1
|
Hu Y, Zhang Y, He J, Rao H, Zhang D, Shen Z, Zhou C. ANO1: central role and clinical significance in non-neoplastic and neoplastic diseases. Front Immunol 2025; 16:1570333. [PMID: 40356890 PMCID: PMC12067801 DOI: 10.3389/fimmu.2025.1570333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/20/2025] [Indexed: 05/15/2025] Open
Abstract
Anoctamin 1 (ANO1), also known as TMEM16A, is a multifunctional protein that serves as a calcium-activated chloride channel (CaCC). It is ubiquitously expressed across various tissues, including epithelial cells, smooth muscle cells, and neurons, where it is integral to physiological processes such as epithelial secretion, smooth muscle contraction, neural conduction, and cell proliferation and migration. Dysregulation of ANO1 has been linked to the pathogenesis of numerous diseases. Extensive research has established its involvement in non-neoplastic conditions such as asthma, hypertension, and gastrointestinal (GI) dysfunction. Moreover, ANO1 has garnered significant attention for its role in the development and progression of cancers, including head and neck cancer, breast cancer, and lung cancer, where its overexpression correlates with increased tumor growth, metastasis, and poor prognosis. Additionally, ANO1 regulates multiple signaling pathways, including the epidermal growth factor receptor (EGFR) pathway, the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway, and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway, among others. These pathways are pivotal in regulating cell proliferation, migration, and invasion. Given its central role in these processes, ANO1 has emerged as a promising diagnostic biomarker and therapeutic target. Recent advancements in ANO1 research have highlighted its potential in disease diagnosis and treatment. Strategies targeting ANO1, such as small molecule modulators or gene-silencing techniques, have shown preclinical promise in both non-neoplastic and neoplastic diseases. This review explores the latest findings in ANO1 research, focusing on its mechanistic involvement in disease progression, its regulation, and its therapeutic potential. Modulating ANO1 activity may offer novel therapeutic strategies for effectively treating ANO1-associated diseases.
Collapse
Affiliation(s)
- Yanghao Hu
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Yifei Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Jiali He
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Huihuang Rao
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Duomi Zhang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Chongchang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
2
|
Ghosh S, Alkawadri T, McGarvey LP, Hollywood MA, Thornbury KD, Sergeant GP. Role of voltage-gated Ca 2+ channels and Ano1 Ca 2+-activated Cl - channels in M2 muscarinic receptor-dependent contractions of murine airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2025; 328:L301-L312. [PMID: 39772966 DOI: 10.1152/ajplung.00188.2024] [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: 06/20/2024] [Revised: 12/13/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Cholinergic tone is elevated in obstructive lung conditions such as chronic obstructive pulmonary disease (COPD) and asthma, but the cellular mechanisms underlying cholinergic contractions of airway smooth muscle (ASM) are still unclear. Some studies report an important role for L-type Ca2+ channels (LTCC) and Ano1 Ca2+-activated Cl- channels (CACC) in these responses, but others dispute their importance. Cholinergic contractions of ASM involve activation of M3Rs, however, stimulation of M2Rs exerts a profound hypersensitization of these responses. Here, we show that M2R-dependent potentiation of cholinergic nerve-evoked contractions of ASM was reversed by the LTCC blocker nifedipine and the Ano1 CACC inhibitors Ani9 and CaCCinh-A01. Carbachol induced sustained contractions of ASM that were converted into oscillatory contractions when M3Rs were blocked with 4-DAMP. The 4-DAMP-resistant contractions were absent in preparations taken from M2R knockout (KO) mice. The remaining M2R-dependent responses, observed in wild-type (WT) mice, were abolished by nifedipine and Ani9. Inhibition of sarcoplasmic endoplasmic reticulum Ca2+ ATPases (SERCA) with thapsigargin increased the amplitude of contractions induced by electrical field stimulation (EFS) and these effects were also reversed by nifedipine and Ani9. Thapsigargin also potentiated contractions of ASM induced by the LTCC activator FPL64176. Therefore, contractions of ASM that involved Ca2+ influx via LTCC were enhanced by inhibition of SERCA. Immunocytochemistry experiments revealed prominent SERCA staining around the periphery of ASM cells. These data indicate that M2R-dependent contractions of ASM involve Ano1 CACC and LTCC by a mechanism involving inhibition of buffering of Ca2+ influx by SERCA.NEW & NOTEWORTHY The role of L-type Ca2+ channels and Ano1 Ca2+-activated Cl- channels in cholinergic contractions of airway smooth muscle is disputed. Here, we show that both channels are involved in M2 muscarinic receptor-dependent contractions of murine airway smooth muscle via inhibition of buffering of Ca2+ influx by sarcoplasmic endoplasmic reticulum Ca2+ ATPases.
Collapse
Affiliation(s)
- Srijit Ghosh
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Co. Louth, Ireland
| | - Tuleen Alkawadri
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Co. Louth, Ireland
| | - Lorcan P McGarvey
- School of Medicine, Dentistry, and Biomedical Sciences, Queen's University, Belfast, Northern Ireland
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Co. Louth, Ireland
| | - Keith D Thornbury
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Co. Louth, Ireland
| | - Gerard P Sergeant
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Co. Louth, Ireland
| |
Collapse
|
3
|
Mažerik J, Gondáš E, Dohál M, Smieško L, Jošková M, Fraňová S, Šutovská M. Targeting TMEM16A ion channels suppresses airway hyperreactivity, inflammation, and remodeling in an experimental Guinea pig asthma model. J Pharmacol Sci 2024; 156:239-246. [PMID: 39608849 DOI: 10.1016/j.jphs.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/07/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
Asthma is a chronic inflammatory disease characterized by airway hyperresponsiveness, inflammation, and remodeling. Calcium (Ca2+)-activated chloride (Cl-) channels, such as TMEM16A, are inferred to be involved in asthma. Therefore, the present study investigated the therapeutic potential of TMEM16A inhibition in a guinea pig model of ovalbumin (OVA)-induced allergic asthma. Guinea pigs were treated with a specific blocker, CaCCinh-A01 (10 μM), administered via inhalation. A significant reduction in cough reflex sensitivity and specific airway resistance was observed in animals treated with CaCCinh-A01, highlighting its potential to improve airway function. Despite a reduction in ciliary beating frequency (CBF), CaCCinh-A01 reduced airway mucus viscosity by decreasing the production of mucin-5AC (MUC5AC). The nonspecific reduction in the Th1/Th2 cytokine spectrum following CaCCinh-A01 treatment indicated the suppression of airway inflammation. Additionally, markers associated with airway remodeling were diminished, suggesting that CaCCinh-A01 may counteract structural changes in airway tissues. Therefore, inhibition appears to mitigate the pathological aspects of asthma, including airway hyperresponsiveness, inflammation, and remodeling. However, further studies are required to comprehensively evaluate the potential of TMEM16A as a therapeutic target for asthma.
Collapse
Affiliation(s)
- Jozef Mažerik
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 11161/4B, Martin, Slovakia.
| | - Eduard Gondáš
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 11161/4B, Martin, Slovakia
| | - Matúš Dohál
- Biomedical Centre, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 11161/4C, Martin, Slovakia
| | - Lukáš Smieško
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 11161/4B, Martin, Slovakia
| | - Marta Jošková
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 11161/4B, Martin, Slovakia
| | - Soňa Fraňová
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 11161/4B, Martin, Slovakia
| | - Martina Šutovská
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 11161/4B, Martin, Slovakia
| |
Collapse
|
4
|
Kinsella JA, Debant M, Parsonage G, Morley LC, Bajarwan M, Revill C, Foster R, Beech DJ. Pharmacology of PIEZO1 channels. Br J Pharmacol 2024; 181:4714-4732. [PMID: 39402010 DOI: 10.1111/bph.17351] [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: 02/06/2024] [Revised: 06/15/2024] [Accepted: 08/25/2024] [Indexed: 11/05/2024] Open
Abstract
PIEZO1 is a eukaryotic membrane protein that assembles as trimers to form calcium-permeable, non-selective cation channels with exquisite capabilities for mechanical force sensing and transduction of force into effect in diverse cell types that include blood cells, endothelial cells, epithelial cells, fibroblasts and stem cells and diverse systems that include bone, lymphatics and muscle. The channel has wide-ranging roles and is considered as a target for novel therapeutics in ailments spanning cancers and cardiovascular, dental, gastrointestinal, hepatobiliary, infectious, musculoskeletal, nervous system, ocular, pregnancy, renal, respiratory and urological disorders. The identification of PIEZO1 modulators is in its infancy but useful experimental tools emerged for activating, and to a lesser extent inhibiting, the channels. Elementary structure-activity relationships are known for the Yoda series of small molecule agonists, which show the potential for diverse physicochemical and pharmacological properties. Intriguing effects of Yoda1 include the stimulated removal of excess cerebrospinal fluid. Despite PIEZO1's broad expression, opportunities are suggested for selective positive or negative modulation without intolerable adverse effects. Here we provide a focused, non-systematic, narrative review of progress with this pharmacology and discuss potential future directions for research in the area.
Collapse
Affiliation(s)
- Jacob A Kinsella
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
- School of Chemistry, University of Leeds, Leeds, UK
| | - Marjolaine Debant
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Gregory Parsonage
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lara C Morley
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Muath Bajarwan
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | | | | | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
5
|
Liu XM, Li J, Chen D, Li H, Qin XY, Wang YX, Gu YZ, Li N, Zhou LG, Feng M. Ano1 regulates embryo transport by modulating intracellular calcium levels in oviduct smooth muscle. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167059. [PMID: 38336104 DOI: 10.1016/j.bbadis.2024.167059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/26/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Oviductal smooth muscle exhibits spontaneous rhythmic contraction (SRC) and controls the passage of the ova at the exact time, but its mechanistic regulation remains to be determined. In this study, female mice with Ano1SMKO (smooth muscle-specific deletion of Ano1) had reduced fertility. Deficiency of Ano1 in mice resulted in impaired oviductal SRC function and reduced calcium signaling in individual smooth muscle cells in the oviduct. The Ano1 antagonist T16Ainh-A01 dose-dependently inhibited SRCs and [Ca2+]i in the oviducts of humans and mice. A similar inhibitory effect of SRCs and [Ca2+]i was observed after treatment with nifedipine. In our study, ANO1 acted primarily as an activator or amplifier in [Ca2+]i and contraction of tubal smooth muscle cells. We found that tubal SRC was markedly attenuated in patients with ectopic pregnancy. Then, our study was designed to determine whether chloride channel Ano1-mediated smooth muscle motility is associated with tubal SRC. Our findings reveal a new mechanism for the regulation of tubal motility that may be associated with abnormal pregnancies such as ectopic pregnancies.
Collapse
Affiliation(s)
- Xiao-Man Liu
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China; Institute of Clinical Microbiology, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, PR China
| | - Juan Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Defang Chen
- Office of Operation Management Committee, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, PR China
| | - Hao Li
- Department of Blood Transfusion, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Xiao-Yan Qin
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Yun-Xia Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Yong-Zhong Gu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Na Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Li-Guang Zhou
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China
| | - Mei Feng
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, PR China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, PR China; Institute of Clinical Microbiology, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, PR China.
| |
Collapse
|
6
|
Ousingsawat J, Centeio R, Reyne N, McCarron A, Cmielewski P, Schreiber R, diStefano G, Römermann D, Seidler U, Donnelley M, Kunzelmann K. Inhibition of mucus secretion by niclosamide and benzbromarone in airways and intestine. Sci Rep 2024; 14:1464. [PMID: 38233410 PMCID: PMC10794189 DOI: 10.1038/s41598-024-51397-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
The Ca2+ activated Cl- channel TMEM16A (anoctamin 1; ANO1) is expressed in secretory epithelial cells of airways and intestine. Previous studies provided evidence for a role of ANO1 in mucus secretion. In the present study we investigated the effects of the two ANO1-inhibitors niclosamide (Niclo) and benzbromarone (Benz) in vitro and in vivo in mouse models for cystic fibrosis (CF) and asthma. In human CF airway epithelial cells (CFBE), Ca2+ increase and activation of ANO1 by adenosine triphosphate (ATP) or ionomycin was strongly inhibited by 200 nM Niclo and 1 µM Benz. In asthmatic mice airway mucus secretion was inhibited by intratracheal instillation of Niclo or Benz. In homozygous F508del-cftr mice, intestinal mucus secretion and infiltration by CD45-positive cells was inhibited by intraperitoneal injection of Niclo (13 mg/kg/day for 7 days). In homozygous F508del-cftr rats intestinal mucus secretion was inhibited by oral application of Benz (5 mg/kg/day for 60 days). Taken together, well tolerated therapeutic concentrations of niclosamide and benzbromarone corresponding to plasma levels of treated patients, inhibit ANO1 and intracellular Ca2+ signals and may therefore be useful in inhibiting mucus hypersecretion and mucus obstruction in airways and intestine of patients suffering from asthma and CF, respectively.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University Street 31, 93053, Regensburg, Germany
| | - Raquel Centeio
- Physiological Institute, University of Regensburg, University Street 31, 93053, Regensburg, Germany
| | - Nicole Reyne
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Alexandra McCarron
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Patricia Cmielewski
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, University Street 31, 93053, Regensburg, Germany
| | - Gabriella diStefano
- Department of Gastroenterology, Hannover Medical School, 30625, Hannover, Germany
| | - Dorothee Römermann
- Department of Gastroenterology, Hannover Medical School, 30625, Hannover, Germany
| | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, 30625, Hannover, Germany
| | - Martin Donnelley
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, University Street 31, 93053, Regensburg, Germany.
