1
|
Jaudas F, Bartenschlager F, Shashikadze B, Santamaria G, Reichart D, Schnell A, Stöckl JB, Degroote RL, Cambra JM, Graeber SY, Bähr A, Kartmann H, Stefanska M, Liu H, Naumann-Bartsch N, Bruns H, Berges J, Hanselmann L, Stirm M, Krebs S, Deeg CA, Blum H, Schulz C, Zawada D, Janda M, Caballero-Posadas I, Kunzelmann K, Moretti A, Laugwitz KL, Kupatt C, Saalmüller A, Fröhlich T, Wolf E, Mall MA, Mundhenk L, Gerner W, Klymiuk N. Perinatal dysfunction of innate immunity in cystic fibrosis. Sci Transl Med 2025; 17:eadk9145. [PMID: 39841805 DOI: 10.1126/scitranslmed.adk9145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 07/13/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025]
Abstract
In patients with cystic fibrosis (CF), repeated cycles of infection and inflammation eventually lead to fatal lung damage. Although diminished mucus clearance can be restored by highly effective CFTR modulator therapy, inflammation and infection often persist. To elucidate the role of the innate immune system in CF etiology, we investigated a CF pig model and compared these results with those for preschool children with CF. In newborn CF pigs, we observed changes in lung immune cell composition before the onset of infection that were dominated by increased monocyte infiltration, whereas neutrophil numbers remained constant. Flow cytometric and transcriptomic profiling revealed that the infiltrating myeloid cells displayed a more immature status. Cells with comparably immature transcriptomic profiles were enriched in the blood of CF pigs at birth as well as in preschool children with CF. This pattern coincided with decreased CD16 expression in the myeloid cells of both pigs and humans, which translated into lower phagocytic activity and reduced production of reactive oxygen species in both species. These results were indicative of a congenital, translationally conserved, and functionally relevant aberration of the immune system in CF. In newborn wild-type pigs, CFTR transcription in immune cells, including lung-derived and circulating monocytes, isolated from the bone marrow, thymus, spleen, and blood was below the detection limits of highly sensitive assays, suggesting an indirect etiology of the observed effects. Our findings highlight the need for additional immunological treatments to target innate immune deficits in patients with CF.
Collapse
Affiliation(s)
- Florian Jaudas
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | | | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Gianluca Santamaria
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro 88100, Italy
| | - Daniel Reichart
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Alexander Schnell
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Erlangen 91054, Germany
| | - Jan Bernd Stöckl
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Roxane L Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Planegg 82152, Germany
| | - Josep M Cambra
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Simon Y Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin 13353, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site, Berlin 13353, Germany
| | - Andrea Bähr
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Heike Kartmann
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Monika Stefanska
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | - Huan Liu
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
| | - Nora Naumann-Bartsch
- Department for Pediatric and Adolescent Medicine, University Hospital Erlangen, Erlangen 91054, Germany
| | - Heiko Bruns
- Department of Pediatrics and Adolescent Medicine, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen 91054, Germany
| | - Johannes Berges
- Department of Pediatrics and Adolescent Medicine, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen 91054, Germany
| | - Lea Hanselmann
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin 14163, Germany
| | - Michael Stirm
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Cornelia A Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, Planegg 82152, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, LMU Munich, Munich 81377, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Department of Immunopharmacology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
| | - Melanie Janda
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
| | | | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg 93053, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
