1
|
Ousingsawat J, Schreiber R, Kunzelmann K. Functional Interdependence of Anoctamins May Influence Conclusions from Overexpression Studies. Int J Mol Sci 2024; 25:9998. [PMID: 39337485 PMCID: PMC11432102 DOI: 10.3390/ijms25189998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/09/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Anoctamin 6 (ANO6, TMEM16F) is a phospholipid (PL) scramblase that moves PLs between both plasma membrane (PM) leaflets and operates as an ion channel. It plays a role in development and is essential for hemostasis, bone mineralization and immune defense. However, ANO6 has also been shown to regulate cellular Ca2+ signaling and PM compartments, thereby controlling the expression of ion channels such as CFTR. Given these pleiotropic effects, we investigated the functional interdependence of the ubiquitous ANO6 with other commonly co-expressed anoctamins. As most expression studies on anoctamins use HEK293 human embryonic kidney cells, we compared ion currents, PL scrambling and Ca2+ signals induced by the overexpression of anoctamins in HEK293 wild-type parental and ANO6-knockout cells. The data suggest that the endogenous expression of ANO6 significantly affects the results obtained from overexpressed anoctamins, particularly after increasing intracellular Ca2+. Thus, a significant interdependence of anoctamins may influence the interpretation of data obtained from the functional analysis of overexpressed anoctamins.
Collapse
Affiliation(s)
| | | | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, University Street 31, D-93053 Regensburg, Germany; (J.O.); (R.S.)
| |
Collapse
|
2
|
Schreiber R, Ousingsawat J, Kunzelmann K. The anoctamins: Structure and function. Cell Calcium 2024; 120:102885. [PMID: 38642428 DOI: 10.1016/j.ceca.2024.102885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/22/2024]
Abstract
When activated by increase in intracellular Ca2+, anoctamins (TMEM16 proteins) operate as phospholipid scramblases and as ion channels. Anoctamin 1 (ANO1) is the Ca2+-activated epithelial anion-selective channel that is coexpressed together with the abundant scramblase ANO6 and additional intracellular anoctamins. In salivary and pancreatic glands, ANO1 is tightly packed in the apical membrane and secretes Cl-. Epithelia of airways and gut use cystic fibrosis transmembrane conductance regulator (CFTR) as an apical Cl- exit pathway while ANO1 supports Cl- secretion mainly by facilitating activation of luminal CFTR and basolateral K+ channels. Under healthy conditions ANO1 modulates intracellular Ca2+ signals by tethering the endoplasmic reticulum, and except of glands its direct secretory contribution as Cl- channel might be small, compared to CFTR. In the kidneys ANO1 supports proximal tubular acid secretion and protein reabsorption and probably helps to excrete HCO3-in the collecting duct epithelium. However, under pathological conditions as in polycystic kidney disease, ANO1 is strongly upregulated and may cause enhanced proliferation and cyst growth. Under pathological condition, ANO1 and ANO6 are upregulated and operate as secretory channel/phospholipid scramblases, partly by supporting Ca2+-dependent processes. Much less is known about the role of other epithelial anoctamins whose potential functions are discussed in this review.
Collapse
Affiliation(s)
- Rainer Schreiber
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, University street 31, D-93053 Regensburg, Germany.
| |
Collapse
|
3
|
Kunzelmann K, Centeio R, Ousingsawat J, Talbi K, Seidler U, Schreiber R. SLC26A9 in airways and intestine: secretion or absorption? Channels (Austin) 2023; 17:2186434. [PMID: 36866602 PMCID: PMC9988340 DOI: 10.1080/19336950.2023.2186434] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
SLC26A9 is one out of 11 proteins that belong to the SLC26A family of anion transporters. Apart from expression in the gastrointestinal tract, SLC26A9 is also found in the respiratory system, in male tissues and in the skin. SLC26A9 has gained attention because of its modifier role in the gastrointestinal manifestation of cystic fibrosis (CF). SLC26A9 appears to have an impact on the extent of intestinal obstruction caused by meconium ileus. SLC26A9 supports duodenal bicarbonate secretion, but was assumed to provide a basal Cl- secretory pathway in airways. However, recent results show that basal airway Cl- secretion is due to cystic fibrosis conductance regulator (CFTR), while SLC26A9 may rather secrete HCO3-, thereby maintaining proper airway surface liquid (ASL) pH. Moreover, SLC26A9 does not secrete but probably supports reabsorption of fluid particularly in the alveolar space, which explains early death by neonatal distress in Slc26a9-knockout animals. While the novel SLC26A9 inhibitor S9-A13 helped to unmask the role of SLC26A9 in the airways, it also provided evidence for an additional role in acid secretion by gastric parietal cells. Here we discuss recent data on the function of SLC26A9 in airways and gut, and how S9-A13 may be useful in unraveling the physiological role of SLC26A9.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität, Universitätsstraße 31, Regensburg, Germany
- CONTACT Karl Kunzelmann
| | - Raquel Centeio
- Institut für Physiologie, Universität, Universitätsstraße 31, Regensburg, Germany
| | - Jiraporn Ousingsawat
- Institut für Physiologie, Universität, Universitätsstraße 31, Regensburg, Germany
| | - Khaoula Talbi
- Institut für Physiologie, Universität, Universitätsstraße 31, Regensburg, Germany
| | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - Rainer Schreiber
- Institut für Physiologie, Universität, Universitätsstraße 31, Regensburg, Germany
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [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
|
5
|
Lin J, Gettings SM, Talbi K, Schreiber R, Taggart MJ, Preller M, Kunzelmann K, Althaus M, Gray MA. Pharmacological inhibitors of the cystic fibrosis transmembrane conductance regulator exert off-target effects on epithelial cation channels. Pflugers Arch 2023; 475:167-179. [PMID: 36205782 PMCID: PMC9849171 DOI: 10.1007/s00424-022-02758-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 02/01/2023]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) anion channel and the epithelial Na+ channel (ENaC) play essential roles in transepithelial ion and fluid transport in numerous epithelial tissues. Inhibitors of both channels have been important tools for defining their physiological role in vitro. However, two commonly used CFTR inhibitors, CFTRinh-172 and GlyH-101, also inhibit non-CFTR anion channels, indicating they are not CFTR specific. However, the potential off-target effects of these inhibitors on epithelial cation channels has to date not been addressed. Here, we show that both CFTR blockers, at concentrations routinely employed by many researchers, caused a significant inhibition of store-operated calcium entry (SOCE) that was time-dependent, poorly reversible and independent of CFTR. Patch clamp experiments showed that both CFTRinh-172 and GlyH-101 caused a significant block of Orai1-mediated whole cell currents, establishing that they likely reduce SOCE via modulation of this Ca2+ release-activated Ca2+ (CRAC) channel. In addition to off-target effects on calcium channels, both inhibitors significantly reduced human αβγ-ENaC-mediated currents after heterologous expression in Xenopus oocytes, but had differential effects on δβγ-ENaC function. Molecular docking identified two putative binding sites in the extracellular domain of ENaC for both CFTR blockers. Together, our results indicate that caution is needed when using these two CFTR inhibitors to dissect the role of CFTR, and potentially ENaC, in physiological processes.
Collapse
Affiliation(s)
- JinHeng Lin
- grid.1006.70000 0001 0462 7212Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH UK ,grid.4991.50000 0004 1936 8948Present Address: Department of Pharmacology, University of Oxford, Oxford, OX1 3QT UK
| | - Sean M. Gettings
- grid.1006.70000 0001 0462 7212School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU UK
| | - Khaoula Talbi
- grid.7727.50000 0001 2190 5763Physiological Institute, University of Regensburg, 93053 Regensburg, Germany
| | - Rainer Schreiber
- grid.7727.50000 0001 2190 5763Physiological Institute, University of Regensburg, 93053 Regensburg, Germany
| | - Michael J. Taggart
- grid.1006.70000 0001 0462 7212Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH UK
| | - Matthias Preller
- grid.425058.e0000 0004 0473 3519Department of Natural Sciences/Institute for Functional Gene Analytics, Structural Biology Group, Bonn-Rhein-Sieg University of Applied Sciences, 53359 Rheinbach, Germany
| | - Karl Kunzelmann
- grid.7727.50000 0001 2190 5763Physiological Institute, University of Regensburg, 93053 Regensburg, Germany
| | - Mike Althaus
- grid.1006.70000 0001 0462 7212School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU UK ,grid.425058.e0000 0004 0473 3519Present Address: Department of Natural Sciences /Institute for Functional Gene Analytics, Ion Transport Physiology Group, Bonn-Rhein-Sieg University of Applied Sciences, 53359 Rheinbach, Germany
| | - Michael A. Gray
- grid.1006.70000 0001 0462 7212Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH UK
| |
Collapse
|
6
|
Madácsy T, Varga Á, Papp N, Tél B, Pallagi P, Szabó V, Kiss A, Fanczal J, Rakonczay Z, Tiszlavicz L, Rázga Z, Hohwieler M, Kleger A, Gray M, Hegyi P, Maléth J. Impaired regulation of PMCA activity by defective CFTR expression promotes epithelial cell damage in alcoholic pancreatitis and hepatitis. Cell Mol Life Sci 2022; 79:265. [PMID: 35484438 PMCID: PMC11073305 DOI: 10.1007/s00018-022-04287-1] [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: 01/05/2022] [Revised: 03/09/2022] [Accepted: 04/04/2022] [Indexed: 11/28/2022]
Abstract
Alcoholic pancreatitis and hepatitis are frequent, potentially lethal diseases with limited treatment options. Our previous study reported that the expression of CFTR Cl- channel is impaired by ethanol in pancreatic ductal cells leading to more severe alcohol-induced pancreatitis. In addition to determining epithelial ion secretion, CFTR has multiple interactions with other proteins, which may influence intracellular Ca2+ signaling. Thus, we aimed to investigate the impact of ethanol-mediated CFTR damage on intracellular Ca2+ homeostasis in pancreatic ductal epithelial cells and cholangiocytes. Human and mouse pancreas and liver samples and organoids were used to study ion secretion, intracellular signaling, protein expression and interaction. The effect of PMCA4 inhibition was analyzed in a mouse model of alcohol-induced pancreatitis. The decreased CFTR expression impaired PMCA function and resulted in sustained intracellular Ca2+ elevation in ethanol-treated and mouse and human pancreatic organoids. Liver samples derived from alcoholic hepatitis patients and ethanol-treated mouse liver organoids showed decreased CFTR expression and function, and impaired PMCA4 activity. PMCA4 co-localizes and physically interacts with CFTR on the apical membrane of polarized epithelial cells, where CFTR-dependent calmodulin recruitment determines PMCA4 activity. The sustained intracellular Ca2+ elevation in the absence of CFTR inhibited mitochondrial function and was accompanied with increased apoptosis in pancreatic epithelial cells and PMCA4 inhibition increased the severity of alcohol-induced AP in mice. Our results suggest that improving Ca2+ extrusion in epithelial cells may be a potential novel therapeutic approach to protect the exocrine pancreatic function in alcoholic pancreatitis and prevent the development of cholestasis in alcoholic hepatitis.
Collapse
Affiliation(s)
- Tamara Madácsy
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
| | - Árpád Varga
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
| | - Noémi Papp
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
| | - Bálint Tél
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Petra Pallagi
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
| | - Viktória Szabó
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
| | - Aletta Kiss
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
| | - Júlia Fanczal
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary
| | - Zoltan Rakonczay
- Department of Pathophysiology, University of Szeged, Szeged, 6720, Hungary
| | | | - Zsolt Rázga
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Meike Hohwieler
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Mike Gray
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, England
| | - Péter Hegyi
- Institute for Translational Medicine, University of Pécs, Pécs, Hungary
- Centre for Translational Medicine and Division for Pancreatic Disorders, Semmelweis University, Budapest, Hungary
| | - József Maléth
- Department of Medicine, University of Szeged, Szeged, 6720, Hungary.
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Szeged, 6720, Hungary.
