 and may control osmic gradients in SGs.
Collapse
Affiliation(s)
- Ryouichi Satou
- Department of Epidemiology and Public Health, Tokyo Dental College, Chiyodaku, Tokyo 101-0061, Japan
| | - Yoshiyuki Shibukawa
- Department of Physiology, Tokyo Dental College, Tokyo, Chiyodaku, Tokyo 101-0061, Japan
| | - Maki Kimura
- Department of Physiology, Tokyo Dental College, Tokyo, Chiyodaku, Tokyo 101-0061, Japan
| | - Naoki Sugihara
- Department of Epidemiology and Public Health, Tokyo Dental College, Chiyodaku, Tokyo 101-0061, Japan
| |
Collapse
|
18
|
Aung T, Asam C, Haerteis S. Ion channels in sarcoma: pathophysiology and treatment options. Pflugers Arch 2019; 471:1163-1171. [PMID: 31377822 DOI: 10.1007/s00424-019-02299-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022]
Abstract
Sarcomas are characterized by aggressive growth and a high metastasis potentially leading in most cases to a lethal outcome. These malignant tumors of the connective tissue have a high heterogeneity with numerous genetic mutations resulting in more than 100 types of sarcoma that can be grouped into two main kinds: soft tissue sarcoma and bone sarcoma. Sarcomas are often diagnosed at late disease stage, whereas a guaranteed diagnosis of the sarcoma type is fundamental for successful therapy. However, there is no appropriate therapy available. Therefore, the need for new therapies, which prolong survival and improve quality of life, is high. In the last two decades, the role of ion channels in cancer has emerged. Ion channels seem to be an ideal target for anti-tumor therapies. However, different cancer types have their own altered ion channel pattern, and the knowledge about the tumor-associated ion channel expression is fundamental. Here, we focus on the role of different ion channels in sarcoma, their pathophysiology, and possible treatment options.
Collapse
Affiliation(s)
- Thiha Aung
- Abteilung für Plastische, Hand- und Wiederherstellungschirurgie, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Claudia Asam
- Lehrstuhl für Molekulare und Zelluläre Anatomie, Universität Regensburg, 93053, Regensburg, Germany
| | - Silke Haerteis
- Lehrstuhl für Molekulare und Zelluläre Anatomie, Universität Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
19
|
Tembo M, Wozniak KL, Bainbridge RE, Carlson AE. Phosphatidylinositol 4,5-bisphosphate (PIP 2) and Ca 2+ are both required to open the Cl - channel TMEM16A. J Biol Chem 2019; 294:12556-12564. [PMID: 31266809 DOI: 10.1074/jbc.ra118.007128] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 07/01/2019] [Indexed: 12/27/2022] Open
Abstract
Transmembrane member 16A (TMEM16A) is a widely expressed Ca2+-activated Cl- channel with various physiological functions ranging from mucosal secretion to regulating smooth muscle contraction. Understanding how TMEM16A controls these physiological processes and how its dysregulation may cause disease requires a detailed understanding of how cellular processes and second messengers alter TMEM16A channel gating. Here we assessed the regulation of TMEM16A gating by recording Ca2+-evoked Cl- currents conducted by endogenous TMEM16A channels expressed in Xenopus laevis oocytes, using the inside-out configuration of the patch clamp technique. During continuous application of Ca2+, we found that TMEM16A-conducted currents decay shortly after patch excision. Such current rundown is common among channels regulated by phosphatidylinositol 4,5-bisphosphate (PIP2). Thus, we sought to investigate a possible role of PIP2 in TMEM16A gating. Consistently, synthetic PIP2 rescued the current after rundown, and the application of PIP2 modulating agents altered the speed kinetics of TMEM16A current rundown. First, two PIP2 sequestering agents, neomycin and anti-PIP2, applied to the intracellular surface of excised patches sped up TMEM16A current rundown to nearly twice as fast. Conversely, rephosphorylation of phosphatidylinositol (PI) derivatives into PIP2 using Mg-ATP or inhibiting dephosphorylation of PIP2 using β-glycerophosphate slowed rundown by nearly 3-fold. Our results reveal that TMEM16A regulation is more complicated than it initially appeared; not only is Ca2+ necessary to signal TMEM16a opening, but PIP2 is also required. These findings improve our understanding of how the dysregulation of these pathways may lead to disease and suggest that targeting these pathways could have utility for potential therapies.
Collapse
Affiliation(s)
- Maiwase Tembo
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Katherine L Wozniak
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Rachel E Bainbridge
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Anne E Carlson
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.
| |
Collapse
|
20
|
Muise ES, Guan HP, Liu J, Nawrocki AR, Yang X, Wang C, Rodríguez CG, Zhou D, Gorski JN, Kurtz MM, Feng D, Leavitt KJ, Wei L, Wilkening RR, Apgar JM, Xu S, Lu K, Feng W, Li Y, He H, Previs SF, Shen X, van Heek M, Souza SC, Rosenbach MJ, Biftu T, Erion MD, Kelley DE, Kemp DM, Myers RW, Sebhat IK. Pharmacological AMPK activation induces transcriptional responses congruent to exercise in skeletal and cardiac muscle, adipose tissues and liver. PLoS One 2019; 14:e0211568. [PMID: 30811418 PMCID: PMC6392219 DOI: 10.1371/journal.pone.0211568] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
Physical activity promotes metabolic and cardiovascular health benefits that derive in part from the transcriptional responses to exercise that occur within skeletal muscle and other organs. There is interest in discovering a pharmacologic exercise mimetic that could imbue wellness and alleviate disease burden. However, the molecular physiology by which exercise signals the transcriptional response is highly complex, making it challenging to identify a single target for pharmacological mimicry. The current studies evaluated the transcriptome responses in skeletal muscle, heart, liver, and white and brown adipose to novel small molecule activators of AMPK (pan-activators for all AMPK isoforms) compared to that of exercise. A striking level of congruence between exercise and pharmacological AMPK activation was observed across the induced transcriptome of these five tissues. However, differences in acute metabolic response between exercise and pharmacologic AMPK activation were observed, notably for acute glycogen balances and related to the energy expenditure induced by exercise but not pharmacologic AMPK activation. Nevertheless, intervention with repeated daily administration of short-acting activation of AMPK was found to mitigate hyperglycemia and hyperinsulinemia in four rodent models of metabolic disease and without the cardiac glycogen accretion noted with sustained pharmacologic AMPK activation. These findings affirm that activation of AMPK is a key node governing exercise mediated transcription and is an attractive target as an exercise mimetic.