- Chair of Regenerative Medicine in Cardiovascular Disease, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
| | - Christian Kupatt
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
| | - Armin Saalmüller
- Institute of Immunology, University of Veterinary Medicine, Vienna 1210, Austria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich 81377, Germany
| | - Eckhard Wolf
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
- Gene Center Munich, LMU Munich, Munich 81377, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin 13353, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site, Berlin 13353, Germany
| | - Lars Mundhenk
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin 14163, Germany
| | - Wilhelm Gerner
- Institute of Immunology, University of Veterinary Medicine, Vienna 1210, Austria
| | - Nikolai Klymiuk
- First Department of Medicine, Cardiology, TUM University Hospital, Technical University of Munich, School of Medicine and Health, Munich 81675, Germany
- Center of Innovative Medical Models (CiMM), LMU Munich, Oberschleissheim 85764, Germany
| |
Collapse
|
2
|
Schnell A, Tamm S, Hedtfeld S, Rodriguez Gonzalez C, Hoerning A, Lachmann N, Stanke F, Dittrich AM, Munder A. Analysis of CFTR mRNA and Protein in Peripheral Blood Mononuclear Cells via Quantitative Real-Time PCR and Western Blot. Int J Mol Sci 2024; 25:6367. [PMID: 38928073 PMCID: PMC11203434 DOI: 10.3390/ijms25126367] [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: 12/21/2023] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
The Cystic Fibrosis Conductance Transmembrane Regulator gene encodes for the CFTR ion channel, which is responsible for the transport of chloride and bicarbonate across the plasma membrane. Mutations in the gene result in impaired ion transport, subsequently leading to perturbed secretion in all exocrine glands and, therefore, the multi-organ disease cystic fibrosis (CF). In recent years, several studies have reported on CFTR expression in immune cells as demonstrated by immunofluorescence, flow cytometry, and immunoblotting. However, these data are mainly restricted to single-cell populations and show significant variation depending on the methodology used. Here, we investigated CFTR transcription and protein expression using standardized protocols in a comprehensive panel of immune cells. Methods: We applied a high-resolution Western blot protocol using a combination of highly specific monoclonal CFTR antibodies that have been optimized for the detection of CFTR in epithelial cells and healthy primary immune cell subpopulations sorted by flow cytometry and used immortalized cell lines as controls. The specificity of CFTR protein detection was controlled by peptide competition and enzymatic Peptide-N-Glycosidase-F (PNGase) digest. CFTR transcripts were analyzed using quantitative real-time PCR and normalized to the level of epithelial T84 cells as a reference. Results: CFTR mRNA expression could be shown for primary CD4+ T cells, NK cells, as well as differentiated THP-1 and Jurkat T cells. In contrast, we failed to detect CFTR transcripts for CD14+ monocytes and undifferentiated THP-1 cells, as well as for B cells and CD8+ T cells. Prominent immunoreactive bands were detectable by immunoblotting with the combination of four CFTR antibodies targeting different epitopes of the CFTR protein. However, in biosamples of non-epithelial origin, these CFTR-like protein bands could be unmasked as false positives through peptide competition or PNGase digest, meaning that the observed mRNA transcripts were not necessarily translated into CFTR proteins, which could be detected via immunoblotting. Our results confirm that mRNA expression in immune cells is many times lower than in that cells of epithelial origin. The immunoreactive signals in immune cells turned out to be false positives, and may be provoked by the presence of a high-affinity protein with a similar epitope. Non-specific binding (e.g., Fab-interaction with glycosyl branches) might also contribute to false positive signals. Our findings highlight the necessity of accurate controls, such as CFTR-negative cells, as well as peptide competition and glycolytic digest in order to identify genuine CFTR protein by immunoblotting. Our data suggest, furthermore, that CFTR protein expression data from techniques such as histology, for which the absence of a molecular weight or other independent control prevents the unmasking of false positive immunoreactive signals, must be interpreted carefully as well.