- HCEMM-USZ Molecular Gastroenterology Research Group, University of Szeged, Szeged, 6720, Hungary.
| |
Collapse
|
7
|
Khuituan P, Huipao N, Jeanmard N, Thantongsakul S, Promjun W, Chuthong S, Tipbunjong C, Peerakietkhajorn S. Sargassum plagiophyllum Extract Enhances Colonic Functions and Modulates Gut Microbiota in Constipated Mice. Nutrients 2022; 14:496. [PMID: 35276855 PMCID: PMC8838385 DOI: 10.3390/nu14030496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 12/11/2022] Open
Abstract
Constipation is a symptom that is widely found in the world’s population. Various dietary supplementations are used to relieve and prevent constipation. Seaweed is widely used for its health benefits. In this study, we aimed to investigate the effects of Sargassum plagiophyllum extract (SPE) on functions of the gastrointestinal tract and gut microbiota. The results show that SPE pretreatment increased the frequency of gut contraction, leading to reduce gut transit time. SPE pretreatment also significantly increased the secretion of Cl− and reduced Na+ absorption, increasing fecal water content in constipated mice (p < 0.05). In addition, the Bifidobacteria population in cecal contents was significantly higher in constipated mice pretreated with 500 mg/kg SPE for 14 days than in untreated constipated mice (p < 0.05). Our findings suggest that SPE can prevent constipation in loperamide-induced mice. This study may be useful for the development of human food supplements from S. plagiophyllum, which prevent constipation.
Collapse
Affiliation(s)
- Pissared Khuituan
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand; (P.K.); (N.H.); (C.T.)
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
| | - Nawiya Huipao
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand; (P.K.); (N.H.); (C.T.)
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
| | - Nilobon Jeanmard
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Sitthiwach Thantongsakul
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Warittha Promjun
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Suwarat Chuthong
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Chittipong Tipbunjong
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand; (P.K.); (N.H.); (C.T.)
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
| | - Saranya Peerakietkhajorn
- Gut Biology and Microbiota Research Unit, Prince of Songkla University, Songkhla 90110, Thailand; (N.J.); (S.T.); (W.P.); (S.C.)
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
8
|
Wilczyński B, Dąbrowska A, Saczko J, Kulbacka J. The Role of Chloride Channels in the Multidrug Resistance. MEMBRANES 2021; 12:38. [PMID: 35054564 PMCID: PMC8781147 DOI: 10.3390/membranes12010038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/23/2021] [Indexed: 12/19/2022]
Abstract
Nowadays, one of medicine's main and most challenging aims is finding effective ways to treat cancer. Unfortunately, although there are numerous anti-cancerous drugs, such as cisplatin, more and more cancerous cells create drug resistance. Thus, it is equally important to find new medicines and research the drug resistance phenomenon and possibilities to avoid this mechanism. Ion channels, including chloride channels, play an important role in the drug resistance phenomenon. Our article focuses on the chloride channels, especially the volume-regulated channels (VRAC) and CLC chloride channels family. VRAC induces multidrug resistance (MDR) by causing apoptosis connected with apoptotic volume decrease (AVD) and VRAC are responsible for the transport of anti-cancerous drugs such as cisplatin. VRACs are a group of heterogenic complexes made from leucine-rich repetition with 8A (LRRC8A) and a subunit LRRC8B-E responsible for the properties. There are probably other subunits, which can create those channels, for example, TTYH1 and TTYH2. It is also known that the ClC family is involved in creating MDR in mainly two mechanisms-by changing the cell metabolism or acidification of the cell. The most researched chloride channel from this family is the CLC-3 channel. However, other channels are playing an important role in inducing MDR as well. In this paper, we review the role of chloride channels in MDR and establish the role of the channels in the MDR phenomenon.
Collapse
Affiliation(s)
- Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, L. Pasteura 1, 50-367 Wroclaw, Poland; (B.W.); (A.D.)
| | - Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, L. Pasteura 1, 50-367 Wroclaw, Poland; (B.W.); (A.D.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
9
|
Zhong L, Gleason EL. Adenylate Cyclase 1 Links Calcium Signaling to CFTR-Dependent Cytosolic Chloride Elevations in Chick Amacrine Cells. Front Cell Neurosci 2021; 15:726605. [PMID: 34456687 PMCID: PMC8385318 DOI: 10.3389/fncel.2021.726605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/19/2021] [Indexed: 01/03/2023] Open
Abstract
The strength and sign of synapses involving ionotropic GABA and glycine receptors are dependent upon the Cl- gradient. We have shown that nitric oxide (NO) elicits the release of Cl- from internal acidic stores in retinal amacrine cells (ACs); temporarily altering the Cl- gradient and the strength or even sign of incoming GABAergic or glycinergic synapses. The underlying mechanism for this effect of NO requires the cystic fibrosis transmembrane regulator (CFTR) but the link between NO and CFTR activation has not been determined. Here, we test the hypothesis that NO-dependent Ca2+ elevations activate the Ca2+-dependent adenylate cyclase 1 (AdC1) leading to activation of protein kinase A (PKA) whose activity is known to open the CFTR channel. Using the reversal potential of GABA-gated currents to monitor cytosolic Cl-, we established the requirement for Ca2+ elevations. Inhibitors of AdC1 suppressed the NO-dependent increases in cytosolic Cl- whereas inhibitors of other AdC subtypes were ineffective suggesting that AdC1 is involved. Inhibition of PKA also suppressed the action of NO. To address the sufficiency of this pathway in linking NO to elevations in cytosolic Cl-, GABA-gated currents were measured under internal and external zero Cl- conditions to isolate the internal Cl- store. Activators of the cAMP pathway were less effective than NO in producing GABA-gated currents. However, coupling the cAMP pathway activators with the release of Ca2+ from stores produced GABA-gated currents indistinguishable from those stimulated with NO. Together, these results demonstrate that cytosolic Ca2+ links NO to the activation of CFTR and the elevation of cytosolic Cl-.
Collapse
Affiliation(s)
- Li Zhong
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Evanna L Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
10
|
CLCA1 Regulates Airway Mucus Production and Ion Secretion Through TMEM16A. Int J Mol Sci 2021; 22:ijms22105133. [PMID: 34066250 PMCID: PMC8151571 DOI: 10.3390/ijms22105133] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
TMEM16A, a Ca2+-activated chloride channel (CaCC), and its regulator, CLCA1, are associated with inflammatory airway disease and goblet cell metaplasia. CLCA1 is a secreted protein with protease activity that was demonstrated to enhance membrane expression of TMEM16A. Expression of CLCA1 is particularly enhanced in goblet cell metaplasia and is associated with various lung diseases. However, mice lacking expression of CLCA1 showed the same degree of mucous cell metaplasia and airway hyperreactivity as asthmatic wild-type mice. To gain more insight into the role of CLCA1, we applied secreted N-CLCA1, produced in vitro, to mice in vivo using intratracheal instillation. We observed no obvious upregulation of TMEM16A membrane expression by CLCA1 and no differences in ATP-induced short circuit currents (Iscs). However, intraluminal mucus accumulation was observed by treatment with N-CLCA1 that was not seen in control animals. The effects of N-CLCA1 were augmented in ovalbumin-sensitized mice. Mucus production induced by N-CLCA1 in polarized BCi-NS1 human airway epithelial cells was dependent on TMEM16A expression. IL-13 upregulated expression of CLCA1 and enhanced mucus production, however, without enhancing purinergic activation of Isc. In contrast to polarized airway epithelial cells and mouse airways, which express very low levels of TMEM16A, nonpolarized airway cells express large amounts of TMEM16A protein and show strong CaCC. The present data show an only limited contribution of TMEM16A to airway ion secretion but suggest a significant role of both CLCA1 and TMEM16A for airway mucus secretion.
Collapse
|
11
|
Rimessi A, Vitto VAM, Patergnani S, Pinton P. Update on Calcium Signaling in Cystic Fibrosis Lung Disease. Front Pharmacol 2021; 12:581645. [PMID: 33776759 PMCID: PMC7990772 DOI: 10.3389/fphar.2021.581645] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder characterized by mutations in the cystic fibrosis transmembrane conductance regulator gene, which causes multifunctional defects that preferentially affect the airways. Abnormal viscosity of mucus secretions, persistent pathogen infections, hyperinflammation, and lung tissue damage compose the classical pathological manifestation referred to as CF lung disease. Among the multifunctional defects associated with defective CFTR, increasing evidence supports the relevant role of perturbed calcium (Ca2+) signaling in the pathophysiology of CF lung disease. The Ca2+ ion is a critical player in cell functioning and survival. Its intracellular homeostasis is maintained by a fine balance between channels, transporters, and exchangers, mediating the influx and efflux of the ion across the plasma membrane and the intracellular organelles. An abnormal Ca2+ profile has been observed in CF cells, including airway epithelial and immune cells, with heavy repercussions on cell function, viability, and susceptibility to pathogens, contributing to proinflammatory overstimulation, organelle dysfunction, oxidative stress, and excessive cytokines release in CF lung. This review discusses the role of Ca2+ signaling in CF and how its dysregulation in airway epithelial and immune cells contributes to hyperinflammation in the CF lung. Finally, we provide an outlook on the therapeutic options that target the Ca2+ signaling to treat the CF lung disease.
Collapse
Affiliation(s)
- Alessandro Rimessi
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.,Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy
| | - Veronica A M Vitto
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.,Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy
| |
Collapse
|
12
|
Abdel Hameid R, Cormet-Boyaka E, Kuebler WM, Uddin M, Berdiev BK. SARS-CoV-2 may hijack GPCR signaling pathways to dysregulate lung ion and fluid transport. Am J Physiol Lung Cell Mol Physiol 2021; 320:L430-L435. [PMID: 33434105 PMCID: PMC7938641 DOI: 10.1152/ajplung.00499.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The tropism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a virus responsible for the ongoing coronavirus disease 2019 (COVID-19) pandemic, toward the host cells is determined, at least in part, by the expression and distribution of its cell surface receptor, angiotensin-converting enzyme 2 (ACE2). The virus further exploits the host cellular machinery to gain access into the cells; its spike protein is cleaved by a host cell surface transmembrane serine protease 2 (TMPRSS2) shortly after binding ACE2, followed by its proteolytic activation at a furin cleavage site. The virus primarily targets the epithelium of the respiratory tract, which is covered by a tightly regulated airway surface liquid (ASL) layer that serves as a primary defense mechanism against respiratory pathogens. The volume and viscosity of this fluid layer is regulated and maintained by a coordinated function of different transport pathways in the respiratory epithelium. We argue that SARS-CoV-2 may potentially alter evolutionary conserved second-messenger signaling cascades via activation of G protein-coupled receptors (GPCRs) or by directly modulating G protein signaling. Such signaling may in turn adversely modulate transepithelial transport processes, especially those involving cystic fibrosis transmembrane conductance regulator (CFTR) and epithelial Na+ channel (ENaC), thereby shifting the delicate balance between anion secretion and sodium absorption, which controls homeostasis of this fluid layer. As a result, activation of the secretory pathways including CFTR-mediated Cl− transport may overwhelm the absorptive pathways, such as ENaC-dependent Na+ uptake, and initiate a pathophysiological cascade leading to lung edema, one of the most serious and potentially deadly clinical manifestations of COVID-19.
Collapse
Affiliation(s)
- Reem Abdel Hameid
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | | | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mohammed Uddin
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bakhrom K Berdiev
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
13
|
Grebert C, Becq F, Vandebrouck C. Phospholipase C controls chloride-dependent short-circuit current in human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2020; 320:L205-L219. [PMID: 33236921 DOI: 10.1152/ajplung.00437.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chloride secretion by airway epithelial cells is primordial for water and ion homeostasis and airways surface prevention of infections. This secretion is impaired in several human diseases, including cystic fibrosis, a genetic pathology due to CFTR gene mutations leading to chloride channel defects. A potential therapeutic approach is aiming at increasing chloride secretion either by correcting the mutated CFTR itself or by stimulating non-CFTR chloride channels at the plasma membrane. Here, we studied the role of phospholipase C in regulating the transepithelial chloride secretion in human airway epithelial 16HBE14o- and CFBE cells over-expressing wild type (WT)- or F508del-CFTR. Western blot analysis shows expression of the three endogenous phospholipase C (PLC) isoforms, namely, PLCδ1, PLCγ1, and PLCβ3 in 16HBE14o- cells. In 16HBE14o- cells, we performed Ussing chamber experiments after silencing each of these PLC isoforms or using the PLC inhibitor U73122 or its inactive analogue U73343. Our results show the involvement of PLCβ3 and PLCγ1 in CFTR-dependent short-circuit current activated by forskolin, but not of PLCδ1. In CFBE-WT CFTR and corrected CFBE-F508del CFTR cells, PLCβ3 silencing also inhibits CFTR-dependent current activated by forskolin and UTP-activated calcium-dependent chloride channels (CaCC). Our study supports the importance of PLC in maintaining CFTR-dependent chloride secretion over time, getting maximal CFTR-dependent current and increasing CaCC activation in bronchial epithelial cells.
