1
|
Luo S, Rollins S, Schmitz-Abe K, Tam A, Li Q, Shi J, Lin J, Wang R, Agrawal PB. The solute carrier family 26 member 9 modifies rapidly progressing cystic fibrosis associated with homozygous F508del CFTR mutation. Clin Chim Acta 2024; 561:119765. [PMID: 38852790 DOI: 10.1016/j.cca.2024.119765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND AND AIMS Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations to the CF transmembrane conductance regulator (CFTR). Symptoms and severity of the disease can be quite variable suggesting modifier genes play an important role. MATERIALS AND METHODS Exome sequencing was performed on six individuals carrying homozygous deltaF508 for CFTR genotype but present with rapidly progressing CF (RPCF). Data was analyzed using an unbiased genome-wide genetic burden test against 3076 controls. Single cell RNA sequencing data from LungMAP was utilized to evaluate unique and co-expression of candidate genes, and structural modeling to evaluate the deleterious effects of identified candidate variants. RESULTS We have identified solute carrier family 26 member 9 (SLC26A9) as a modifier gene to be associated with RPCF. Two rare missense SLC26A9 variants were discovered in three of six individuals deemed to have RPCF: c.229G > A; p.G77S (present in two patients), and c.1885C > T; p.P629S. Co-expression of SLC26A9 and CFTR mRNA is limited across different lung cell types, with the highest level of co-expression seen in human (6.3 %) and mouse (9.0 %) alveolar type 2 (AT2) cells. Structural modeling suggests deleterious effects of these mutations as they are in critical protein domains which might affect the anion transport capability of SLC26A9. CONCLUSION The enrichment of rare and potentially deleterious SLC26A9 mutations in patients with RPCF suggests SLC26A9 may act as an alternative anion transporter in CF and is a modifier gene associated with this lung phenotype.
Collapse
Affiliation(s)
- Shiyu Luo
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stuart Rollins
- Division of Pulmonary Medicine, Boston Children's Hospital, USA; Department of Medicine, Harvard Medical School, USA
| | - Klaus Schmitz-Abe
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amy Tam
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Qifei Li
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jiahai Shi
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jasmine Lin
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ruobing Wang
- Division of Pulmonary Medicine, Boston Children's Hospital, USA; Department of Medicine, Harvard Medical School, USA; Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02115, USA.
| | - Pankaj B Agrawal
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Lee D, Hong JH. Chloride/Multiple Anion Exchanger SLC26A Family: Systemic Roles of SLC26A4 in Various Organs. Int J Mol Sci 2024; 25:4190. [PMID: 38673775 PMCID: PMC11050216 DOI: 10.3390/ijms25084190] [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: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Solute carrier family 26 member 4 (SLC26A4) is a member of the SLC26A transporter family and is expressed in various tissues, including the airway epithelium, kidney, thyroid, and tumors. It transports various ions, including bicarbonate, chloride, iodine, and oxalate. As a multiple-ion transporter, SLC26A4 is involved in the maintenance of hearing function, renal function, blood pressure, and hormone and pH regulation. In this review, we have summarized the various functions of SLC26A4 in multiple tissues and organs. Moreover, the relationships between SLC26A4 and other channels, such as cystic fibrosis transmembrane conductance regulator, epithelial sodium channel, and sodium chloride cotransporter, are highlighted. Although the modulation of SLC26A4 is critical for recovery from malfunctions of various organs, development of specific inducers or agonists of SLC26A4 remains challenging. This review contributes to providing a better understanding of the role of SLC26A4 and development of therapeutic approaches for the SLC26A4-associated hearing loss and SLC26A4-related dysfunction of various organs.
Collapse
Affiliation(s)
| | - Jeong Hee Hong
- Department of Health Sciences and Technology, GAIHST (Gachon Advanced Institute for Health Sciences and Technology), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| |
Collapse
|
3
|
Ramananda Y, Naren AP, Arora K. Functional Consequences of CFTR Interactions in Cystic Fibrosis. Int J Mol Sci 2024; 25:3384. [PMID: 38542363 PMCID: PMC10970640 DOI: 10.3390/ijms25063384] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 09/01/2024] Open
Abstract
Cystic fibrosis (CF) is a fatal autosomal recessive disorder caused by the loss of function mutations within a single gene for the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). CFTR is a chloride channel that regulates ion and fluid transport across various epithelia. The discovery of CFTR as the CF gene and its cloning in 1989, coupled with extensive research that went into the understanding of the underlying biological mechanisms of CF, have led to the development of revolutionary therapies in CF that we see today. The highly effective modulator therapies have increased the survival rates of CF patients and shifted the epidemiological landscape and disease prognosis. However, the differential effect of modulators among CF patients and the presence of non-responders and ineligible patients underscore the need to develop specialized and customized therapies for a significant number of patients. Recent advances in the understanding of the CFTR structure, its expression, and defined cellular compositions will aid in developing more precise therapies. As the lifespan of CF patients continues to increase, it is becoming critical to clinically address the extra-pulmonary manifestations of CF disease to improve the quality of life of the patients. In-depth analysis of the molecular signature of different CF organs at the transcriptional and post-transcriptional levels is rapidly advancing and will help address the etiological causes and variability of CF among patients and develop precision medicine in CF. In this review, we will provide an overview of CF disease, leading to the discovery and characterization of CFTR and the development of CFTR modulators. The later sections of the review will delve into the key findings derived from single-molecule and single-cell-level analyses of CFTR, followed by an exploration of disease-relevant protein complexes of CFTR that may ultimately define the etiological course of CF disease.
Collapse
Affiliation(s)
- Yashaswini Ramananda
- Department of Pediatrics, Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anjaparavanda P. Naren
- Department of Pediatrics, Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kavisha Arora
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
4
|
Zajac M, Lepissier A, Dréano E, Chevalier B, Hatton A, Kelly-Aubert M, Guidone D, Planelles G, Edelman A, Girodon E, Hinzpeter A, Crambert G, Pranke I, Galietta LJV, Sermet-Gaudelus I. Putting bicarbonate on the spot: pharmacological insights for CFTR correction in the airway epithelium. Front Pharmacol 2023; 14:1293578. [PMID: 38149052 PMCID: PMC10750368 DOI: 10.3389/fphar.2023.1293578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023] Open
Abstract
Introduction: Cystic fibrosis (CF) is caused by defective Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) proteins. CFTR controls chloride (Cl-) and bicarbonate (HCO3 -) transport into the Airway Surface Liquid (ASL). We investigated the impact of F508del-CFTR correction on HCO3 - secretion by studying transepithelial HCO3 - fluxes. Methods: HCO3 - secretion was measured by pH-stat technique in primary human respiratory epithelial cells from healthy subjects (WT) and people with CF (pwCF) carrying at least one F508del variant. Its changes after CFTR modulation by the triple combination VX445/661/770 and in the context of TNF-α+IL-17 induced inflammation were correlated to ASL pH and transcriptional levels of CFTR and other HCO3 - transporters of airway epithelia such as SLC26A4 (Pendrin), SLC26A9 and NBCe1. Results: CFTR-mediated HCO3 - secretion was not detected in F508del primary human respiratory epithelial cells. It was rescued up to ∼ 80% of the WT level by VX-445/661/770. In contrast, TNF-α+IL-17 normalized transepithelial HCO3 - transport and increased ASL pH. This was related to an increase in SLC26A4 and CFTR transcript levels. VX-445/661/770 induced an increase in pH only in the context of inflammation. Effects on HCO3 - transport were not different between F508del homozygous and F508del compound heterozygous CF airway epithelia. Conclusion: Our studies show that correction of F508del-CFTR HCO3 - is not sufficient to buffer acidic ASL and inflammation is a key regulator of HCO3 - secretion in CF airways. Prediction of the response to CFTR modulators by theratyping should take into account airway inflammation.
Collapse
Affiliation(s)
- Miroslaw Zajac
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Agathe Lepissier
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Elise Dréano
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Benoit Chevalier
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Aurélie Hatton
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mairead Kelly-Aubert
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | - Aleksander Edelman
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Emmanuelle Girodon
- Université de Paris-Cité, Paris, France
- Service de Médecine Génomique des Maladies de Système et d’Organe, Hôpital Cochin, Paris, France
| | - Alexandre Hinzpeter
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Gilles Crambert
- U1138/CNRS ERL 8228, Centre de Recherche des Cordeliers, Paris, France
| | - Iwona Pranke
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Isabelle Sermet-Gaudelus
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- European Reference Network for Rare Diseases, Frankfurt, Belgium
| |
Collapse
|
5
|
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
|
6
|
Jo S, Centeio R, Park J, Ousingsawat J, Jeon DK, Talbi K, Schreiber R, Ryu K, Kahlenberg K, Somoza V, Delpiano L, Gray MA, Amaral MD, Railean V, Beekman JM, Rodenburg LW, Namkung W, Kunzelmann K. The SLC26A9 inhibitor S9-A13 provides no evidence for a role of SLC26A9 in airway chloride secretion but suggests a contribution to regulation of ASL pH and gastric proton secretion. FASEB J 2022; 36:e22534. [PMID: 36183361 DOI: 10.1096/fj.202200313rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/11/2022] [Accepted: 08/24/2022] [Indexed: 11/11/2022]
Abstract
The solute carrier 26 family member A9 (SLC26A9) is an epithelial anion transporter that is assumed to contribute to airway chloride secretion and surface hydration. Whether SLC26A9 or CFTR is responsible for airway Cl- transport under basal conditions is still unclear, due to the lack of a specific inhibitor for SLC26A9. In the present study, we report a novel potent and specific inhibitor for SLC26A9, identified by screening of a drug-like molecule library and subsequent chemical modifications. The most potent compound S9-A13 inhibited SLC26A9 with an IC50 of 90.9 ± 13.4 nM. S9-A13 did not inhibit other members of the SLC26 family and had no effects on Cl- channels such as CFTR, TMEM16A, or VRAC. S9-A13 inhibited SLC26A9 Cl- currents in cells that lack expression of CFTR. It also inhibited proton secretion by HGT-1 human gastric cells. In contrast, S9-A13 had minimal effects on ion transport in human airway epithelia and mouse trachea, despite clear expression of SLC26A9 in the apical membrane of ciliated cells. In both tissues, basal and stimulated Cl- secretion was due to CFTR, while acidification of airway surface liquid by S9-A13 suggests a role of SLC26A9 for airway bicarbonate secretion.