Collapse
Affiliation(s)
- Eric S. Muise
- Genetics and Pharmacogenomics Department, MRL, Kenilworth, NJ, United States of America
- * E-mail: (ESM); (IKS)
| | - Hong-Ping Guan
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Jinqi Liu
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Andrea R. Nawrocki
- In Vivo Pharmacology Department, MRL, Kenilworth, NJ, United States of America
| | - Xiaodong Yang
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Chuanlin Wang
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Carlos G. Rodríguez
- In Vivo Pharmacology Department, MRL, Kenilworth, NJ, United States of America
| | - Dan Zhou
- In Vivo Pharmacology Department, MRL, Kenilworth, NJ, United States of America
| | - Judith N. Gorski
- In Vivo Pharmacology Department, MRL, Kenilworth, NJ, United States of America
| | - Marc M. Kurtz
- In Vitro PharmacologyDepartment, MRL, NJ, United States of America
| | - Danqing Feng
- Medicinal ChemistryDepartment, MRL, Kenilworth, NJ, United States of America
| | - Kenneth J. Leavitt
- Medicinal ChemistryDepartment, MRL, Kenilworth, NJ, United States of America
| | - Lan Wei
- Medicinal ChemistryDepartment, MRL, Kenilworth, NJ, United States of America
| | - Robert R. Wilkening
- Medicinal ChemistryDepartment, MRL, Kenilworth, NJ, United States of America
| | - James M. Apgar
- Medicinal ChemistryDepartment, MRL, Kenilworth, NJ, United States of America
| | - Shiyao Xu
- PPDM Preclinical ADME Department, MRL, Kenilworth, NJ, United States of America
| | - Ku Lu
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Wen Feng
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Ying Li
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Huaibing He
- PPDM Preclinical ADME Department, MRL, Kenilworth, NJ, United States of America
| | - Stephen F. Previs
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Xiaolan Shen
- SALAR Department, MRL, Kenilworth, NJ, United States of America
| | - Margaret van Heek
- In Vivo Pharmacology Department, MRL, Kenilworth, NJ, United States of America
| | - Sandra C. Souza
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Mark J. Rosenbach
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Tesfaye Biftu
- Medicinal ChemistryDepartment, MRL, Kenilworth, NJ, United States of America
| | - Mark D. Erion
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - David E. Kelley
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Daniel M. Kemp
- Biology-Discovery Department, MRL, Kenilworth, NJ, United States of America
| | - Robert W. Myers
- In Vitro PharmacologyDepartment, MRL, NJ, United States of America
| | - Iyassu K. Sebhat
- Medicinal ChemistryDepartment, MRL, Kenilworth, NJ, United States of America
- * E-mail: (ESM); (IKS)
| |
Collapse
|
21
|
Ji Q, Guo S, Wang X, Pang C, Zhan Y, Chen Y, An H. Recent advances in TMEM16A: Structure, function, and disease. J Cell Physiol 2018; 234:7856-7873. [PMID: 30515811 DOI: 10.1002/jcp.27865] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022]
Abstract
TMEM16A (also known as anoctamin 1, ANO1) is the molecular basis of the calcium-activated chloride channels, with ten transmembrane segments. Recently, atomic structures of the transmembrane domains of mouse TMEM16A (mTMEM16A) were determined by single-particle electron cryomicroscopy. This gives us a solid ground to discuss the electrophysiological properties and functions of TMEM16A. TMEM16A is reported to be dually regulated by Ca2+ and voltage. In addition, the dysfunction of TMEM16A has been found to be involved in many diseases including cystic fibrosis, various cancers, hypertension, and gastrointestinal motility disorders. TMEM16A is overexpressed in many cancers, including gastrointestinal stromal tumors, gastric cancer, head and neck squamous cell carcinoma (HNSCC), colon cancer, pancreatic ductal adenocarcinoma, and esophageal cancer. Furthermore, overexpression of TMEM16A is related to the occurrence, proliferation, and migration of tumor cells. To date, several studies have shown that many natural compounds and synthetic compounds have regulatory effects on TMEM16A. These small molecule compounds might be novel drugs for the treatment of diseases caused by TMEM16A dysfunction in the future. In addition, recent studies have shown that TMEM16A plays different roles in different diseases through different signal transduction pathways. This review discusses the topology, electrophysiological properties, modulators and functions of TMEM16A in mediates nociception, gastrointestinal dysfunction, hypertension, and cancer and focuses on multiple regulatory mechanisms regarding TMEM16A.
Collapse
Affiliation(s)
- Qiushuang Ji
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Shuai Guo
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Xuzhao Wang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Chunli Pang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Yong Zhan
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, China
| |
Collapse
|
22
|
Zhao Y, Song K, Zhang Y, Xu H, Zhang X, Wang L, Fan C, Jiang G, Wang E. TMEM17 promotes malignant progression of breast cancer via AKT/GSK3β signaling. Cancer Manag Res 2018; 10:2419-2428. [PMID: 30122991 PMCID: PMC6080873 DOI: 10.2147/cmar.s168723] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Current knowledge of TMEM17, a recently identified protein of the transmembrane (TMEM) family, is limited, especially with respect to its expression and biological functions in malignant tumors. This study analyzed TMEM17 expression in invasive breast cancer tissue and breast cell lines and its relevance to clinicopathological factors, and investigated the mechanisms underlying the biological effects of TMEM17 on breast cancer cells. Patients and methods TMEM17 protein expression was determined in 20 freshly harvested specimens (tumor and paired normal tissues) by Western blotting. Immunohistochemical analysis was performed to determine the expression and subcellular localization of TMEM17 in samples from 167 patients (mean age, 49 years) diagnosed with invasive ductal carcinoma (38 with triple-negative breast cancer; 129 with non-triple-negative breast cancer) who underwent complete resection in the First Affiliated Hospital of China Medical University between 2011 and 2013. Furthermore, TMEM17 was knocked down by small interfering RNAs in breast cancer cell lines. Results TMEM17 was found to be significantly upregulated in breast cancer tissues compared to the corresponding normal breast tissues by Western blotting (p=0.015). Immunohistochemical analysis revealed that TMEM was significantly upregulated in invasive breast cancer cells compared to adjacent normal breast duct glandular epithelial cells (10.78% vs 76.05%, p<0.001), and its expression was closely related to the patient’s T-stage (p=0.022), advanced TNM stages (p=0.007), and lymph node metastasis (p=0.012). After TMEM17 knockdown or overexpression in breast cancer cell lines, TMEM17 upregulated p-AKT, p-GSK3β, active β-catenin, and Snail, and downstream target proteins c-myc and cyclin D1, and downregulated E-cadherin, resulting in increased cancer cell proliferation, invasion, and migration. These effects were reversed by the AKT inhibitor LY294002. Conclusion Our results indicate that TMEM17 is upregulated in breast cancer tissues and can promote malignant progression of breast cancer cells by activating the AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Kuiyuan Song
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Yong Zhang
- Departments of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Hongtao Xu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Xiupeng Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Liang Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Chuifeng Fan
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Guiyang Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| | - Enhua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China,
| |
Collapse
|
23
|
Guo R, Huang X, Jin X, Yang J. [Diltiazem inhibits proliferation and motility of hepatocellular cells in vitro by downregulating calcium-activated chloride channel TMEM16A]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:818-823. [PMID: 33168514 DOI: 10.3969/j.issn.1673-4254.2018.07.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To assess the inhibitory effect of diltiazem, a calcium channel inhibitor, on the proliferation and mobility of human hepatocellular carcinoma cells in vitro and explore the possible mechanism. METHODS Two human hepatocellular carcinoma cell lines, MHCC97H and 7402, were treated with different concentrations (0-400 μmol/L) of diltiazem for 12, 24, or 48 h, and the changes in the cell proliferation and mobility were observed with MTT assay and wound healing assay, respectively. The changes in the expressions of calcium-activated chloride channel TMEM16A at mRNA and protein levels in the treated cells were detected using RT-PCR and immunocytochemistry. RESULTS Treatment with diltiazem obviously inhibited the proliferation and suppressed the mobility of MHCC97H and 7402 cells in a time- and concentration-dependent manner (P < 0.05). Treatment with 100 μmol/L diltiazem for 24 h significantly inhibited the proliferation of MHCC97H cells and down-regulated the mRNA and protein levels of TMEM16A. In 7402 cells, diltiazem treatment at 50 μmol/L for 48 h resulted in the most significant inhibitory effect on the cell proliferation and TMEM16A expressions. CONCLUSIONS Diltiazem can transiently inhibit the invasion of hepatocellular carcinoma cells in vitro possibly by down-regulating the expression of TMEM16A at both the mRNA and protein levels.