Collapse
Affiliation(s)
- Chloé Grebert
- Laboratoire Signalisation et Transports Ioniques Membranaires, Université de Poitiers, Poitiers, France
| | - Frédéric Becq
- Laboratoire Signalisation et Transports Ioniques Membranaires, Université de Poitiers, Poitiers, France
| | - Clarisse Vandebrouck
- Laboratoire Signalisation et Transports Ioniques Membranaires, Université de Poitiers, Poitiers, France
| |
Collapse
|
14
|
Shan W, Hu Y, Ding J, Yang X, Lou J, Du Q, Liao Q, Luo L, Xu J, Xie R. Advances in Ca 2+ modulation of gastrointestinal anion secretion and its dysregulation in digestive disorders (Review). Exp Ther Med 2020; 20:8. [PMID: 32934673 PMCID: PMC7471861 DOI: 10.3892/etm.2020.9136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/22/2020] [Indexed: 11/29/2022] Open
Abstract
Intracellular calcium (Ca2+) is a critical cell signaling component in gastrointestinal (GI) physiology. Cytosolic calcium ([Ca2+]cyt), as a secondary messenger, controls GI epithelial fluid and ion transport, mucus and neuropeptide secretion, as well as synaptic transmission and motility. The key roles of Ca2+ signaling in other types of secretory cell (including those in the airways and salivary glands) are well known. However, its action in GI epithelial secretion and the underlying molecular mechanisms have remained to be fully elucidated. The present review focused on the role of [Ca2+]cyt in GI epithelial anion secretion. Ca2+ signaling regulates the activities of ion channels and transporters involved in GI epithelial ion and fluid transport, including Cl- channels, Ca2+-activated K+ channels, cystic fibrosis (CF) transmembrane conductance regulator and anion/HCO3- exchangers. Previous studies by the current researchers have focused on this field over several years, providing solid evidence that Ca2+ signaling has an important role in the regulation of GI epithelial anion secretion and uncovering underlying molecular mechanisms. The present review is largely based on previous studies by the current researchers and provides an overview of the currently known molecular mechanisms of GI epithelial anion secretion with an emphasis on Ca2+-mediated ion secretion and its dysregulation in GI disorders. In addition, previous studies by the current researchers demonstrated that different regulatory mechanisms are in place for GI epithelial HCO3- and Cl- secretion. An increased understanding of the roles of Ca2+ signaling and its targets in GI anion secretion may lead to the development of novel strategies to inhibit GI diseases, including the enhancement of fluid secretion in CF and protection of the GI mucosa in ulcer diseases.
Collapse
Affiliation(s)
- Weixi Shan
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yanxia Hu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jianhong Ding
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Xiaoxu Yang
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jun Lou
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qian Du
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qiushi Liao
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Lihong Luo
- Department of Oncology and Geriatrics, Traditional Chinese Medicine Hospital of Chishui City, Guizhou 564700, P.R. China
| | - Jingyu Xu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Rui Xie
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
15
|
Wang Y, Zhao J, Cai Y, Ballard HJ. Cystic fibrosis transmembrane conductance regulator-dependent bicarbonate entry controls rat cardiomyocyte ATP release via pannexin1 through mitochondrial signalling and caspase activation. Acta Physiol (Oxf) 2020; 230:e13495. [PMID: 32386453 DOI: 10.1111/apha.13495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/27/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022]
Abstract
AIM Cystic fibrosis transmembrane conductance regulator (CFTR) is expressed in the heart, but its function there is unclear. CFTR regulates an ATP release pore in many tissues, but the identity and regulatory mechanism of the pore are unknown. We investigated the role of CFTR in ATP release from primary cardiomyocytes and ventricular wall in vivo. METHODS Proteins involved in the signalling pathway for ATP release during simulated ischaemia (lactic acid treatment) were investigated using inhibitors and siRNA; colocalization was identified by coimmunofluorescence and proximity ligation assays; changes in near-membrane pH and calcium were identified with total internal reflection microscopy; in vivo ATP release was investigated using interstitial microdialysis of rat heart. RESULTS Lactic acid-induced CFTR-dependent ATP release from cultured cardiomyocytes and left ventricle in vivo. Lactic acid entry elevated near-membrane calcium, which involved Na/H- and Na/Ca-exchangers colocalized with CFTR. Calcium entry-induced CFTR activation, which involved cAMP, protein kinase A, FAK, Pyk2 and Src. Removal of extracellular bicarbonate abolished cardiomyocyte ATP release induced by lactic acid or CFTR activators. Bicarbonate stimulated cytochrome c expression, cytochrome c release and ATP release from isolated cardiomyocyte mitochondria. Pannexin 1 (Panx1) colocalized with CFTR. Lactic acid increased cardiomyocyte caspase activity: caspase inhibitors or Panx1 siRNA abolished cardiomyocyte ATP release, while pannexin inhibition abolished cardiac ATP release in vivo. CONCLUSION During simulated ischaemia, CFTR-dependent bicarbonate entry stimulated ATP and cytochrome c release from mitochondria; in the cytoplasm, cytochrome c-activated caspase 3, which in turn activated Panx1, and ATP was released through the opened Panx1 channel.
Collapse
Affiliation(s)
- Yongshun Wang
- School of Biomedical Sciences The University of Hong Kong Pokfulam Hong Kong
| | - Junjun Zhao
- School of Biomedical Sciences The University of Hong Kong Pokfulam Hong Kong
| | - Yin Cai
- Department of Anaesthesiology The University of Hong Kong Pokfulam Hong Kong
| | - Heather J. Ballard
- School of Biomedical Sciences The University of Hong Kong Pokfulam Hong Kong
| |
Collapse
|
16
|
Cyst growth in ADPKD is prevented by pharmacological and genetic inhibition of TMEM16A in vivo. Nat Commun 2020; 11:4320. [PMID: 32859916 PMCID: PMC7455562 DOI: 10.1038/s41467-020-18104-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
In autosomal dominant polycystic kidney disease (ADPKD) multiple bilateral renal cysts gradually enlarge, leading to a decline in renal function. Transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and TMEM16A (anoctamin 1) are known to drive cyst enlargement. Here we demonstrate that loss of Pkd1 increased expression of TMEM16A and CFTR and Cl- secretion in murine kidneys, with TMEM16A essentially contributing to cyst growth. Upregulated TMEM16A enhanced intracellular Ca2+ signaling and proliferation of Pkd1-deficient renal epithelial cells. In contrast, increase in Ca2+ signaling, cell proliferation and CFTR expression was not observed in Pkd1/Tmem16a double knockout mice. Knockout of Tmem16a or inhibition of TMEM16A in vivo by the FDA-approved drugs niclosamide and benzbromarone, as well as the TMEM16A-specific inhibitor Ani9 largely reduced cyst enlargement and abnormal cyst cell proliferation. The present data establish a therapeutic concept for the treatment of ADPKD.
Collapse
|
17
|
Cabrini G, Rimessi A, Borgatti M, Lampronti I, Finotti A, Pinton P, Gambari R. Role of Cystic Fibrosis Bronchial Epithelium in Neutrophil Chemotaxis. Front Immunol 2020; 11:1438. [PMID: 32849500 PMCID: PMC7427443 DOI: 10.3389/fimmu.2020.01438] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
A hallmark of cystic fibrosis (CF) chronic respiratory disease is an extensive neutrophil infiltrate in the mucosa filling the bronchial lumen, starting early in life for CF infants. The genetic defect of the CF Transmembrane conductance Regulator (CFTR) ion channel promotes dehydration of the airway surface liquid, alters mucus properties, and decreases mucociliary clearance, favoring the onset of recurrent and, ultimately, chronic bacterial infection. Neutrophil infiltrates are unable to clear bacterial infection and, as an adverse effect, contribute to mucosal tissue damage by releasing proteases and reactive oxygen species. Moreover, the rapid cellular turnover of lumenal neutrophils releases nucleic acids that further alter the mucus viscosity. A prominent role in the recruitment of neutrophil in bronchial mucosa is played by CF bronchial epithelial cells carrying the defective CFTR protein and are exposed to whole bacteria and bacterial products, making pharmacological approaches to regulate the exaggerated neutrophil chemotaxis in CF a relevant therapeutic target. Here we revise: (a) the major receptors, kinases, and transcription factors leading to the expression, and release of neutrophil chemokines in bronchial epithelial cells; (b) the role of intracellular calcium homeostasis and, in particular, the calcium crosstalk between endoplasmic reticulum and mitochondria; (c) the epigenetic regulation of the key chemokines; (d) the role of mutant CFTR protein as a co-regulator of chemokines together with the host-pathogen interactions; and (e) different pharmacological strategies to regulate the expression of chemokines in CF bronchial epithelial cells through novel drug discovery and drug repurposing.
Collapse
Affiliation(s)
- Giulio Cabrini
- Center for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy.,Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Alessandro Rimessi
- Center for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy.,Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Monica Borgatti
- Center for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy.,Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Ilaria Lampronti
- Center for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy.,Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Alessia Finotti
- Center for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy.,Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Center for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy.,Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberto Gambari
- Center for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Ferrara, Italy.,Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
18
|
Transport properties in CFTR-/- knockout piglets suggest normal airway surface liquid pH and enhanced amiloride-sensitive Na + absorption. Pflugers Arch 2020; 472:1507-1519. [PMID: 32712714 PMCID: PMC7476968 DOI: 10.1007/s00424-020-02440-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/12/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Previous analysis of CFTR-knockout (CFTR-/-) in piglets has provided important insights into the pathology of cystic fibrosis. However, controversies exist as to the true contribution of CFTR to the pH balance in airways and intestine. We therefore compared ion transport properties in newborn wild-type (CFTR+/+) and CFTR-knockout (CFTR-/- piglets). Tracheas of CFTR-/- piglets demonstrated typical cartilage malformations and muscle cell bundles. CFTR-/- airway epithelial cells showed enhanced lipid peroxidation, suggesting inflammation early in life. CFTR was mainly expressed in airway submucosal glands and was absent in lungs of CFTR-/- piglets, while expression of TMEM16A was uncompromised. mRNA levels for TMEM16A, TMEM16F, and αβγENaC were unchanged in CFTR-/- airways, while mRNA for SLC26A9 appeared reduced. CFTR was undetectable in epithelial cells of CFTR-/- airways and intestine. Small intestinal epithelium of CFTR-/- piglets showed mucus accumulation. Secretion of both electrolytes and mucus was activated by stimulation with prostaglandin E2 and ATP in the intestine of CFTR+/+, but not of CFTR-/- animals. pH was measured inside small bronchi using a pH microelectrode and revealed no difference between CFTR+/+ and CFTR-/- piglets. Intracellular pH in porcine airway epithelial cells revealed only a small contribution of CFTR to bicarbonate secretion, which was absent in cells from CFTR-/- piglets. In contrast to earlier reports, our data suggest a minor impact of CFTR on ASL pH. In contrast, enhanced amiloride-sensitive Na+ absorption may contribute to lung pathology in CFTR-/- piglets, along with a compromised CFTR- and TMEM16A-dependent Cl- transport.