Collapse
Affiliation(s)
- Sungwoo Jo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, South Korea
| | - Raquel Centeio
- Physiological Institute, University of Regensburg, Regensburg, Germany
| | - Jinhong Park
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, South Korea
| | | | - Dong-Kyu Jeon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, South Korea
| | - Khaoula Talbi
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, Regensburg, Germany
| | - Kunhi Ryu
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, South Korea
| | - Kristin Kahlenberg
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Veronika Somoza
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Livia Delpiano
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Michael A Gray
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Margarida D Amaral
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Violeta Railean
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisbon, Portugal
| | - Jeffrey M Beekman
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, Netherlands
| | - Lisa W Rodenburg
- Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, Netherlands
| | - Wan Namkung
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, South Korea
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, Regensburg, Germany
| |
Collapse
|
7
|
Differential CFTR-Interactome Proximity Labeling Procedures Identify Enrichment in Multiple SLC Transporters. Int J Mol Sci 2022; 23:ijms23168937. [PMID: 36012204 PMCID: PMC9408702 DOI: 10.3390/ijms23168937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Proteins interacting with CFTR and its mutants have been intensively studied using different experimental approaches. These studies provided information on the cellular processes leading to proper protein folding, routing to the plasma membrane, recycling, activation and degradation. Recently, new approaches have been developed based on the proximity labeling of protein partners or proteins in close vicinity and their subsequent identification by mass spectrometry. In this study, we evaluated TurboID- and APEX2-based proximity labeling of WT CFTR and compared the obtained data to those reported in databases. The CFTR-WT interactome was then compared to that of two CFTR (G551D and W1282X) mutants and the structurally unrelated potassium channel KCNK3. The two proximity labeling approaches identified both known and additional CFTR protein partners, including multiple SLC transporters. Proximity labeling approaches provided a more comprehensive picture of the CFTR interactome and improved our knowledge of the CFTR environment.
Collapse
|
8
|
Gong J, He G, Wang C, Bartlett C, Panjwani N, Mastromatteo S, Lin F, Keenan K, Avolio J, Halevy A, Shaw M, Esmaeili M, Côté-Maurais G, Adam D, Bégin S, Bjornson C, Chilvers M, Reisman J, Price A, Parkins M, van Wylick R, Berthiaume Y, Bilodeau L, Mateos-Corral D, Hughes D, Smith MJ, Morrison N, Brusky J, Tullis E, Stephenson AL, Quon BS, Wilcox P, Leung WM, Solomon M, Sun L, Brochiero E, Moraes TJ, Gonska T, Ratjen F, Rommens JM, Strug LJ. Genetic evidence supports the development of SLC26A9 targeting therapies for the treatment of lung disease. NPJ Genom Med 2022; 7:28. [PMID: 35396391 PMCID: PMC8993824 DOI: 10.1038/s41525-022-00299-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/04/2022] [Indexed: 12/19/2022] Open
Abstract
Over 400 variants in the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) are CF-causing. CFTR modulators target variants to improve lung function, but marked variability in response exists and current therapies do not address all CF-causing variants highlighting unmet needs. Alternative epithelial ion channel/transporters such as SLC26A9 could compensate for CFTR dysfunction, providing therapeutic targets that may benefit all individuals with CF. We investigate the relationship between rs7512462, a marker of SLC26A9 activity, and lung function pre- and post-treatment with CFTR modulators in Canadian and US CF cohorts, in the general population, and in those with chronic obstructive pulmonary disease (COPD). Rs7512462 CC genotype is associated with greater lung function in CF individuals with minimal function variants (for which there are currently no approved therapies; p = 0.008); and for gating (p = 0.033) and p.Phe508del/ p.Phe508del (p = 0.006) genotypes upon treatment with CFTR modulators. In parallel, human nasal epithelia with CC and p.Phe508del/p.Phe508del after Ussing chamber analysis of a combination of approved and experimental modulator treatments show greater CFTR function (p = 0.0022). Beyond CF, rs7512462 is associated with peak expiratory flow in a meta-analysis of the UK Biobank and Spirometa Consortium (p = 2.74 × 10-44) and provides p = 0.0891 in an analysis of COPD case-control status in the UK Biobank defined by spirometry. These findings support SLC26A9 as a therapeutic target to improve lung function for all people with CF and in individuals with other obstructive lung diseases.
Collapse
Affiliation(s)
- Jiafen Gong
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Gengming He
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Cheng Wang
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Claire Bartlett
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Naim Panjwani
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Scott Mastromatteo
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Fan Lin
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Katherine Keenan
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Julie Avolio
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anat Halevy
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michelle Shaw
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mohsen Esmaeili
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Guillaume Côté-Maurais
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Damien Adam
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Stéphanie Bégin
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | | | - Mark Chilvers
- British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Joe Reisman
- The Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - April Price
- The Children's Hospital, London Health Science Centre, London, ON, Canada
| | | | | | - Yves Berthiaume
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Lara Bilodeau
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec City, QC, Canada
| | | | | | - Mary J Smith
- Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Nancy Morrison
- Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| | - Janna Brusky
- Department of Pediatrics, University of Saskatchewan, Saskatoon, SK, Canada
| | | | | | | | | | | | - Melinda Solomon
- Respiratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Lei Sun
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada
| | - Emmanuelle Brochiero
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Theo J Moraes
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Respiratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Tanja Gonska
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada
| | - Felix Ratjen
- Program in Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Johanna M Rommens
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lisa J Strug
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada.
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, ON, Canada.
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Ousingsawat J, Centeio R, Schreiber R, Kunzelmann K. Expression of SLC26A9 in Airways and Its Potential Role in Asthma. Int J Mol Sci 2022; 23:ijms23062998. [PMID: 35328418 PMCID: PMC8950296 DOI: 10.3390/ijms23062998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
SLC26A9 is an epithelial anion transporter with a poorly defined function in airways. It is assumed to contribute to airway chloride secretion and airway surface hydration. However, immunohistochemistry showing precise localization of SLC26A9 in airways is missing. Some studies report localization near tight junctions, which is difficult to reconcile with a chloride secretory function of SLC26A9. We therefore performed immunocytochemistry of SLC26A9 in sections of human and porcine lungs. Obvious apical localization of SLC26A9 was detected in human and porcine superficial airway epithelia, whereas submucosal glands did not express SLC26A9. The anion transporter was located exclusively in ciliated epithelial cells. Highly differentiated BCi-NS1 human airway epithelial cells grown on permeable supports also expressed SLC26A9 in the apical membrane of ciliated epithelial cells. BCi-NS1 cells expressed the major Cl− transporting proteins CFTR, TMEM16A and SLC26A9 in about equal proportions and produced short-circuit currents activated by increases in intracellular cAMP or Ca2+. Both CFTR and SLC26A9 contribute to basal chloride currents in non-stimulated BCi-NS1 airway epithelia, with CFTR being the dominating Cl− conductance. In wtCFTR-expressing CFBE human airway epithelial cells, SLC26A9 was partially located in the plasma membrane, whereas CFBE cells expressing F508del-CFTR showed exclusive cytosolic localization of SLC26A9. Membrane localization of SLC26A9 and basal chloride currents were augmented by interleukin 13 in wild-type CFTR-expressing cells, but not in cells expressing the most common disease-causing mutant F508del-CFTR. The data suggest an upregulation of SLC26A9-dependent chloride secretion in asthma, but not in the presence of F508del-CFTR.
Collapse
Affiliation(s)
| | | | | | - Karl Kunzelmann
- Correspondence: ; Tel.: +49-(0)941-943-4302; Fax: +49-(0)941-943-4315
| |
Collapse
|
10
|
SLC26A9 as a Potential Modifier and Therapeutic Target in Cystic Fibrosis Lung Disease. Biomolecules 2022; 12:biom12020202. [PMID: 35204703 PMCID: PMC8961553 DOI: 10.3390/biom12020202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 11/16/2022] Open
Abstract
SLC26A9 belongs to the solute carrier family 26 (SLC26), which comprises membrane proteins involved in ion transport mechanisms. On the basis of different preliminary findings, including the phenotype of SlC26A9-deficient mice and its possible role as a gene modifier of the human phenotype and treatment response, SLC26A9 has emerged as one of the most interesting alternative targets for the treatment of cystic fibrosis (CF). However, despite relevant clues, some open issues and controversies remain. The lack of specific pharmacological modulators, the elusive expression reported in the airways, and its complex relationships with CFTR and the CF phenotype prevent us from conclusively understanding the contribution of SLC26A9 in human lung physiology and its real potential as a therapeutic target in CF. In this review, we summarized the various studies dealing with SLC26A9 expression, molecular structure, and function as an anion channel or transporter; its interaction and functional relationships with CFTR; and its role as a gene modifier and tried to reconcile them in order to highlight the current understanding and the gap in knowledge regarding the contribution of SLC26A9 to human lung physiology and CF disease and treatment.
Collapse
|
11
|
Whittamore JM, Hatch M. Oxalate Flux Across the Intestine: Contributions from Membrane Transporters. Compr Physiol 2021; 12:2835-2875. [PMID: 34964122 DOI: 10.1002/cphy.c210013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Epithelial oxalate transport is fundamental to the role occupied by the gastrointestinal (GI) tract in oxalate homeostasis. The absorption of dietary oxalate, together with its secretion into the intestine, and degradation by the gut microbiota, can all influence the excretion of this nonfunctional terminal metabolite in the urine. Knowledge of the transport mechanisms is relevant to understanding the pathophysiology of hyperoxaluria, a risk factor in kidney stone formation, for which the intestine also offers a potential means of treatment. The following discussion presents an expansive review of intestinal oxalate transport. We begin with an overview of the fate of oxalate, focusing on the sources, rates, and locations of absorption and secretion along the GI tract. We then consider the mechanisms and pathways of transport across the epithelial barrier, discussing the transcellular, and paracellular components. There is an emphasis on the membrane-bound anion transporters, in particular, those belonging to the large multifunctional Slc26 gene family, many of which are expressed throughout the GI tract, and we summarize what is currently known about their participation in oxalate transport. In the final section, we examine the physiological stimuli proposed to be involved in regulating some of these pathways, encompassing intestinal adaptations in response to chronic kidney disease, metabolic acid-base disorders, obesity, and following gastric bypass surgery. There is also an update on research into the probiotic, Oxalobacter formigenes, and the basis of its unique interaction with the gut epithelium. © 2021 American Physiological Society. Compr Physiol 11:1-41, 2021.