Collapse
Affiliation(s)
- Rui Guo
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiaozhong Huang
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xueyuan Jin
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jun Yang
- Department of Pathology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
24
|
Hahn A, Salomon JJ, Leitz D, Feigenbutz D, Korsch L, Lisewski I, Schrimpf K, Millar-Büchner P, Mall MA, Frings S, Möhrlen F. Expression and function of Anoctamin 1/TMEM16A calcium-activated chloride channels in airways of in vivo mouse models for cystic fibrosis research. Pflugers Arch 2018; 470:1335-1348. [DOI: 10.1007/s00424-018-2160-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/30/2018] [Accepted: 05/23/2018] [Indexed: 01/17/2023]
|
25
|
Therapeutic Approaches to Acquired Cystic Fibrosis Transmembrane Conductance Regulator Dysfunction in Chronic Bronchitis. Ann Am Thorac Soc 2018; 13 Suppl 2:S169-76. [PMID: 27115953 DOI: 10.1513/annalsats.201509-601kv] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease is a common cause of morbidity and a rising cause of mortality worldwide. Its rising impact indicates the ongoing unmet need for novel and effective therapies. Previous work has established a pathophysiological link between the chronic bronchitis phenotype of chronic obstructive pulmonary disease and cystic fibrosis as well as phenotypic similarities between these two airways diseases. An extensive body of evidence has established that cigarette smoke and its constituents contribute to acquired dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) protein in the airways, pointing to a mechanistic link with smoking-related and chronic bronchitis. Recent interest surrounding new drugs that target both mutant and wild-type CFTR channels has paved the way for a new treatment opportunity addressing the mucus defect in chronic bronchitis. We review the clinical and pathologic evidence for modulating CFTR to address acquired CFTR dysfunction and pragmatic issues surrounding clinical trials as well as a discussion of other ion channels that may represent alternative therapeutic targets.
Collapse
|
26
|
Zhang M, Gao CX, Wang YP, Ma KT, Li L, Yin JW, Dai ZG, Wang S, Si JQ. The association between the expression of PAR2 and TMEM16A and neuropathic pain. Mol Med Rep 2017; 17:3744-3750. [PMID: 29257338 PMCID: PMC5802179 DOI: 10.3892/mmr.2017.8295] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022] Open
Abstract
Chronic constriction injury (CCI) of the sciatic nerve may induce dorsal root ganglion (DRG) neuronal hyperexcitability and behaviorally expressed hyperalgesia. CCI is a model of neuropathic pain. To investigate the association between the expression of protease activated receptor 2 (PAR2), TMEM16A and neuropathic pain, the expression of PAR2 and TMEM16A proteins in the DRG neurons of rats following CCI of the sciatic nerve was investigated. Following the creation of the CCI model, the thermal withdrawal latency (TWL) was examined by a hot plate test. An immunofluorescence assay and western blot assay were performed to determine the expression of PAR2 and TMEM16A proteins in the ipsilateral L4–6 DRG neurons. The concentration of inositol 1,4,5-triphosphate (IP3) in the L4–6 DRG was determined by ELISA. In the CCI-D7 (7 days after CCI) and CCI-D14 (14 days after CCI) treatment groups, the TWL of rats was significantly shorter than that in the sham operated group (P<0.01; n=12). The expression of PAR2 and TMEM16A proteins in the CCI-D7 and CCI-D14 groups were significantly upregulated compared with the sham operated group (P<0.05; n=12). Additionally, it was revealed that PAR2 and TMEM16A were co-expressed in DRG neurons. It was also observed that IP3 significantly increased in the CCI-D7 and CCI-D14 groups compared with the sham operation group (P<0.05; n=6) as PAR2 and TMEM16A also increased. These findings suggest that the upregulation of PAR2 and TMEM16A in DRG neurons, the co-expression of the two proteins and increasing IP3 are critical to the development of neuropathic pain.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Cun-Xiang Gao
- Department of Urology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Yan-Ping Wang
- Houbo College, Xinjiang Medical University, Karamay, Xinjiang 834000, P.R. China
| | - Ke-Tao Ma
- Electrophysiological Laboratory, Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Electrophysiological Laboratory, Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jiang-Wen Yin
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Zhi-Gang Dai
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jun-Qiang Si
- Electrophysiological Laboratory, Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
27
|
Wang H, Zou L, Ma K, Yu J, Wu H, Wei M, Xiao Q. Cell-specific mechanisms of TMEM16A Ca 2+-activated chloride channel in cancer. Mol Cancer 2017; 16:152. [PMID: 28893247 PMCID: PMC5594453 DOI: 10.1186/s12943-017-0720-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/01/2017] [Indexed: 02/08/2023] Open
Abstract
TMEM16A (known as anoctamin 1) Ca2+-activated chloride channel is overexpressed in many tumors. TMEM16A overexpression can be caused by gene amplification in many tumors harboring 11q13 amplification. TMEM16A expression is also controlled in many cancer cells via transcriptional regulation, epigenetic regulation and microRNAs. In addition, TMEM16A activates different signaling pathways in different cancers, e.g. the EGFR and CAMKII signaling in breast cancer, the p38 and ERK1/2 signaling in hepatoma, the Ras-Raf-MEK-ERK1/2 signaling in head and neck squamous cell carcinoma and bladder cancer, and the NFκB signaling in glioma. Furthermore, TMEM16A overexpression has been reported to promote, inhibit, or produce no effects on cell proliferation and migration in different cancer cells. Since TMEM16A exerts different roles in different cancer cells via activation of distinct signaling pathways, we try to develop the idea that TMEM16A regulates cancer cell proliferation and migration in a cell-dependent mechanism. The cell-specific role of TMEM16A may depend on the cellular environment that is predetermined by TMEM16A overexpression mechanisms specific for a particular cancer type. TMEM16A may exert its cell-specific role via its associated protein networks, phosphorylation by different kinases, and involvement of different signaling pathways. In addition, we discuss the role of TMEM16A channel activity in cancer, and its clinical use as a prognostic and predictive marker in different cancers. This review highlights the cell-type specific mechanisms of TMEM16A in cancer, and envisions the promising use of TMEM16A inhibitors as a potential treatment for TMEM16A-overexpressing cancers.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Liang Zou
- Department of Anesthesiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 China
| | - Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Jiankun Yu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| |
Collapse
|
28
|
Tran Q, Park J, Lee H, Hong Y, Hong S, Park S, Park J, Kim SH. TMEM39A and Human Diseases: A Brief Review. Toxicol Res 2017; 33:205-209. [PMID: 28744351 PMCID: PMC5523561 DOI: 10.5487/tr.2017.33.3.205] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/13/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
Transmembrane Protein 39A (TMEM39A) is a member of TMEM family. The understanding about this protein is still limited. The earlier studies indicated that TMEM39A was a key mediator of autoimmune disease. TMEM39A seems to be involved in systemic lupus erythematosus and multiple sclerosis in numerous of populations. All of these works stop at insufficient information by using gene functioning methods such as: Genome-wide association studies (GWASs) and/or follow-up study. It is the fact that the less understood of TMEM39A actually is the attraction to the scientist in near future. In this review the current knowledge about TMEM39A and its possible roles in cell biology, physiology and pathology will be described.