Collapse
|
19
|
Mondejar-Parreño G, Perez-Vizcaino F, Cogolludo A. Kv7 Channels in Lung Diseases. Front Physiol 2020; 11:634. [PMID: 32676036 PMCID: PMC7333540 DOI: 10.3389/fphys.2020.00634] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/18/2020] [Indexed: 12/23/2022] Open
Abstract
Lung diseases constitute a global health concern causing disability. According to WHO in 2016, respiratory diseases accounted for 24% of world population mortality, the second cause of death after cardiovascular diseases. The Kv7 channels family is a group of voltage-dependent K+ channels (Kv) encoded by KCNQ genes that are involved in various physiological functions in numerous cell types, especially, cardiac myocytes, smooth muscle cells, neurons, and epithelial cells. Kv7 channel α-subunits are regulated by KCNE1–5 ancillary β-subunits, which modulate several characteristics of Kv7 channels such as biophysical properties, cell-location, channel trafficking, and pharmacological sensitivity. Kv7 channels are mainly expressed in two large groups of lung tissues: pulmonary arteries (PAs) and bronchial tubes. In PA, Kv7 channels are expressed in pulmonary artery smooth muscle cells (PASMCs); while in the airway (trachea, bronchus, and bronchioles), Kv7 channels are expressed in airway smooth muscle cells (ASMCs), airway epithelial cells (AEPs), and vagal airway C-fibers (VACFs). The functional role of Kv7 channels may vary depending on the cell type. Several studies have demonstrated that the impairment of Kv7 channel has a strong impact on pulmonary physiology contributing to the pathophysiology of different respiratory diseases such as cystic fibrosis, asthma, chronic obstructive pulmonary disease, chronic coughing, lung cancer, and pulmonary hypertension. Kv7 channels are now recognized as playing relevant physiological roles in many tissues, which have encouraged the search for Kv7 channel modulators with potential therapeutic use in many diseases including those affecting the lung. Modulation of Kv7 channels has been proposed to provide beneficial effects in a number of lung conditions. Therefore, Kv7 channel openers/enhancers or drugs acting partly through these channels have been proposed as bronchodilators, expectorants, antitussives, chemotherapeutics and pulmonary vasodilators.
Collapse
Affiliation(s)
- Gema Mondejar-Parreño
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Angel Cogolludo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| |
Collapse
|
20
|
Ahmadi S, Wu YS, Li M, Ip W, Lloyd-Kuzik A, Di Paola M, Du K, Xia S, Lew A, Bozoky Z, Forman-Kay J, Bear CE, Gonska T. Augmentation of Cystic Fibrosis Transmembrane Conductance Regulator Function in Human Bronchial Epithelial Cells via SLC6A14-Dependent Amino Acid Uptake. Implications for Treatment of Cystic Fibrosis. Am J Respir Cell Mol Biol 2020; 61:755-764. [PMID: 31189070 DOI: 10.1165/rcmb.2019-0094oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
SLC6A14-mediated l-arginine transport has been shown to augment the residual anion channel activity of the major mutant, F508del-CFTR, in the murine gastrointestinal tract. It is not yet known if this transporter augments residual and pharmacological corrected F508del-CFTR in primary airway epithelia. We sought to determine the role of l-arginine uptake via SLC6A14 in modifying F508del-CFTR channel activity in airway cells from patients with cystic fibrosis (CF). Human bronchial epithelial (HBE) cells from lung explants of patients without CF (HBE) and those with CF (CF-HBE) were used for H3-flux, airway surface liquid, and Ussing chamber studies. We used α-methyltryptophan as a specific inhibitor for SLC6A14. CFBE41o-, a commonly used CF airway cell line, was employed for studying the mechanism of the functional interaction between SLC6A14 and F508del-CFTR. SLC6A14 is functionally expressed in CF-HBE cells. l-arginine uptake via SLC6A14 augmented F508del-CFTR function at baseline and after treatment with lumacaftor. SLC6A14-mediated l-arginine uptake also increased the airway surface liquid in CF-HBE cells. Using CFBE41o cells, we showed that the positive SLC6A14 effect was mainly dependent on the nitric oxide (NO) synthase activity, nitrogen oxides, including NO, and phosphorylation by protein kinase G. These finding were confirmed in CF-HBE, as inducible NO synthase inhibition abrogated the functional interaction between SLC6A14 and pharmacological corrected F508del-CFTR. In summary, SLC6A14-mediated l-arginine transport augments residual F508del-CFTR channel function via a noncanonical, NO pathway. This effect is enhanced with increasing pharmacological rescue of F508del-CFTR to the membrane. The current study demonstrates how endogenous pathways can be used for the development of companion therapy in CF.
Collapse
Affiliation(s)
- Saumel Ahmadi
- Department of Physiology.,Programme in Molecular Medicine.,Programme in Genetics and Genome Biology, and
| | - Yu-Sheng Wu
- Department of Physiology.,Programme in Molecular Medicine
| | - Mingyuan Li
- Programme in Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Wan Ip
- Programme in Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrew Lloyd-Kuzik
- Department of Physiology.,Programme in Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Kai Du
- Department of Biochemistry, and
| | - Sunny Xia
- Department of Physiology.,Programme in Molecular Medicine
| | | | | | - Julie Forman-Kay
- Department of Biochemistry, and.,Programme in Molecular Medicine
| | - Christine E Bear
- Department of Physiology.,Department of Biochemistry, and.,Programme in Molecular Medicine
| | - Tanja Gonska
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada; and.,Programme in Translational Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
21
|
TMEM16A: An Alternative Approach to Restoring Airway Anion Secretion in Cystic Fibrosis? Int J Mol Sci 2020; 21:ijms21072386. [PMID: 32235608 PMCID: PMC7177896 DOI: 10.3390/ijms21072386] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/18/2022] Open
Abstract
The concept that increasing airway hydration leads to improvements in mucus clearance and lung function in cystic fibrosis has been clinically validated with osmotic agents such as hypertonic saline and more convincingly with cystic fibrosis transmembrane conductance regulator (CFTR) repair therapies. Although rapidly becoming the standard of care in cystic fibrosis (CF), current CFTR modulators do not treat all patients nor do they restore the rate of decline in lung function to normal levels. As such, novel approaches are still required to ensure all with CF have effective therapies. Although CFTR plays a fundamental role in the regulation of fluid secretion across the airway mucosa, there are other ion channels and transporters that represent viable targets for future therapeutics. In this review article we will summarise the current progress with CFTR-independent approaches to restoring mucosal hydration, including epithelial sodium channel (ENaC) blockade and modulators of SLC26A9. A particular emphasis is given to modulation of the airway epithelial calcium-activated chloride channel (CaCC), TMEM16A, as there is controversy regarding whether it should be positively or negatively modulated. This is discussed in light of a recent report describing for the first time bona fide TMEM16A potentiators and their positive effects upon epithelial fluid secretion and mucus clearance.
Collapse
|
22
|
Genovese M, Borrelli A, Venturini A, Guidone D, Caci E, Viscido G, Gambardella G, di Bernardo D, Scudieri P, Galietta LJV. TRPV4 and purinergic receptor signalling pathways are separately linked in airway epithelia to CFTR and TMEM16A chloride channels. J Physiol 2019; 597:5859-5878. [PMID: 31622498 DOI: 10.1113/jp278784] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/09/2019] [Indexed: 01/09/2023] Open
Abstract
KEY POINTS Eact is a putative pharmacological activator of TMEM16A. Eact is strongly effective in recombinant Fischer rat thyroid (FRT) cells but not in airway epithelial cells with endogenous TMEM16A expression. Transcriptomic analysis, gene silencing and functional studies in FRT cells reveal that Eact is actually an activator of the Ca2+ -permeable TRPV4 channel. In airway epithelial cells TRPV4 and TMEM16A are expressed in separate cell types. Intracellular Ca2+ elevation by TRPV4 stimulation leads to CFTR channel activation. ABSTRACT TMEM16A is a Ca2+ -activated Cl- channel expressed in airway epithelial cells, particularly under conditions of mucus hypersecretion. To investigate the role of TMEM16A, we used Eact, a putative TMEM16A pharmacological activator. However, in contrast to purinergic stimulation, we found little effect of Eact on bronchial epithelial cells under conditions of high TMEM16A expression. We hypothesized that Eact is an indirect activator of TMEM16A. By a combination of approaches, including short-circuit current recordings, bulk and single cell RNA sequencing, intracellular Ca2+ imaging and RNA interference, we found that Eact is actually an activator of the Ca2+ -permeable TRPV4 channel and that the modest effect of this compound in bronchial epithelial cells is due to a separate expression of TMEM16A and TRPV4 in different cell types. Importantly, we found that TRPV4 stimulation induced activation of the CFTR Cl- channel. Our study reveals the existence of separate Ca2+ signalling pathways linked to different Cl- secretory processes.
Collapse
Affiliation(s)
- Michele Genovese
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Anna Borrelli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Arianna Venturini
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Emanuela Caci
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Gaetano Viscido
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Paolo Scudieri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Department of Translational Medical Sciences (DISMET), Università di Napoli Federico II, Napoli, Italy
| |
Collapse
|
23
|
Yeh KM, Johansson O, Le H, Rao K, Markus I, Perera DS, Lubowski DZ, King DW, Zhang L, Chen H, Liu L. Cystic fibrosis transmembrane conductance regulator modulates enteric cholinergic activities and is abnormally expressed in the enteric ganglia of patients with slow transit constipation. J Gastroenterol 2019; 54:994-1006. [PMID: 31392489 DOI: 10.1007/s00535-019-01610-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/31/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cystic fibrosis transmembrane conductance regulator (CFTR) was recently found in the enteric nervous system, where its role is unclear. We aimed to identify which enteric neuronal structures express CFTR, whether CFTR modulates enteric neurotransmission and if altered CFTR expression is associated with slow transit constipation (STC). METHODS Immunofluorescence double labeling was performed to localize CFTR with various neuronal and glial cell markers in the human colon. The immunoreactivity (IR) of CFTR and choline acetyltransferase (ChAT) on myenteric plexus of control and STC colon was quantitatively analyzed. In control colonic muscle strips, electrical field stimulation (EFS) evoked contractile responses and the release of acetylcholine (ACh) was measured in the presence of the CFTR channel inhibitor, CFTR(inh)-172. RESULTS CFTR-IR was densely localized to myenteric ganglia, where it was co-localized with neuronal markers HuC/D and β-tubulin, and glial marker S-100 but little with glial fibrillary acidic protein. Vesicular ACh transport was almost exclusively co-localized with CFTR, but neurons expressing nitric oxide synthase were CFTR negative. Significant reductions of CFTR-IR (P < 0.01) and ChAT-IR (P < 0.05) were observed on myenteric ganglia of STC compared to control. Pre-treatment of colonic muscle strips with CFTR(inh)-172 (10 µM) significantly inhibited EFS-evoked contractile responses (P < 0.01) and ACh release (P < 0.05). CONCLUSIONS Co-localization of CFTR-IR with cholinergic markers, inhibition of EFS-induced colonic muscle contractility and ACh release by CFTR(inh)-172 suggest that CFTR modulates enteric cholinergic neurotransmission. The downregulation of CFTR and ChAT in myenteric ganglia of STC correlated with the impaired contractile responses to EFS.
Collapse
Affiliation(s)
- Ka Ming Yeh
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Olle Johansson
- Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Huy Le
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Karan Rao
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Irit Markus
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | | | | | | | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Hongzhuan Chen
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Liu
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
24
|
Delpiano L, Gray MA. Location, location, location: lessons from airway epithelial anion channels. J Physiol 2019; 597:5739-5740. [PMID: 31654403 DOI: 10.1113/jp279125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Livia Delpiano
- Epithelial Research Group, Institute for Cell & Molecular Biosciences, The Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Michael A Gray
- Epithelial Research Group, Institute for Cell & Molecular Biosciences, The Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
25
|
Kuan SP, Liao YSJ, Davis KM, Messer JG, Zubcevic J, Aguirre JI, Reznikov LR. Attenuated Amiloride-Sensitive Current and Augmented Calcium-Activated Chloride Current in Marsh Rice Rat (Oryzomys palustris) Airways. iScience 2019; 19:737-748. [PMID: 31491720 PMCID: PMC6731178 DOI: 10.1016/j.isci.2019.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/22/2019] [Accepted: 08/05/2019] [Indexed: 11/29/2022] Open
Abstract
Prolonged heat and sea salt aerosols pose a challenge for the mammalian airway, placing the protective airway surface liquid (ASL) at risk for desiccation. Thus, mammals inhabiting salt marshes might have acquired adaptations for ASL regulation. We studied the airways of the rice rat, a rodent that inhabits salt marshes. We discovered negligible Na+ transport through the epithelial sodium channel (ENaC). In contrast, carbachol induced a large Cl- secretory current that was blocked by the calcium-activated chloride channel (CaCC) inhibitor CaCCinhi-A01. Decreased mRNA expression of α, β, and γ ENaC, and increased mRNA expression of the CaCC transmembrane member 16A, distinguished the rice rat airway. Rice rat airway cultures also secreted fluid in response to carbachol and displayed an exaggerated expansion of the ASL volume when challenged with 3.5% NaCl. These data suggest that the rice rat airway might possess unique ion transport adaptations to facilitate survival in the salt marsh environment.