Collapse
Affiliation(s)
- Jonathan M Whittamore
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Marguerite Hatch
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
SLC26A9 is selected for endoplasmic reticulum associated degradation (ERAD) via Hsp70-dependent targeting of the soluble STAS domain. Biochem J 2021; 478:4203-4220. [PMID: 34821356 PMCID: PMC8826537 DOI: 10.1042/bcj20210644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022]
Abstract
SLC26A9, a member of the solute carrier protein family, transports chloride ions across various epithelia. SLC26A9 also associates with other ion channels and transporters linked to human health, and in some cases these heterotypic interactions are essential to support the biogenesis of both proteins. Therefore, understanding how this complex membrane protein is initially folded might provide new therapeutic strategies to overcome deficits in the function of SLC26A9 partners, one of which is associated with Cystic Fibrosis. To this end, we developed a novel yeast expression system for SLC26A9. This facile system has been used extensively with other ion channels and transporters to screen for factors that oversee protein folding checkpoints. As commonly observed for other channels and transporters, we first noted that a substantial fraction of SLC26A9 is targeted for endoplasmic reticulum associated degradation (ERAD), which destroys folding-compromised proteins in the early secretory pathway. We next discovered that ERAD selection requires the Hsp70 chaperone, which can play a vital role in ERAD substrate selection. We then created SLC26A9 mutants and found that the transmembrane-rich domain of SLC26A9 was quite stable, whereas the soluble cytosolic STAS domain was responsible for Hsp70-dependent ERAD. To support data obtained in the yeast model, we were able to recapitulate Hsp70-facilitated ERAD of the STAS domain in human tissue culture cells. These results indicate that a critical barrier to nascent membrane protein folding can reside within a specific soluble domain, one that is monitored by components associated with the ERAD machinery.
Collapse
|
13
|
Synergy in Cystic Fibrosis Therapies: Targeting SLC26A9. Int J Mol Sci 2021; 22:ijms222313064. [PMID: 34884866 PMCID: PMC8658147 DOI: 10.3390/ijms222313064] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
SLC26A9, a constitutively active Cl- transporter, has gained interest over the past years as a relevant disease modifier in several respiratory disorders including Cystic Fibrosis (CF), asthma, and non-CF bronchiectasis. SLC26A9 contributes to epithelial Cl- secretion, thus preventing mucus obstruction under inflammatory conditions. Additionally, SLC26A9 was identified as a CF gene modifier, and its polymorphisms were shown to correlate with the response to drugs modulating CFTR, the defective protein in CF. Here, we aimed to investigate the relationship between SLC26A9 and CFTR, and its role in CF pathogenesis. Our data show that SLC26A9 expression contributes to enhanced CFTR expression and function. While knocking-down SLC26A9 in human bronchial cells leads to lower wt- and F508del-CFTR expression, function, and response to CFTR correctors, the opposite occurs upon its overexpression, highlighting SLC26A9 relevance for CF. Accordingly, F508del-CFTR rescue by the most efficient correctors available is further enhanced by increasing SLC26A9 expression. Interestingly, SLC26A9 overexpression does not increase the PM expression of non-F508del CFTR traffic mutants, namely those unresponsive to corrector drugs. Altogether, our data indicate that SLC26A9 stabilizes CFTR at the ER level and that the efficacy of CFTR modulator drugs may be further enhanced by increasing its expression.
Collapse
|
14
|
Larsen MB, Choi JJ, Wang X, Myerburg MM, Frizzell RA, Bertrand CA. Separating the contributions of SLC26A9 and CFTR to anion secretion in primary human bronchial epithelia. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1147-L1160. [PMID: 34668421 PMCID: PMC8715023 DOI: 10.1152/ajplung.00563.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 10/01/2021] [Accepted: 10/15/2021] [Indexed: 11/22/2022] Open
Abstract
Aberrant anion secretion across the bronchial epithelium is associated with airway disease, most notably in cystic fibrosis. Although the cystic fibrosis transmembrane conductance regulator (CFTR) is recognized as the primary source of airway anion secretion, alternative anion transport mechanisms play a contributing role. An alternative anion transporter of growing interest is SLC26A9, a constitutively active chloride channel that has been shown to interact with CFTR and may also contribute to bicarbonate secretion. Interest in SLC26A9 has been fueled by genome-wide association studies that suggest it is a significant modifier of CF disease severity. Despite this growing evidence that SLC26A9 plays an important role in the airway, its presence and function in bronchial epithelia remain poorly understood, in part, because its activity is difficult to separate from the activity of CFTR. Here, we present results using primary human bronchial epithelia (HBE) from multiple patient sources to confirm that SLC26A9 mRNA is present in HBE and that its constitutive channel activity is unaffected by knockdown of CFTR. Furthermore, SLC26A9 and CFTR show differential responses to common inhibitors of anion secretion. Finally, we assess the impact of bicarbonate on the activity of SLC26A9 and CFTR. These results confirm that SLC26A9 is the primary source of constitutive anion secretion across HBE, and should inform future studies focused on activation of SLC26A9 as an alternative anion channel in CF. These results should provide a strong foundation to investigate how single-nucleotide polymorphisms in SLC26A9 modulate airway disease.
Collapse
Affiliation(s)
- Mads B Larsen
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jeannie J Choi
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xiaohui Wang
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael M Myerburg
- Department of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Raymond A Frizzell
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Carol A Bertrand
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Laselva O, Guerra L, Castellani S, Favia M, Di Gioia S, Conese M. Small-molecule drugs for cystic fibrosis: Where are we now? Pulm Pharmacol Ther 2021; 72:102098. [PMID: 34793977 DOI: 10.1016/j.pupt.2021.102098] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/24/2021] [Accepted: 11/12/2021] [Indexed: 01/05/2023]
Abstract
The cystic fibrosis (CF) lung disease is due to the lack/dysfunction of the CF Transmembrane Conductance Regulator (CFTR), a chloride channel expressed by epithelial cells as the main regulator of ion and fluid homeostasis. More than 2000 genetic variation in the CFTR gene are known, among which those with identified pathomechanism have been divided into six VI mutation classes. A major advancement in the pharmacotherapy of CF has been the development of small-molecule drugs hitting the root of the disease, i.e. the altered ion and fluid transport through the airway epithelium. These drugs, called CFTR modulators, have been advanced to the clinics to treat nearly 90% of CF patients, including the CFTR potentiator ivacaftor, approved for residual function mutations (Classes III and IV), and combinations of correctors (lumacaftor, tezacaftor, elexacaftor) and ivacaftor for patients bearing at least one the F508del mutation, the most frequent mutation belonging to class II. To cover the 10% of CF patients without etiological therapies, other novel small-molecule CFTR modulators are in evaluation of their effectiveness in all the CFTR mutation classes: read-through agents for Class I, correctors, potentiators and amplifiers from different companies for Class II-V, stabilizers for Class VI. In alternative, other solute carriers, such as SLC26A9 and SLC6A14, are the focus of intensive investigation. Finally, other molecular targets are being evaluated for patients with no approved CFTR modulator therapy or as means of enhancing CFTR modulatory therapy, including small molecules forming ion channels, inhibitors of the ENaC sodium channel and potentiators of the calcium-activated chloride channel TMEM16A. This paper aims to give an up-to-date overview of old and novel CFTR modulators as well as of novel strategies based on small-molecule drugs. Further investigations in in-vivo and cell-based models as well as carrying out large prospective studies will be required to determine if novel CFTR modulators, stabilizers, amplifiers, and the ENaC inhibitors or TMEM16A potentiators will further improve the clinical outcomes in CF management.
Collapse
Affiliation(s)
- Onofrio Laselva
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Stefano Castellani
- Department of Medical Sciences and Human Oncology, University of Bari, Bari, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
16
|
Pereira C, Mazein A, Farinha CM, Gray MA, Kunzelmann K, Ostaszewski M, Balaur I, Amaral MD, Falcao AO. CyFi-MAP: an interactive pathway-based resource for cystic fibrosis. Sci Rep 2021; 11:22223. [PMID: 34782688 PMCID: PMC8592983 DOI: 10.1038/s41598-021-01618-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is a life-threatening autosomal recessive disease caused by more than 2100 mutations in the CF transmembrane conductance regulator (CFTR) gene, generating variability in disease severity among individuals with CF sharing the same CFTR genotype. Systems biology can assist in the collection and visualization of CF data to extract additional biological significance and find novel therapeutic targets. Here, we present the CyFi-MAP-a disease map repository of CFTR molecular mechanisms and pathways involved in CF. Specifically, we represented the wild-type (wt-CFTR) and the F508del associated processes (F508del-CFTR) in separate submaps, with pathways related to protein biosynthesis, endoplasmic reticulum retention, export, activation/inactivation of channel function, and recycling/degradation after endocytosis. CyFi-MAP is an open-access resource with specific, curated and continuously updated information on CFTR-related pathways available online at https://cysticfibrosismap.github.io/ . This tool was developed as a reference CF pathway data repository to be continuously updated and used worldwide in CF research.
Collapse
Affiliation(s)
- Catarina Pereira
- Faculty of Sciences, BioISI-Biosystems Integrative Sciences Institute, University of Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
- LASIGE, Faculty of Sciences, University of Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| | - Alexander Mazein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, 4367, Belvaux, Luxembourg
- CIRI UMR5308, CNRS-ENS-UCBL-INSERM, European Institute for Systems Biology and Medicine, Université de Lyon, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Carlos M Farinha
- Faculty of Sciences, BioISI-Biosystems Integrative Sciences Institute, University of Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| | - Michael A Gray
- Biosciences Institute, University Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | | | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, 4367, Belvaux, Luxembourg
| | - Irina Balaur
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, 4367, Belvaux, Luxembourg
- CIRI UMR5308, CNRS-ENS-UCBL-INSERM, European Institute for Systems Biology and Medicine, Université de Lyon, 50 Avenue Tony Garnier, 69007, Lyon, France
| | - Margarida D Amaral
- Faculty of Sciences, BioISI-Biosystems Integrative Sciences Institute, University of Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| | - Andre O Falcao
- Faculty of Sciences, BioISI-Biosystems Integrative Sciences Institute, University of Lisboa, Campo Grande, 1749-016, Lisbon, Portugal.