Collapse
Affiliation(s)
- Quangdon Tran
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Jisoo Park
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Hyunji Lee
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Youngeun Hong
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Suntaek Hong
- Laboratory of Cancer Cell Biology, Department of Biochemistry, School of Medicine, Gachon University, Incheon, Korea
| | - Sungjin Park
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Jongsun Park
- Department of Pharmacology and Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Seon-Hwan Kim
- Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
29
|
Satou R, Sato M, Kimura M, Ishizuka Y, Tazaki M, Sugihara N, Shibukawa Y. Temporal Expression Patterns of Clock Genes and Aquaporin 5/Anoctamin 1 in Rat Submandibular Gland Cells. Front Physiol 2017; 8:320. [PMID: 28588500 PMCID: PMC5440558 DOI: 10.3389/fphys.2017.00320] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/03/2017] [Indexed: 01/20/2023] Open
Abstract
Circadian rhythms are essential for health and regulate various physiological functions. These rhythms are regulated by a negative-feedback loop involving clock genes in the suprachiasmatic nucleus (SCN) and peripheral tissues. The rate of secretion of salivary substances, ions, and water follows a circadian rhythm, however, the relationship between the molecular mechanism of salivary secretion and peripheral circadian rhythm is not yet clear. Anoctamin 1 (ANO1, also known as TMEM16A) and Aquaporin 5 (AQP5) play an important role in the transport of ions and water in the submandibular glands (SGs). We examined the interaction between the rhythmic expression pattern of the clock genes, Ano1 and Aqp5, in rat whole SGs as well as isolated acinar and ductal cells. Circadian rhythmic expression for Bmal1, Per1, Per2, Clock, Cry1, Cry2, Rorα, and Rev-erbα mRNAs, also called the clock genes, was observed in rat SGs by semi-quantitative RT-PCR analysis. We also observed rhythmic patterns in Ano1 and Aqp5 mRNA expression. The expression of ANO1 protein also showed circadian rhythm, as confirmed by western blot analysis. We could not observe any time delay between the peak expression of ANO1 protein and its mRNA. Expression levels of the clock gene mRNAs in the ductal cells was higher than that in acinar cells, however, rhythmic oscillations were observed in both. Our results suggest that SGs have peripheral clocks, and rhythmic expressions of Ano1 and Aqp5 along with the clock genes, may play an important role in the circadian regulation of salivary secretion.
Collapse
Affiliation(s)
- Ryouichi Satou
- Department of Epidemiology and Public Health, Tokyo Dental CollegeTokyo, Japan
| | - Masaki Sato
- Department of Physiology, Tokyo Dental CollegeTokyo, Japan
| | - Maki Kimura
- Department of Physiology, Tokyo Dental CollegeTokyo, Japan
| | - Yoichi Ishizuka
- Department of Epidemiology and Public Health, Tokyo Dental CollegeTokyo, Japan
| | | | - Naoki Sugihara
- Department of Epidemiology and Public Health, Tokyo Dental CollegeTokyo, Japan
| | | |
Collapse
|
30
|
Kamaleddin MA. Molecular, biophysical, and pharmacological properties of calcium-activated chloride channels. J Cell Physiol 2017; 233:787-798. [PMID: 28121009 DOI: 10.1002/jcp.25823] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/15/2022]
Abstract
Calcium-activated chloride channels (CaCCs) are a family of anionic transmembrane ion channels. They are mainly responsible for the movement of Cl- and other anions across the biological membranes, and they are widely expressed in different tissues. Since the Cl- flow into or out of the cell plays a crucial role in hyperpolarizing or depolarizing the cells, respectively, the impact of intracellular Ca2+ concentration on these channels is attracting a lot of attentions. After summarizing the molecular, biophysical, and pharmacological properties of CaCCs, the role of CaCCs in normal cellular functions will be discussed, and I will emphasize how dysregulation of CaCCs in pathological conditions can account for different diseases. A better understanding of CaCCs and a pivotal regulatory role of Ca2+ can shed more light on the therapeutic strategies for different neurological disorders that arise from chloride dysregulation, such as asthma, cystic fibrosis, and neuropathic pain.
Collapse
Affiliation(s)
- Mohammad Amin Kamaleddin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Angstadt JD, Giordano JR, Goncalves AJ. 9-Phenanthrol modulates postinhibitory rebound and afterhyperpolarizing potentials in an excitatory motor neuron of the medicinal leech. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2017; 203:613-633. [DOI: 10.1007/s00359-017-1178-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/19/2022]
|
32
|
Vikhareva EA, Zamoyski VL, Grigoriev VV. Modification of Calcium-Activated Chloride Currents in Cerebellar Purkinje Neurons. Bull Exp Biol Med 2017; 162:709-713. [PMID: 28429218 DOI: 10.1007/s10517-017-3694-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 11/30/2022]
Abstract
The whole-cell voltage clamp technique was employed to record the total ionic currents in rat cerebellar Purkinje neurons. When intrapipette solution contained 120 mM KCl, replacement of the standard external physiological saline with Na-free solution resulted in appearance of inward tail current after the end of the depolarizing pulse. When intrapipette potassium ions were replaced for cesium ones, the tail currents were observed even in the presence of normal Na+ concentration (140 mM) in the external solution. Tail currents were not observed when external solution contained no Cl- and/or Ca2+ ions. Niflumic acid (25-100 μM) blocked these currents by 80-100%. Complete replacement of external Na+ for Tris ions pronouncedly augmented the amplitude and duration of the tail currents. These findings suggest that the tail transients in rat cerebellar Purkinje neurons are calcium-activated chloride currents whose amplitude and kinetics depend on ionic composition of the extracellular and intracellular solutions.