| |
Collapse
|
7
|
Pongkorpsakol P, Yimnual C, Satianrapapong W, Worakajit N, Kaewin S, Saetang P, Rukachaisirikul V, Muanprasat C. Discovery of Fungus-Derived Nornidulin as a Novel TMEM16A Inhibitor: A Potential Therapy to Inhibit Mucus Secretion in Asthma. J Exp Pharmacol 2023; 15:449-466. [PMID: 38026233 PMCID: PMC10657771 DOI: 10.2147/jep.s427594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Inhibition of Ca2+-activated transmembrane protein 16A (TMEM16A) Cl- channels has been proposed to alleviate mucus secretion in asthma. In this study, we identified a novel class of TMEM16A inhibitors from natural sources in airway epithelial Calu-3 cells and determine anti-asthmatic efficacy of the most potent candidate in a mouse model of asthma. Methods For electrophysiological analyses, IL-4-primed Calu-3 cell monolayers were mounted in Ussing chamber and treated with various fungus-derived depsidones prior to the addition of UTP, ionomycin, thapsigargin, or Eact to stimulate TMEM16A Cl- current. Ca2+-induced mucus secretion in Calu-3 cell monolayers was assessed by determining MUC5AC protein remaining in the cells using immunofluorescence staining. OVA-induced female BALB/c mice was used as an animal model of asthma. After the course of induction, cellular and mucus components in bronchoalveolar lavage were analyzed. Lungs were fixed and undergone with H&E and PAS staining for the evaluation of airway inflammation and mucus production, respectively. Results The screening of fungus-derived depsidones revealed that nornidulin completely abolished the UTP-activated TMEM16A current in Calu-3 cell monolayers with the IC50 and a maximal effect being at ~0.8 µM and 10 µM, respectively. Neither cell viability nor barrier function was affected by nornidulin. Mechanistically, nornidulin (10 µM) suppressed Cl- currents induced by ionomycin (a Ca2+-specific ionophore), thapsigargin (an inhibitor of the endoplasmic reticulum Ca2+ ATPase), and Eact (a putative TMEM16A activator) without interfering with intracellular Ca2+ ([Ca2+]i) levels. These results suggest that nornidulin exerts its effect without changing [Ca2+]i, possibly through direct effect on TMEM16A. Interestingly, nornidulin (at 10 µM) reduced Ca2+-dependent mucus release in the Calu-3 cell monolayers. In addition, nornidulin (20 mg/kg) inhibited bronchoalveolar mucus secretion without impeding airway inflammation in ovalbumin-induced asthmatic mice. Discussion and Conclusion Our study revealed that nornidulin is a novel TMEM16A inhibitor that suppresses mucus secretion without compromising immunologic activity. Further development of nornidulin may provide a new remedy for asthma or other diseases associated with allergic mucus hypersecretion without causing opportunistic infections.
Collapse
Affiliation(s)
- Pawin Pongkorpsakol
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Chantapol Yimnual
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | | | - Nichakorn Worakajit
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suchada Kaewin
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Praphatsorn Saetang
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Vatcharin Rukachaisirikul
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| |
Collapse
|
8
|
Figueiredo IAD, Ferreira SRD, Fernandes JM, Silva BA, Vasconcelos LHC, Cavalcante FA. A review of the pathophysiology and the role of ion channels on bronchial asthma. Front Pharmacol 2023; 14:1236550. [PMID: 37841931 PMCID: PMC10568497 DOI: 10.3389/fphar.2023.1236550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Asthma is one of the main non-communicable chronic diseases and affects a huge portion of the population. It is a multifactorial disease, classified into several phenotypes, being the allergic the most frequent. The pathophysiological mechanism of asthma involves a Th2-type immune response, with high concentrations of allergen-specific immunoglobulin E, eosinophilia, hyperreactivity and airway remodeling. These mechanisms are orchestrated by intracellular signaling from effector cells, such as lymphocytes and eosinophils. Ion channels play a fundamental role in maintaining the inflammatory response on asthma. In particular, transient receptor potential (TRP), stock-operated Ca2+ channels (SOCs), Ca2+-activated K+ channels (IKCa and BKCa), calcium-activated chloride channel (TMEM16A), cystic fibrosis transmembrane conductance regulator (CFTR), piezo-type mechanosensitive ion channel component 1 (PIEZO1) and purinergic P2X receptor (P2X). The recognition of the participation of these channels in the pathological process of asthma is important, as they become pharmacological targets for the discovery of new drugs and/or pharmacological tools that effectively help the pharmacotherapeutic follow-up of this disease, as well as the more specific mechanisms involved in worsening asthma.
Collapse
Affiliation(s)
- Indyra Alencar Duarte Figueiredo
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Sarah Rebeca Dantas Ferreira
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Jayne Muniz Fernandes
- Graduação em Farmácia, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Luiz Henrique César Vasconcelos
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Fabiana de Andrade Cavalcante
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| |
Collapse
|
9
|
Kunzelmann K, Ousingsawat J, Kraus A, Park JH, Marquardt T, Schreiber R, Buchholz B. Pathogenic Relationships in Cystic Fibrosis and Renal Diseases: CFTR, SLC26A9 and Anoctamins. Int J Mol Sci 2023; 24:13278. [PMID: 37686084 PMCID: PMC10487509 DOI: 10.3390/ijms241713278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The Cl--transporting proteins CFTR, SLC26A9, and anoctamin (ANO1; ANO6) appear to have more in common than initially suspected, as they all participate in the pathogenic process and clinical outcomes of airway and renal diseases. In the present review, we will therefore concentrate on recent findings concerning electrolyte transport in the airways and kidneys, and the role of CFTR, SLC26A9, and the anoctamins ANO1 and ANO6. Special emphasis will be placed on cystic fibrosis and asthma, as well as renal alkalosis and polycystic kidney disease. In essence, we will summarize recent evidence indicating that CFTR is the only relevant secretory Cl- channel in airways under basal (nonstimulated) conditions and after stimulation by secretagogues. Information is provided on the expressions of ANO1 and ANO6, which are important for the correct expression and function of CFTR. In addition, there is evidence that the Cl- transporter SLC26A9 expressed in the airways may have a reabsorptive rather than a Cl--secretory function. In the renal collecting ducts, bicarbonate secretion occurs through a synergistic action of CFTR and the Cl-/HCO3- transporter SLC26A4 (pendrin), which is probably supported by ANO1. Finally, in autosomal dominant polycystic kidney disease (ADPKD), the secretory function of CFTR in renal cyst formation may have been overestimated, whereas ANO1 and ANO6 have now been shown to be crucial in ADPKD and therefore represent new pharmacological targets for the treatment of polycystic kidney disease.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Physiological Institute, University of Regensburg, University Street 31, 93053 Regensburg, Germany; (J.O.); (R.S.)
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University Street 31, 93053 Regensburg, Germany; (J.O.); (R.S.)
| | - Andre Kraus
- Department of Nephrology and Hypertension, Friedrich Alexander University Erlangen Nuremberg, 91054 Erlangen, Germany; (A.K.); (B.B.)
| | - Julien H. Park
- Department of Pediatrics, University Hospital Münster, 48149 Münster, Germany; (J.H.P.); (T.M.)
| | - Thorsten Marquardt
- Department of Pediatrics, University Hospital Münster, 48149 Münster, Germany; (J.H.P.); (T.M.)
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, University Street 31, 93053 Regensburg, Germany; (J.O.); (R.S.)
| | - Björn Buchholz
- Department of Nephrology and Hypertension, Friedrich Alexander University Erlangen Nuremberg, 91054 Erlangen, Germany; (A.K.); (B.B.)
| |
Collapse
|
10
|
Gao J, Yin H, Dong Y, Wang X, Liu Y, Wang K. A Novel Role of Uricosuric Agent Benzbromarone in BK Channel Activation and Reduction of Airway Smooth Muscle Contraction. Mol Pharmacol 2023; 103:241-254. [PMID: 36669879 DOI: 10.1124/molpharm.122.000638] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/28/2022] [Accepted: 12/19/2022] [Indexed: 01/21/2023] Open
Abstract
The uricosuric drug benzbromarone, widely used for treatment of gout, hyperpolarizes the membrane potential of airway smooth muscle cells, but how it works remains unknown. Here we show a novel role of benzbromarone in activation of large conductance calcium-activated K+ channels. Benzbromarone results in dose-dependent activation of macroscopic big potassium (BK) currents about 1.7- to 14.5-fold with an EC50 of 111 μM and shifts the voltage-dependent channel activation to a more hyperpolarizing direction about 10 to 54 mV in whole-cell patch clamp recordings. In single-channel recordings, benzbromarone decreases single BKα channel closed dwell time and increases the channel open probability. Coexpressing β1 subunit also enhances BK activation by benzbromarone with an EC50 of 67 μM and a leftward shift of conductance-voltage (G-V) curve about 44 to 138 mV. Site-directed mutagenesis reveals that a motif of three amino acids 329RKK331 in the cytoplasmic linker between S6 and C-terminal regulator of potassium conductance (RCK) gating ring mediates the pharmacological activation of BK channels by benzbromarone. Further ex vivo assay shows that benzbromarone causes reduction of tracheal strip contraction. Taken together, our findings demonstrate that uricosuric benzbromarone activates BK channels through molecular mechanism of action involving the channel RKK motif of S6-RCK linker. Pharmacological activation of BK channel by benzbromarone causes reduction of tracheal strip contraction, holding a repurposing potential for asthma and pulmonary arterial hypertension or BK channelopathies. SIGNIFICANCE STATEMENT: We describe a novel role of uricosuric agent benzbromarone in big potassium (BK) channel activation and relaxation of airway smooth muscle contraction. In this study, we find that benzbromarone is an activator of the large-conductance Ca2+- and voltage-activated K+ channel (BK channel), which serves numerous cellular functions, including control of smooth muscle contraction. Pharmacological activation of BK channel by the FDA-approved drug benzbromarone may hold repurposing potential for treatment of asthma and pulmonary arterial hypertension or BK channelopathies.
Collapse
Affiliation(s)
- Jian Gao
- Department of Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China (J.G., X.W.); Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China (H.Y., Y.D., Y.L., K.W.); and Institute of Innovative Drugs, Qingdao University, Qingdao, China (Y.L., K.W.)
| | - Hao Yin
- Department of Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China (J.G., X.W.); Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China (H.Y., Y.D., Y.L., K.W.); and Institute of Innovative Drugs, Qingdao University, Qingdao, China (Y.L., K.W.)
| | - Yanqun Dong
- Department of Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China (J.G., X.W.); Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China (H.Y., Y.D., Y.L., K.W.); and Institute of Innovative Drugs, Qingdao University, Qingdao, China (Y.L., K.W.)
| | - Xintong Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China (J.G., X.W.); Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China (H.Y., Y.D., Y.L., K.W.); and Institute of Innovative Drugs, Qingdao University, Qingdao, China (Y.L., K.W.)
| | - Yani Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China (J.G., X.W.); Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China (H.Y., Y.D., Y.L., K.W.); and Institute of Innovative Drugs, Qingdao University, Qingdao, China (Y.L., K.W.)
| | - KeWei Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China (J.G., X.W.); Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China (H.Y., Y.D., Y.L., K.W.); and Institute of Innovative Drugs, Qingdao University, Qingdao, China (Y.L., K.W.)
| |
Collapse
|
11
|
Dwivedi R, Drumm BT, Alkawadri T, Martin SL, Sergeant GP, Hollywood MA, Thornbury KD. The TMEM16A blockers benzbromarone and MONNA cause intracellular Ca2+-release in mouse bronchial smooth muscle cells. Eur J Pharmacol 2023; 947:175677. [PMID: 36967079 DOI: 10.1016/j.ejphar.2023.175677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
We investigated effects of TMEM16A blockers benzbromarone, MONNA, CaCCinhA01 and Ani9 on isometric contractions in mouse bronchial rings and on intracellular calcium in isolated bronchial myocytes. Separate concentrations of carbachol (0.1-10 μM) were applied for 10 min periods to bronchial rings, producing concentration-dependent contractions that were well maintained throughout each application period. Benzbromarone (1 μM) markedly reduced the contractions with a more pronounced effect on their sustained component (at 10 min) compared to their initial component (at 2 min). Iberiotoxin (0.3 μM) enhanced the contractions, but they were still blocked by benzbromarone. MONNA (3 μM) and CaCCinhA01 (10 μM) had similar effects to benzbromarone, but were less potent. In contrast, Ani9 (10 μM) had no effect on carbachol-induced contractions. Confocal imaging revealed that benzbromarone (0.3 μM), MONNA (1 μM) and CaCCinhA01 (10 μM) increased intracellular calcium in isolated myocytes loaded with Fluo-4AM. In contrast, Ani9 (10 μM) had no effect on intracellular calcium. Benzbromarone and MONNA also increased calcium in calcium-free extracellular solution, but failed to do so when intracellular stores were discharged with caffeine (10 mM). Caffeine was unable to cause further discharge of the store when applied in the presence of benzbromarone. Ryanodine (100 μM) blocked the ability of benzbromarone (0.3 μM) to increase calcium, while tetracaine (100 μM) reversibly reduced the rise in calcium induced by benzbromarone. We conclude that benzbromarone and MONNA caused intracellular calcium release, probably by opening ryanodine receptors. Their ability to block carbachol contractions was likely due to this off-target effect.