- LASIGE, Faculty of Sciences, University of Lisboa, Campo Grande, 1749-016, Lisbon, Portugal.
| |
Collapse
|
17
|
Huang EN, Quach H, Lee JA, Dierolf J, Moraes TJ, Wong AP. A Developmental Role of the Cystic Fibrosis Transmembrane Conductance Regulator in Cystic Fibrosis Lung Disease Pathogenesis. Front Cell Dev Biol 2021; 9:742891. [PMID: 34708042 PMCID: PMC8542926 DOI: 10.3389/fcell.2021.742891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022] Open
Abstract
The cystic fibrosis (CF) transmembrane conductance regulator (CFTR) protein is a cAMP-activated anion channel that is critical for regulating fluid and ion transport across the epithelium. This process is disrupted in CF epithelia, and patients harbouring CF-causing mutations experience reduced lung function as a result, associated with the increased rate of mortality. Much progress has been made in CF research leading to treatments that improve CFTR function, including small molecule modulators. However, clinical outcomes are not necessarily mutation-specific as individuals harboring the same genetic mutation may present with varying disease manifestations and responses to therapy. This suggests that the CFTR protein may have alternative functions that remain under-appreciated and yet can impact disease. In this mini review, we highlight some notable research implicating an important role of CFTR protein during early lung development and how mutant CFTR proteins may impact CF airway disease pathogenesis. We also discuss recent novel cell and animal models that can now be used to identify a developmental cause of CF lung disease.
Collapse
Affiliation(s)
- Elena N Huang
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Henry Quach
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jin-A Lee
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Joshua Dierolf
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Theo J Moraes
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.,Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Amy P Wong
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
The Application of Bicarbonate Recovers the Chemical-Physical Properties of Airway Surface Liquid in Cystic Fibrosis Epithelia Models. BIOLOGY 2021; 10:biology10040278. [PMID: 33805545 PMCID: PMC8065534 DOI: 10.3390/biology10040278] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 01/24/2023]
Abstract
Cystic fibrosis (CF) is a genetic disease associated with the defective function of the cystic fibrosis transmembrane conductance regulator (CFTR) protein that causes obstructive disease and chronic bacterial infections in airway epithelia. Deletion of phenylalanine at position 508, p.F508del, the most frequent mutation among CF patients, causes a folding and traffic defect, resulting in a dramatic reduction in the CFTR expression. To investigate whether the direct application of bicarbonate could modify the properties of the airway surface liquid (ASL), we measured the micro-viscosity, fluid transport and pH of human bronchial epithelial cells monolayers. We have demonstrated that the treatment of a CF-epithelia with an iso-osmotic solution containing bicarbonate is capable of reducing both, the ASL viscosity and the apical fluid re-absorption. We suggest the possibility of design a supportive treatment based on topical application of bicarbonate, or any other alkaline buffer.
Collapse
|
19
|
Zajac M, Dreano E, Edwards A, Planelles G, Sermet-Gaudelus I. Airway Surface Liquid pH Regulation in Airway Epithelium Current Understandings and Gaps in Knowledge. Int J Mol Sci 2021; 22:3384. [PMID: 33806154 PMCID: PMC8037888 DOI: 10.3390/ijms22073384] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Knowledge on the mechanisms of acid and base secretion in airways has progressed recently. The aim of this review is to summarize the known mechanisms of airway surface liquid (ASL) pH regulation and their implication in lung diseases. Normal ASL is slightly acidic relative to the interstitium, and defects in ASL pH regulation are associated with various respiratory diseases, such as cystic fibrosis. Basolateral bicarbonate (HCO3-) entry occurs via the electrogenic, coupled transport of sodium (Na+) and HCO3-, and, together with carbonic anhydrase enzymatic activity, provides HCO3- for apical secretion. The latter mainly involves CFTR, the apical chloride/bicarbonate exchanger pendrin and paracellular transport. Proton (H+) secretion into ASL is crucial to maintain its relative acidity compared to the blood. This is enabled by H+ apical secretion, mainly involving H+/K+ ATPase and vacuolar H+-ATPase that carry H+ against the electrochemical potential gradient. Paracellular HCO3- transport, the direction of which depends on the ASL pH value, acts as an ASL protective buffering mechanism. How the transepithelial transport of H+ and HCO3- is coordinated to tightly regulate ASL pH remains poorly understood, and should be the focus of new studies.
Collapse
Affiliation(s)
- Miroslaw Zajac
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Elise Dreano
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
| | - Aurelie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA;
| | - Gabrielle Planelles
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Laboratoire de Physiologie rénale et Tubulopathies, CNRS ERL 8228, 75006 Paris, France
| | - Isabelle Sermet-Gaudelus
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Centre de Référence Maladies Rares, Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, 75015 Paris, France
- Clinical Trial Network, European Cystic Fibrosis Society, BT2 Belfast, Ireland
- European Respiratory Network Lung, 75006 Paris, France
| |
Collapse
|
20
|
Eastman AC, Pace RG, Dang H, Aksit MA, Vecchio-Pagán B, Lam ATN, O'Neal WK, Blackman SM, Knowles MR, Cutting GR. SLC26A9 SNP rs7512462 is not associated with lung disease severity or lung function response to ivacaftor in cystic fibrosis patients with G551D-CFTR. J Cyst Fibros 2021; 20:851-856. [PMID: 33674211 DOI: 10.1016/j.jcf.2021.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND The CFTR modulator ivacaftor has been variably effective in treating individuals with cystic fibrosis (CF) who harbor CFTR gating variants such as G551D, as well as other classes of CFTR variants when used with other modulators. Because CFTR genotype does not fully explain this variability, defining genetic modifiers of response to modulator therapy is of particular interest to the field of individualized CF drug therapy. Previous studies have proposed that a variant in SLC26A9 (rs7512462) is associated with lung disease severity and with response to treatment with ivacaftor in individuals with CF who carry G551D or gating variants. METHODS Given the implications for CF treatment, we re-examined the reported associations in three cohorts; patients enrolled in the Twin and Siblings study at Johns Hopkins University, the CF modifier study at the University of North Carolina at Chapel Hill, and the prospective G551D Observational (GOAL) study. The GOAL study was specifically designed to measure lung function response to ivacaftor. RESULTS We find no association between SLC26A9 (rs7512462) genotype and lung disease severity (n = 272) or change in lung function at one-, three-, and six-month intervals following ivacaftor treatment(n = 141) in individuals with CF who carry at least one G551D variant. CONCLUSIONS Our inability to replicate this association indicates that rs7512462 genotype should not be used in treatment decisions.
Collapse
Affiliation(s)
- Alice C Eastman
- Department of Genetic Medicine, Johns Hopkins University (JHU), Baltimore, MD, 21205, USA
| | - Rhonda G Pace
- University of North Carolina at Chapel Hill (UNC), Chapel Hill, NC, 27599, USA
| | - Hong Dang
- University of North Carolina at Chapel Hill (UNC), Chapel Hill, NC, 27599, USA
| | - Melis Atalar Aksit
- Department of Genetic Medicine, Johns Hopkins University (JHU), Baltimore, MD, 21205, USA
| | - Briana Vecchio-Pagán
- Department of Genetic Medicine, Johns Hopkins University (JHU), Baltimore, MD, 21205, USA
| | - Anh-Thu N Lam
- Department of Genetic Medicine, Johns Hopkins University (JHU), Baltimore, MD, 21205, USA
| | - Wanda K O'Neal
- University of North Carolina at Chapel Hill (UNC), Chapel Hill, NC, 27599, USA
| | - Scott M Blackman
- Department of Genetic Medicine, Johns Hopkins University (JHU), Baltimore, MD, 21205, USA
| | - Michael R Knowles
- University of North Carolina at Chapel Hill (UNC), Chapel Hill, NC, 27599, USA.
| | - Garry R Cutting
- Department of Genetic Medicine, Johns Hopkins University (JHU), Baltimore, MD, 21205, USA.
| |
Collapse
|
21
|
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: 0.3] [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
|
22
|
Choice of Differentiation Media Significantly Impacts Cell Lineage and Response to CFTR Modulators in Fully Differentiated Primary Cultures of Cystic Fibrosis Human Airway Epithelial Cells. Cells 2020; 9:cells9092137. [PMID: 32967385 PMCID: PMC7565948 DOI: 10.3390/cells9092137] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
In vitro cultures of primary human airway epithelial cells (hAECs) grown at air–liquid interface have become a valuable tool to study airway biology under normal and pathologic conditions, and for drug discovery in lung diseases such as cystic fibrosis (CF). An increasing number of different differentiation media, are now available, making comparison of data between studies difficult. Here, we investigated the impact of two common differentiation media on phenotypic, transcriptomic, and physiological features of CF and non-CF epithelia. Cellular architecture and density were strongly impacted by the choice of medium. RNA-sequencing revealed a shift in airway cell lineage; one medium promoting differentiation into club and goblet cells whilst the other enriched the growth of ionocytes and multiciliated cells. Pathway analysis identified differential expression of genes involved in ion and fluid transport. Physiological assays (intracellular/extracellular pH, Ussing chamber) specifically showed that ATP12A and CFTR function were altered, impacting pH and transepithelial ion transport in CF hAECs. Importantly, the two media differentially affected functional responses to CFTR modulators. We argue that the effect of growth conditions should be appropriately determined depending on the scientific question and that our study can act as a guide for choosing the optimal growth medium for specific applications.