Collapse
Affiliation(s)
- E A Vikhareva
- Laboratory of Neuroreception, Institute of Physiologically Active Substances, Russian Academy of Sciences, Chernogolovka, Moscow, Region, Russia
| | - V L Zamoyski
- Laboratory of Neuroreception, Institute of Physiologically Active Substances, Russian Academy of Sciences, Chernogolovka, Moscow, Region, Russia.
| | - V V Grigoriev
- Laboratory of Neuroreception, Institute of Physiologically Active Substances, Russian Academy of Sciences, Chernogolovka, Moscow, Region, Russia
| |
Collapse
|
33
|
Abstract
Pendred syndrome is an autosomal recessive disorder that is classically defined by the combination of sensorineural deafness/hearing impairment, goiter, and an abnormal organification of iodide with or without hypothyroidism. The hallmark of the syndrome is the impaired hearing, which is associated with inner ear malformations such as an enlarged vestibular aqueduct (EVA). The thyroid phenotype is variable and may be modified by the nutritional iodine intake. Pendred syndrome is caused by biallelic mutations in the SLC26A4/PDS gene, which encodes the multifunctional anion exchanger pendrin. Pendrin has affinity for chloride, iodide, and bicarbonate, among other anions. In the inner ear, pendrin functions as a chloride/bicarbonate exchanger that is essential for maintaining the composition and the potential of the endolymph. In the thyroid, pendrin is expressed at the apical membrane of thyroid cells facing the follicular lumen. Functional studies have demonstrated that pendrin can mediate iodide efflux in heterologous cells. This, together with the thyroid phenotype observed in humans (goiter, impaired iodine organification) suggests that pendrin could be involved in iodide efflux into the lumen, one of the steps required for thyroid hormone synthesis. Iodide efflux can, however, also occur in the absence of pendrin suggesting that other exchangers or channels are involved. It has been suggested that Anoctamin 1 (ANO1/TMEM16A), a calcium-activated anion channel, which is also expressed at the apical membrane of thyrocytes, could participate in mediating apical efflux. In the kidney, pendrin is involved in bicarbonate secretion and chloride reabsorption. While there is no renal phenotype under basal conditions, severe metabolic alkalosis has been reported in Pendred syndrome patients exposed to an increased alkali load. This review provides an overview on the clinical spectrum of Pendred syndrome, the functional data on pendrin with a focus on its potential role in the thyroid, as well as the controversy surrounding the relative physiological roles of pendrin and anoctamin.
Collapse
Affiliation(s)
- Jean-Louis Wémeau
- Université de Lille 2, Centre Hospitalier Régional Universitaire de Lille, Clinique Endocrinologique Marc-Linquette, 59037 Lille, France.
| | - Peter Kopp
- Northwestern University, Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Tarry 15, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|
34
|
Yu B, Jiang Y, Jin L, Ma T, Yang H. Role of Quercetin in Modulating Chloride Transport in the Intestine. Front Physiol 2016; 7:549. [PMID: 27932986 PMCID: PMC5120089 DOI: 10.3389/fphys.2016.00549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022] Open
Abstract
Epithelial chloride channels provide the pathways for fluid secretion in the intestine. Cystic fibrosis transmembrane conductance regulator (CFTR) and calcium-activated chloride channels (CaCCs) are the main chloride channels in the luminal membrane of enterocytes. These transmembrane proteins play important roles in many physiological processes. In this study, we have identified a flavonoid quercetin as a modulator of CaCC chloride channel activity. Fluorescence quenching assay showed that quercetin activated Cl− transport in a dose-dependent manner, with EC50 ~37 μM. Short-circuit current analysis confirmed that quercetin activated CaCC-mediated Cl− currents in HT-29 cells that can be abolished by CaCCinh-A01. Ex vivo studies indicated that application of quercetin to mouse ileum and colon on serosal side resulted in activation of CFTR and CaCC-mediated Cl− currents. Notably, we found that quercetin exhibited inhibitory effect against ANO1 chloride channel activity in ANO1-expressing FRT cells and decreased mouse intestinal motility. Quercetin-stimulated short-circuit currents in mouse ileum was multi-component, which included elevation of Ca2+ concentration through L-type calcium channel and activation of basolateral NKCC, Na+/K+-ATPase, and K+ channels. In vivo studies further revealed that quercetin promoted fluid secretion in mouse ileum. The modulatory effect of quercetin on CaCC chloirde channels may therefore represent a potential therapeutic strategy for treating CaCC-related diseases like constipation, secretory diarrhea and hypertension. The inverse effects of quercetin on CaCCs provided evidence that ANO1 and intestinal epithelial CaCCs are different calcium-activated chloride channels.
Collapse
Affiliation(s)
- Bo Yu
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| | - Yu Jiang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| | - Lingling Jin
- College of Basic Medical Sciences, Dalian Medical University Dalian, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University Dalian, China
| | - Hong Yang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| |
Collapse
|
35
|
Delpire E, Wolfe L, Flores B, Koumangoye R, Schornak CC, Omer S, Pusey B, Lau C, Markello T, Adams DR. A patient with multisystem dysfunction carries a truncation mutation in human SLC12A2, the gene encoding the Na-K-2Cl cotransporter, NKCC1. Cold Spring Harb Mol Case Stud 2016; 2:a001289. [PMID: 27900370 PMCID: PMC5111002 DOI: 10.1101/mcs.a001289] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/10/2016] [Indexed: 12/30/2022] Open
Abstract
This study describes a 13-yr-old girl with orthostatic intolerance, respiratory weakness, multiple endocrine abnormalities, pancreatic insufficiency, and multiorgan failure involving the gut and bladder. Exome sequencing revealed a de novo, loss-of-function allele in SLC12A2, the gene encoding the Na-K-2Cl cotransporter-1. The 11-bp deletion in exon 22 results in frameshift (p.Val1026Phefs*2) and truncation of the carboxy-terminal tail of the cotransporter. Preliminary studies in heterologous expression systems demonstrate that the mutation leads to a nonfunctional transporter, which is expressed and trafficked to the plasma membrane alongside wild-type NKCC1. The truncated protein, visible at higher molecular sizes, indicates either enhanced dimerization or misfolded aggregate. No significant dominant-negative effect was observed. K+ transport experiments performed in fibroblasts from the patient showed reduced total and NKCC1-mediated K+ influx. The absence of a bumetanide effect on K+ influx in patient fibroblasts only under hypertonic conditions suggests a deficit in NKCC1 regulation. We propose that disruption in NKCC1 function might affect sensory afferents and/or smooth muscle cells, as their functions depend on NKCC1 creating a Cl- gradient across the plasma membrane. This Cl- gradient allows the γ-aminobutyric acid (GABA) receptor or other Cl- channels to depolarize the membrane affecting processes such as neurotransmission or cell contraction. Under this hypothesis, disrupted sensory and smooth muscle function in a diverse set of tissues could explain the patient's phenotype.