Collapse
|
12
|
Jimenez C, Hawn MB, Akin E, Leblanc N. Translational potential of targeting Anoctamin-1-Encoded Calcium-Activated chloride channels in hypertension. Biochem Pharmacol 2022; 206:115320. [PMID: 36279919 DOI: 10.1016/j.bcp.2022.115320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Calcium-activated chloride channels (CaCC) provide a depolarizing stimulus to a variety of tissues through chloride efflux in response to a rise in internal Ca2+ and voltage. One of these channels, Anoctamin-1 (ANO1 or TMEM16A) is now recognized to play a central role in promoting smooth muscle tone in various types of blood vessels. Its role in hypertension, and thus the therapeutic promise of targeting ANO1, is less straightforward. This review gives an overview of our current knowledge about the potential role ANO1 may play in hypertension within the systemic, portal, and pulmonary vascular systems and the importance of this information when pursuing potential treatment strategies. While the role of ANO1 is well-established in several forms of pulmonary hypertension, its contributions to both the generation of vascular tone and its role in hypertension within the systemic and portal systems are much less clear. This, combined with ANO1's various roles throughout a multitude of tissues throughout the body, command caution when targeting ANO1 as a therapeutic target and may require tissue-selective strategies.
Collapse
Affiliation(s)
- Connor Jimenez
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Matthew B Hawn
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Elizabeth Akin
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA
| | - Normand Leblanc
- Department of Pharmacology and Center of Biomedical Research Excellence (COBRE) for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, Nevada 89557, USA.
| |
Collapse
|
13
|
Sinha M, Zabini D, Guntur D, Nagaraj C, Enyedi P, Olschewski H, Kuebler WM, Olschewski A. Chloride channels in the lung: Challenges and perspectives for viral infections, pulmonary arterial hypertension, and cystic fibrosis. Pharmacol Ther 2022; 237:108249. [PMID: 35878810 DOI: 10.1016/j.pharmthera.2022.108249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 10/16/2022]
Abstract
Fine control over chloride homeostasis in the lung is required to maintain membrane excitability, transepithelial transport as well as intra- and extracellular ion and water homeostasis. Over the last decades, a growing number of chloride channels and transporters have been identified in the cells of the pulmonary vasculature and the respiratory tract. The importance of these proteins is underpinned by the fact that impairment of their physiological function is associated with functional dysregulation, structural remodeling, or hereditary diseases of the lung. This paper reviews the field of chloride channels and transporters in the lung and discusses chloride channels in disease processes such as viral infections including SARS-CoV- 2, pulmonary arterial hypertension, cystic fibrosis and asthma. Although chloride channels have become a hot research topic in recent years, remarkably few of them have been targeted by pharmacological agents. As such, we complement the putative pathophysiological role of chloride channels here with a summary of their therapeutic potential.
Collapse
Affiliation(s)
- Madhushri Sinha
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Diana Zabini
- Department of Physiology, Neue Stiftingtalstrasse 6/V, 8010 Graz, Austria.
| | - Divya Guntur
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Neue Stiftingtalstraße 6, 8010 Graz, Austria.
| | - Peter Enyedi
- Department of Physiology, Semmelweis University, Tűzoltó utca 37-47, 1094 Budapest, Hungary.
| | - Horst Olschewski
- Department of Internal Medicine, Division of Pulmonology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Andrea Olschewski
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Neue Stiftingtalstraße 6, 8010 Graz, Austria.
| |
Collapse
|
14
|
Chen D, Li H, Wang X, Zhang L, Ji Z, Zhang J. Hypertriglyceridemia impairs urethral spontaneous tone through down-regulation of ANO1 in mouse urethral smooth muscle cells. Urology 2022; 165:157-163. [DOI: 10.1016/j.urology.2022.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022]
|
15
|
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
|
16
|
Mucus Release and Airway Constriction by TMEM16A May Worsen Pathology in Inflammatory Lung Disease. Int J Mol Sci 2021; 22:ijms22157852. [PMID: 34360618 PMCID: PMC8346050 DOI: 10.3390/ijms22157852] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Activation of the Ca2+ activated Cl− channel TMEM16A is proposed as a treatment in inflammatory airway disease. It is assumed that activation of TMEM16A will induce electrolyte secretion, and thus reduce airway mucus plugging and improve mucociliary clearance. A benefit of activation of TMEM16A was shown in vitro and in studies in sheep, but others reported an increase in mucus production and airway contraction by activation of TMEM16A. We analyzed expression of TMEM16A in healthy and inflamed human and mouse airways and examined the consequences of activation or inhibition of TMEM16A in asthmatic mice. TMEM16A was found to be upregulated in the lungs of patients with asthma or cystic fibrosis, as well as in the airways of asthmatic mice. Activation or potentiation of TMEM16A by the compounds Eact or brevenal, respectively, induced acute mucus release from airway goblet cells and induced bronchoconstriction in mice in vivo. In contrast, niclosamide, an inhibitor of TMEM16A, blocked mucus production and mucus secretion in vivo and in vitro. Treatment of airway epithelial cells with niclosamide strongly inhibited expression of the essential transcription factor of Th2-dependent inflammation and goblet cell differentiation, SAM pointed domain-containing ETS-like factor (SPDEF). Activation of TMEM16A in people with inflammatory airway diseases is likely to induce mucus secretion along with airway constriction. In contrast, inhibitors of TMEM16A may suppress pulmonary Th2 inflammation, goblet cell metaplasia, mucus production, and bronchoconstriction, partially by inhibiting expression of SPDEF.
Collapse
|
17
|
Yimnual C, Satitsri S, Ningsih BNS, Rukachaisirikul V, Muanprasat C. A fungus-derived purpactin A as an inhibitor of TMEM16A chloride channels and mucin secretion in airway epithelial cells. Biomed Pharmacother 2021; 139:111583. [PMID: 33901875 DOI: 10.1016/j.biopha.2021.111583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/20/2022] Open
Abstract
TMEM16A is a Ca2+-activated Cl- channel involved in mucus secretion in inflamed airways and proposed as a drug target for diseases associated with mucus hypersecretion including asthma. This study aimed to identify novel inhibitors of TMEM16A-mediated Cl- secretion in airway epithelial cells from a collection of compounds isolated from fungi indigenous in Thailand and examine its potential utility in mitigating airway mucus secretion using Calu-3 cells as a study model. Screening of > 400 fungal metabolites revealed purpactin A isolated from a soil-derived fungus Penicillium aculeatum PSU-RSPG105 as an inhibitor of TMEM16A-mediated Cl- transport with an IC50 value of ~2 µM. A consistent inhibitory effect of purpactin A on TMEM16A were observed regardless of TMEM16A activators or in the presence of an inhibitor of Ca2+/calmodulin-dependent protein kinase II (CaMKII), a negative regulator of TMEM16A. In addition, purpactin A did not affect cell viability, epithelial barrier integrity and activities of membrane transport proteins essential for maintaining airway hydration including CFTR Cl- channels and apical BK K+ channels. Intriguingly, purpactin A prevented a Ca2+-induced mucin release in cytokine-treated airway cells. Taken together, purpactin A represents the first class of TMEM16A inhibitor derived from fungus, which may be beneficial for the treatment of diseases associated with mucus hypersecretion.
Collapse
Affiliation(s)
- Chantapol Yimnual
- Department of Physiology, Faculty of Science, Mahidol University, Rajathevi, Bangkok 10400, Thailand; Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn 10540, Thailand
| | - Saravut Satitsri
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn 10540, Thailand
| | - Baiq Nila Sari Ningsih
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Vatcharin Rukachaisirikul
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn 10540, Thailand.
| |
Collapse
|
18
|
Henckels KA, Fong D, Phillips JE. Development of a QPatch-Automated Electrophysiology Assay for Identifying TMEM16A Small-Molecule Inhibitors. Assay Drug Dev Technol 2021; 18:134-147. [PMID: 32319819 PMCID: PMC7268545 DOI: 10.1089/adt.2019.962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The calcium-activated chloride channel, TMEM16A, is involved in airway hydration and bronchoconstriction and is a promising target for respiratory disease. Drug development efforts around channels require an electrophysiology-based assay for identifying inhibitors or activators. TMEM16A has proven to be a difficult channel to record on automated electrophysiology platforms due to its propensity for rundown. We developed an automated, whole-cell, electrophysiology assay on the QPatch-48 to evaluate small-molecule inhibitors of TMEM16A. In this assay, currents remained stable for a duration of roughly 11 min, allowing for the cumulative addition of five concentrations of compounds and resulted in reproducible IC50s. The absence of rundown was likely due to a low internal free-calcium level of 250 nM, which was high enough to produce large currents, but also maintained the voltage dependence of the channel. Current amplitude averaged 6 nA using the single-hole QPlate and the channel maintained outward rectification throughout the recording. Known TMEM16A inhibitors were tested and their IC50s aligned with those reported in the literature using manual patch-clamp. Once established, this assay was used to validate novel TMEM16A inhibitors that were identified in our high-throughput fluorescent-based assay, as well as to assist in structure–activity relationship efforts by the chemists. Overall, we demonstrate an easy to operate, reproducible, automated electrophysiology assay using the QPatch-48 for TMEM16A drug development efforts.
Collapse
Affiliation(s)
- Kathryn A Henckels
- Department of Process Development, Amgen, Inc., Thousand Oaks, California, USA
| | - David Fong
- Department of Inflammation Discovery Research, Amgen, Inc., Thousand Oaks, California, USA
| | - Jonathan E Phillips
- Department of Inflammation Discovery Research, Amgen, Inc., Thousand Oaks, California, USA
| |
Collapse
|
19
|
Liu Y, Liu Z, Wang K. The Ca 2+-activated chloride channel ANO1/TMEM16A: An emerging therapeutic target for epithelium-originated diseases? Acta Pharm Sin B 2021; 11:1412-1433. [PMID: 34221860 PMCID: PMC8245819 DOI: 10.1016/j.apsb.2020.12.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/19/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Anoctamin 1 (ANO1) or TMEM16A gene encodes a member of Ca2+ activated Cl– channels (CaCCs) that are critical for physiological functions, such as epithelial secretion, smooth muscle contraction and sensory signal transduction. The attraction and interest in ANO1/TMEM16A arise from a decade long investigations that abnormal expression or dysfunction of ANO1 is involved in many pathological phenotypes and diseases, including asthma, neuropathic pain, hypertension and cancer. However, the lack of specific modulators of ANO1 has impeded the efforts to validate ANO1 as a therapeutic target. This review focuses on the recent progress made in understanding of the pathophysiological functions of CaCC ANO1 and the current modulators used as pharmacological tools, hopefully illustrating a broad spectrum of ANO1 channelopathy and a path forward for this target validation.
Collapse
Key Words
- ANO1
- ANO1, anoctamin-1
- ASM, airway smooth muscle
- Ang II, angiotensin II
- BBB, blood–brain barrier
- CAMK, Ca2+/calmodulin-dependent protein kinase
- CF, cystic fibrosis
- CFTR, cystic fibrosis transmembrane conductance regulator
- Ca2+-activated Cl– channels (CaCCs)
- CaCCinh-A01
- CaCCs, Ca2+ activated chloride channels
- Cancer
- Cystic fibrosis
- DRG, dorsal root ganglion
- Drug target
- EGFR, epidermal growth factor receptor
- ENaC, epithelial sodium channels
- ER, endoplasmic reticulum
- ESCC, esophageal squamous cell carcinoma
- FRT, fisher rat thyroid
- GI, gastrointestinal
- GIST, gastrointestinal stromal tumor
- GPCR, G-protein coupled receptor
- HNSCC, head and neck squamous cell carcinoma
- HTS, high-throughput screening
- ICC, interstitial cells of Cajal
- IPAH, idiopathic pulmonary arterial hypertension
- MAPK, mitogen-activated protein kinase
- NF-κB, nuclear factor κB
- PAH, pulmonary arterial hypertension
- PAR2, protease activated receptor 2
- PASMC, pulmonary artery smooth muscle cells
- PIP2, phosphatidylinositol 4,5-bisphosphate
- PKD, polycystic kidney disease
- T16Ainh-A01
- TGF-β, transforming growth factor-β
- TMEM16A
- VGCC, voltage gated calcium channel
- VRAC, volume regulated anion channel
- VSMC, vascular smooth muscle cells
- YFP, yellow fluorescent protein
Collapse
Affiliation(s)
- Yani Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, Qingdao 266021, China
| | - Zongtao Liu
- Department of Clinical Laboratory, Qingdao Third People's Hospital, Qingdao 266041, China
| | - KeWei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, Qingdao 266021, China
- Corresponding authors.
| |
Collapse
|
20
|
Cao X, Zhou Z, Tian Y, Liu Z, Cheng KO, Chen X, Hu W, Wong YM, Li X, Zhang H, Hu R, Huang P. Opposing roles of E3 ligases TRIM23 and TRIM21 in regulation of ion channel ANO1 protein levels. J Biol Chem 2021; 296:100738. [PMID: 33957127 PMCID: PMC8191318 DOI: 10.1016/j.jbc.2021.100738] [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] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 04/19/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Anoctamin-1 (ANO1) (TMEM16A) is a calcium-activated chloride channel that plays critical roles in diverse physiological processes, such as sensory transduction and epithelial secretion. ANO1 levels have been shown to be altered under physiological and pathological conditions, although the molecular mechanisms that control ANO1 protein levels remain unclear. The ubiquitin–proteasome system is known to regulate the levels of numerous ion channels, but little information is available regarding whether and how ubiquitination regulates levels of ANO1. Here, we showed that two E3 ligases, TRIM23 and TRIM21, physically interact with the C terminus of ANO1. In vitro and in vivo assays demonstrated that whereas TRIM23 ubiquitinated ANO1 leading to its stabilization, TRIM21 ubiquitinated ANO1 and induced its degradation. Notably, ANO1 regulation by TRIM23 and TRIM21 is involved in chemical-induced pain sensation, salivary secretion, and heart-rate control in mice, and TRIM23 also mediates ANO1 upregulation induced by epidermal growth factor treatment. Our results suggest that these two antagonistic E3 ligases act together to control ANO1 expression and function. Our findings reveal a previously unrecognized mechanism for regulating ANO1 protein levels and identify a potential molecular link between ANO1 regulation, epidermal growth factor, and other signaling pathways.