Collapse
|
23
|
Bajko J, Duguid M, Altmann S, Hurlbut GD, Kaczmarek JS. Pendrin stimulates a chloride absorption pathway to increase CFTR-mediated chloride secretion from Cystic Fibrosis airway epithelia. FASEB Bioadv 2020; 2:526-537. [PMID: 32923987 PMCID: PMC7475303 DOI: 10.1096/fba.2020-00012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 03/24/2020] [Accepted: 06/26/2020] [Indexed: 01/04/2023] Open
Abstract
Cystic Fibrosis (CF), an inherited multi-system disease, is caused by mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) that disrupt its ability to secrete anions from epithelia. Recovery of functional anion secretion may be curative for CF, so different components of the ion transport machinery have become attractive therapeutic targets. Several members of the SLC26 ion transporter family have been linked to epithelial ion flux, some through putative functional interactions with CFTR. Using a small-scale qPCR screen, we confirmed that the anion transporter SLC26A4 (pendrin) is downregulated in CF. Upregulation of pendrin using interleukins IL-4 or IL-13 increased Cl- secretion through CFTR in human bronchial epithelial cell (HBEC) derived epithelia differentiated in vitro and measured in the Ussing Chamber. Inhibition or knockdown of pendrin prevented this increased secretion. Increased CFTR activity was not driven by increases in CFTR protein or upstream regulatory pathway components. When basolateral Cl- absorption through NKCC1 was inhibited, a pendrin-dependent Cl- absorption pathway allowing CFTR to continue secreting Cl- from the epithelium was revealed. Although CFTR is often considered the bottleneck in the transepithelial Cl- transport pathway, these studies indicate that basolateral Cl- permeability becomes limiting as CFTR activity increases. Therefore, an increase of epithelial Cl- absorption via pendrin might have additional therapeutic benefit in combination with CFTR modulators.
Collapse
|
24
|
Chi X, Jin X, Chen Y, Lu X, Tu X, Li X, Zhang Y, Lei J, Huang J, Huang Z, Zhou Q, Pan X. Structural insights into the gating mechanism of human SLC26A9 mediated by its C-terminal sequence. Cell Discov 2020; 6:55. [PMID: 32818062 PMCID: PMC7417587 DOI: 10.1038/s41421-020-00193-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/07/2020] [Indexed: 11/25/2022] Open
Abstract
The human SLC26 transporter family exhibits various transport characteristics, and family member SLC26A9 performs multiple roles, including acting as Cl-/HCO3- exchangers, Cl- channels, and Na+ transporters. Some mutations of SLC26A9 are correlated with abnormalities in respiration and digestion systems. As a potential target colocalizing with CFTR in cystic fibrosis patients, SLC26A9 is of great value in drug development. Here, we present a cryo-EM structure of the human SLC26A9 dimer at 2.6 Å resolution. A segment at the C-terminal end is bound to the entry of the intracellular vestibule of the putative transport pathway, which has been proven by electrophysiological experiments to be a gating modulator. Multiple chloride and sodium ions are resolved in the high-resolution structure, identifying novel ion-binding pockets for the first time. Together, our structure takes important steps in elucidating the structural features and regulatory mechanism of SLC26A9, with potential significance in the treatment of cystic fibrosis.
Collapse
Affiliation(s)
- Ximin Chi
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024 China
| | - Xueqin Jin
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Yun Chen
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024 China
| | - Xiaoli Lu
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024 China
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191 China
| | - Xiaorong Li
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024 China
| | - Yuanyuan Zhang
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024 China
| | - Jianlin Lei
- Technology Center for Protein Sciences, Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Jing Huang
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024 China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191 China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, 100191 China
| | - Qiang Zhou
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024 China
| | - Xiaojing Pan
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| |
Collapse
|
25
|
Laselva O, Bartlett C, Popa A, Ouyang H, Gunawardena TNA, Gonska T, Moraes TJ, Bear CE. Emerging preclinical modulators developed for F508del-CFTR have the potential to be effective for ORKAMBI resistant processing mutants. J Cyst Fibros 2020; 20:106-119. [PMID: 32741662 DOI: 10.1016/j.jcf.2020.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/07/2020] [Accepted: 07/22/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND F508del is prototypical of Class 2 CFTR mutations associated with protein misprocessing and reduced function. Corrector compounds like lumacaftor partially rescue the processing defect of F508del-CFTR whereas potentiators like ivacaftor, enhance its channel activity once trafficked to the cell surface. We asked if emerging modulators developed for F508del-CFTR can rescue Class 2 mutations previously shown to be poorly responsive to lumacaftor and ivacaftor. METHODS Rescue of mutant CFTRs by the correctors: AC1, AC2-1 or AC2-2 and the potentiator, AP2, was studied in HEK-293 cells and in primary human nasal epithelial (HNE) cultures, using a membrane potential assay and Ussing chamber, respectively. RESULTS In HEK-293 cells, we found that a particular combination of corrector molecules (AC1 plus AC2-1) and a potentiator (AP2) was effective in rescuing both the misprocessing and reduced function of M1101K and G85E respectively. These findings were recapitulated in patient-derived nasal cultures, although another corrector combination, AC1 plus AC2-2 also improved misprocessing in these primary tissues. Interestingly, while this corrector combination only led to a modest increase in the abundance of mature N1303K-CFTR it did enable its functional expression in the presence of the potentiator, AP2, in part, because the nominal corrector, AC2-2 also exhibits potentiator activity. CONCLUSIONS Strategic combinations of novel modulators can potentially rescue Class 2 mutants thought to be relatively unresponsive to lumacaftor and ivacaftor.
Collapse
Affiliation(s)
- Onofrio Laselva
- Programme in Molecular Medicine, Hospital for Sick Children, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - Claire Bartlett
- Programme in Translational Medicine, Hospital for Sick Children, Toronto, Canada
| | - Alec Popa
- Programme in Molecular Medicine, Hospital for Sick Children, Toronto, Canada
| | - Hong Ouyang
- Programme in Translational Medicine, Hospital for Sick Children, Toronto, Canada
| | | | - Tanja Gonska
- Programme in Translational Medicine, Hospital for Sick Children, Toronto, Canada; Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Theo J Moraes
- Programme in Translational Medicine, Hospital for Sick Children, Toronto, Canada; Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Christine E Bear
- Programme in Molecular Medicine, Hospital for Sick Children, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada; Department of Biochemistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
26
|
Lam ATN, Aksit MA, Vecchio-Pagan B, Shelton CA, Osorio DL, Anzmann AF, Goff LA, Whitcomb DC, Blackman SM, Cutting GR. Increased expression of anion transporter SLC26A9 delays diabetes onset in cystic fibrosis. J Clin Invest 2020; 130:272-286. [PMID: 31581148 DOI: 10.1172/jci129833] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/25/2019] [Indexed: 12/16/2022] Open
Abstract
Diabetes is a common complication of cystic fibrosis (CF) that affects approximately 20% of adolescents and 40%-50% of adults with CF. The age at onset of CF-related diabetes (CFRD) (marked by clinical diagnosis and treatment initiation) is an important measure of the disease process. DNA variants associated with age at onset of CFRD reside in and near SLC26A9. Deep sequencing of the SLC26A9 gene in 762 individuals with CF revealed that 2 common DNA haplotypes formed by the risk variants account for the association with diabetes. Single-cell RNA sequencing (scRNA-Seq) indicated that SLC26A9 is predominantly expressed in pancreatic ductal cells and frequently coexpressed with CF transmembrane conductance regulator (CFTR) along with transcription factors that have binding sites 5' of SLC26A9. These findings were replicated upon reanalysis of scRNA-Seq data from 4 independent studies. DNA fragments derived from the 5' region of SLC26A9-bearing variants from the low-risk haplotype generated 12%-20% higher levels of expression in PANC-1 and CFPAC-1 cells compared with the high- risk haplotype. Taken together, our findings indicate that an increase in SLC26A9 expression in ductal cells of the pancreas delays the age at onset of diabetes, suggesting a CFTR-agnostic treatment for a major complication of CF.
Collapse
Affiliation(s)
- Anh-Thu N Lam
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Melis A Aksit
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Briana Vecchio-Pagan
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Applied Physics Laboratory, Johns Hopkins University, Laurel, Maryland, USA
| | - Celeste A Shelton
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Ariel Precision Medicine, Pittsburgh, Pennsylvania, USA
| | - Derek L Osorio
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Arianna F Anzmann
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Loyal A Goff
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Scott M Blackman
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Garry R Cutting
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
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: 3.0] [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
|
28
|
Sato Y, Thomas DY, Hanrahan JW. The anion transporter SLC26A9 localizes to tight junctions and is degraded by the proteasome when co-expressed with F508del-CFTR. J Biol Chem 2019; 294:18269-18284. [PMID: 31645438 PMCID: PMC6885613 DOI: 10.1074/jbc.ra119.010192] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/21/2019] [Indexed: 01/05/2023] Open
Abstract
Mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) disrupt epithelial secretion and cause cystic fibrosis (CF). Available CFTR modulators provide only modest clinical benefits, so alternative therapeutic targets are being explored. The anion-conducting transporter solute carrier family 26 member 9 (SLC26A9) is a promising candidate, but its functional expression is drastically reduced in cells that express the most common CF-associated CFTR variant, F508del-CFTR, through mechanisms that remain incompletely understood. Here, we examined the metabolic stability and location of SLC26A9 and its relationship to CFTR. Compared with SLC26A9 levels in BHK cells expressing SLC26A9 alone or with WT-CFTR, co-expression of SLC26A9 with F508del-CFTR reduced total and plasma membrane levels of SLC26A9. Proteasome inhibitors increased SLC26A9 immunofluorescence in primary human bronchial epithelial cells (pHBEs) homozygous for F508del-CFTR but not in non-CF pHBEs, suggesting that F508del-CFTR enhances proteasomal SLC26A9 degradation. Apical SLC26A9 expression increased when F508del-CFTR trafficking was partially corrected by low temperature or with the CFTR modulator VX-809. The immature glycoforms of SLC26A9 and CFTR co-immunoprecipitated, consistent with their interaction in the endoplasmic reticulum (ER). Transfection with increasing amounts of WT-CFTR cDNA progressively increased SLC26A9 levels in F508del-CFTR-expressing cells, suggesting that WT-CFTR competes with F508del-CFTR for SLC26A9 binding. Immunofluorescence staining of endogenous SLC26A9 and transfection of a 3HA-tagged construct into well-differentiated cells revealed that SLC26A9 is mostly present at tight junctions. We conclude that SLC26A9 interacts with CFTR in both the ER and Golgi and that its interaction with F508del-CFTR increases proteasomal SLC26A9 degradation.