Collapse
Affiliation(s)
- Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Lynne Wolfe
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Bianca Flores
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Rainelli Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Cara C Schornak
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Salma Omer
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Barbara Pusey
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Christopher Lau
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Thomas Markello
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - David R Adams
- Undiagnosed Diseases Program, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
36
|
Ma K, Wang H, Yu J, Wei M, Xiao Q. New Insights on the Regulation of Ca 2+ -Activated Chloride Channel TMEM16A. J Cell Physiol 2016; 232:707-716. [PMID: 27682822 DOI: 10.1002/jcp.25621] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022]
Abstract
TMEM16A, also known as anoctamin 1, is a recently identified Ca2+ -activated chloride channel and the first member of a 10-member TMEM16 family. TMEM16A dysfunction is implicated in many diseases such as cancer, hypertension, and cystic fibrosis. TMEM16A channels are well known to be dually regulated by voltage and Ca2+ . In addition, recent studies have revealed that TMEM16A channels are regulated by many molecules such as calmodulin, protons, cholesterol, and phosphoinositides, and a diverse range of stimuli such as thermal and mechanical stimuli. A better understanding of the regulatory mechanisms of TMEM16A is important to understand its physiological and pathological role. Recently, the crystal structure of a TMEM16 family member from the fungus Nectria haematococcaten (nhTMEM16) is discovered, and provides valuable information for studying the structure and function of TMEM16A. In this review, we discuss the structure and function of TMEM16A channels based on the crystal structure of nhTMEM16A and focus on the regulatory mechanisms of TMEM16A channels. J. Cell. Physiol. 232: 707-716, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Jiankun Yu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| |
Collapse
|
37
|
Jiang Y, Yu B, Yang H, Ma T. Shikonin Inhibits Intestinal Calcium-Activated Chloride Channels and Prevents Rotaviral Diarrhea. Front Pharmacol 2016; 7:270. [PMID: 27601995 PMCID: PMC4993765 DOI: 10.3389/fphar.2016.00270] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/10/2016] [Indexed: 01/26/2023] Open
Abstract
Secretory diarrhea remains a global health burden and causes major mortality in children. There have been some focuses on antidiarrheal therapies that may reduce fluid losses and intestinal motility in diarrheal diseases. In the present study, we identified shikonin as an inhibitor of TMEM16A chloride channel activity using cell-based fluorescent-quenching assay. The IC50 value of shikonin was 6.5 μM. Short-circuit current measurements demonstrated that shikonin inhibited Eact-induced Cl(-) current in a dose-dependent manner, with IC50 value of 1.5 μM. Short-circuit current measurement showed that shikonin exhibited inhibitory effect against CCh-induced Cl(-) currents in mouse colonic epithelia but did not affect cytoplasmic Ca(2+) concentration as well as the other major enterocyte chloride channel conductance regulator. Characterization study found that shikonin inhibited basolateral K(+) channel activity without affecting Na(+)/K(+)-ATPase activities. In vivo studies revealed that shikonin significantly delayed intestinal motility in mice and reduced stool water content in a neonatal mice model of rotaviral diarrhea without affecting the viral infection process in vivo. Taken together, the results suggested that shikonin inhibited enterocyte calcium-activated chloride channels, the inhibitory effect was partially through inhbition of basolateral K(+) channel activity, and shikonin could be a lead compound in the treatment of rotaviral secretory diarrhea.
Collapse
Affiliation(s)
- Yu Jiang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| | - Bo Yu
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| | - Hong Yang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Sciences, Liaoning Normal University Dalian, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University Dalian, China
| |
Collapse
|
38
|
Deng L, Yang J, Chen H, Ma B, Pan K, Su C, Xu F, Zhang J. Knockdown of TMEM16A suppressed MAPK and inhibited cell proliferation and migration in hepatocellular carcinoma. Onco Targets Ther 2016; 9:325-33. [PMID: 26834491 PMCID: PMC4716773 DOI: 10.2147/ott.s95985] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
TMEM16A plays an important role in cell proliferation in various cancers. However, less was known about the expression and role of TMEM16A in hepatocellular carcinoma. We screened the expression of TMEM16A in patients' hepatocellular carcinoma tissues, and also analyzed the biological function of hepatocellular carcinoma cells by knockdown of TMEM16A, as well as the expression of MAPK signaling proteins, including p38, p-p38, ERK1/2, p-ERK1/2, JNK, and p-JNK, and cell cycle regulatory protein cyclin D1 in TMEM16A siRNA-transfected SMMC-7721 cells by Western blot. Our results showed that TMEM16A was overexpressed in hepatocellular carcinoma tissues. Inhibition of TMEM16A suppressed the cell proliferation, migration, and invasion, and cell cycle progression but did not influence the cell apoptosis. TMEM16A siRNA-suppressed cancer cell proliferation and tumor growth were accompanied by a reduction of p38 and ERK1/2 activation and cyclin D1 induction, and were not influenced by other tested MAPK signaling proteins. In addition, inhibition of TMEM16A suppressed tumorigenicity in vivo. TMEM16A is overexpressed in hepatocellular carcinoma, and that inhibition of TMEM16A suppressed MAPK and growth of hepatocellular carcinoma. TMEM16A could be a potentially novel therapeutic target for human cancers, including hepatocellular carcinoma.
Collapse
Affiliation(s)
- Liang Deng
- Department of Hepatobiliary Surgery, The Eastern Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jihong Yang
- Department of General Surgery, The Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Hongwu Chen
- Department of Emergency, The Eastern Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Bo Ma
- Department of Gastroenterology, The Eastern Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Kangming Pan
- Department of Hepatobiliary Surgery, The Eastern Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Caikun Su
- Department of Hepatobiliary Surgery, The Eastern Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Fengfeng Xu
- Department of Hepatobiliary Surgery, The Eastern Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jihong Zhang
- Department of Hepatobiliary Surgery, The Eastern Hospital of the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
39
|
Vykhareva EA, Zamoyski VL, Grigoriev VV, Bachurin SO. Calcium-dependent chloride current in rat cerebellar Purkinje cell membranes. DOKL BIOCHEM BIOPHYS 2016; 465:386-8. [PMID: 26728731 DOI: 10.1134/s1607672915060125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Indexed: 11/23/2022]
Abstract
The presence of calcium-dependent potential-activated chloride currents in the membranes of freshly isolated rat cerebellar Purkinje cells (12-15 days) was shown by the whole-cell patch clamp technique. Chloride currents appeared in a sodium-free external solution and reversibly disappeared in the absence of external chloride and calcium ions.
Collapse
Affiliation(s)
- E A Vykhareva
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Severnyi proezd 1, Chernogolovka, Moscow oblast, 142432, Russia.
| | - V L Zamoyski
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Severnyi proezd 1, Chernogolovka, Moscow oblast, 142432, Russia
| | - V V Grigoriev
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Severnyi proezd 1, Chernogolovka, Moscow oblast, 142432, Russia
| | - S O Bachurin
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Severnyi proezd 1, Chernogolovka, Moscow oblast, 142432, Russia
| |
Collapse
|
40
|
Sun Y, Birnbaumer L, Singh BB. TRPC1 regulates calcium-activated chloride channels in salivary gland cells. J Cell Physiol 2015; 230:2848-56. [PMID: 25899321 PMCID: PMC4872598 DOI: 10.1002/jcp.25017] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/14/2015] [Indexed: 12/19/2022]
Abstract
Calcium-activated chloride channel (CaCC) plays an important role in modulating epithelial secretion. It has been suggested that in salivary tissues, sustained fluid secretion is dependent on Ca(2+) influx that activates ion channels such as CaCC to initiate Cl(-) efflux. However direct evidence as well as the molecular identity of the Ca(2+) channel responsible for activating CaCC in salivary tissues is not yet identified. Here we provide evidence that in human salivary cells, an outward rectifying Cl(-) current was activated by increasing [Ca(2+)]i, which was inhibited by the addition of pharmacological agents niflumic acid (NFA), an antagonist of CaCC, or T16Ainh-A01, a specific TMEM16a inhibitor. Addition of thapsigargin (Tg), that induces store-depletion and activates TRPC1-mediated Ca(2+) entry, potentiated the Cl(-) current, which was inhibited by the addition of a non-specific TRPC channel blocker SKF96365 or removal of external Ca(2+). Stimulation with Tg also increased plasma membrane expression of TMEM16a protein, which was also dependent on Ca(2+) entry. Importantly, in salivary cells, TRPC1 silencing, but not that of TRPC3, inhibited CaCC especially upon store depletion. Moreover, primary acinar cells isolated from submandibular gland also showed outward rectifying Cl(-) currents upon increasing [Ca(2+)]i. These Cl(-) currents were again potentiated with the addition of Tg, but inhibited in the presence of T16Ainh-A01. Finally, acinar cells isolated from the submandibular glands of TRPC1 knockout mice showed significant inhibition of the outward Cl(-) currents without decreasing TMEM16a expression. Together the data suggests that Ca(2+) entry via the TRPC1 channels is essential for the activation of CaCC.