Collapse
Affiliation(s)
- Xu Cao
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Zijing Zhou
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Ye Tian
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Zhengzhao Liu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China; State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; Xiangya Hospital, Central South University, Changsha, China
| | - Kar On Cheng
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Xibing Chen
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Wenbao Hu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Yuk Ming Wong
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Xiaofen Li
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Ronggui Hu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China; School of Life Science, Hangzhou Institute for Advance Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Pingbo Huang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China; Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China; State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China; HKUST Shenzhen Research Institute, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China; Hong Kong Branch of Guangdong Southern Marine Science and Engineering Laboratory (Guangzhou), Hong Kong University of Science and Technology, Hong Kong, People's Republic of China.
| |
Collapse
|
21
|
ANO7: Insights into topology, function, and potential applications as a biomarker and immunotherapy target. Tissue Cell 2021; 72:101546. [PMID: 33940566 DOI: 10.1016/j.tice.2021.101546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/21/2021] [Accepted: 04/11/2021] [Indexed: 01/01/2023]
Abstract
Anoctamin 7 (ANO7) is a member of the transmembrane protein TMEM16 family. It has a conservative topology similar to other members in this family, such as the typical eight-transmembrane domain, but it also has unique features. Although the ion channel role of ANO7 has been well accepted, evolutionary analyses and relevant studies suggest that ANO7 may be a multi-facet protein in function. Studies have shown that ANO7 may also function as a scramblase. ANO7 is highly expressed in prostate cancer as well as normal prostate tissues. A considerable amount of evidence has confirmed that ANO7 is associated with human physiology and pathology, particularly with the development of prostate cancer, which makes ANO7 a good candidate as a diagnostic and prognostic biomarker. In addition, ANO7 may be a potential target for prostate cancer immunotherapy. Antibody-based or T cell-mediated immunotherapies against prostate cancer by targeting ANO7 have been highly anticipated. ANO7 may also correlate with several other types of cancers or diseases, where further studies are warranted.
Collapse
|
22
|
Okonski R, Zheng YM, Di Mise A, Wang YX. Reciprocal Correlations of Inflammatory and Calcium Signaling in Asthma Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:319-331. [PMID: 33788200 DOI: 10.1007/978-3-030-63046-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Asthma is a chronic disease characterized by airway hyperresponsiveness, which can be caused by exposure to an allergen, spasmogen, or be induced by exercise. Despite its prevalence, the exact mechanisms by which the airway becomes hyperresponsive in asthma are not fully understood. There is evidence that myosin light-chain kinase is overexpressed, with a concomitant downregulation of myosin light-chain phosphatase in the airway smooth muscle, leading to sustained contraction. Additionally, the sarco/endoplasmic reticulum ATPase may be affected by inflammatory cytokines, such as IL-4, IL-5, IL-13, and TNF-α, which are all associated with asthmatic airway inflammation. IL-13 and TNF-α seem to promote sodium/calcium exchanger 1 overexpression as well. Anyhow, the exact mechanisms beyond these dysregulations need to be clarified. Of note, multiple studies show an association between asthma and the ORMLD3 gene, opening new perspectives to future potential gene therapies. Currently, several treatments are available for asthma, although many of them have systemic side effects, or are not effective in patients with severe asthma. Furthering our knowledge on the molecular and pathophysiological mechanisms of asthma plays a pivotal role for the development of new and more targeted treatments for patients who cannot totally benefit from the current therapies.
Collapse
Affiliation(s)
- Ryan Okonski
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA. .,Department of Biosciences, Biotechnologies e Biopharmaceutics, University of Bari, Bari, Italy.
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA.
| |
Collapse
|
23
|
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: 8] [Impact Index Per Article: 2.0] [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
|
24
|
Drumm BT, Thornbury KD, Hollywood MA, Sergeant GP. Role of Ano1 Ca 2+-activated Cl - channels in generating urethral tone. Am J Physiol Renal Physiol 2021; 320:F525-F536. [PMID: 33554780 DOI: 10.1152/ajprenal.00520.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Urinary continence is maintained in the lower urinary tract by the contracture of urethral sphincters, including smooth muscle of the internal urethral sphincter. These contractions occlude the urethral lumen, preventing urine leakage from the bladder to the exterior. Over the past 20 years, research on the ionic conductances that contribute to urethral smooth muscle contractility has greatly accelerated. A debate has emerged over the role of interstitial cell of Cajal (ICC)-like cells in the urethra and their expression of Ca2+-activated Cl- channels encoded by anoctamin-1 [Ano1; transmembrane member 16 A (Tmem16a) gene]. It has been proposed that Ano1 channels expressed in urethral ICC serve as a source of depolarization for smooth muscle cells, increasing their excitability and contributing to tone. Although a clear role for Ano1 channels expressed in ICC is evident in other smooth muscle organs, such as the gastrointestinal tract, the role of these channels in the urethra is unclear, owing to differences in the species (rabbit, rat, guinea pig, sheep, and mouse) examined and experimental approaches by different groups. The importance of clarifying this situation is evident as effective targeting of Ano1 channels may lead to new treatments for urinary incontinence. In this review, we summarize the key findings from different species on the role of ICC and Ano1 channels in urethral contractility. Finally, we outline proposals for clarifying this controversial and important topic by addressing how cell-specific optogenetic and inducible cell-specific genetic deletion strategies coupled with advances in Ano1 channel pharmacology may clarify this area in future studies.NEW & NOTEWORTHY Studies from the rabbit have shown that anoctamin-1 (Ano1) channels expressed in urethral interstitial cells of Cajal (ICC) serve as a source of depolarization for smooth muscle cells, increasing excitability and tone. However, the role of urethral Ano1 channels is unclear, owing to differences in the species examined and experimental approaches. We summarize findings from different species on the role of urethral ICC and Ano1 channels in urethral contractility and outline proposals for clarifying this topic using cell-specific optogenetic approaches.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Keith D Thornbury
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Gerard P Sergeant
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| |
Collapse
|
25
|
Qu M, Lu P, Bellve K, Fogarty K, Lifshitz L, Shi F, Zhuge R. Smooth muscle cell-specific TMEM16A deletion does not alter Ca2+ signaling, uterine contraction, gestation length, or litter size in mice†. Biol Reprod 2020; 101:318-327. [PMID: 31175367 DOI: 10.1093/biolre/ioz096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/15/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022] Open
Abstract
Ion channels in myometrial cells play critical roles in spontaneous and agonist-induced uterine contraction during the menstrual cycle, pregnancy maintenance, and parturition; thus, identifying the genes of ion channels in these cells and determining their roles are essential to understanding the biology of reproduction. Previous studies with in vitro functional and pharmacological approaches have produced controversial results regarding the presence and role of TMEM16A Ca2+-activated Cl- channels in myometrial cells. To unambiguously determine the function of this channel in these cells, we employed a genetic approach by using smooth muscle cell-specific TMEM16A deletion (i.e. TMEM16ASMKO) mice. We found that myometrial cells from TMEM16ASMKO mice generated the same pattern and magnitude in Ca2+ signals upon stimulation with KCl, oxytocin, and PGF2α compared to the isogenic control myometrial cells. At the uterine tissue level, TMEM16A deletion also did not cause detectable changes in either spontaneous or agonist (i.e. KCl, oxytocin, and PGF2α)-induced contractions. Moreover, in vivo the TMEM16ASMKO mice gave birth at full term with the same litter size as genetically identical control mice. Finally, TMEM16A immunostaining in both control and TMEM16ASMKO mice revealed that this protein was highly expressed in the endometrial stroma, but did not co-localize with a smooth muscle specific marker MYH11. Collectively, these results unequivocally demonstrate that TMEM16A does not serve as a pacemaking channel for spontaneous uterine contraction, neither does it function as a depolarizing channel for agonist-evoked uterine contraction. Yet these two functions could underlie the normal gestation length and litter size in the TMEM16ASMKO mice.
Collapse
Affiliation(s)
- Mingzi Qu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Department of Microbiology & Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ping Lu
- Department of Microbiology & Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Karl Bellve
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kevin Fogarty
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Lawrence Lifshitz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Fangxiong Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ronghua Zhuge
- Department of Microbiology & Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
26
|
Gazzola M, Flamand N, Bossé Y. [Extracellular molecules controlling the contraction of airway smooth muscle and their potential contribution to bronchial hyperresponsiveness]. Rev Mal Respir 2020; 37:462-473. [PMID: 32487422 DOI: 10.1016/j.rmr.2020.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION A significant portion of symptoms in some lung diseases results from an excessive constriction of airways due to the contraction of smooth muscle and bronchial hyperresponsiveness. A better understanding of the extracellular molecules that control smooth muscle contractility is necessary to identify the underlying causes of the problem. STATE OF KNOWLEDGE Almost a hundred molecules, some of which newly identified, influence the contractility of airway smooth muscle. While some molecules activate the contraction, others activate the relaxation, thus acting directly as bronchoconstrictors and bronchodilators, respectively. Other molecules do not affect contraction directly but rather influence it indirectly by modifying the effect of bronchoconstrictors and bronchodilators. These are called bronchomodulators. Some of these bronchomodulators increase the contractile effect of bronchoconstrictors and could thus contribute to bronchial hyperresponsiveness. PROSPECTS Considering the high number of molecules potentially involved, as well as the level of functional overlap between some of them, identifying the extracellular molecules responsible for excessive airway constriction in a patient is a major contemporary challenge.
Collapse
Affiliation(s)
| | | | - Y Bossé
- Université Laval, Québec, Canada.
| |
Collapse
|
27
|
Centeio R, Cabrita I, Benedetto R, Talbi K, Ousingsawat J, Schreiber R, Sullivan JK, Kunzelmann K. Pharmacological Inhibition and Activation of the Ca 2+ Activated Cl - Channel TMEM16A. Int J Mol Sci 2020; 21:ijms21072557. [PMID: 32272686 PMCID: PMC7177308 DOI: 10.3390/ijms21072557] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/26/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
TMEM16A is a Ca2+ activated Cl− channel with important functions in airways, intestine, and other epithelial organs. Activation of TMEM16A is proposed as a therapy in cystic fibrosis (CF) to reinstall airway Cl− secretion and to enhance airway surface liquid (ASL). This CFTR-agnostic approach is thought to improve mucociliary clearance and lung function in CF. This could indeed improve ASL, however, mucus release and airway contraction may also be induced by activators of TMEM16A, particularly in inflamed airways of patients with asthma, COPD, or CF. Currently, both activators and inhibitors of TMEM16A are developed and examined in different types of tissues. Here we compare activation and inhibition of endogenous and overexpressed TMEM16A and analyze potential off-target effects. The three well-known blockers benzbromarone, niclosamide, and Ani9 inhibited both TMEM16A and ATP-induced Ca2+ increase by variable degrees, depending on the cell type. Niclosamide, while blocking Ca2+ activated TMEM16A, also induced a subtle but significant Ca2+ store release and inhibited store-operated Ca2+ influx. Niclosamide, benzbromarone and Ani9 also affected TMEM16F whole cell currents, indicating limited specificity for these inhibitors. The compounds Eact, cinnamaldehyde, and melittin, as well as the phosphatidylinositol diC8-PIP2 are the reported activators of TMEM16A. However, the compounds were unable to activate endogenous TMEM16A in HT29 colonic epithelial cells. In contrast, TMEM16A overexpressed in HEK293 cells was potently stimulated by these activators. We speculate that overexpressed TMEM16A might have a better accessibility to intracellular Ca2+, which causes spontaneous activity even at basal intracellular Ca2+ concentrations. Small molecules may therefore potentiate pre-stimulated TMEM16A currents, but may otherwise fail to activate silent endogenous TMEM16A.
Collapse
Affiliation(s)
- Raquel Centeio
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Inês Cabrita
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Roberta Benedetto
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Khaoula Talbi
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
| | | | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany; (R.C.); (I.C.); (R.B.); (K.T.); (J.O.); (R.S.)
- * Correspondence: ; Tel.: +49-(0)941-943-4302; Fax: +49-(0)941-943-4315
| |
Collapse
|
28
|
Danielsson J, Kuforiji AS, Yocum GT, Zhang Y, Xu D, Gallos G, Emala CW. Agonism of the TMEM16A calcium-activated chloride channel modulates airway smooth muscle tone. Am J Physiol Lung Cell Mol Physiol 2019; 318:L287-L295. [PMID: 31747299 DOI: 10.1152/ajplung.00552.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
TMEM16A (anoctamin 1) is an important calcium-activated chloride channel in airway smooth muscle (ASM). We have previously shown that TMEM16A antagonists such as benzbromarone relax ASM and have proposed TMEM16A antagonists as novel therapies for asthma treatment. However, TMEM16A is also expressed on airway epithelium, and TMEM16A agonists are being investigated as novel therapies for cystic fibrosis. There are theoretical concerns that agonism of TMEM16A on ASM could lead to bronchospasm, making them detrimental as airway therapeutics. The TMEM16A agonist Eact induced a significant contraction of human ASM and guinea pig tracheal rings in an ex vivo organ bath model. Pretreatment with two different TMEM16A antagonists, benzbromarone or T16Ainh-A01, completely attenuated these Eact-induced contractions. Pretreatment with Eact alone augmented the maximum acetylcholine contraction. Pretreatment of A/J mice in vivo with nebulized Eact caused an augmentation of methacholine-induced increases in airway resistance measured by the forced oscillatory technique (flexiVent). Pretreatment with the TMEM16A antagonist benzbromarone significantly attenuated methacholine-induced increases in airway resistance. In in vitro cellular studies, TMEM16A was found to be expressed more abundantly in ASM compared with epithelial cells in culture (8-fold higher in ASM). Eact caused an increase in intracellular calcium in human ASM cells that was completely attenuated by pretreatment with benzbromarone. Eact acutely depolarized the plasma membrane potential of ASM cells, which was attenuated by benzbromarone or nifedipine. The TMEM16A agonist Eact modulates ASM contraction in both ex vivo and in vivo models, suggesting that agonism of TMEM16A may lead to clinically relevant bronchospasm.