Collapse
Affiliation(s)
- Shin-Ping Kuan
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Yan-Shin J Liao
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Katelyn M Davis
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Jonathan G Messer
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Jasenka Zubcevic
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - J Ignacio Aguirre
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Leah R Reznikov
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
26
|
Cabrita I, Benedetto R, Schreiber R, Kunzelmann K. Niclosamide repurposed for the treatment of inflammatory airway disease. JCI Insight 2019; 4:128414. [PMID: 31391337 DOI: 10.1172/jci.insight.128414] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/02/2019] [Indexed: 12/22/2022] Open
Abstract
Inflammatory airway diseases, such as asthma, cystic fibrosis (CF), and chronic obstructive pulmonary disease (COPD), are characterized by mucus hypersecretion and airway plugging. In both CF and asthma, enhanced expression of the Ca2+-activated Cl- channel TMEM16A is detected in mucus-producing club/goblet cells and airway smooth muscle. TMEM16A contributes to mucus hypersecretion and bronchoconstriction, which are both inhibited by blockers of TMEM16A, such as niflumic acid. Here we demonstrate that the FDA-approved drug niclosamide, a potent inhibitor of TMEM16A identified by high-throughput screening, is an inhibitor of both TMEM16A and TMEM16F. In asthmatic mice, niclosamide reduced mucus production and secretion, as well as bronchoconstriction, and showed additional antiinflammatory effects. Using transgenic asthmatic mice, we found evidence that TMEM16A and TMEM16F are required for normal mucus production/secretion, which may be due to their effects on intracellular Ca2+ signaling. TMEM16A and TMEM16F support exocytic release of mucus and inflammatory mediators, both of which are blocked by niclosamide. Thus, inhibition of mucus and cytokine release, bronchorelaxation, and reported antibacterial effects make niclosamide a potentially suitable drug for the treatment of inflammatory airway diseases, such as CF, asthma, and COPD.
Collapse
|
27
|
Abu-Arish A, Pandžić E, Kim D, Tseng HW, Wiseman PW, Hanrahan JW. Agonists that stimulate secretion promote the recruitment of CFTR into membrane lipid microdomains. J Gen Physiol 2019; 151:834-849. [PMID: 31048413 PMCID: PMC6572005 DOI: 10.1085/jgp.201812143] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 04/05/2019] [Indexed: 01/20/2023] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a tightly regulated anion channel that mediates secretion by epithelia and is mutated in the disease cystic fibrosis. CFTR forms macromolecular complexes with many proteins; however, little is known regarding its associations with membrane lipids or the regulation of its distribution and mobility at the cell surface. We report here that secretagogues (agonists that stimulate secretion) such as the peptide hormone vasoactive intestinal peptide (VIP) and muscarinic agonist carbachol increase CFTR aggregation into cholesterol-dependent clusters, reduce CFTR lateral mobility within and between membrane microdomains, and trigger the fusion of clusters into large (3.0 µm2) ceramide-rich platforms. CFTR clusters are closely associated with motile cilia and with the enzyme acid sphingomyelinase (ASMase) that is constitutively bound on the cell surface. Platform induction is prevented by pretreating cells with cholesterol oxidase to disrupt lipid rafts or by exposure to the ASMase functional inhibitor amitriptyline or the membrane-impermeant reducing agent 2-mercaptoethanesulfonate. Platforms are reversible, and their induction does not lead to an increase in apoptosis; however, blocking platform formation does prevent the increase in CFTR surface expression that normally occurs during VIP stimulation. These results demonstrate that CFTR is colocalized with motile cilia and reveal surprisingly robust regulation of CFTR distribution and lateral mobility, most likely through autocrine redox activation of extracellular ASMase. Formation of ceramide-rich platforms containing CFTR enhances transepithelial secretion and likely has other functions related to inflammation and mucosal immunity.
Collapse
Affiliation(s)
- Asmahan Abu-Arish
- Department of Physiology, McGill University, Montréal, Canada
- Department of Physics, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
| | - Elvis Pandžić
- Department of Physics, McGill University, Montréal, Canada
| | - Dusik Kim
- Department of Physiology, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
| | - Hsin Wei Tseng
- Department of Physiology, McGill University, Montréal, Canada
| | - Paul W Wiseman
- Department of Physics, McGill University, Montréal, Canada
- Department of Chemistry, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
| | - John W Hanrahan
- Department of Physiology, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
- McGill University Health Centre Research Institute, Montréal, Canada
| |
Collapse
|
28
|
Rimessi A, Bezzerri V, Salvatori F, Tamanini A, Nigro F, Dechecchi MC, Santangelo A, Prandini P, Munari S, Provezza L, Garreau de Loubresse N, Muller J, Ribeiro CMP, Lippi G, Gambari R, Pinton P, Cabrini G. PLCB3 Loss of Function Reduces Pseudomonas aeruginosa-Dependent IL-8 Release in Cystic Fibrosis. Am J Respir Cell Mol Biol 2019; 59:428-436. [PMID: 29668297 DOI: 10.1165/rcmb.2017-0267oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The lungs of patients with cystic fibrosis (CF) are characterized by an exaggerated inflammation driven by secretion of IL-8 from bronchial epithelial cells and worsened by Pseudomonas aeruginosa infection. To identify novel antiinflammatory molecular targets, we previously performed a genetic study of 135 genes of the immune response, which identified the c.2534C>T (p.S845L) variant of phospholipase C-β3 (PLCB3) as being significantly associated with mild progression of pulmonary disease. Silencing PLCB3 revealed that it potentiates the Toll-like receptor's inflammatory signaling cascade originating from CF bronchial epithelial cells. In the present study, we investigated the role of the PLCB3-S845L variant together with two synthetic mutants paradigmatic of impaired catalytic activity or lacking functional activation in CF bronchial epithelial cells. In experiments in which cells were exposed to P. aeruginosa, the supernatant of mucopurulent material from the airways of patients with CF or different agonists revealed that PLCB3-S845L has defects of 1) agonist-induced Ca2+ release from endoplasmic reticulum and rise of Ca2+ concentration, 2) activation of conventional protein kinase C isoform β, and 3) induction of IL-8 release. These results, besides identifying S845L as a loss-of-function variant, strengthen the importance of targeting PLCB3 to mitigate the CF inflammatory response in bronchial epithelial cells without blunting the immune response.
Collapse
Affiliation(s)
| | - Valentino Bezzerri
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Francesca Salvatori
- 3 Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Anna Tamanini
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Federica Nigro
- 1 Department of Morphology, Surgery and Experimental Medicine and
| | - Maria Cristina Dechecchi
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Alessandra Santangelo
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Paola Prandini
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Silvia Munari
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Lisa Provezza
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Nicolas Garreau de Loubresse
- 4 Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Jean Muller
- 5 Laboratoire de diagnostic génétique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France; and
| | - Carla M P Ribeiro
- 6 Department of Medicine, and.,7 Department of Cell Biology and Physiology, Cystic Fibrosis Research Center, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Giuseppe Lippi
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Roberto Gambari
- 3 Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- 1 Department of Morphology, Surgery and Experimental Medicine and
| | - Giulio Cabrini
- 2 Laboratory of Molecular Pathology, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| |
Collapse
|
29
|
Zhang F, Wan H, Yang X, He J, Lu C, Yang S, Tuo B, Dong H. Molecular mechanisms of caffeine-mediated intestinal epithelial ion transports. Br J Pharmacol 2019; 176:1700-1716. [PMID: 30808064 DOI: 10.1111/bph.14640] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/10/2019] [Accepted: 01/31/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE As little is known about the effect of caffeine, one of the most widely consumed substances worldwide, on intestinal function, we aimed to study its action on intestinal anion secretion and the underlying molecular mechanisms. EXPERIMENTAL APPROACH Anion secretion and channel expression were examined in mouse duodenal epithelium by Ussing chambers and immunocytochemistry. Ca2+ imaging was also performed in intestinal epithelial cells (IECs). KEY RESULTS Caffeine (10 mM) markedly increased mouse duodenal short-circuit current (Isc ), which was attenuated by a removal of either Cl- or HCO3 - , Ca2+ -free serosal solutions and selective blockers of store-operated Ca2+ channels (SOC/Ca2+ release-activated Ca2+ channels), and knockdown of Orai1 channels on the serosal side of duodenal tissues. Caffeine induced SOC entry in IEC, which was inhibited by ruthenium red and selective blockers of SOC. Caffeine-stimulated duodenal Isc was inhibited by the endoplasmic reticulum Ca2+ chelator (N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine), selective blockers (ruthenium red and dantrolene) of ryanodine receptors (RyR), and of Ca2+ -activated Cl- channels (niflumic acid and T16A). There was synergism between cAMP and Ca2+ signalling, in which cAMP/PKA promoted caffeine/Ca2+ -mediated anion secretion. Expression of STIM1 and Orai1 was detected in mouse duodenal mucosa and human IECs. The Orai1 proteins were primarily co-located with the basolateral marker Na+ , K+ -ATPase. CONCLUSIONS AND IMPLICATIONS Caffeine stimulated intestinal anion secretion mainly through the RyR/Orai1/Ca2+ signalling pathway. There is synergism between cAMP/PKA and caffeine/Ca2+ -mediated anion secretion. Our findings suggest that a caffeine-mediated RyR/Orai1/Ca2+ pathway could provide novel potential drug targets to control intestinal anion secretion.
Collapse
Affiliation(s)
- Fenglian Zhang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hanxing Wan
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xin Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jialin He
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Lu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi, China
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Department of Medicine, School of Medicine, University of California, San Diego, California, USA
| |
Collapse
|
30
|
Patel W, Moore PJ, Sassano MF, Lopes-Pacheco M, Aleksandrov AA, Amaral MD, Tarran R, Gray MA. Increases in cytosolic Ca 2+ induce dynamin- and calcineurin-dependent internalisation of CFTR. Cell Mol Life Sci 2019; 76:977-994. [PMID: 30547226 PMCID: PMC6394554 DOI: 10.1007/s00018-018-2989-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/28/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-regulated, apical anion channel that regulates ion and fluid transport in many epithelia including the airways. We have previously shown that cigarette smoke (CS) exposure to airway epithelia causes a reduction in plasma membrane CFTR expression which correlated with a decrease in airway surface hydration. The effect of CS on CFTR was dependent on an increase in cytosolic Ca2+. However, the underlying mechanism for this Ca2+-dependent, internalisation of CFTR is unknown. To gain a better understanding of the effect of Ca2+ on CFTR, we performed whole cell current recordings to study the temporal effect of raising cytosolic Ca2+ on CFTR function. We show that an increase in cytosolic Ca2+ induced a time-dependent reduction in whole cell CFTR conductance, which was paralleled by a loss of cell surface CFTR expression, as measured by confocal and widefield fluorescence microscopy. The decrease in CFTR conductance and cell surface expression were both dynamin-dependent. Single channel reconstitution studies showed that raising cytosolic Ca2+ per se had no direct effect on CFTR. In fact, the loss of CFTR plasma membrane activity correlated with activation of calcineurin, a Ca2+-dependent phosphatase, suggesting that dephosphorylation of CFTR was linked to the loss of surface expression. In support of this, the calcineurin inhibitor, cyclosporin A, prevented the Ca2+-induced decrease in cell surface CFTR. These results provide a hitherto unrecognised role for cytosolic Ca2+ in modulating the residency of CFTR at the plasma membrane through a dynamin- and calcineurin-dependent mechanism.