Collapse
Affiliation(s)
- Yukiko Sato
- Department of Physiology, McGill University, Montréal, Québec H3G 1Y6, Canada; Cystic Fibrosis Translational Research Center, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - David Y Thomas
- Cystic Fibrosis Translational Research Center, McGill University, Montréal, Québec H3G 1Y6, Canada; Department of Biochemistry, McGill University, Montréal, Québec H3A 1A3, Canada
| | - John W Hanrahan
- Department of Physiology, McGill University, Montréal, Québec H3G 1Y6, Canada; Cystic Fibrosis Translational Research Center, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
29
|
Hou X, Wu Q, Rajagopalan C, Zhang C, Bouhamdan M, Wei H, Chen X, Zaman K, Li C, Sun X, Chen S, Frizzell RA, Sun F. CK19 stabilizes CFTR at the cell surface by limiting its endocytic pathway degradation. FASEB J 2019; 33:12602-12615. [PMID: 31450978 PMCID: PMC9292138 DOI: 10.1096/fj.201901050r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/30/2019] [Indexed: 01/07/2023]
Abstract
Protein interactions that stabilize the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) at the apical membranes of epithelial cells have not yet been fully elucidated. We identified keratin 19 (CK19 or K19) as a novel CFTR-interacting protein. CK19 overexpression stabilized both wild-type (WT)-CFTR and Lumacaftor (VX-809)-rescued F508del-CFTR (where F508del is the deletion of the phenylalanine residue at position 508) at the plasma membrane (PM), promoting Cl- secretion across human bronchial epithelial (HBE) cells. CK19 prevention of Rab7A-mediated lysosomal degradation was a key mechanism in apical CFTR stabilization. Unexpectedly, CK19 expression was decreased by ∼40% in primary HBE cells from homogenous F508del patients with CF relative to non-CF controls. CK19 also positively regulated multidrug resistance-associated protein 4 expression at the PM, suggesting that this keratin may regulate the apical expression of other ATP-binding cassette proteins as well as CFTR.-Hou, X., Wu, Q., Rajagopalan, C., Zhang, C., Bouhamdan, M., Wei, H., Chen, X., Zaman, K., Li, C., Sun, X., Chen, S., Frizzell, R. A., Sun, F. CK19 stabilizes CFTR at the cell surface by limiting its endocytic pathway degradation.
Collapse
Affiliation(s)
- Xia Hou
- Department of PhysiologyWayne State University School of MedicineDetroitMichiganUSA
- Department of Biochemistry and Molecular BiologyJiamusi University School of Basic MedicineJiamusiChina
| | - Qingtian Wu
- Department of PhysiologyWayne State University School of MedicineDetroitMichiganUSA
- Department of Biochemistry and Molecular BiologyJiamusi University School of Basic MedicineJiamusiChina
| | - Carthic Rajagopalan
- Department of PhysiologyWayne State University School of MedicineDetroitMichiganUSA
| | - Chunbing Zhang
- Department of Biochemistry and Molecular BiologyJiamusi University School of Basic MedicineJiamusiChina
| | - Mohamad Bouhamdan
- Department of PhysiologyWayne State University School of MedicineDetroitMichiganUSA
| | - Hongguang Wei
- Department of PhysiologyWayne State University School of MedicineDetroitMichiganUSA
| | - Xuequn Chen
- Department of PhysiologyWayne State University School of MedicineDetroitMichiganUSA
| | - Khalequz Zaman
- Department of Pediatric Respiratory MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Chunying Li
- Center for Molecular and Translational Medicine, Georgia State UniversityAtlantaGeorgiaUSA
| | - Xiaonan Sun
- Center for Molecular and Translational Medicine, Georgia State UniversityAtlantaGeorgiaUSA
| | - Song Chen
- Institute of Medical Biotechnology, Jiangsu College of NursingHuai'anChina
| | - Raymond A. Frizzell
- Department of Pediatrics
- Department of Cell BiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Fei Sun
- Department of PhysiologyWayne State University School of MedicineDetroitMichiganUSA
| |
Collapse
|
30
|
Touré A. Importance of SLC26 Transmembrane Anion Exchangers in Sperm Post-testicular Maturation and Fertilization Potential. Front Cell Dev Biol 2019; 7:230. [PMID: 31681763 PMCID: PMC6813192 DOI: 10.3389/fcell.2019.00230] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/26/2019] [Indexed: 12/17/2022] Open
Abstract
In mammals, sperm cells produced within the testis are structurally differentiated but remain immotile and are unable to fertilize the oocyte unless they undergo a series of maturation events during their transit in the male and female genital tracts. This post-testicular functional maturation is known to rely on the micro-environment of both male and female genital tracts, and is tightly controlled by the pH of their luminal milieus. In particular, within the epididymis, the establishment of a low bicarbonate (HCO3–) concentration contributes to luminal acidification, which is necessary for sperm maturation and subsequent storage in a quiescent state. Following ejaculation, sperm is exposed to the basic pH of the female genital tract and bicarbonate (HCO3–), calcium (Ca2+), and chloride (Cl–) influxes induce biochemical and electrophysiological changes to the sperm cells (cytoplasmic alkalinization, increased cAMP concentration, and protein phosphorylation cascades), which are indispensable for the acquisition of fertilization potential, a process called capacitation. Solute carrier 26 (SLC26) members are conserved membranous proteins that mediate the transport of various anions across the plasma membrane of epithelial cells and constitute important regulators of pH and HCO3– concentration. Most SLC26 members were shown to physically interact and cooperate with the cystic fibrosis transmembrane conductance regulator channel (CFTR) in various epithelia, mainly by stimulating its Cl– channel activity. Among SLC26 members, the function of SLC26A3, A6, and A8 were particularly investigated in the male genital tract and the sperm cells. In this review, we will focus on SLC26s contributions to ionic- and pH-dependent processes during sperm post-testicular maturation. We will specify the current knowledge regarding their functions, based on data from the literature generated by means of in vitro and in vivo studies in knock-out mouse models together with genetic studies of infertile patients. We will also discuss the limits of those studies, the current research gaps and identify some key points for potential developments in this field.
Collapse
Affiliation(s)
- Aminata Touré
- INSERM U1016, Centre National de la Recherche Scientifique, UMR 8104, Institut Cochin, Université de Paris, Paris, France
| |
Collapse
|
31
|
A physiologically-motivated model of cystic fibrosis liquid and solute transport dynamics across primary human nasal epithelia. J Pharmacokinet Pharmacodyn 2019; 46:457-472. [PMID: 31494805 DOI: 10.1007/s10928-019-09649-0] [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: 03/22/2019] [Accepted: 08/02/2019] [Indexed: 11/27/2022]
Abstract
Cystic fibrosis (CF) disease is caused by mutations affecting the gene coding for the cystic fibrosis transmembrane conductance regulator (CFTR), an anion channel expressed in the mucosal side of epithelial tissue. In the airway, dysfunctional CFTR results in a transepithelial osmotic imbalance leading to hyperabsorption of airway surface liquid mucostasis, chronic inflammation, and eventual respiratory failure. Human nasal epithelial cell cultures from healthy and CF donors were used to perform studies of liquid and solute transport dynamics at an air/liquid interface in order to emulate the in vivo airway. Then, these results were used to inform a quantitative systems pharmacology model of airway epithelium describing electrically and chemically driven transcellular ionic transport, contributions of both convective and diffusive paracellular solute transport, and osmotically driven transepithelial water dynamics. Model predictions showed CF cultures, relative to non-CF ones, have increased apical and basolateral water permeabilities, and increase paracellular permeability and transepithelial chemical driving force for a radiolabeled tracer used to track small molecule absorption. These results provide a computational platform to better understand and probe the mechanisms behind the liquid hyperabsorption and small molecule retention profiles observed in the CF airway.
Collapse
|
32
|
Walter JD, Sawicka M, Dutzler R. Cryo-EM structures and functional characterization of murine Slc26a9 reveal mechanism of uncoupled chloride transport. eLife 2019; 8:46986. [PMID: 31339488 PMCID: PMC6656431 DOI: 10.7554/elife.46986] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022] Open
Abstract
The epithelial anion transporter SLC26A9 contributes to airway surface hydration and gastric acid production. Colocalizing with CFTR, SLC26A9 has been proposed as a target for the treatment of cystic fibrosis. To provide molecular details of its transport mechanism, we present cryo-EM structures and a functional characterization of murine Slc26a9. These structures define the general architecture of eukaryotic SLC26 family members and reveal an unusual mode of oligomerization which relies predominantly on the cytosolic STAS domain. Our data illustrates conformational transitions of Slc26a9, supporting a rapid alternate-access mechanism which mediates uncoupled chloride transport with negligible bicarbonate or sulfate permeability. The characterization of structure-guided mutants illuminates the properties of the ion transport path, including a selective anion binding site located in the center of a mobile module within the transmembrane domain. This study thus provides a structural foundation for the understanding of the entire SLC26 family and potentially facilitates their therapeutic exploitation. Many processes in the human body are regulated by chloride and other charged particles (known as ions) moving in and out of cells. Each cell is surrounded by a membrane barrier, which prevents ions from entering or exiting. Therefore, to control the levels of ions inside the cell, specific proteins in the membrane act as channels or transporters to provide routes for the ions to pass through the membrane. Channel proteins form pores that, when open, allow a steady stream of ions to pass through the membrane. Transporter proteins, on the other hand, generally contain a pocket that is only accessible from one side of the membrane. When individual ions enter this pocket the transporter changes shape. This causes the entrance of the pocket to close and then re-open on the other side of the membrane. Inside the lung, an ion channel known as CFTR provides a route for chloride ions to move out of cells, which helps clear harmful material from the airways. Mutations affecting this protein cause the mucus lining the airways to become very sticky, leading to a severe disease known as cystic fibrosis. CFTR works together with another protein that is also found in the membrane, called SLC26A9. Previous studies have suggested that SLC26A9 also allows chloride ions to pass through the membrane. It was not clear, however, if SLC26A9 operates as an ion channel or a transporter protein, or how the protein is arranged in the membrane. Now, Walter, Sawicka and Dutzler combined two techniques known as cryo-electron microscopy and patch-clamp electrophysiology to reveal the detailed three-dimensional structure of the mouse version of SLC26A9, which is highly similar to the human form. The experiments found that mouse SLC26A9 proteins form pairs in the membrane referred to as homodimers, which arranged themselves in an unexpected way. Further investigation into the structure of these homodimers suggests that despite having many channel-like properties, SLC26A9 operates as a fast transporter, rather than a true channel. These findings help us understand the role of SLC26A9 and other similar proteins in the lung and other parts of the body. In the future it may be possible to develop drugs that target SLC26A9 to treat cystic fibrosis and other severe lung diseases.