Collapse
Affiliation(s)
| | - Aisha S Kuforiji
- Department of Anesthesiology, Columbia University, New York, New York
| | - Gene T Yocum
- Department of Anesthesiology, Columbia University, New York, New York
| | - Yi Zhang
- Department of Anesthesiology, Columbia University, New York, New York
| | - Dingbang Xu
- Department of Anesthesiology, Columbia University, New York, New York
| | - George Gallos
- Department of Anesthesiology, Columbia University, New York, New York
| | - Charles W Emala
- Department of Anesthesiology, Columbia University, New York, New York
| |
Collapse
|
29
|
Roberts G, Almqvist C, Boyle R, Crane J, Hogan SP, Marsland B, Saglani S, Woodfolk JA. Developments in the field of allergy in 2017 through the eyes of Clinical and Experimental Allergy. Clin Exp Allergy 2019; 48:1606-1621. [PMID: 30489681 DOI: 10.1111/cea.13318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this article, we described the development in the field of allergy as described by Clinical and Experimental Allergy in 2017. Experimental models of allergic disease, basic mechanisms, clinical mechanisms, allergens, asthma and rhinitis and clinical allergy are all covered.
Collapse
Affiliation(s)
- G Roberts
- Faculty of Medicine, Clinical and Experimental Sciences and Human Development and Health, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Isle of Wight, UK
| | - C Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - R Boyle
- Department of Paediatrics, Imperial College London, London, UK
| | - J Crane
- Department of Medicine, University of Otago Wellington, Wellington, New Zealand
| | - S P Hogan
- Mary H Weiser Food Allergy Center, Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - B Marsland
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - S Saglani
- National Heart & Lung Institute, Imperial College London, London, UK
| | - J A Woodfolk
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
30
|
Liu PY, Zhang Z, Liu Y, Tang XL, Shu S, Bao XY, Zhang Y, Gu Y, Xu Y, Cao X. TMEM16A Inhibition Preserves Blood-Brain Barrier Integrity After Ischemic Stroke. Front Cell Neurosci 2019; 13:360. [PMID: 31447648 PMCID: PMC6691060 DOI: 10.3389/fncel.2019.00360] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/23/2019] [Indexed: 12/23/2022] Open
Abstract
The inflammatory response plays a pivotal role in Blood–Brain Barrier (BBB) destruction following ischemic brain injury. Enhanced leukocyte adhesion to vascular endothelial cells is an essential event in the inflammatory process. TMEM16A, a newly discovered protein regulating calcium-activated chloride channels, is widely expressed in eukaryotes. Recent studies have suggested that upregulated expression of TMEM16A is associated with the occurrence and development of many diseases. However, the role of TMEM16A in regulating BBB integrity after ischemic stroke has not been fully investigated. In this study, we found that TMEM16A is mainly expressed in brain endothelial cells and upregulated after ischemic stroke in the mouse brain. Caccinh-A01, an TMEM16A inhibitor that reduced its upregulation, attenuated brain infarct size and neurological deficits after ischemic stroke. ICAM-1 and MPO expression and BBB permeability were decreased after TMEM16A inhibitor administration. In addition, TMEM16A silencing rescued oxygen-glucose deprivation/reoxygenation (OGD/R)-induced transendothelial permeability in vitro accompanied by decreased ICAM-1 expression and leukocyte adhesion. Furthermore, our mechanistic study showed that TMEM16A knockdown alleviated NF-κB activation and nuclear translocation, indicating that TMEM16A knockdown downregulated OGD/R-induced ICAM-1 expression in an NF-κB-dependent manner. Finally, NF-κB inhibitor treatment also alleviated OGD/ R-induced BBB permeability, confirming that activated NF-κB and increased ICAM-1 are essential factors involved in ischemia-induced BBB damage. Thus, our research provides a promising treatment strategy against BBB destruction after ischemic stroke, and TMEM16A may become a potential target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Pin-Yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Xue-Lian Tang
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Shu Shu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Xin-Yu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yue Gu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| |
Collapse
|
31
|
Tonic inhibition of murine proximal colon is due to nitrergic suppression of Ca 2+ signaling in interstitial cells of Cajal. Sci Rep 2019; 9:4402. [PMID: 30867452 PMCID: PMC6416298 DOI: 10.1038/s41598-019-39729-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/30/2019] [Indexed: 12/18/2022] Open
Abstract
Spontaneous excitability and contractions of colonic smooth muscle cells (SMCs) are normally suppressed by inputs from inhibitory motor neurons, a behavior known as tonic inhibition. The post-junctional cell(s) mediating tonic inhibition have not been elucidated. We investigated the post-junctional cells mediating tonic inhibition in the proximal colon and whether tonic inhibition results from suppression of the activity of Ano1 channels, which are expressed exclusively in interstitial cells of Cajal (ICC). We found that tetrodotoxin (TTX), an inhibitor of nitric oxide (NO) synthesis, L-NNA, and an inhibitor of soluble guanylyl cyclase, ODQ, greatly enhanced colonic contractions. Ano1 antagonists, benzbromarone and Ani9 inhibited the effects of TTX, L-NNA and ODQ. Ano1 channels are activated by Ca2+ release from the endoplasmic reticulum (ER) in ICC, and blocking Ca2+ release with a SERCA inhibitor (thapsigargin) or a store-operated Ca2+ entry blocker (GSK 7975 A) reversed the effects of TTX, L-NNA and ODQ. Ca2+ imaging revealed that TTX, L-NNA and ODQ increased Ca2+ transient firing in colonic ICC. Our results suggest that tonic inhibition in the proximal colon occurs through suppression of Ca2+ release events in ICC. Suppression of Ca2+ release in ICC limits the open probability of Ano1 channels, reducing the excitability of electrically-coupled SMCs.
Collapse
|
32
|
Miner K, Labitzke K, Liu B, Wang P, Henckels K, Gaida K, Elliott R, Chen JJ, Liu L, Leith A, Trueblood E, Hensley K, Xia XZ, Homann O, Bennett B, Fiorino M, Whoriskey J, Yu G, Escobar S, Wong M, Born TL, Budelsky A, Comeau M, Smith D, Phillips J, Johnston JA, McGivern JG, Weikl K, Powers D, Kunzelmann K, Mohn D, Hochheimer A, Sullivan JK. Drug Repurposing: The Anthelmintics Niclosamide and Nitazoxanide Are Potent TMEM16A Antagonists That Fully Bronchodilate Airways. Front Pharmacol 2019; 10:51. [PMID: 30837866 PMCID: PMC6382696 DOI: 10.3389/fphar.2019.00051] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/18/2019] [Indexed: 01/21/2023] Open
Abstract
There is an unmet need in severe asthma where approximately 40% of patients exhibit poor β-agonist responsiveness, suffer daily symptoms and show frequent exacerbations. Antagonists of the Ca2+-activated Cl- channel, TMEM16A, offers a new mechanism to bronchodilate airways and block the multiple contractiles operating in severe disease. To identify TMEM16A antagonists we screened a library of ∼580,000 compounds. The anthelmintics niclosamide, nitazoxanide, and related compounds were identified as potent TMEM16A antagonists that blocked airway smooth muscle depolarization and contraction. To evaluate whether TMEM16A antagonists resist use- and inflammatory-desensitization pathways limiting β-agonist action, we tested their efficacy under harsh conditions using maximally contracted airways or airways pretreated with a cytokine cocktail. Stunningly, TMEM16A antagonists fully bronchodilated airways, while the β-agonist isoproterenol showed only partial effects. Thus, antagonists of TMEM16A and repositioning of niclosamide and nitazoxanide represent an important additional treatment for patients with severe asthma and COPD that is poorly controlled with existing therapies. It is of note that drug repurposing has also attracted wide interest in niclosamide and nitazoxanide as a new treatment for cancer and infectious disease. For the first time we identify TMEM16A as a molecular target for these drugs and thus provide fresh insights into their mechanism for the treatment of these disorders in addition to respiratory disease.
Collapse
Affiliation(s)
- Kent Miner
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Katja Labitzke
- Department of Therapeutic Discovery, Amgen Inc., Regensburg, Germany
| | - Benxian Liu
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Paul Wang
- Department of Therapeutic Discovery, Amgen Inc., Thousand Oaks, CA, United States
| | - Kathryn Henckels
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Kevin Gaida
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Robin Elliott
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Jian Jeffrey Chen
- Department of Medicinal Chemistry, Amgen Inc., Thousand Oaks, CA, United States
| | - Longbin Liu
- Department of Medicinal Chemistry, Amgen Inc., Thousand Oaks, CA, United States
| | - Anh Leith
- Department of Inflammation Research, Amgen Inc., Seattle, WA, United States
| | - Esther Trueblood
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Seattle, WA, United States
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, United States
- Department of Comparative Biology and Safety Sciences, Amgen Inc., South San Francisco, CA, United States
| | - Kelly Hensley
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Seattle, WA, United States
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, United States
- Department of Comparative Biology and Safety Sciences, Amgen Inc., South San Francisco, CA, United States
| | - Xing-Zhong Xia
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Oliver Homann
- Genome Analysis Unit, Amgen Inc., South San Francisco, CA, United States
| | - Brian Bennett
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Mike Fiorino
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - John Whoriskey
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Gang Yu
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Sabine Escobar
- Department of Inflammation Research, Amgen Inc., Seattle, WA, United States
| | - Min Wong
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Teresa L. Born
- Department of Inflammation Research, Amgen Inc., Seattle, WA, United States
| | - Alison Budelsky
- Department of Inflammation Research, Amgen Inc., Seattle, WA, United States
| | - Mike Comeau
- Department of Inflammation Research, Amgen Inc., Seattle, WA, United States
| | - Dirk Smith
- Department of Inflammation Research, Amgen Inc., Seattle, WA, United States
| | - Jonathan Phillips
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - James A. Johnston
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Joseph G. McGivern
- Department of Therapeutic Discovery, Amgen Inc., Thousand Oaks, CA, United States
| | - Kerstin Weikl
- Department of Therapeutic Discovery, Amgen Inc., Regensburg, Germany
| | - David Powers
- Department of Therapeutic Discovery, Amgen Inc., Thousand Oaks, CA, United States
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Deanna Mohn
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| | | | - John K. Sullivan
- Department of Inflammation Research, Amgen Inc., Thousand Oaks, CA, United States
| |
Collapse
|
33
|
Kunzelmann K, Ousingsawat J, Cabrita I, Doušová T, Bähr A, Janda M, Schreiber R, Benedetto R. TMEM16A in Cystic Fibrosis: Activating or Inhibiting? Front Pharmacol 2019; 10:3. [PMID: 30761000 PMCID: PMC6362895 DOI: 10.3389/fphar.2019.00003] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
The inflammatory airway disease cystic fibrosis (CF) is characterized by airway obstruction due to mucus hypersecretion, airway plugging, and bronchoconstriction. The cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel is dysfunctional in CF, leading to defects in epithelial transport. Although CF pathogenesis is still disputed, activation of alternative Cl- channels is assumed to improve lung function in CF. Two suitable non-CFTR Cl- channels are present in the airway epithelium, the Ca2+ activated channel TMEM16A and SLC26A9. Activation of these channels is thought to be feasible to improve hydration of the airway mucus and to increase mucociliary clearance. Interestingly, both channels are upregulated during inflammatory lung disease. They are assumed to support fluid secretion, necessary to hydrate excess mucus and to maintain mucus clearance. During inflammation, however, TMEM16A is upregulated particularly in mucus producing cells, with only little expression in ciliated cells. Recently it was shown that knockout of TMEM16A in ciliated cells strongly compromises Cl- conductance and attenuated mucus secretion, but does not lead to a CF-like lung disease and airway plugging. Along this line, activation of TMEM16A by denufosol, a stable purinergic ligand, failed to demonstrate any benefit to CF patients in earlier studies. It rather induced adverse effects such as cough. A number of studies suggest that TMEM16A is essential for mucus secretion and possibly also for mucus production. Evidence is now provided for a crucial role of TMEM16A in fusion of mucus-filled granules with the apical plasma membrane and cellular exocytosis. This is probably due to local Ca2+ signals facilitated by TMEM16A. Taken together, TMEM16A supports fluid secretion by ciliated airway epithelial cells, but also maintains excessive mucus secretion during inflammatory airway disease. Because TMEM16A also supports airway smooth muscle contraction, inhibition rather than activation of TMEM16A might be the appropriate treatment for CF lung disease, asthma and COPD. As a number of FDA-approved and well-tolerated drugs have been shown to inhibit TMEM16A, evaluation in clinical trials appears timely.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | | | - Inês Cabrita
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Tereza Doušová
- Department of Pediatrics, Second Faculty of Medicine, University Hospital Motol, Charles University in Prague, Prague, Czechia
| | - Andrea Bähr
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Munich, Germany
- Innere Medizin I, Klinikum Rechts der Isar der TU München, München, Germany
| | - Melanie Janda
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| |
Collapse
|
34
|
Benedetto R, Cabrita I, Schreiber R, Kunzelmann K. TMEM16A is indispensable for basal mucus secretion in airways and intestine. FASEB J 2018; 33:4502-4512. [PMID: 30586313 DOI: 10.1096/fj.201801333rrr] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Transmembrane member 16A (TMEM16A) is the Ca2+-activated chloride channel in airways and intestine. It has been associated with goblet cell metaplasia, as expression of TMEM16A is strongly up-regulated in cystic fibrosis and asthma during mucus hypersecretion. However, the possible role of TMEM16A for mucus production or mucus secretion remains obscure, and whether TMEM16A controls the function of intestinal goblet cells is entirely unknown. Basal mucus secretion in lungs occurs through low levels of ATP in the airway surface liquid. Here, we report for the first time that TMEM16A is essential for basal secretion of mucus in airways and intestine. Airway-ciliated and intestinal epithelial-specific knockout of TMEM16A ( TMEM16Aflox/floxFoxJ1, TMEM16Aflox/floxVil1) leads to accumulation of mucus in airway club (Clara) cells and intestinal goblet cells, respectively. Acute ATP-induced mucus secretion by airway club cells is inhibited when TMEM16A is knocked out in ciliated cells, possibly as a result of compromised release of prosecretory cytokines. Knockdown or inhibition of TMEM16A in human Calu3 airway epithelial cells indicates compromised IL-8 release. In intestinal goblet cells lacking expression of TMEM16A, mucus accumulates as a result of compromised ATP-induced secretion. In contrast, cholinergic mucus secretion by compound exocytosis is independent of TMEM16A. The data demonstrate a previously unrecognized role of TMEM16A for membrane exocytosis and describe a novel, ATP-driven pathway for intestinal mucus secretion. We conclude that ATP-dependent mucus secretion in both airways and intestine requires TMEM16A. The present results may form the basis for a novel, therapeutic approach for the treatment of mucus hypersecretion in inflammatory airway and intestinal disease.-Benedetto, R., Cabrita, I., Schreiber, R., Kunzelmann, K. TMEM16A is indispensable for basal mucus secretion in airways and intestine.