Collapse
Affiliation(s)
- Waseema Patel
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick J Moore
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Flori Sassano
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miquéias Lopes-Pacheco
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| | - Andrei A Aleksandrov
- Department of Biochemistry and Biophysics, Cystic Fibrosis Research and Treatment Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Margarida D Amaral
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| | - Robert Tarran
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, Cystic Fibrosis Research and Treatment Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael A Gray
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
31
|
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: 55] [Impact Index Per Article: 9.2] [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
|
32
|
Schreiber R, Buchholz B, Kraus A, Schley G, Scholz J, Ousingsawat J, Kunzelmann K. Lipid Peroxidation Drives Renal Cyst Growth In Vitro through Activation of TMEM16A. J Am Soc Nephrol 2019; 30:228-242. [PMID: 30606785 DOI: 10.1681/asn.2018010039] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 11/19/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Transepithelial chloride- secretion, through the chloride channels cystic fibrosis transmembrane conductance regulator (CFTR) and TMEM16A (anoctamin 1), drives cyst enlargement in polycystic kidney disease (PKD). Polycystic kidneys are hypoxic, and oxidative stress activates TMEM16A. However, mechanisms for channel activation in PKD remain obscure. METHODS Using tissue samples from patients with autosomal dominant PKD, embryonic kidney cultures, and an MDCK in vitro cyst model, we assessed peroxidation of plasma membrane phospholipids in human and mouse polycystic kidneys. We also used electrophysiologic Ussing chamber and patch clamp experiments to analyze activation of TMEM16A and growth of renal cysts. RESULTS Peroxidation of phospholipids in human and mouse kidneys as well as MDCK cysts in vitro is probably due to enhanced levels of reactive oxygen species. Lipid peroxidation correlated with increased cyst volume as shown in renal cultures and MDCK cysts in three-dimensional cultures. Reactive oxygen species and lipid peroxidation strongly activated TMEM16A, leading to depletion of calcium ion stores and store-operated calcium influx. Activation of TMEM16A- and CFTR-dependent chloride secretion strongly augmented cyst growth. Exposure to scavengers of reactive oxygen species, such as glutathione, coenzyme Q10, or idebenone (a synthetic coenzyme Q10 homolog), as well as inhibition of oxidative lipid damage by ferrostatin-1 largely reduced activation of TMEM16A. Inhibition of TMEM16A reduced proliferation and fluid secretion in vitro. CONCLUSIONS These findings indicate that activation of TMEM16A by lipid peroxidation drives growth of renal cysts. We propose direct inhibition of TMEM16A or inhibition of lipid peroxidation as potentially powerful therapeutic approaches to delay cyst development in PKD.
Collapse
Affiliation(s)
- Rainer Schreiber
- Department of Physiology, University of Regensburg, Regensburg, Germany; and
| | - Björn Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andre Kraus
- Department of Nephrology and Hypertension, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gunnar Schley
- Department of Nephrology and Hypertension, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Scholz
- Department of Nephrology and Hypertension, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | | | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, Regensburg, Germany; and
| |
Collapse
|
33
|
Hoppe S, Breves G, Klinger S. Calcium-induced chloride secretion is decreased by Resveratrol in ileal porcine tissue. BMC Res Notes 2018; 11:719. [PMID: 30309374 PMCID: PMC6182809 DOI: 10.1186/s13104-018-3825-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/08/2018] [Indexed: 11/16/2022] Open
Abstract
Objective Chloride (Cl−) secretion is crucial for intestinal fluid secretion. Therefore, effects of the polyphenol Resveratrol (RSV) on Cl− secretion have been investigated. In a previous study, we observed effects of RSV on forskolin-induced Cl− secretion in the porcine jejunum but not the ileum although RSV itself induced a transepithelial ion current that may represent Cl− secretion in the ileum. The aim of this study was to gain further insights regarding the effects of RSV on characteristics of Cl− secretion in the porcine ileum using the Ussing chamber technique (recording of short circuit currents (Isc) as a measure for epithelial net ion transfer). Results RSV increased the Isc in the porcine ileum but not in the porcine jejunum as is already known. This increase was absent in a Cl−-free buffer system, indicating that RSV indeed induces Cl− secretion. However, the carbachol-induced Isc was significantly inhibited by RSV indicating an inhibition of Ca2+-induced Cl− secretion. The cellular basis for these contradictory, segment specific results of RSV on Cl− secretion has to be subjected to further studies. The results also underline, that is difficult to generalize effects of RSV between different intestinal locations, organs, cell culture models or species. Electronic supplementary material The online version of this article (10.1186/s13104-018-3825-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Susanne Hoppe
- Department of Physiology, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173, Hannover, Germany
| | - Gerhard Breves
- Department of Physiology, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173, Hannover, Germany
| | - Stefanie Klinger
- Department of Physiology, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173, Hannover, Germany.
| |
Collapse
|
34
|
Rottgen TS, Nickerson AJ, Minor EA, Stewart AB, Harold AD, Rajendran VM. Dextran sulfate sodium-induced chronic colitis attenuates Ca 2+-activated Cl - secretion in murine colon by downregulating TMEM16A. Am J Physiol Cell Physiol 2018; 315:C10-C20. [PMID: 29561662 PMCID: PMC6087728 DOI: 10.1152/ajpcell.00328.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/13/2018] [Accepted: 03/13/2018] [Indexed: 12/29/2022]
Abstract
Attenuated Ca2+-activated Cl- secretion has previously been observed in the model of dextran sulfate sodium (DSS)-induced colitis. Prior studies have implicated dysfunctional muscarinic signaling from basolateral membranes as the potential perpetrator leading to decreased Ca2+-activated Cl- secretion. However, in our chronic model of DSS-colitis, cholinergic receptor muscarinic 3 ( Chrm3) transcript (1.028 ± 0.12 vs. 1.029 ± 0.27, P > 0.05) and CHRM3 protein expression (1.021 ± 0.24 vs. 0.928 ± 0.09, P > 0.05) were unchanged. Therefore, we hypothesized that decreased carbachol (CCH)-stimulated Cl- secretion in DSS-induced colitis could be attributed to a loss of Ca2+-activated Cl- channels (CaCC) in apical membranes of colonic epithelium. To establish this chemically-induced colitis, Balb/C mice were exposed to 4% DSS for five alternating weeks to stimulate a more moderate, chronic colitis. Upon completion of the protocol, whole thickness sections of colon were mounted in an Ussing chamber under voltage-clamp conditions. DSS-induced colitis demonstrated a complete inhibition of basolateral administration of CCH-stimulated Cl- secretion that actually displayed a reversal in polarity (15.40 ± 2.22 μA/cm2 vs. -2.47 ± 0.25 μA/cm2). Western blotting of potential CaCCs, quantified by densitometric analysis, demonstrated no change in bestrophin-2 and cystic fibrosis transmembrane regulator, whereas anoctamin-1 [ANO1, transmembrane protein 16A (TMEM16A)] was significantly downregulated (1.001 ± 0.13 vs. 0.510 ± 0.12, P < 0.05). Our findings indicate that decreased expression of TMEM16A in DSS-induced colitis contributes to the decreased Ca2+-activated Cl- secretion in murine colon.
Collapse
Affiliation(s)
- Trey S Rottgen
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Andrew J Nickerson
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Emily A Minor
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | | | - Abby D Harold
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Vazhaikkurichi M Rajendran
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| |
Collapse
|
35
|
Muimo R, Alothaid HM, Mehta A. NM23 proteins: innocent bystanders or local energy boosters for CFTR? J Transl Med 2018; 98:272-282. [PMID: 29251738 DOI: 10.1038/labinvest.2017.121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 08/25/2017] [Accepted: 09/12/2017] [Indexed: 12/17/2022] Open
Abstract
NM23 proteins NDPK-A and -B bind to the cystic fibrosis (CF) protein CFTR in different ways from kinases such as PKA, CK2 and AMPK or linkers to cell calcium such as calmodulin and annexins. NDPK-A (not -B) interacts with CFTR through reciprocal AMPK binding/control, whereas NDPK-B (not -A) binds directly to CFTR. NDPK-B can activate G proteins without ligand-receptor coupling, so perhaps NDPK-B's binding influences energy supply local to a nucleotide-binding site (NBD1) needed for CFTR to function. Curiously, CFTR (ABC-C7) is a member of the ATP-binding cassette (ABC) protein family that does not obey 'clan rules'; CFTR channels anions and is not a pump, regulates disparate processes, is itself regulated by multiple means and is so pleiotropic that it acts as a hub that orchestrates calcium signaling through its consorts such as calmodulin/annexins. Furthermore, its multiple partners make CFTR dance to different tunes in different cellular and subcellular locations as it recycles from the plasma membrane to endosomes. CFTR function in airway apical membranes is inhibited by smoking which has been dubbed 'acquired CF'. CFTR alone among family members possesses a trap for other proteins that it unfurls as a 'fish-net' and which bears consensus phosphorylation sites for many protein kinases, with PKA being the most canonical. Recently, the site of CFTR's commonest mutation has been proposed as a knock-in mutant that alters allosteric control of kinase CK2 by log orders of activity towards calmodulin and other substrates after CFTR fragmentation. This link from CK2 to calmodulin that binds the R region invokes molecular paths that control lumen formation, which is incomplete in the tracheas of some CF-affected babies. Thus, we are poised to understand the many roles of NDPK-A and -B in CFTR function and, especially lumen formation, which is defective in the gut and lungs of many CF babies.
Collapse
Affiliation(s)
- Richmond Muimo
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, UK
| | - Hani Mm Alothaid
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, UK
| | - Anil Mehta
- Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
36
|
Yang X, Wen G, Tuo B, Zhang F, Wan H, He J, Yang S, Dong H. Molecular mechanisms of calcium signaling in the modulation of small intestinal ion transports and bicarbonate secretion. Oncotarget 2017; 9:3727-3740. [PMID: 29423078 PMCID: PMC5790495 DOI: 10.18632/oncotarget.23197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/01/2017] [Indexed: 01/13/2023] Open
Abstract
Background and Purpose: Although Ca2+ signaling may stimulate small intestinal ion secretion, little is known about its critical role and the molecular mechanisms of Ca2+-mediated biological action. Key Results Activation of muscarinic receptors by carbachol(CCh) stimulated mouse duodenal Isc, which was significantly inhibited in Ca2+-free serosal solution and by several selective store-operated Ca2+ channels(SOC) blockers added to the serosal side of duodenal tissues. Furthermore, we found that CRAC/Orai channels may represent the molecular candidate of SOC in intestinal epithelium. CCh increased intracellular Ca2+ but not cAMP, and Ca2+ signaling mediated duodenal Cl- and HCO3- secretion in wild type mice but not in CFTR knockout mice. CCh induced duodenal ion secretion and stimulated PI3K/Akt activity in duodenal epithelium, all of which were inhibited by selective PI3K inhibitors with different structures. CCh-induced Ca2+ signaling also stimulated the phosphorylation of CFTR proteins and their trafficking to the plasma membrane of duodenal epithelial cells, which were inhibited again by selective PI3K inhibitors. Materials and Methods Functional, biochemical and morphological experiments were performed to examine ion secretion, PI3K/Akt and CFTR activity of mouse duodenal epithelium. Ca2+ imaging was performed on HT-29 cells. Conclusions and Implications Ca2+ signaling plays a critical role in intestinal ion secretion via CRAC/Orai-mediated SOCE mechanism on the serosal side of epithelium. We also demonstrated the molecular mechanisms of Ca2+ signaling in CFTR-mediated secretion via novel PI3K/Akt pathway. Our findings suggest new perspectives for drug targets to protect the upper GI tract and control liquid homeostasis in the small intestine.
Collapse
Affiliation(s)
- Xin Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Guorong Wen
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China
| | - Fenglian Zhang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Hanxing Wan
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Jialin He
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Shiming Yang
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi 563003, China
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.,Department of Medicine, School of Medicine, University of California, San Diego, CA 92093, USA
| |
Collapse
|
37
|
Benedetto R, Ousingsawat J, Wanitchakool P, Zhang Y, Holtzman MJ, Amaral M, Rock JR, Schreiber R, Kunzelmann K. Epithelial Chloride Transport by CFTR Requires TMEM16A. Sci Rep 2017; 7:12397. [PMID: 28963502 PMCID: PMC5622110 DOI: 10.1038/s41598-017-10910-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/16/2017] [Indexed: 12/15/2022] Open
Abstract
Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) is the secretory chloride/bicarbonate channel in airways and intestine that is activated through ATP binding and phosphorylation by protein kinase A, but fails to operate in cystic fibrosis (CF). TMEM16A (also known as anoctamin 1, ANO1) is thought to function as the Ca2+ activated secretory chloride channel independent of CFTR. Here we report that tissue specific knockout of the TMEM16A gene in mouse intestine and airways not only eliminates Ca2+-activated Cl− currents, but unexpectedly also abrogates CFTR-mediated Cl− secretion and completely abolishes cAMP-activated whole cell currents. The data demonstrate fundamentally new roles of TMEM16A in differentiated epithelial cells: TMEM16A provides a mechanism for enhanced ER Ca2+ store release, possibly engaging Store Operated cAMP Signaling (SOcAMPS) and activating Ca2+ regulated adenylyl cyclases. TMEM16A is shown to be essential for proper activation and membrane expression of CFTR. This intimate regulatory relationship is the cause for the functional overlap of CFTR and Ca2+-dependent chloride transport.