Collapse
Affiliation(s)
- Justin D Walter
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Marta Sawicka
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Raimund Dutzler
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Hanrahan JW, Sato Y, Carlile GW, Jansen G, Young JC, Thomas DY. Cystic Fibrosis: Proteostatic correctors of CFTR trafficking and alternative therapeutic targets. Expert Opin Ther Targets 2019; 23:711-724. [PMID: 31169041 DOI: 10.1080/14728222.2019.1628948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Cystic fibrosis (CF) is the most frequent lethal orphan disease and is caused by mutations in the CFTR gene. The most frequent mutation F508del-CFTR affects multiple organs; infections and subsequent infections and complications in the lung lead to death. Areas covered: This review focuses on new targets and mechanisms that are attracting interest for the development of CF therapies. The F508del-CFTR protein is retained in the endoplasmic reticulum (ER) but has some function if it can traffic to the plasma membrane. Cell-based assays have been used to screen chemical libraries for small molecule correctors that restore its trafficking. Pharmacological chaperones are correctors that bind directly to the F508del-CFTR mutant and promote its folding and trafficking. Other correctors fall into a heterogeneous class of proteostasis modulators that act indirectly by altering cellular homeostasis. Expert opinion: Pharmacological chaperones have so far been the most successful correctors of F508del-CFTR trafficking, but their level of correction means that more than one corrector is required. Proteostasis modulators have low levels of correction but hold promise because some can correct several different CFTR mutations. Identification of their cellular targets and the potential for development may lead to new therapies for CF.
Collapse
Affiliation(s)
- John W Hanrahan
- a Department of Physiology , McGill University , Montréal , QC , Canada.,c Research Institute of the McGill University Health Centre , McGill University , Montréal , QC , Canada
| | - Yukiko Sato
- a Department of Physiology , McGill University , Montréal , QC , Canada.,b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada
| | - Graeme W Carlile
- b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada.,d Department of Biochemistry , McGill University , Montréal , QC , Canada
| | - Gregor Jansen
- d Department of Biochemistry , McGill University , Montréal , QC , Canada
| | - Jason C Young
- b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada.,d Department of Biochemistry , McGill University , Montréal , QC , Canada
| | - David Y Thomas
- b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada.,d Department of Biochemistry , McGill University , Montréal , QC , Canada.,e Department of Human Genetics , McGill University , Montréal , QC , Canada
| |
Collapse
|
34
|
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: 11.0] [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
|
35
|
Dutta RK, Chinnapaiyan S, Rasmussen L, Raju SV, Unwalla HJ. A Neutralizing Aptamer to TGFBR2 and miR-145 Antagonism Rescue Cigarette Smoke- and TGF-β-Mediated CFTR Expression. Mol Ther 2018; 27:442-455. [PMID: 30595527 PMCID: PMC6369566 DOI: 10.1016/j.ymthe.2018.11.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/16/2018] [Accepted: 11/27/2018] [Indexed: 11/26/2022] Open
Abstract
Transforming growth factor β (TGF-β), signaling induced by cigarette smoke (CS), plays an important role in the progression of airway diseases, like chronic bronchitis associated with chronic obstructive pulmonary disease (COPD), and in smokers. Chronic bronchitis is characterized by reduced mucociliary clearance (MCC). Cystic fibrosis transmembrane conductance regulator (CFTR) plays an important role in normal MCC. TGF-β and CS (via TGF-β) promote acquired CFTR dysfunction by suppressing CFTR biogenesis and function. Understanding the mechanism by which CS promotes CFTR dysfunction can identify therapeutic leads to reverse CFTR suppression and rescue MCC. TGF-β alters the microRNAome of primary human bronchial epithelium. TGF-β and CS upregulate miR-145-5p expression to suppress CFTR and the CFTR modifier, SLC26A9. miR-145-5p upregulation with a concomitant CFTR and SLC26A9 suppression was validated in CS-exposed mouse models. While miR-145-5p antagonism rescued the effects of TGF-β in bronchial epithelial cells following transfection, an aptamer to block TGF-β signaling rescues CS- and TGF-β-mediated suppression of CFTR biogenesis and function in the absence of any transfection reagent. These results demonstrate that miR-145-5p plays a significant role in acquired CFTR dysfunction by CS, and they validate a clinically feasible strategy for delivery by inhalation to locally modulate TGF-β signaling in the airway and rescue CFTR biogenesis and function.
Collapse
Affiliation(s)
- Rajib K Dutta
- Department of Immunology and Nanomedicine, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Srinivasan Chinnapaiyan
- Department of Immunology and Nanomedicine, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Lawrence Rasmussen
- Division of Pulmonary, Allergy, and Critical Care Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - S Vamsee Raju
- Division of Pulmonary, Allergy, and Critical Care Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hoshang J Unwalla
- Department of Immunology and Nanomedicine, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
| |
Collapse
|
36
|
Balázs A, Mall MA. Role of the SLC26A9 Chloride Channel as Disease Modifier and Potential Therapeutic Target in Cystic Fibrosis. Front Pharmacol 2018; 9:1112. [PMID: 30327603 PMCID: PMC6174851 DOI: 10.3389/fphar.2018.01112] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022] Open
Abstract
The solute carrier family 26, member 9 (SLC26A9) is an epithelial chloride channel that is expressed in several organs affected in patients with cystic fibrosis (CF) including the lungs, the pancreas, and the intestine. Emerging evidence suggests SLC26A9 as a modulator of wild-type and mutant CFTR function, and as a potential alternative target to circumvent the basic ion transport defect caused by deficient CFTR-mediated chloride transport in CF. In this review, we summarize in vitro studies that revealed multifaceted molecular and functional interactions between SLC26A9 and CFTR that may be implicated in normal transepithelial chloride secretion in health, as well as impaired chloride/fluid transport in CF. Further, we focus on recent genetic association studies and investigations utilizing genetically modified mouse models that identified SLC26A9 as a disease modifier and supported an important role of this alternative chloride channel in the pathophysiology of several organ manifestations in CF, as well as other chronic lung diseases such as asthma and non-CF bronchiectasis. Collectively, these findings and the overlapping endogenous expression with CFTR suggest SLC26A9 an attractive novel therapeutic target that may be exploited to restore epithelial chloride secretion in patients with CF irrespective of their CFTR genotype. In addition, pharmacological activation of SLC26A9 may help to augment the effect of CFTR modulator therapies in patients with CF carrying responsive mutations such as the most common disease-causing mutation F508del-CFTR. However, future research and development including the identification of compounds that activate SLC26A9-mediated chloride transport are needed to explore this alternative chloride channel as a therapeutic target in CF and potentially other muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Anita Balázs
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research, Giessen, Germany
| | - Marcus A Mall
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research, Giessen, Germany
| |
Collapse
|
37
|
Philippe R, Urbach V. Specialized Pro-Resolving Lipid Mediators in Cystic Fibrosis. Int J Mol Sci 2018; 19:ijms19102865. [PMID: 30241412 PMCID: PMC6213393 DOI: 10.3390/ijms19102865] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 12/22/2022] Open
Abstract
In cystic fibrosis (CF), impaired airway surface hydration (ASL) and mucociliary clearance that promote chronic bacterial colonization, persistent inflammation, and progressive structural damage to the airway wall architecture are typically explained by ion transport abnormalities related to the mutation of the gene coding for the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) channel. However, the progressive and unrelenting inflammation of the CF airway begins early in life, becomes persistent, and is excessive relative to the bacterial burden. Intrinsic abnormalities of the inflammatory response in cystic fibrosis have been suggested but the mechanisms involved remain poorly understood. This review aims to give an overview of the recent advances in the understanding of the defective resolution of inflammation in CF including the abnormal production of specialized pro-resolving lipid mediators (lipoxin and resolvin) and their impact on the pathogenesis of the CF airway disease.
Collapse
Affiliation(s)
- Réginald Philippe
- INSERM, U1151, Institut Necker Enfants Malades, 75993 Paris, France.
| | - Valerie Urbach
- INSERM, U1151, Institut Necker Enfants Malades, 75993 Paris, France.
| |
Collapse
|
38
|
Liu X, Li T, Tuo B. Physiological and Pathophysiological Relevance of the Anion Transporter Slc26a9 in Multiple Organs. Front Physiol 2018; 9:1197. [PMID: 30233393 PMCID: PMC6127633 DOI: 10.3389/fphys.2018.01197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/08/2018] [Indexed: 02/05/2023] Open
Abstract
Transepithelial Cl- and HCO3- transport is crucial for the function of all epithelia, and HCO3- is a biological buffer that maintains acid-base homeostasis. In most epithelia, a series of Cl-/HCO3- exchangers and Cl- channels that mediate Cl- absorption and HCO3- secretion have been detected in the luminal and basolateral membranes. Slc26a9 belongs to the solute carrier 26 (Slc26) family of anion transporters expressed in the epithelia of multiple organs. This review summarizes the expression pattern and functional diversity of Slc26a9 in different systems based on all investigations performed thus far. Furthermore, the physical and functional interactions between Slc26a9 and cystic fibrosis transmembrane conductance regulator (CFTR) are discussed due to their overlapping expression pattern in multiple organs. Finally, we focus on the relationship between slc26a9 mutations and disease onset. An understanding of the physiological and pathophysiological relevance of Slc26a9 in multiple organs offers new possibilities for disease therapy.