Collapse
Affiliation(s)
- Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Inês Cabrita
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| |
Collapse
|
35
|
Wang Y, Wang A, Zhang M, Zeng H, Lu Y, Liu L, Li J, Deng L. Artesunate attenuates airway resistance in vivo and relaxes airway smooth muscle cells in vitro via bitter taste receptor-dependent calcium signalling. Exp Physiol 2018; 104:231-243. [PMID: 30379382 DOI: 10.1113/ep086824] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 10/18/2018] [Indexed: 01/20/2023]
Abstract
NEW FINDINGS What is the central question of this study? The aim of this study was to evaluate artesunate for its use as a bronchodilator in asthma treatment. What is the main finding and its importance? We found that artesunate reduces airway resistance in both normal and ovalbumin-treated Balb/c mice in vivo. Artesunate reduces traction force while inducing Ca2+ influx into cultured airway smooth muscle cells in vitro, most probably via the bitter taste receptor. These findings provide important evidence at both animal and cellular levels that artesunate might potentially be used as a bronchodilator for treating obstructive airway diseases, such as asthma. ABSTRACT Following the surprising discovery that bitter taste receptors (TAS2Rs) expressed in the lung and can be stimulated to relax airway smooth muscle cells (ASMCs), there is great interest in searching for a bitter taste receptor agonist as a new bronchodilator for asthma therapy. Among the great many other natural bitter substances, artesunate is of special interest to be evaluated for this purpose because of its pharmacological value as a derivative from the well-known anti-malarial, artemisinin. Therefore, in this study we treated either normal or ovalbumin (OVA)-induced asthmatic Balb/c mice in vivo with artesunate (30, 60 or 120 μg) via aerosol inhalation. Subsequently, we measured the airway resistance of the mice in the presence or absence of artesunate. In addition, we treated either mouse or human ASMCs cultured in vitro with artesunate (0.25-2.0 mM) and then measured the traction force and [Ca2+ ]i flux of the cells in the presence or absence of artesunate. The results demonstrate that artesunate attenuated airway resistance in a dose-dependent manner in both the normal and the OVA-treated mice, but more potently in the latter. The in vivo efficacy of artesunate at 120 μg was comparable to that of the conventional bronchodilator, salbutamol, at 3 μg in terms of the reduction in airway resistance. Artesunate also reduced traction force and induced an increase in [Ca2+ ]i in the cultured ASMCs, which was mediated, at least in part, by TAS2R signalling in the human ASMCs. These results together suggest that artesunate might potentially be a cheap and safe bronchodilator to complement the current therapy of asthma.
Collapse
Affiliation(s)
- Yue Wang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China.,School of Pharmaceutical Engineering and Life Science & School of Nursing, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Aili Wang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China.,School of Pharmaceutical Engineering and Life Science & School of Nursing, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Ming Zhang
- Department of Thoracic and Cardiovascular Surgery, Changzhou No. 2 People's Hospital, Affiliated to Nanjing Medical University, Changzhou, Jiangsu, 213003, China
| | - Huilong Zeng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Yun Lu
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Lei Liu
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Jingjing Li
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, 213164, China.,School of Pharmaceutical Engineering and Life Science & School of Nursing, Changzhou University, Changzhou, Jiangsu, 213164, China
| |
Collapse
|
36
|
Bradley E, Large RJ, Bihun VV, Mullins ND, Hollywood MA, Sergeant GP, Thornbury KD. Inhibitory effects of openers of large-conductance Ca 2+-activated K + channels on agonist-induced phasic contractions in rabbit and mouse bronchial smooth muscle. Am J Physiol Cell Physiol 2018; 315:C818-C829. [PMID: 30257105 DOI: 10.1152/ajpcell.00068.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Airway smooth muscle expresses abundant BKCa channels, but their role in regulating contractions remains controversial. This study examines the effects of two potent BKCa channel openers on agonist-induced phasic contractions in rabbit and mouse bronchi. First, we demonstrated the ability of 10 μM GoSlo-SR5-130 to activate BKCa channels in inside-out patches from rabbit bronchial myocytes, where it shifted the activation V1/2 by -88 ± 11 mV (100 nM Ca2+, n = 7). In mouse airway smooth muscle cells, GoSlo-SR5-130 dose dependently shifted V1/2 by 12-83 mV over a concentration range of 1-30 μM. Compound X, a racemic mixture of two enantiomers, reported to be potent BKCa channel openers, shifted V1/2 by 20-79 mV over a concentration range of 0.3-3 μM. In rabbit bronchial rings, exposure to histamine (1 μM) induced phasic contractions after a delay of ~35 min. These were abolished by GoSlo-SR5-130 (30 μM). Nifedipine (100 nM) and CaCCinhA01 (10 μM), a TMEM16A blocker, also abolished histamine-induced phasic contractions. In mouse bronchi, similar phasic contractions were evoked by exposure to U46619 (100 nM) and carbachol (100 nM). In each case, these were inhibited by concentrations of GoSlo-SR5-130 and compound X that shifted the activation V1/2 of BKCa channels in the order of -80 mV. In conclusion, membrane potential-dependent regulation of L-type Ca2+ channels appears to be important for histamine-, U46619-, and carbachol-induced phasic contractions in airway smooth muscle. Contractions can be abolished by BKCa channel openers, suggesting that these channels are potential targets for treating some causes of airway obstruction.
Collapse
Affiliation(s)
- Eamonn Bradley
- Smooth Muscle Research Centre, Dundalk Institute of Technology , Dundalk , Ireland
| | - Roddy J Large
- Smooth Muscle Research Centre, Dundalk Institute of Technology , Dundalk , Ireland
| | | | - Nicolas D Mullins
- Smooth Muscle Research Centre, Dundalk Institute of Technology , Dundalk , Ireland
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology , Dundalk , Ireland
| | - Gerard P Sergeant
- Smooth Muscle Research Centre, Dundalk Institute of Technology , Dundalk , Ireland
| | - Keith D Thornbury
- Smooth Muscle Research Centre, Dundalk Institute of Technology , Dundalk , Ireland
| |
Collapse
|
37
|
Xia Y, Xia L, Lou L, Jin R, Shen H, Li W. Transient Receptor Potential Channels and Chronic Airway Inflammatory Diseases: A Comprehensive Review. Lung 2018; 196:505-516. [PMID: 30094794 DOI: 10.1007/s00408-018-0145-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/04/2018] [Indexed: 12/22/2022]
Abstract
Chronic airway inflammatory diseases remain a major problem worldwide, such that there is a need for additional therapeutic targets and novel drugs. Transient receptor potential (TRP) channels are a group of non-selective cation channels expressed throughout the body that are regulated by various stimuli. TRP channels have been identified in numerous cell types in the respiratory tract, including sensory neurons, airway epithelial cells, airway smooth muscle cells, and fibroblasts. Different types of TRP channels induce cough in sensory neurons via the vagus nerve. Permeability and cytokine production are also regulated by TRP channels in airway epithelial cells, and these channels also contribute to the modulation of bronchoconstriction. TRP channels may cooperate with other TRP channels, or act in concert with calcium-dependent potassium channels and calcium-activated chloride channel. Hence, TRP channels could be the potential therapeutic targets for chronic airway inflammatory diseases. In this review, we aim to discuss the expression profiles and physiological functions of TRP channels in the airway, and the roles they play in chronic airway inflammatory diseases.
Collapse
Affiliation(s)
- Yang Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| | - Lexin Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Lingyun Lou
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Rui Jin
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
38
|
Zak JD, Grimaud J, Li RC, Lin CC, Murthy VN. Calcium-activated chloride channels clamp odor-evoked spike activity in olfactory receptor neurons. Sci Rep 2018; 8:10600. [PMID: 30006552 PMCID: PMC6045664 DOI: 10.1038/s41598-018-28855-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/02/2018] [Indexed: 02/06/2023] Open
Abstract
The calcium-activated chloride channel anoctamin-2 (Ano2) is thought to amplify transduction currents in olfactory receptor neurons (ORNs), a hypothesis supported by previous studies in dissociated neurons from Ano2−/− mice. Paradoxically, despite a reduction in transduction currents in Ano2−/− ORNs, their spike output for odor stimuli may be higher. We examined the role of Ano2 in ORNs in their native environment in freely breathing mice by imaging activity in ORN axons as they arrive in the olfactory bulb glomeruli. Odor-evoked responses in ORN axons of Ano2−/− animals were consistently larger for a variety of odorants and concentrations. In an open arena, Ano2−/− animals took longer to approach a localized odor source than Ano2+/+ animals, revealing clear olfactory behavioral deficits. Our studies provide the first in vivo evidence toward an alternative or additional role for Ano2 in the olfactory transduction cascade, where it may serve as a feedback mechanism to clamp ORN spike output.
Collapse
Affiliation(s)
- Joseph D Zak
- Department of Molecular and Cellular Biology & Center for Brain Science, Harvard University, 16 Divinity Ave, Cambridge, MA, 02138, USA
| | - Julien Grimaud
- Department of Molecular and Cellular Biology & Center for Brain Science, Harvard University, 16 Divinity Ave, Cambridge, MA, 02138, USA.,Molecules, Cells & Organisms Program, Harvard University, Cambridge, 02138, MA, USA
| | - Rong-Chang Li
- Johns Hopkins University School of Medicine, Preclinical Teaching Building Room 905A, 725 North Wolfe Street, MD, 21205, Baltimore, USA
| | - Chih-Chun Lin
- Johns Hopkins University School of Medicine, Preclinical Teaching Building Room 905A, 725 North Wolfe Street, MD, 21205, Baltimore, USA
| | - Venkatesh N Murthy
- Department of Molecular and Cellular Biology & Center for Brain Science, Harvard University, 16 Divinity Ave, Cambridge, MA, 02138, USA.
| |
Collapse
|
39
|
Lérias JR, Pinto MC, Botelho HM, Awatade NT, Quaresma MC, Silva IAL, Wanitchakool P, Schreiber R, Pepperkok R, Kunzelmann K, Amaral MD. A novel microscopy-based assay identifies extended synaptotagmin-1 (ESYT1) as a positive regulator of anoctamin 1 traffic. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:421-431. [PMID: 29154949 DOI: 10.1016/j.bbamcr.2017.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 01/14/2023]
Abstract
An attractive possibility to treat Cystic Fibrosis (CF), a severe condition caused by dysfunctional CFTR, an epithelial anion channel, is through the activation of alternative (non-CFTR) anion channels. Anoctamin 1 (ANO1) was demonstrated to be a Ca2+-activated chloride channel (CaCC) and thus of high potential to replace CFTR. Despite that ANO1 is expressed in human lung CF tissue, it is present at the cell surface at very low levels. In addition, little is known about regulation of ANO1 traffic, namely which factors promote its plasma membrane (PM) localization. Here, we generated a novel cellular model, expressing an inducible 3HA-ANO1-eGFP construct, and validated its usage as a microscopy tool to monitor for ANO1 traffic. We demonstrate the robustness and specificity of this cell-based assay, by the identification of siRNAs acting both as ANO1 traffic enhancer and inhibitor, targeting respectively COPB1 and ESYT1 (extended synaptotagmin-1), the latter involved in coupling of the endoplasmic reticulum to the PM at specific microdomains. We further show that knockdown of ESYT1 (and family members ESYT2 and ESYT3) significantly decreased ANO1 current density. This ANO1 cell-based assay constitutes an important tool to be further used in high-throughput screens and drug discovery of high relevance for CF and cancer.