Collapse
Affiliation(s)
- Yuyang Sun
- Department of Basic Sciences, School of Medicine Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Lutz Birnbaumer
- Laboratory of Signal Transduction, NIHES, NIH, Research Triangle Park, North Carolina
| | - Brij B Singh
- Department of Basic Sciences, School of Medicine Health Sciences, University of North Dakota, Grand Forks, North Dakota
| |
Collapse
|
41
|
Silveira JC, Kopp PA. Pendrin and anoctamin as mediators of apical iodide efflux in thyroid cells. Curr Opin Endocrinol Diabetes Obes 2015; 22:374-80. [PMID: 26313899 DOI: 10.1097/med.0000000000000188] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW Thyroid hormones are essential for normal development, growth, and metabolism. Their synthesis occurs in thyroid follicles and requires an adequate iodide supply and a sequence of regulated biochemical steps. The uptake of iodide into thyrocytes is well characterized, but its efflux at the apical membrane is poorly understood. This review discusses potential mechanisms underlying iodide efflux with emphasis on recent developments and controversies. RECENT FINDINGS The functional characterization of pendrin (PDS/SLC26A4), a multifunctional anion exchanger, suggested that it could be involved in mediating iodide efflux. This is supported by the phenotype of patients with Pendred syndrome (deafness, goiter, partial iodide organification defect), which is caused by biallelic mutations in the SLC26A4 gene, as well as functional studies. However, apical iodide efflux is also possible in the absence of pendrin, implicating the presence of at least another channel. Recently, Anoctamin 1 (TMEM16A), a calcium-activated anion channel has been identified at the apical membrane of thyrocytes and functional studies suggest that it may play a predominant role in mediating iodide efflux. SUMMARY Anoctamin and pendrin are two plausible candidates as mediators of apical iodide efflux. Their relative affinity for iodide and their exact physiological role await, however, further characterization.
Collapse
Affiliation(s)
- Jamile C Silveira
- aDivision of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA bDepartment of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
42
|
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
|
43
|
Matchkov VV, Boedtkjer DM, Aalkjaer C. The role of Ca2+ activated Cl− channels in blood pressure control. Curr Opin Pharmacol 2015; 21:127-37. [DOI: 10.1016/j.coph.2015.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 12/17/2022]
|
44
|
Amjad A, Hernandez-Clavijo A, Pifferi S, Maurya DK, Boccaccio A, Franzot J, Rock J, Menini A. Conditional knockout of TMEM16A/anoctamin1 abolishes the calcium-activated chloride current in mouse vomeronasal sensory neurons. ACTA ACUST UNITED AC 2015; 145:285-301. [PMID: 25779870 PMCID: PMC4380210 DOI: 10.1085/jgp.201411348] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
TMEM16A is an essential component of Ca2+-activated Cl− currents in mouse vomeronasal sensory neurons. Pheromones are substances released from animals that, when detected by the vomeronasal organ of other individuals of the same species, affect their physiology and behavior. Pheromone binding to receptors on microvilli on the dendritic knobs of vomeronasal sensory neurons activates a second messenger cascade to produce an increase in intracellular Ca2+ concentration. Here, we used whole-cell and inside-out patch-clamp analysis to provide a functional characterization of currents activated by Ca2+ in isolated mouse vomeronasal sensory neurons in the absence of intracellular K+. In whole-cell recordings, the average current in 1.5 µM Ca2+ and symmetrical Cl− was −382 pA at −100 mV. Ion substitution experiments and partial blockade by commonly used Cl− channel blockers indicated that Ca2+ activates mainly anionic currents in these neurons. Recordings from inside-out patches from dendritic knobs of mouse vomeronasal sensory neurons confirmed the presence of Ca2+-activated Cl− channels in the knobs and/or microvilli. We compared the electrophysiological properties of the native currents with those mediated by heterologously expressed TMEM16A/anoctamin1 or TMEM16B/anoctamin2 Ca2+-activated Cl− channels, which are coexpressed in microvilli of mouse vomeronasal sensory neurons, and found a closer resemblance to those of TMEM16A. We used the Cre–loxP system to selectively knock out TMEM16A in cells expressing the olfactory marker protein, which is found in mature vomeronasal sensory neurons. Immunohistochemistry confirmed the specific ablation of TMEM16A in vomeronasal neurons. Ca2+-activated currents were abolished in vomeronasal sensory neurons of TMEM16A conditional knockout mice, demonstrating that TMEM16A is an essential component of Ca2+-activated Cl− currents in mouse vomeronasal sensory neurons.
Collapse
Affiliation(s)
- Asma Amjad
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| | - Andres Hernandez-Clavijo
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| | - Simone Pifferi
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| | - Devendra Kumar Maurya
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| | - Anna Boccaccio
- Istituto di Biofisica, National Research Council, 16149 Genova, Italy
| | - Jessica Franzot
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| | - Jason Rock
- Department of Anatomy, University of California, San Francisco, School of Medicine, San Francisco, CA 94143
| | - Anna Menini
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Trieste, Italy
| |
Collapse
|
45
|
Lin J, Jiang Y, Li L, Liu Y, Tang H, Jiang D. TMEM16A mediates the hypersecretion of mucus induced by Interleukin-13. Exp Cell Res 2015; 334:260-9. [PMID: 25770012 DOI: 10.1016/j.yexcr.2015.02.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 02/26/2015] [Accepted: 02/28/2015] [Indexed: 10/23/2022]
Abstract
Previous studies showed that the Ca(2+)-activated Cl(-) channel (CaCC) was involved in the pathogenesis of mucus hypersecretion induced by Interleukin-13 (IL-13). However, the mechanisms underlying the process were unknown. Recently, transmembrane protein 16A (TMEM16A) was identified as the channel underlying the CaCC current. The aim of the current study was to investigate whether the TMEM16A channel is part of the mechanism underlying IL-13-induced mucus hypersecretion. We observed that both TMEM16A mRNA and protein expression were significantly up-regulated after treatment with IL-13 in human bronchial epithelial 16 (HBE 16) cells, which correlated with an increase in mucus production. Additionally, mucus hypersecretion in rat airways was induced by intratracheal instillation of IL-13 and similar increases were observed in the expression of TMEM16A mRNA and protein in the bronchial epithelium. Niflumic acid (NA), a selective antagonist of CaCC, markedly blocked IL-13-induced mucin (MUC) 5AC mRNA and protein production in vivo and in vitro. Further investigation with HBE16 cells revealed that TMEM16A overexpression clearly promoted mucus production, IκBα phosphorylation, and p65 accumulation in the nucleus. The loss of TMEM16A resulted in inhibition of mucus production, and the TMEM16A-mediated production of MUC5AC was significantly blocked by a nuclear factor-kappa B (NF-κB) inhibitor. Therefore, the TMEM16A channel acts upstream of NF-κB in the regulation of mucus production. This is the first demonstration that the TMEM16A-NF-κB pathway is positively involved in IL-13-induced mucus production, which provides novel insight into the molecular mechanism of mucin overproduction.