Collapse
Affiliation(s)
- Roberta Benedetto
- Physiological institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany
| | - Jiraporn Ousingsawat
- Physiological institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany
| | - Podchanart Wanitchakool
- Physiological institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany
| | - Yong Zhang
- Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael J Holtzman
- Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Margarida Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, C8, 1749-016, Lisboa, Portugal
| | - Jason R Rock
- Department of Anatomy, University of California, San Francisco, USA
| | - Rainer Schreiber
- Physiological institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany
| | - Karl Kunzelmann
- Physiological institute, University of Regensburg, University street 31, D-93053, Regensburg, Germany.
| |
Collapse
|
38
|
Krishnan V, Maddox JW, Rodriguez T, Gleason E. A role for the cystic fibrosis transmembrane conductance regulator in the nitric oxide-dependent release of Cl - from acidic organelles in amacrine cells. J Neurophysiol 2017; 118:2842-2852. [PMID: 28835528 DOI: 10.1152/jn.00511.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/16/2022] Open
Abstract
γ-Amino butyric acid (GABA) and glycine typically mediate synaptic inhibition because their ligand-gated ion channels support the influx of Cl- However, the electrochemical gradient for Cl- across the postsynaptic plasma membrane determines the voltage response of the postsynaptic cell. Typically, low cytosolic Cl- levels support inhibition, whereas higher levels of cytosolic Cl- can suppress inhibition or promote depolarization. We previously reported that nitric oxide (NO) releases Cl- from acidic organelles and transiently elevates cytosolic Cl-, making the response to GABA and glycine excitatory. In this study, we test the hypothesis that the cystic fibrosis transmembrane conductance regulator (CFTR) is involved in the NO-dependent efflux of organellar Cl- We first establish the mRNA and protein expression of CFTR in our model system, cultured chick retinal amacrine cells. Using whole cell voltage-clamp recordings of currents through GABA-gated Cl- channels, we examine the effects of pharmacological inhibition of CFTR on the NO-dependent release of internal Cl- To interfere with the expression of CFTR, we used clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 genome editing. We find that both pharmacological inhibition and CRISPR/Cas9-mediated knockdown of CFTR block the ability of NO to release Cl- from internal stores. These results demonstrate that CFTR is required for the NO-dependent efflux of Cl- from acidic organelles.NEW & NOTEWORTHY Although CFTR function has been studied extensively in the context of epithelia, relatively little is known about its function in neurons. We show that CFTR is involved in an NO-dependent release of Cl- from acidic organelles. This internal function of CFTR is particularly relevant to neuronal physiology because postsynaptic cytosolic Cl- levels determine the outcome of GABA- and glycinergic synaptic signaling. Thus the CFTR may play a role in regulating synaptic transmission.
Collapse
Affiliation(s)
- Vijai Krishnan
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - J Wesley Maddox
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Tyler Rodriguez
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
39
|
Hahn A, Faulhaber J, Srisawang L, Stortz A, Salomon JJ, Mall MA, Frings S, Möhrlen F. Cellular distribution and function of ion channels involved in transport processes in rat tracheal epithelium. Physiol Rep 2017; 5:e13290. [PMID: 28642338 PMCID: PMC5492199 DOI: 10.14814/phy2.13290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 01/17/2023] Open
Abstract
Transport of water and electrolytes in airway epithelia involves chloride-selective ion channels, which are controlled either by cytosolic Ca2+ or by cAMP The contributions of the two pathways to chloride transport differ among vertebrate species. Because rats are becoming more important as animal model for cystic fibrosis, we have examined how Ca2+- dependent and cAMP- dependent Cl- secretion is organized in the rat tracheal epithelium. We examined the expression of the Ca2+-gated Cl- channel anoctamin 1 (ANO1), the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel, the epithelial Na+ channel ENaC, and the water channel aquaporin 5 (AQP5) in rat tracheal epithelium. The contribution of ANO1 channels to nucleotide-stimulated Cl- secretion was determined using the channel blocker Ani9 in short-circuit current recordings obtained from primary cultures of rat tracheal epithelial cells in Ussing chambers. We found that ANO1, CFTR and AQP5 proteins were expressed in nonciliated cells of the tracheal epithelium, whereas ENaC was expressed in ciliated cells. Among nonciliated cells, ANO1 occurred together with CFTR and Muc5b and, in addition, in a different cell type without CFTR and Muc5b. Bioelectrical studies with the ANO1-blocker Ani9 indicated that ANO1 mediated the secretory response to the nucleotide uridine-5'-triphosphate. Our data demonstrate that, in rat tracheal epithelium, Cl- secretion and Na+ absorption are routed through different cell types, and that ANO1 channels form the molecular basis of Ca2+-dependent Cl- secretion in this tissue. These characteristic features of Cl--dependent secretion reveal similarities and distinct differences to secretory processes in human airways.
Collapse
Affiliation(s)
- Anne Hahn
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Johannes Faulhaber
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Lalita Srisawang
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Andreas Stortz
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Johanna J Salomon
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC) German Center for Lung Research (DZL) University of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC) German Center for Lung Research (DZL) University of Heidelberg, Heidelberg, Germany
| | - Stephan Frings
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Frank Möhrlen
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
40
|
Abstract
Cystic fibrosis is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that encodes a chloride channel located in the apical membrane of epithelia cells. The cAMP signaling pathway and protein phosphorylation are known to be primary controlling mechanisms for channel function. In this study, we present an alternative activation pathway that involves calcium-activated calmodulin binding of the intrinsically disordered regulatory (R) region of CFTR. Beyond their potential therapeutic value, these data provide insights into the intersection of calcium signaling with control of ion homeostasis and the ways in which the local CFTR microdomain organizes itself. Cystic fibrosis results from mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel, leading to defective apical chloride transport. Patients also experience overactivation of inflammatory processes, including increased calcium signaling. Many investigations have described indirect effects of calcium signaling on CFTR or other calcium-activated chloride channels; here, we investigate the direct response of CFTR to calmodulin-mediated calcium signaling. We characterize an interaction between the regulatory region of CFTR and calmodulin, the major calcium signaling molecule, and report protein kinase A (PKA)-independent CFTR activation by calmodulin. We describe the competition between calmodulin binding and PKA phosphorylation and the differential effects of this competition for wild-type CFTR and the major F508del mutant, hinting at potential therapeutic strategies. Evidence of CFTR binding to isolated calmodulin domains/lobes suggests a mechanism for the role of CFTR as a molecular hub. Together, these data provide insights into how loss of active CFTR at the membrane can have additional consequences besides impaired chloride transport.
Collapse
|
41
|
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: 77] [Impact Index Per Article: 9.6] [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
|
42
|
Chin S, Hung M, Bear CE. Current insights into the role of PKA phosphorylation in CFTR channel activity and the pharmacological rescue of cystic fibrosis disease-causing mutants. Cell Mol Life Sci 2017; 74:57-66. [PMID: 27722768 PMCID: PMC11107731 DOI: 10.1007/s00018-016-2388-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 12/21/2022]
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) channel gating is predominantly regulated by protein kinase A (PKA)-dependent phosphorylation. In addition to regulating CFTR channel activity, PKA phosphorylation is also involved in enhancing CFTR trafficking and mediating conformational changes at the interdomain interfaces of the protein. The major cystic fibrosis (CF)-causing mutation is the deletion of phenylalanine at position 508 (F508del); it causes many defects that affect CFTR trafficking, stability, and gating at the cell surface. Due to the multiple roles of PKA phosphorylation, there is growing interest in targeting PKA-dependent signaling for rescuing the trafficking and functional defects of F508del-CFTR. This review will discuss the effects of PKA phosphorylation on wild-type CFTR, the consequences of CF mutations on PKA phosphorylation, and the development of therapies that target PKA-mediated signaling.
Collapse
Affiliation(s)
- Stephanie Chin
- Programme of Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Maurita Hung
- Programme of Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Christine E Bear
- Programme of Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Canada.
- Department of Biochemistry, University of Toronto, Toronto, Canada.
- Department of Physiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
43
|
Wanitchakool P, Ousingsawat J, Sirianant L, MacAulay N, Schreiber R, Kunzelmann K. Cl - channels in apoptosis. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2016; 45:599-610. [PMID: 27270446 DOI: 10.1007/s00249-016-1140-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 05/06/2016] [Accepted: 05/14/2016] [Indexed: 12/28/2022]
Abstract
A remarkable feature of apoptosis is the initial massive cell shrinkage, which requires opening of ion channels to allow release of K+, Cl-, and organic osmolytes to drive osmotic water movement and cell shrinkage. This article focuses on the role of the Cl- channels LRRC8, TMEM16/anoctamin, and cystic fibrosis transmembrane conductance regulator (CFTR) in cellular apoptosis. LRRC8A-E has been identified as a volume-regulated anion channel expressed in many cell types. It was shown to be required for regulatory and apoptotic volume decrease (RVD, AVD) in cultured cell lines. Its presence also determines sensitivity towards cytostatic drugs such as cisplatin. Recent data point to a molecular and functional relationship of LRRC8A and anoctamins (ANOs). ANO6, 9, and 10 (TMEM16F, J, and K) augment apoptotic Cl- currents and AVD, but it remains unclear whether these anoctamins operate as Cl- channels or as regulators of other apoptotic Cl- channels, such as LRRC8. CFTR has been known for its proapoptotic effects for some time, and this effect may be based on glutathione release from the cell and increase in cytosolic reactive oxygen species (ROS). Although we find that CFTR is activated by cell swelling, it is possible that CFTR serves RVD/AVD through accumulation of ROS and activation of independent membrane channels such as ANO6. Thus activation of ANO6 will support cell shrinkage and induce additional apoptotic events, such as membrane phospholipid scrambling.
Collapse
Affiliation(s)
- Podchanart Wanitchakool
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Lalida Sirianant
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Nanna MacAulay
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, 93053, Regensburg, Germany.
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Renal collecting ducts maintain NaCl homeostasis by fine-tuning urinary excretion to balance dietary salt intake. This review focuses on recent studies on transcellular Cl secretion by collecting ducts, its regulation and its role in cyst growth in autosomal dominant polycystic kidney disease (ADPKD). RECENT FINDINGS Lumens of nonperfused rat medullary collecting ducts collapse in control media but expand with fluid following treatment with cAMP, demonstrating the capacity for both salt absorption and secretion. Recently, inhibition of apical epithelial Na channels (ENaC) unmasked Cl secretion in perfused mouse cortical collecting ducts (CCDs), involving Cl uptake by basolateral NKCC1 and efflux through apical Cl channels. AVP, the key hormone for osmoregulation, promotes cystic fibrosis transmembrane conductance regulator (CFTR)-mediated Cl secretion. In addition, prostaglandin E2 stimulates Cl secretion through both CFTR and Ca-activated Cl channels. SUMMARY Renal Cl secretion has been commonly overlooked because of the overwhelming capacity for the nephron to reabsorb NaCl from the glomerular filtrate. In ADPKD, Cl secretion plays a central role in the accumulation of cyst fluid and the remarkable size of the cystic kidneys. Investigation of renal Cl secretion may provide a better understanding of NaCl homeostasis and identify new approaches to reduce cyst growth in PKD.