Collapse
Affiliation(s)
- Joana R Lérias
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Madalena C Pinto
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany; Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Hugo M Botelho
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Nikhil T Awatade
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Margarida C Quaresma
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Iris A L Silva
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Podchanart Wanitchakool
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Rainer Schreiber
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | - Margarida D Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016 Lisboa, Portugal; Cell Biology and Biophysics Unit and Advanced Light Microscopy Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany.
| |
Collapse
|
40
|
Zhao QY, Peng YB, Luo XJ, Luo X, Xu H, Wei MY, Jiang QJ, Li WE, Ma LQ, Xu JC, Liu XC, Zang DA, She YS, Zhu H, Shen J, Zhao P, Xue L, Yu MF, Chen W, Zhang P, Fu X, Chen J, Nie X, Shen C, Chen S, Chen S, Chen J, Hu S, Zou C, Qin G, Fang Y, Ding J, Ji G, Zheng YM, Song T, Wang YX, Liu QH. Distinct Effects of Ca 2+ Sparks on Cerebral Artery and Airway Smooth Muscle Cell Tone in Mice and Humans. Int J Biol Sci 2017; 13:1242-1253. [PMID: 29104491 PMCID: PMC5666523 DOI: 10.7150/ijbs.21475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/10/2017] [Indexed: 11/21/2022] Open
Abstract
The effects of Ca2+ sparks on cerebral artery smooth muscle cells (CASMCs) and airway smooth muscle cells (ASMCs) tone, as well as the underlying mechanisms, are not clear. In this investigation, we elucidated the underlying mechanisms of the distinct effects of Ca2+ sparks on cerebral artery smooth muscle cells (CASMCs) and airway smooth muscle cells (ASMCs) tone. In CASMCs, owing to the functional loss of Ca2+-activated Cl- (Clca) channels, Ca2+ sparks activated large-conductance Ca2+-activated K+ channels (BKs), resulting in a decreases in tone against a spontaneous depolarization-caused high tone in the resting state. In ASMCs, Ca2+ sparks induced relaxation through BKs and contraction via Clca channels. However, the integrated result was contraction because Ca2+ sparks activated BKs prior to Clca channels and Clca channels-induced depolarization was larger than BKs-caused hyperpolarization. However, the effects of Ca2+ sparks on both cell types were determined by L-type voltage-dependent Ca2+ channels (LVDCCs). In addition, compared with ASMCs, CASMCs had great and higher amplitude Ca2+ sparks, a higher density of BKs, and higher Ca2+ and voltage sensitivity of BKs. These differences enhanced the ability of Ca2+ sparks to decrease CASMC and to increase ASMC tone. The higher Ca2+ and voltage sensitivity of BKs in CASMCs than ASMCs were determined by the β1 subunits. Moreover, Ca2+ sparks showed the similar effects on human CASMC and ASMC tone. In conclusions, Ca2+ sparks decrease CASMC tone and increase ASMC tone, mediated by BKs and Clca channels, respectively, and finally determined by LVDCCs.
Collapse
Affiliation(s)
- Qing-Yang Zhao
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Yong-Bo Peng
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xiao-Jing Luo
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xi Luo
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Hao Xu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Ming-Yu Wei
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Qiu-Ju Jiang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Wen-Er Li
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Li-Qun Ma
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Jin-Chao Xu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xiao-Cao Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Dun-An Zang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Yu-San She
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - He Zhu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Jinhua Shen
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Ping Zhao
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Lu Xue
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Meng-Fei Yu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Weiwei Chen
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Ping Zhang
- Department of Cerebral Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Xiangning Fu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Jingyu Chen
- Wuxi &Jiangsu Key Laboratory of Organ Transplantation, Department of Cardiothoracic Surgery, Lung Transplant Group, Wuxi People's Hospital, Nanjing Medical University, Jiangsu, China
| | - Xiaowei Nie
- Wuxi &Jiangsu Key Laboratory of Organ Transplantation, Department of Cardiothoracic Surgery, Lung Transplant Group, Wuxi People's Hospital, Nanjing Medical University, Jiangsu, China
| | - Chenyou Shen
- Wuxi &Jiangsu Key Laboratory of Organ Transplantation, Department of Cardiothoracic Surgery, Lung Transplant Group, Wuxi People's Hospital, Nanjing Medical University, Jiangsu, China
| | - Shu Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Shanshan Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Jingcao Chen
- Department of Cerebral Surgery, Zhongnan Hospital, Wuhan University Medical College, Wuhan, 430071, Hubei, China
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Chunbin Zou
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine & School of Engineering, University of Alabama Birmingham, AL, 35294, USA
| | - Ying Fang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiuping Ding
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, 12208, USA
| | - Tengyao Song
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, 12208, USA
| | - Qing-Hua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| |
Collapse
|
41
|
Inflammatory mediators mediate airway smooth muscle contraction through a G protein-coupled receptor-transmembrane protein 16A-voltage-dependent Ca 2+ channel axis and contribute to bronchial hyperresponsiveness in asthma. J Allergy Clin Immunol 2017; 141:1259-1268.e11. [PMID: 28754608 DOI: 10.1016/j.jaci.2017.05.053] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/27/2017] [Accepted: 05/19/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Allergic inflammation has long been implicated in asthmatic hyperresponsiveness of airway smooth muscle (ASM), but its underlying mechanism remains incompletely understood. Serving as G protein-coupled receptor agonists, several inflammatory mediators can induce membrane depolarization, contract ASM, and augment cholinergic contractile response. We hypothesized that the signal cascade integrating on membrane depolarization by the mediators might involve asthmatic hyperresponsiveness. OBJECTIVE We sought to investigate the signaling transduction of inflammatory mediators in ASM contraction and assess its contribution in the genesis of hyperresponsiveness. METHODS We assessed the capacity of inflammatory mediators to induce depolarization currents by electrophysiological analysis. We analyzed the phenotypes of transmembrane protein 16A (TMEM16A) knockout mice, applied pharmacological reagents, and measured the Ca2+ signal during ASM contraction. To study the role of the depolarization signaling in asthmatic hyperresponsiveness, we measured the synergistic contraction by methacholine and inflammatory mediators both ex vivo and in an ovalbumin-induced mouse model. RESULTS Inflammatory mediators, such as 5-hydroxytryptamin, histamine, U46619, and leukotriene D4, are capable of inducing Ca2+-activated Cl- currents in ASM cells, and these currents are mediated by TMEM16A. A combination of multiple analysis revealed that a G protein-coupled receptor-TMEM16A-voltage-dependent Ca2+ channel signaling axis was required for ASM contraction induced by inflammatory mediators. Block of TMEM16A activity may significantly inhibit the synergistic contraction of acetylcholine and the mediators and hence reduces hypersensitivity. CONCLUSIONS A G protein-coupled receptor-TMEM16A-voltage-dependent Ca2+ channel axis contributes to inflammatory mediator-induced ASM contraction and synergistically activated TMEM16A by allergic inflammatory mediators with cholinergic stimuli.
Collapse
|
42
|
Fedigan S, Bradley E, Webb T, Large RJ, Hollywood MA, Thornbury KD, McHale NG, Sergeant GP. Effects of new-generation TMEM16A inhibitors on calcium-activated chloride currents in rabbit urethral interstitial cells of Cajal. Pflugers Arch 2017; 469:1443-1455. [PMID: 28733893 DOI: 10.1007/s00424-017-2028-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/21/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022]
Abstract
Interstitial cells of Cajal (ICC) isolated from the rabbit urethra exhibit Ca2+-activated Cl- currents (I ClCa) that are important for the development of urethral tone. Here, we examined if TMEM16A (ANO1) contributed to this activity by examining the effect of "new-generation" TMEM16A inhibitors, CACCinh-A01 and T16Ainh-A01, on I ClCa recorded from freshly isolated rabbit urethral ICC (RUICC) and on contractions of intact strips of rabbit urethra smooth muscle. Real-time quantitative PCR experiments demonstrated that TMEM16A was highly expressed in rabbit urethra smooth muscle, in comparison to TMEM16B and TMEM16F. Single-cell RT-PCR experiments revealed that only TMEM16A was expressed in freshly isolated RUICC. Depolarization-evoked I ClCa in isolated RUICC, recorded using voltage clamp, were inhibited by CACCinh-A01 and T16Ainh-A01 with IC50 values of 1.2 and 3.4 μM, respectively. Similarly, spontaneous transient inward currents (STICs) recorded from RUICC voltage clamped at -60 mV and spontaneous transient depolarizations (STDs), recorded in current clamp, were also inhibited by CACCinh-A01 and T16Ainh-A01. In contrast, spontaneous Ca2+ waves in isolated RUICC were only partially reduced by CACCinh-A01 and T16Ainh-A01. Finally, neurogenic contractions of strips of rabbit urethra smooth muscle (RUSM), evoked by electric field stimulation (EFS), were also significantly reduced by CACCinh-A01 and T16Ainh-A01. These data are consistent with the idea that TMEM16A is involved with CACCs in RUICC and in contraction of rabbit urethral smooth muscle.
Collapse
Affiliation(s)
- Stephen Fedigan
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Eamonn Bradley
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Timothy Webb
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Roddy J Large
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Mark A Hollywood
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Keith D Thornbury
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Noel G McHale
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Gerard P Sergeant
- The Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland.
| |
Collapse
|
43
|
Keeler AM, Liu D, Zieger M, Xiong L, Salemi J, Bellvé K, Byrne BJ, Fuller DD, ZhuGe R, ElMallah MK. Airway smooth muscle dysfunction in Pompe ( Gaa-/- ) mice. Am J Physiol Lung Cell Mol Physiol 2017; 312:L873-L881. [PMID: 28336814 DOI: 10.1152/ajplung.00568.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 01/01/2023] Open
Abstract
Pompe disease is an autosomal recessive disorder caused by a deficiency of acid α-glucosidase (GAA), an enzyme responsible for hydrolyzing lysosomal glycogen. Deficiency of GAA leads to systemic glycogen accumulation in the lysosomes of skeletal muscle, motor neurons, and smooth muscle. Skeletal muscle and motor neuron pathology are known to contribute to respiratory insufficiency in Pompe disease, but the role of airway pathology has not been evaluated. Here we propose that GAA enzyme deficiency disrupts the function of the trachea and bronchi and this lower airway pathology contributes to respiratory insufficiency in Pompe disease. Using an established mouse model of Pompe disease, the Gaa-/- mouse, we compared histology, pulmonary mechanics, airway smooth muscle (ASM) function, and calcium signaling between Gaa-/- and age-matched wild-type (WT) mice. Lysosomal glycogen accumulation was observed in the smooth muscle of both the bronchi and the trachea in Gaa-/- but not WT mice. Furthermore, Gaa-/- mice had hyporesponsive airway resistance and bronchial ring contraction to the bronchoconstrictive agents methacholine (MCh) and potassium chloride (KCl) and to a bronchodilator (albuterol). Finally, calcium signaling during bronchiolar smooth muscle contraction was impaired in Gaa-/- mice indicating impaired extracellular calcium influx. We conclude that GAA enzyme deficiency leads to glycogen accumulation in the trachea and bronchi and impairs the ability of lower ASM to regulate calcium and respond appropriately to bronchodilator or constrictors. Accordingly, ASM dysfunction may contribute to respiratory impairments in Pompe disease.
Collapse
Affiliation(s)
- Allison M Keeler
- Division of Pulmonary Medicine, Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Donghai Liu
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Marina Zieger
- Division of Pulmonary Medicine, Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Lang Xiong
- Division of Pulmonary Medicine, Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jeffrey Salemi
- Division of Pulmonary Medicine, Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Karl Bellvé
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Barry J Byrne
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, Florida; and
| | - David D Fuller
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Ronghua ZhuGe
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Mai K ElMallah
- Division of Pulmonary Medicine, Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; .,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
44
|
Kondo M, Tsuji M, Hara K, Arimura K, Yagi O, Tagaya E, Takeyama K, Tamaoki J. Chloride ion transport and overexpression of TMEM16A in a guinea-pig asthma model. Clin Exp Allergy 2017; 47:795-804. [PMID: 28109183 DOI: 10.1111/cea.12887] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 12/09/2016] [Accepted: 12/18/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND TMEM16A, a Ca-activated Cl channel, regulates various physiological functions such as mucin secretion. However, the role of TMEM16A in hyper-secretion in asthma is not fully understood. OBJECTIVE The aim of this study is to evaluate Cl ion transport via TMEM16A and determine the localization of TMEM16A in a guinea-pig asthma model. METHODS Guinea-pigs were sensitized with ovalbumin (OVA) i.p. on Days 1 and 8. On Day 22, we assessed OVA challenge-induced Cl ion transport in the sensitized tracheas ex vivo in an Ussing chamber, compared with the non-sensitized tracheas. We then examined the effect of T16Ainh-A01, a TMEM16A inhibitor, on the increase in Cl ion transport. The tracheal epithelium was immunostained with an anti-TMEM16A antibody. Epithelial cells from guinea-pig tracheas were cultured at the air-liquid interface in the presence of IL-13 for in vitro study. We studied the effect of TMEM16A inhibitors on Ca-dependent agonist, uridine triphosphate (UTP)-induced increases in Cl ion transport in the cultured cells. The cells were immunostained with an anti-TMEM16A antibody, an anti-MUC5AC antibody and an anti-α-tubulin antibody. RESULTS OVA challenge induced an increase in short circuit current within 1 min in the OVA-sensitized tracheas but not in the non-sensitized tracheas, which was inhibited by pretreatment of T16Ainh-A01. Sensitized tracheas showed goblet cell metaplasia with more positive TMEM16A immunostaining, particularly in the apical portion compared with the non-sensitized tracheas. The in vitro UTP-induced increase in Cl ion transport was strongly inhibited by pretreatment with T16Ainh-A01, benzbromarone, and niflumic acid. TMEM16A was positively immunostained at the apical portion and in the MUC5AC-positive area in IL-13-induced goblet cell metaplasia. CONCLUSIONS Antigen challenge and Ca-dependent agonist treatment increased Cl ion transport via the overexpression of TMEM16A in goblet cell metaplasia in a guinea-pig asthma model. TMEM16A inhibitors may be useful for the treatment of hyper-secretion in asthma.