Collapse
Affiliation(s)
- Jiachen Lin
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Youfan Jiang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Li Li
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yanan Liu
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Hui Tang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Depeng Jiang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
46
|
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
|
47
|
Jiang Y, Yu B, Fang F, Cao H, Ma T, Yang H. Modulation of Chloride Channel Functions by the Plant Lignan Compounds Kobusin and Eudesmin. FRONTIERS IN PLANT SCIENCE 2015; 6:1041. [PMID: 26635857 PMCID: PMC4658577 DOI: 10.3389/fpls.2015.01041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/09/2015] [Indexed: 05/05/2023]
Abstract
Plant lignans are diphenolic compounds widely present in vegetables, fruits, and grains. These compounds have been demonstrated to have protective effect against cancer, hypertension and diabetes. In the present study, we showed that two lignan compounds, kobusin and eudesmin, isolated from Magnoliae Flos, could modulate intestinal chloride transport mediated by cystic fibrosis transmembrane conductance regulator (CFTR) and calcium-activated chloride channels (CaCCs). The compounds activated CFTR channel function in both FRT cells and in HT-29 cells. The modulating effects of kobusin and eudesmin on the activity of CaCCgie (CaCC expressed in gastrointestinal epithelial cells) were also investigated, and the result showed that both compounds could stimulate CaCCgie-mediated short-circuit currents and the stimulation was synergistic with ATP. In ex vivo studies, both compounds activated CFTR and CaCCgie chloride channel activities in mouse colonic epithelia. Remarkably, the compounds showed inhibitory effects toward ANO1/CaCC-mediated short-circuit currents in ANO1/CaCC-expressing FRT cells, with IC50 values of 100 μM for kobusin and 200 μM for eudesmin. In charcoal transit study, both compounds mildly reduced gastrointestinal motility in mice. Taken together, these results revealed a new kind of activity displayed by the lignan compounds, one that is concerned with the modulation of chloride channel function.
Collapse
Affiliation(s)
- Yu Jiang
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University Dalian, China
| | - Bo Yu
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University Dalian, China
| | - Fang Fang
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University Dalian, China
| | - Huanhuan Cao
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University Dalian, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University Dalian, China
| | - Hong Yang
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University Dalian, China
| |
Collapse
|
48
|
Sui Y, Sun M, Wu F, Yang L, Di W, Zhang G, Zhong L, Ma Z, Zheng J, Fang X, Ma T. Inhibition of TMEM16A expression suppresses growth and invasion in human colorectal cancer cells. PLoS One 2014; 9:e115443. [PMID: 25541940 PMCID: PMC4277312 DOI: 10.1371/journal.pone.0115443] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 11/23/2014] [Indexed: 12/12/2022] Open
Abstract
Metastasis leads to poor prognosis in colorectal cancer patients, and there is a growing need for new therapeutic targets. TMEM16A (ANO1, DOG1 or TAOS2) has recently been identified as a calcium-activated chloride channel (CaCC) and is reported to be overexpressed in several malignancies; however, its expression and function in colorectal cancer (CRC) remains unclear. In this study, we found expression of TMEM16A mRNA and protein in high-metastatic-potential SW620, HCT116 and LS174T cells, but not in primary HCT8 and SW480 cells, using RT-PCR, western blotting and immunofluorescence labeling. Patch-clamp recordings detected CaCC currents regulated by intracellular Ca(2+) and voltage in SW620 cells. Knockdown of TMEM16A by short hairpin RNAs (shRNA) resulted in the suppression of growth, migration and invasion of SW620 cells as detected by MTT, wound-healing and transwell assays. Mechanistically, TMEM16A depletion was accompanied by the dysregulation of phospho-MEK, phospho-ERK1/2 and cyclin D1 expression. Flow cytometry analysis showed that SW620 cells were inhibited from the G1 to S phase of the cell cycle in the TMEM16A shRNA group compared with the control group. In conclusion, our results indicate that TMEM16A CaCC is involved in growth, migration and invasion of metastatic CRC cells and provide evidence for TMEM16A as a potential drug target for treating metastatic colorectal carcinoma.
Collapse
Affiliation(s)
- Yujie Sui
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Meiyan Sun
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Fei Wu
- Department of Gynecology and Obstetrics, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Longfei Yang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Weihua Di
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Guizhen Zhang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Lili Zhong
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Zhiming Ma
- Department of General Surgery, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Jinhao Zheng
- Department of General Surgery, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Xuedong Fang
- Department of General Surgery, Jilin University Bethune Second Hospital, Changchun, P. R. China
- Department of General Surgery, China-Japan Friendship Hospital of Jilin University, Changchun, P. R. China
- * E-mail: (XDF); (THM)
| | - Tonghui Ma
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
- College of Basic Medical Sciences, Dalian Medical University, Dalian, P. R. China
- * E-mail: (XDF); (THM)
| |
Collapse
|
49
|
Jin X, Shah S, Du X, Zhang H, Gamper N. Activation of Ca(2+) -activated Cl(-) channel ANO1 by localized Ca(2+) signals. J Physiol 2014; 594:19-30. [PMID: 25398532 PMCID: PMC4704509 DOI: 10.1113/jphysiol.2014.275107] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022] Open
Abstract
Ca2+‐activated chloride channels (CaCCs) regulate numerous physiological processes including epithelial transport, smooth muscle contraction and sensory processing. Anoctamin‐1 (ANO1, TMEM16A) is a principal CaCC subunit in many cell types, yet our understanding of the mechanisms of ANO1 activation and regulation are only beginning to emerge. Ca2+ sensitivity of ANO1 is rather low and at negative membrane potentials the channel requires several micromoles of intracellular Ca2+ for activation. However, global Ca2+ levels in cells rarely reach such levels and, therefore, there must be mechanisms that focus intracellular Ca2+ transients towards the ANO1 channels. Recent findings indeed indicate that ANO1 channels often co‐localize with sources of intracellular Ca2+ signals. Interestingly, it appears that in many cell types ANO1 is particularly tightly coupled to the Ca2+ release sites of the intracellular Ca2+ stores. Such preferential coupling may represent a general mechanism of ANO1 activation in native tissues.
Collapse
Affiliation(s)
- Xin Jin
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Sihab Shah
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.,Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
50
|
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
|