Collapse
Affiliation(s)
- Xuemei Liu
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi, China.,Digestive Disease Institute of Guizhou Province, Zunyi, China
| | - Taolang Li
- Department of Thyroid and Breast Surgery, Affiliated Hospital, Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi, China.,Digestive Disease Institute of Guizhou Province, Zunyi, China
| |
Collapse
|
39
|
Strug LJ, Stephenson AL, Panjwani N, Harris A. Recent advances in developing therapeutics for cystic fibrosis. Hum Mol Genet 2018; 27:R173-R186. [PMID: 30060192 PMCID: PMC6061831 DOI: 10.1093/hmg/ddy188] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 12/23/2022] Open
Abstract
Despite hope that a cure was imminent when the causative gene was cloned nearly 30 years ago, cystic fibrosis (CF [MIM: 219700]) remains a life-shortening disease affecting more than 70 000 individuals worldwide. However, within the last 6 years the Food and Drug Administration's approval of Ivacaftor, the first drug that corrects the defective cystic fibrosis transmembrane conductance regulator protein [CFTR (MIM: 602421)] in patients with the G551D mutation, marks a watershed in the development of novel therapeutics for this devastating disease. Here we review recent progress in diverse research areas, which all focus on curing CF at the genetic, biochemical or physiological level. In the near future it seems probable that development of mutation-specific therapies will be the focus, since it is unlikely that any one approach will be efficient in correcting the more than 2000 disease-associated variants. We discuss the new drugs and combinations of drugs that either enhance delivery of misfolded CFTR protein to the cell membrane, where it functions as an ion channel, or that activate channel opening. Next we consider approaches to correct the causative genetic lesion at the DNA or RNA level, through repressing stop mutations and nonsense-mediated decay, modulating splice mutations, fixing errors by gene editing or using novel routes to gene replacement. Finally, we explore how modifier genes, loci elsewhere in the genome that modify CF disease severity, may be used to restore a normal phenotype. Progress in all of these areas has been dramatic, generating enthusiasm that CF may soon become a broadly treatable disease.
Collapse
Affiliation(s)
- Lisa J Strug
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anne L Stephenson
- Department of Respirology, Adult Cystic Fibrosis Program, St. Michael’s Hospital, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Naim Panjwani
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
40
|
Corvol H, Mésinèle J, Douksieh IH, Strug LJ, Boëlle PY, Guillot L. SLC26A9 Gene Is Associated With Lung Function Response to Ivacaftor in Patients With Cystic Fibrosis. Front Pharmacol 2018; 9:828. [PMID: 30140228 PMCID: PMC6095007 DOI: 10.3389/fphar.2018.00828] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/09/2018] [Indexed: 11/23/2022] Open
Abstract
Ivacaftor is a drug used to treat cystic fibrosis (CF) patients carrying specific gating CFTR mutations. Interpatient variability in the lung response has been shown to be partly explained by rs7512462 in the Solute Carrier Family 26 Member 9 (SLC26A9) gene. In an independent and larger cohort, we aimed to evaluate whether SLC26A9 variants contribute to the variability of the lung phenotype and if they influence the lung response to ivacaftor. We genotyped the French CF Gene Modifier Study cohort (n = 4,840) to investigate whether SLC26A9 variants were involved in the lung phenotype heterogeneity. Their influence in the response to ivacaftor was tested in the 30 treated patients who met the inclusion criteria: older than 6 years of age, percent-predicted forced expiratory volume measured in 1 s (FEV1pp) in the 3 months before treatment initiation ranging between 40 and 90%. Response to treatment was determined by the change in FEV1pp from baseline, averaged in 15–75 days, and the 1st-year post-treatment. We observed that SLC26A9 variants were not associated with lung function variability in untreated patients and that gain of lung function in patients treated with ivacaftor was similar to clinical trials. We confirmed that rs7512462 was associated with variability in ivacaftor-lung response, with a significant reduction in lung function improvement for patients with the C allele. Other SLC26A9 SNPs also contributed to the ivacaftor-response. Interindividual variability in lung response to ivacaftor is associated with SLC26A9 variants in French CF patients. Pharmacogenomics and personalized medicine will soon be part of CF patient care.
Collapse
Affiliation(s)
- Harriet Corvol
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, UPMC Univ Paris 06, INSERM, Paris, France.,Pneumologie Pédiatrique, APHP, Hôpital Trousseau, Paris, France
| | - Julie Mésinèle
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, UPMC Univ Paris 06, INSERM, Paris, France
| | - Isman-Hassan Douksieh
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, UPMC Univ Paris 06, INSERM, Paris, France.,INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Sorbonne Université, UPMC Univ Paris 06, Paris, France
| | - Lisa J Strug
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Pierre-Yves Boëlle
- INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Sorbonne Université, UPMC Univ Paris 06, Paris, France
| | - Loïc Guillot
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, UPMC Univ Paris 06, INSERM, Paris, France
| |
Collapse
|
41
|
Chin S, Hung M, Won A, Wu YS, Ahmadi S, Yang D, Elmallah S, Toutah K, Hamilton CM, Young RN, Viirre RD, Yip CM, Bear CE. Lipophilicity of the Cystic Fibrosis Drug, Ivacaftor (VX-770), and Its Destabilizing Effect on the Major CF-causing Mutation: F508del. Mol Pharmacol 2018; 94:917-925. [PMID: 29903751 DOI: 10.1124/mol.118.112177] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/13/2018] [Indexed: 12/18/2022] Open
Abstract
Deletion of phenylalanine at position 508 (F508del) in cystic fibrosis transmembrane conductance regulator (CFTR) is the most common cystic fibrosis (CF)-causing mutation. Recently, ORKAMBI, a combination therapy that includes a corrector of the processing defect of F508del-CFTR (lumacaftor or VX-809) and a potentiator of channel activity (ivacaftor or VX-770), was approved for CF patients homozygous for this mutation. However, clinical studies revealed that the effect of ORKAMBI on lung function is modest and it was proposed that this modest effect relates to a negative impact of VX-770 on the stability of F508del-CFTR. In the current studies, we showed that this negative effect of VX-770 at 10 μM correlated with its inhibitory effect on VX-809-mediated correction of the interface between the second membrane spanning domain and the first nucleotide binding domain bearing F508del. Interestingly, we found that VX-770 exerted a similar negative effect on the stability of other membrane localized solute carriers (SLC26A3, SLC26A9, and SLC6A14), suggesting that this negative effect is not specific for F508del-CFTR. We determined that the relative destabilizing effect of a panel of VX-770 derivatives on F508del-CFTR correlated with their predicted lipophilicity. Polarized total internal reflection fluorescence microscopy on a supported lipid bilayer model shows that VX-770, and not its less lipophilic derivative, increased the fluidity of and reorganized the membrane. In summary, our findings show that there is a potential for nonspecific effects of VX-770 on the lipid bilayer and suggest that this effect may account for its destabilizing effect on VX-809- rescued F508del-CFTR.
Collapse
Affiliation(s)
- Stephanie Chin
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Maurita Hung
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Amy Won
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Yu-Sheng Wu
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Saumel Ahmadi
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Donghe Yang
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Salma Elmallah
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Krimo Toutah
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - C Michael Hamilton
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Robert N Young
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Russell D Viirre
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Christopher M Yip
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| | - Christine E Bear
- Department of Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada (S.C., M.H., Y.-S.W., S.A., D.Y., C.E.B.); Departments of Biochemistry (S.C., C.E.B.) and Physiology (M.H., Y.-S.W., S.A., C.E.B.), and Institute of Biomaterials and Biomedical Engineering (A.W., C.M.Y.), University of Toronto, Toronto, Ontario, Canada; Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada (S.E., K.T., R.D.V.); and Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (C.M.H., R.N.Y.)
| |
Collapse
|
42
|
Lim SH, Legere EA, Snider J, Stagljar I. Recent Progress in CFTR Interactome Mapping and Its Importance for Cystic Fibrosis. Front Pharmacol 2018; 8:997. [PMID: 29403380 PMCID: PMC5785726 DOI: 10.3389/fphar.2017.00997] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/26/2017] [Indexed: 12/25/2022] Open
Abstract
Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) is a chloride channel found in secretory epithelia with a plethora of known interacting proteins. Mutations in the CFTR gene cause cystic fibrosis (CF), a disease that leads to progressive respiratory illness and other complications of phenotypic variance resulting from perturbations of this protein interaction network. Studying the collection of CFTR interacting proteins and the differences between the interactomes of mutant and wild type CFTR provides insight into the molecular machinery of the disease and highlights possible therapeutic targets. This mini review focuses on functional genomics and proteomics approaches used for systematic, high-throughput identification of CFTR-interacting proteins to provide comprehensive insight into CFTR regulation and function.
Collapse
Affiliation(s)
- Sang Hyun Lim
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | | | - Jamie Snider
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Igor Stagljar
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
43
|
Li H, Salomon JJ, Sheppard DN, Mall MA, Galietta LJ. Bypassing CFTR dysfunction in cystic fibrosis with alternative pathways for anion transport. Curr Opin Pharmacol 2017; 34:91-97. [PMID: 29065356 DOI: 10.1016/j.coph.2017.10.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/20/2017] [Accepted: 10/04/2017] [Indexed: 12/20/2022]
Abstract
One therapeutic strategy for cystic fibrosis (CF) seeks to restore anion transport to affected epithelia by targeting other apical membrane Cl- channels to bypass dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel. The properties and regulation of the Ca2+-activated Cl- channel TMEM16A argue that long-acting small molecules which target directly TMEM16A are required to overcome CFTR loss. Through genetic studies of lung diseases, SLC26A9, a member of the solute carrier 26 family of anion transporters, has emerged as a promising target to bypass CFTR dysfunction. An alternative strategy to circumvent CFTR dysfunction is to deliver to CF epithelia artificial anion transporters that shuttle Cl- across the apical membrane. Recently, powerful, non-toxic, biologically-active artificial anion transporters have emerged.
Collapse
Affiliation(s)
- Hongyu Li
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Johanna J Salomon
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics, University Hospital Heidelberg, Heidelberg, Germany; Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Marcus A Mall
- Division of Pediatric Pulmonology and Allergy and Cystic Fibrosis Center, Department of Pediatrics, University Hospital Heidelberg, Heidelberg, Germany; Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Luis Jv Galietta
- Telethon Institute for Genetics and Medicine (Tigem), Pozzuoli, Italy.
| |
Collapse
|