Collapse
Affiliation(s)
- M Kondo
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - M Tsuji
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - K Hara
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - K Arimura
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - O Yagi
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - E Tagaya
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - K Takeyama
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - J Tamaoki
- First Department of Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Cabrita I, Benedetto R, Fonseca A, Wanitchakool P, Sirianant L, Skryabin BV, Schenk LK, Pavenstädt H, Schreiber R, Kunzelmann K. Differential effects of anoctamins on intracellular calcium signals. FASEB J 2017; 31:2123-2134. [PMID: 28183802 DOI: 10.1096/fj.201600797rr] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/23/2017] [Indexed: 01/04/2023]
Abstract
The Ca2+-activated Cl- channel TMEM16A [anoctamin (ANO)1] is homologous to yeast Ist2 and has been shown to tether the cortical endoplasmic reticulum (ER) to the plasma membrane. We therefore examined whether ANO1 and other members of the ANO family affect intracellular Ca2+ ([Ca2+]i) signals. It is shown that expression of ANO1 augments Ca2+ store release upon stimulation of GPCRs, whereas knockdown of ANO1, or lack of Ano1 expression in Ano1-/- animals, as shown in an earlier report, inhibits Ca2+ release. ANO6, and -10 show similar effects, whereas expression of ANO4, -8, and -9 attenuate filling of the ER store. The impact of ANO1 and -4 were examined in more detail. ANO1 colocalized and interacted with IP3R, whereas ANO4 colocalized with SERCA Ca2+ pumps and interacted with ORAI-1 channels, respectively. ANO1 Cl currents were rapidly activated exclusively through Ca2+ store release, and remained untouched by influx of extracellular Ca2+ In contrast expression of ANO4 caused a delayed activation of membrane-localized ANO6 channels, solely through store-operated Ca2+ entry via ORAI. Ca2+ signals were inhibited by knocking down expression of endogenous ANO1, -5, -6, and -10 and were also reduced in epithelial cells from Ano10-/- mice. The data suggest that ANOs affect compartmentalized [Ca2+]i signals, which may explain some of the cellular defects related to ANO mutations.-Cabrita, I., Benedetto, R., Fonseca, A., Wanitchakool, P., Sirianant, L., Skryabin, B. V., Schenk, L. K., Pavenstädt, H., Schreiber, R., Kunzelmann, K. Differential effects of anoctamins on intracellular calcium signals.
Collapse
Affiliation(s)
- Inês Cabrita
- Physiological Institute, University of Regensburg, Regensburg, Germany;
| | - Roberta Benedetto
- Physiological Institute, University of Regensburg, Regensburg, Germany
| | - Ana Fonseca
- Physiological Institute, University of Regensburg, Regensburg, Germany
| | | | - Lalida Sirianant
- Physiological Institute, University of Regensburg, Regensburg, Germany
| | - Boris V Skryabin
- Department of Medicine (TRAM), University of Münster, Münster, Germany; and
| | - Laura K Schenk
- Department of Internal Medicine D, Universitätsklinikum Münster, Albert-Schweitzer-Campus, Münster, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, Universitätsklinikum Münster, Albert-Schweitzer-Campus, Münster, Germany
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, Regensburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, Regensburg, Germany;
| |
Collapse
|
46
|
Wang Y, Lu Y, Luo M, Shi X, Pan Y, Zeng H, Deng L. Evaluation of pharmacological relaxation effect of the natural product naringin on in vitro cultured airway smooth muscle cells and in vivo ovalbumin-induced asthma Balb/c mice. Biomed Rep 2016; 5:715-722. [PMID: 28101344 DOI: 10.3892/br.2016.797] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/29/2016] [Indexed: 12/23/2022] Open
Abstract
Asthma has become a common chronic respiratory disease worldwide and its prevalence is predicted to continue increasing in the next decade, particularly in developing countries. A key component in asthma therapy is to alleviate the excessive bronchial airway narrowing ultimately due to airway smooth muscle contraction, which is often facilitated by a smooth muscle relaxant, such as the β2-adrenergic agonists. Recently, bitter taste receptor (TAS2R) agonists, including saccharin and chloroquine, have been found to potently relax the airway smooth muscle cells (ASMCs) via intracellular Ca2+ signaling. This inspires a great interest in screening the vast resource of natural bitter substances for potential bronchodilatory drugs. In the present study, the relaxation effect of naringin, a compound extracted from common grapefruit, on ASMCs cultured in vitro or bronchial airways of Balb/c mice in vivo was evaluated. The results demonstrated that, when exposed to increasing doses of naringin (0.125, 0.25, 0.5 and 1.0 mM), the traction force generated by the cultured ASMCs decreased progressively, while the intracellular calcium flux signaling in the ASMCs increased. When inhaled at increasing doses (15, 30 and 60 µg), naringin also dose-dependently reduced the bronchial airway resistance of the normal and ovalbumin-induced asthma Balb/c mice in response to challenge with methacholine. In conclusion, these findings indicate that naringin was able to effectively relax murine ASMCs in vitro and in vivo, thus suggesting that it is a promising drug agent to be further investigated in the development of novel bronchodilators for the treatment of asthma.
Collapse
Affiliation(s)
- Yue Wang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China; School of Nursing, Changzhou University, Changzhou, Jiangsu 213164, P.R. China; School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Yun Lu
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China; School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Xiaohao Shi
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Yan Pan
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Huilong Zeng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China; School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu 213164, P.R. China; School of Nursing, Changzhou University, Changzhou, Jiangsu 213164, P.R. China; School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu 213164, P.R. China
| |
Collapse
|
47
|
Pera T, Penn RB. Bronchoprotection and bronchorelaxation in asthma: New targets, and new ways to target the old ones. Pharmacol Ther 2016; 164:82-96. [PMID: 27113408 PMCID: PMC4942340 DOI: 10.1016/j.pharmthera.2016.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/07/2016] [Indexed: 01/01/2023]
Abstract
Despite over 50years of inhaled beta-agonists and corticosteroids as the default management or rescue drugs for asthma, recent research suggests that new therapeutic options are likely to emerge. This belief stems from both an improved understanding of what causes and regulates airway smooth muscle (ASM) contraction, and the identification of new targets whose inhibition or activation can relax ASM. In this review we discuss the recent findings that provide new insight into ASM contractile regulation, a revolution in pharmacology that identifies new ways to "tune" G protein-coupled receptors to improve therapeutic efficacy, and the discovery of several novel targets/approaches capable of effecting bronchoprotection or bronchodilation.
Collapse
Affiliation(s)
- Tonio Pera
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| | - Raymond B Penn
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
48
|
Schnúr A, Hegyi P, Rousseau S, Lukacs GL, Veit G. Epithelial Anion Transport as Modulator of Chemokine Signaling. Mediators Inflamm 2016; 2016:7596531. [PMID: 27382190 PMCID: PMC4921137 DOI: 10.1155/2016/7596531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/03/2016] [Accepted: 05/12/2016] [Indexed: 12/16/2022] Open
Abstract
The pivotal role of epithelial cells is to secrete and absorb ions and water in order to allow the formation of a luminal fluid compartment that is fundamental for the epithelial function as a barrier against environmental factors. Importantly, epithelial cells also take part in the innate immune system. As a first line of defense they detect pathogens and react by secreting and responding to chemokines and cytokines, thus aggravating immune responses or resolving inflammatory states. Loss of epithelial anion transport is well documented in a variety of diseases including cystic fibrosis, chronic obstructive pulmonary disease, asthma, pancreatitis, and cholestatic liver disease. Here we review the effect of aberrant anion secretion with focus on the release of inflammatory mediators by epithelial cells and discuss putative mechanisms linking these transport defects to the augmented epithelial release of chemokines and cytokines. These mechanisms may contribute to the excessive and persistent inflammation in many respiratory and gastrointestinal diseases.
Collapse
Affiliation(s)
- Andrea Schnúr
- Department of Physiology, McGill University, Montréal, QC, Canada H3G 1Y6
| | - Péter Hegyi
- Institute for Translational Medicine and 1st Department of Medicine, University of Pécs, Pécs 7624, Hungary
- MTA-SZTE Translational Gastroenterology Research Group, Szeged 6720, Hungary
| | - Simon Rousseau
- The Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada H2X 2P2
| | - Gergely L. Lukacs
- Department of Physiology, McGill University, Montréal, QC, Canada H3G 1Y6
- Department of Biochemistry, McGill University, Montréal, QC, Canada H3G 1Y6
- Groupe de Recherche Axé sur la Structure des Protéines (GRASP), McGill University, Montréal, QC, Canada H3G 1Y6
| | - Guido Veit
- Department of Physiology, McGill University, Montréal, QC, Canada H3G 1Y6
| |
Collapse
|
49
|
Zhang CH, Wang P, Liu DH, Chen CP, Zhao W, Chen X, Chen C, He WQ, Qiao YN, Tao T, Sun J, Peng YJ, Lu P, Zheng K, Craige SM, Lifshitz LM, Keaney JF, Fogarty KE, ZhuGe R, Zhu MS. The molecular basis of the genesis of basal tone in internal anal sphincter. Nat Commun 2016; 7:11358. [PMID: 27101932 PMCID: PMC4844698 DOI: 10.1038/ncomms11358] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/16/2016] [Indexed: 02/06/2023] Open
Abstract
Smooth muscle sphincters exhibit basal tone and control passage of contents through organs such as the gastrointestinal tract; loss of this tone leads to disorders such as faecal incontinence. However, the molecular mechanisms underlying this tone remain unknown. Here, we show that deletion of myosin light-chain kinases (MLCK) in the smooth muscle cells from internal anal sphincter (IAS-SMCs) abolishes basal tone, impairing defecation. Pharmacological regulation of ryanodine receptors (RyRs), L-type voltage-dependent Ca2+ channels (VDCCs) or TMEM16A Ca2+-activated Cl− channels significantly changes global cytosolic Ca2+ concentration ([Ca2+]i) and the tone. TMEM16A deletion in IAS-SMCs abolishes the effects of modulators for TMEM16A or VDCCs on a RyR-mediated rise in global [Ca2+]i and impairs the tone and defecation. Hence, MLCK activation in IAS-SMCs caused by a global rise in [Ca2+]i via a RyR-TMEM16A-VDCC signalling module sets the basal tone. Targeting this module may lead to new treatments for diseases like faecal incontinence. The molecular basis of the basal tone generated by internal anal sphincters (IAS) is largely unknown. Here, the authors show that the tone arises from a global rise in intracellular Ca2+ in smooth muscle cells via a Ryanodine receptor-TMEM16A-L-type Ca2+ channel-MLC kinase pathway, suggesting a potential therapy for IAS motility disorders.
Collapse
Affiliation(s)
- Cheng-Hai Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Pei Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Dong-Hai Liu
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Cai-Ping Chen
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Wei Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Xin Chen
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Chen Chen
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Wei-Qi He
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China.,CAM-SU Genomic Resource Center, Soochow University, Suzhou 215123, China
| | - Yan-Ning Qiao
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Tao Tao
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Jie Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Ya-Jing Peng
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China
| | - Ping Lu
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Kaizhi Zheng
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Siobhan M Craige
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Lawrence M Lifshitz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - John F Keaney
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Kevin E Fogarty
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Ronghua ZhuGe
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing 210061, China.,Innovation Center for Cardiovascular Disorders, Beijing 100029, China
| |
Collapse
|
50
|
Bill A, Gutierrez A, Kulkarni S, Kemp C, Bonenfant D, Voshol H, Duvvuri U, Gaither LA. ANO1/TMEM16A interacts with EGFR and correlates with sensitivity to EGFR-targeting therapy in head and neck cancer. Oncotarget 2016; 6:9173-88. [PMID: 25823819 PMCID: PMC4496210 DOI: 10.18632/oncotarget.3277] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/07/2015] [Indexed: 12/23/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) contributes to the pathogenesis of head&neck squamous cell carcinoma (HNSCC). However, only a subset of HNSCC patients benefit from anti-EGFR targeted therapy. By performing an unbiased proteomics screen, we found that the calcium-activated chloride channel ANO1 interacts with EGFR and facilitates EGFR-signaling in HNSCC. Using structural mutants of EGFR and ANO1 we identified the trans/juxtamembrane domain of EGFR to be critical for the interaction with ANO1. Our results show that ANO1 and EGFR form a functional complex that jointly regulates HNSCC cell proliferation. Expression of ANO1 affected EGFR stability, while EGFR-signaling elevated ANO1 protein levels, establishing a functional and regulatory link between ANO1 and EGFR. Co-inhibition of EGFR and ANO1 had an additive effect on HNSCC cell proliferation, suggesting that co-targeting of ANO1 and EGFR could enhance the clinical potential of EGFR-targeted therapy in HNSCC and might circumvent the development of resistance to single agent therapy. HNSCC cell lines with amplification and high expression of ANO1 showed enhanced sensitivity to Gefitinib, suggesting ANO1 overexpression as a predictive marker for the response to EGFR-targeting agents in HNSCC therapy. Taken together, our results introduce ANO1 as a promising target and/or biomarker for EGFR-directed therapy in HNSCC.
Collapse
Affiliation(s)
- Anke Bill
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Abraham Gutierrez
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Sucheta Kulkarni
- University of Pittsburgh, Medical Center, Department of Otolaryngology, Pittsburgh, PA 15213, USA
| | - Carolyn Kemp
- University of Pittsburgh, Medical Center, Department of Otolaryngology, Pittsburgh, PA 15213, USA
| | - Debora Bonenfant
- Novartis Institutes for Biomedical Research, Basel, CH-4002, Switzerland
| | - Hans Voshol
- Novartis Institutes for Biomedical Research, Basel, CH-4002, Switzerland
| | - Umamaheswar Duvvuri
- University of Pittsburgh, Medical Center, Department of Otolaryngology, Pittsburgh, PA 15213, USA.,VA Pittsburgh HealthCare System, Pittsburgh, PA 15213, USA
| | - L Alex Gaither
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| |
Collapse
|