Collapse
|
45
|
Melis N, Tauc M, Cougnon M, Bendahhou S, Giuliano S, Rubera I, Duranton C. Revisiting CFTR inhibition: a comparative study of CFTRinh -172 and GlyH-101 inhibitors. Br J Pharmacol 2016; 171:3716-27. [PMID: 24758416 PMCID: PMC4128068 DOI: 10.1111/bph.12726] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 04/04/2014] [Accepted: 04/10/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE For decades, inhibitors of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel have been used as tools to investigate the role and function of CFTR conductance in cystic fibrosis research. In the early 2000s, two new and potent inhibitors of CFTR, CFTRinh-172 and GlyH-101, were described and are now widely used to inhibit specifically CFTR. However, despite some evidence, the effects of both drugs on other types of Cl−-conductance have been overlooked. In this context, we explore the specificity and the cellular toxicity of both inhibitors in CFTR-expressing and non–CFTR-expressing cells. EXPERIMENTAL APPROACH Using patch-clamp technique, we tested the effects of CFTRinh-172 and GlyH-101 inhibitors on three distinct types of Cl− currents: the CFTR-like conductance, the volume-sensitive outwardly rectifying Cl− conductance (VSORC) and finally the Ca2+-dependent Cl− conductance (CaCC). We also explored the effect of both inhibitors on cell viability using live/dead and cell proliferation assays in two different cell lines. KEY RESULTS We confirmed that these two compounds were potent inhibitors of the CFTR-mediated Cl− conductance. However,GlyH-101 also inhibited the VSORC conductance and the CaCC at concentrations used to inhibit CFTR. The CFTRinh-172 did not affect the CaCC but did inhibit the VSORC, at concentrations higher than 5 µM. Neither inhibitor (20 µM; 24 h exposure) affected cell viability, but both were cytotoxic at higher concentrations. CONCLUSIONS AND IMPLICATIONS Both inhibitors affected Cl− conductances apart from CFTR. Our results provided insights into their use in mouse models.
Collapse
Affiliation(s)
- N Melis
- University of Nice-Sophia Antipolis, LP2M CNRS-UMR7370, Faculté de médecine, Nice, France
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Submucosal glands contribute to airway surface liquid (ASL), a film that protects all airway surfaces. Glandular mucus comprises electrolytes, water, the gel-forming mucin MUC5B, and hundreds of different proteins with diverse protective functions. Gland volume per unit area of mucosal surface correlates positively with impaction rate of inhaled particles. In human main bronchi, the volume of the glands is ∼ 50 times that of surface goblet cells, but the glands diminish in size and frequency distally. ASL and its trapped particles are removed from the airways by mucociliary transport. Airway glands have a tubuloacinar structure, with a single terminal duct, a nonciliated collecting duct, then branching secretory tubules lined with mucous cells and ending in serous acini. They allow for a massive increase in numbers of mucus-producing cells without replacing surface ciliated cells. Active secretion of Cl(-) and HCO3 (-) by serous cells produces most of the fluid of gland secretions. Glands are densely innervated by tonically active, mutually excitatory airway intrinsic neurons. Most gland mucus is secreted constitutively in vivo, with large, transient increases produced by emergency reflex drive from the vagus. Elevations of [cAMP]i and [Ca(2+)]i coordinate electrolyte and macromolecular secretion and probably occur together for baseline activity in vivo, with cholinergic elevation of [Ca(2+)]i being mainly responsive for transient increases in secretion. Altered submucosal gland function contributes to the pathology of all obstructive diseases, but is an early stage of pathogenesis only in cystic fibrosis.
Collapse
Affiliation(s)
- Jonathan H Widdicombe
- Department of Physiology and Membrane Biology, University of California-Davis, Davis, California; and Department of Psychology and Cystic Fibrosis Research Laboratory, Stanford University, Stanford, California
| | - Jeffrey J Wine
- Department of Physiology and Membrane Biology, University of California-Davis, Davis, California; and Department of Psychology and Cystic Fibrosis Research Laboratory, Stanford University, Stanford, California
| |
Collapse
|
47
|
Nichols JM, Maiellaro I, Abi-Jaoude J, Curci S, Hofer AM. "Store-operated" cAMP signaling contributes to Ca2+-activated Cl- secretion in T84 colonic cells. Am J Physiol Gastrointest Liver Physiol 2015; 309:G670-9. [PMID: 26316590 PMCID: PMC4609931 DOI: 10.1152/ajpgi.00214.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/21/2015] [Indexed: 01/31/2023]
Abstract
Apical cAMP-dependent CFTR Cl(-) channels are essential for efficient vectorial movement of ions and fluid into the lumen of the colon. It is well known that Ca(2+)-mobilizing agonists also stimulate colonic anion secretion. However, CFTR is apparently not activated directly by Ca(2+), and the existence of apical Ca(2+)-dependent Cl(-) channels in the native colonic epithelium is controversial, leaving the identity of the Ca(2+)-activated component unresolved. We recently showed that decreasing free Ca(2+) concentration ([Ca(2+)]) within the endoplasmic reticulum (ER) lumen elicits a rise in intracellular cAMP. This process, which we termed "store-operated cAMP signaling" (SOcAMPS), requires the luminal ER Ca(2+) sensor STIM1 and does not depend on changes in cytosolic Ca(2+). Here we assessed the degree to which SOcAMPS participates in Ca(2+)-activated Cl(-) transport as measured by transepithelial short-circuit current (Isc) in polarized T84 monolayers in parallel with imaging of cAMP and PKA activity using fluorescence resonance energy transfer (FRET)-based reporters in single cells. In Ca(2+)-free conditions, the Ca(2+)-releasing agonist carbachol and Ca(2+) ionophore increased Isc, cAMP, and PKA activity. These responses persisted in cells loaded with the Ca(2+) chelator BAPTA-AM. The effect on Isc was enhanced in the presence of the phosphodiesterase (PDE) inhibitor 3-isobutyl-1-methylxanthine (IBMX), inhibited by the CFTR inhibitor CFTRinh-172 and the PKA inhibitor H-89, and unaffected by Ba(2+) or flufenamic acid. We propose that a discrete component of the "Ca(2+)-dependent" secretory activity in the colon derives from cAMP generated through SOcAMPS. This alternative mode of cAMP production could contribute to the actions of diverse xenobiotic agents that disrupt ER Ca(2+) homeostasis, leading to diarrhea.
Collapse
Affiliation(s)
- Jonathan M. Nichols
- Department of Veterans Affairs Boston Healthcare System and Department of Surgery, Brigham & Women's Hospital and Harvard Medical School, West Roxbury, Massachusetts
| | - Isabella Maiellaro
- Department of Veterans Affairs Boston Healthcare System and Department of Surgery, Brigham & Women's Hospital and Harvard Medical School, West Roxbury, Massachusetts
| | - Joanne Abi-Jaoude
- Department of Veterans Affairs Boston Healthcare System and Department of Surgery, Brigham & Women's Hospital and Harvard Medical School, West Roxbury, Massachusetts
| | - Silvana Curci
- Department of Veterans Affairs Boston Healthcare System and Department of Surgery, Brigham & Women's Hospital and Harvard Medical School, West Roxbury, Massachusetts
| | - Aldebaran M. Hofer
- Department of Veterans Affairs Boston Healthcare System and Department of Surgery, Brigham & Women's Hospital and Harvard Medical School, West Roxbury, Massachusetts
| |
Collapse
|
48
|
Matalon S, Bartoszewski R, Collawn JF. Role of epithelial sodium channels in the regulation of lung fluid homeostasis. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1229-38. [PMID: 26432872 DOI: 10.1152/ajplung.00319.2015] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/25/2015] [Indexed: 01/11/2023] Open
Abstract
In utero, fetal lung epithelial cells actively secrete Cl(-) ions into the lung air spaces while Na(+) ions follow passively to maintain electroneutrality. This process, driven by an electrochemical gradient generated by the Na(+)-K(+)-ATPase, is responsible for the secretion of fetal fluid that is essential for normal lung development. Shortly before birth, a significant upregulation of amiloride-sensitive epithelial channels (ENaCs) on the apical side of the lung epithelial cells results in upregulation of active Na(+) transport. This process is critical for the reabsorption of fetal lung fluid and the establishment of optimum gas exchange. In the adult lung, active Na(+) reabsorption across distal lung epithelial cells limits the degree of alveolar edema in patients with acute lung injury and cardiogenic edema. Cl(-) ions are transported either paracellularly or transcellularly to preserve electroneutrality. An increase in Cl(-) secretion across the distal lung epithelium has been reported following an acute increase in left atrial pressure and may result in pulmonary edema. In contrast, airway epithelial cells secrete Cl(-) through apical cystic fibrosis transmembrane conductance regulator and Ca(2+)-activated Cl(-) channels and absorb Na(+). Thus the coordinated action of Cl(-) secretion and Na(+) absorption is essential for maintenance of the volume of epithelial lining fluid that, in turn, maximizes mucociliary clearance and facilitates clearance of bacteria and debris from the lungs. Any factor that interferes with Na(+) or Cl(-) transport or dramatically upregulates ENaC activity in airway epithelial cells has been associated with lung diseases such as cystic fibrosis or chronic obstructive lung disease. In this review we focus on the role of the ENaC, the mechanisms involved in ENaC regulation, and how ENaC dysregulation can lead to lung pathology.
Collapse
Affiliation(s)
- Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
49
|
Affiliation(s)
- Karl Kunzelmann
- a Institut für Physiologie ; Universität Regensburg ; Regensburg , Germany
| | | |
Collapse
|
50
|
Billet A, Jia Y, Jensen T, Riordan JR, Hanrahan JW. Regulation of the cystic fibrosis transmembrane conductance regulator anion channel by tyrosine phosphorylation. FASEB J 2015; 29:3945-53. [PMID: 26062600 DOI: 10.1096/fj.15-273151] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/26/2015] [Indexed: 11/11/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) channel is activated by PKA phosphorylation of a regulatory domain that interacts dynamically with multiple CFTR domains and with other proteins. The large number of consensus sequences for phosphorylation by PKA has naturally focused most attention on regulation by this kinase. We report here that human CFTR is also phosphorylated by the tyrosine kinases p60c-Src (proto-oncogene tyrosine-protein kinase) and the proline-rich tyrosine kinase 2 (Pyk2), and they can also cause robust activation of quiescent CFTR channels. In excised patch-clamp experiments, CFTR activity during exposure to Src or Pyk2 reached ∼80% of that stimulated by PKA. Exposure to PKA after Src or Pyk2 caused a further increase to the level induced by PKA alone, implying a common limiting step. Channels became spontaneously active when v-Src or the catalytic domain of Pyk2 was coexpressed with CFTR and were further stimulated by the tyrosine phosphatase inhibitor dephostatin. Exogenous Src also activated 15SA-CFTR, a variant that lacks 15 potential PKA sites and has little response to PKA. PKA-independent activation by tyrosine phosphorylation has implications for the mechanism of regulation by the R domain and for the physiologic functions of CFTR.
Collapse
Affiliation(s)
- Arnaud Billet
- *Department of Physiology and Cystic Fibrosis Translational Research Centre, McGill University, Montreal, Quebec, Canada; Department of Biochemistry and Biophysics and Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA; and Research Institute of the McGill University Hospital Centre, Montreal, Quebec, Canada
| | - Yanlin Jia
- *Department of Physiology and Cystic Fibrosis Translational Research Centre, McGill University, Montreal, Quebec, Canada; Department of Biochemistry and Biophysics and Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA; and Research Institute of the McGill University Hospital Centre, Montreal, Quebec, Canada
| | - Tim Jensen
- *Department of Physiology and Cystic Fibrosis Translational Research Centre, McGill University, Montreal, Quebec, Canada; Department of Biochemistry and Biophysics and Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA; and Research Institute of the McGill University Hospital Centre, Montreal, Quebec, Canada
| | - John R Riordan
- *Department of Physiology and Cystic Fibrosis Translational Research Centre, McGill University, Montreal, Quebec, Canada; Department of Biochemistry and Biophysics and Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA; and Research Institute of the McGill University Hospital Centre, Montreal, Quebec, Canada
| | - John W Hanrahan
- *Department of Physiology and Cystic Fibrosis Translational Research Centre, McGill University, Montreal, Quebec, Canada; Department of Biochemistry and Biophysics and Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, USA; and Research Institute of the McGill University Hospital Centre, Montreal, Quebec, Canada
| |
Collapse
|