Collapse
Affiliation(s)
- Alexander Schnell
- Department of Pediatric and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Stephanie Tamm
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Silke Hedtfeld
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Claudio Rodriguez Gonzalez
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Andre Hoerning
- Department of Pediatric and Adolescent Medicine, University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Nico Lachmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany
| | - Frauke Stanke
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| | - Antje Munder
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
3
|
Gillan JL, Chokshi M, Hardisty GR, Clohisey Hendry S, Prasca-Chamorro D, Robinson NJ, Lasota B, Clark R, Murphy L, Whyte MK, Baillie JK, Davidson DJ, Bao G, Gray RD. CAGE sequencing reveals CFTR-dependent dysregulation of type I IFN signaling in activated cystic fibrosis macrophages. SCIENCE ADVANCES 2023; 9:eadg5128. [PMID: 37235648 PMCID: PMC10219589 DOI: 10.1126/sciadv.adg5128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023]
Abstract
An intense, nonresolving airway inflammatory response leads to destructive lung disease in cystic fibrosis (CF). Dysregulation of macrophage immune function may be a key facet governing the progression of CF lung disease, but the underlying mechanisms are not fully understood. We used 5' end centered transcriptome sequencing to profile P. aeruginosa LPS-activated human CF macrophages, showing that CF and non-CF macrophages deploy substantially distinct transcriptional programs at baseline and following activation. This includes a significantly blunted type I IFN signaling response in activated patient cells relative to healthy controls that was reversible upon in vitro treatment with CFTR modulators in patient cells and by CRISPR-Cas9 gene editing to correct the F508del mutation in patient-derived iPSC macrophages. These findings illustrate a previously unidentified immune defect in human CF macrophages that is CFTR dependent and reversible with CFTR modulators, thus providing new avenues in the search for effective anti-inflammatory interventions in CF.
Collapse
Affiliation(s)
- Jonathan L. Gillan
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mithil Chokshi
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Gareth R. Hardisty
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | | | | | - Nicola J. Robinson
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Benjamin Lasota
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Richard Clark
- Edinburgh Clinical Research Facility, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Lee Murphy
- Edinburgh Clinical Research Facility, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Moira K. B. Whyte
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | | | - Donald J. Davidson
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Robert D. Gray
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
4
|
Jaganathan D, Bruscia EM, Kopp BT. Emerging Concepts in Defective Macrophage Phagocytosis in Cystic Fibrosis. Int J Mol Sci 2022; 23:7750. [PMID: 35887098 PMCID: PMC9319215 DOI: 10.3390/ijms23147750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Chronic inflammation and decline in lung function are major reasons for morbidity in CF. Mutant CFTR expressed in phagocytic cells such as macrophages contributes to persistent infection, inflammation, and lung disease in CF. Macrophages play a central role in innate immunity by eliminating pathogenic microbes by a process called phagocytosis. Phagocytosis is required for tissue homeostasis, balancing inflammation, and crosstalk with the adaptive immune system for antigen presentation. This review focused on (1) current understandings of the signaling underlying phagocytic mechanisms; (2) existing evidence for phagocytic dysregulation in CF; and (3) the emerging role of CFTR modulators in influencing CF phagocytic function. Alterations in CF macrophages from receptor initiation to phagosome formation are linked to disease progression in CF. A deeper understanding of macrophages in the context of CFTR and phagocytosis proteins at each step of phagosome formation might contribute to the new therapeutic development of dysregulated innate immunity in CF. Therefore, the review also indicates future areas of research in the context of CFTR and macrophages.
Collapse
Affiliation(s)
- Devi Jaganathan
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Emanuela M. Bruscia
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06510, USA;
| | - Benjamin T. Kopp
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Division of Pulmonary Medicine, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| |
Collapse
|
5
|
Gabillard-Lefort C, Casey M, Glasgow AMA, Boland F, Kerr O, Marron E, Lyons AM, Gunaratnam C, McElvaney NG, Reeves EP. Trikafta Rescues CFTR and Lowers Monocyte P2X7R-Induced Inflammasome Activation in Cystic Fibrosis. Am J Respir Crit Care Med 2022; 205:783-794. [PMID: 35021019 DOI: 10.1164/rccm.202106-1426oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Cystic Fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene and is characterized by sustained inflammation. Adenosine-5'-Triphosphate (ATP) triggers interleukin (IL)-1β secretion via the P2X7 receptor (P2X7R) and activation of the NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome. OBJECTIVES To explore the effect of the CFTR modulator Trikafta (Elexacaftor/Tezacaftor/Ivacaftor) on CFTR expression and the ATP/P2X7R signaling axis in monocytes and on circulating pro-inflammatory markers. METHODS Inflammatory mediators were detected in blood from 42 patients with CF (PWCF) before and after 3 months of Trikafta therapy. Markers of inflammasome activation and IL-1β secretion were measured in monocytes, and following stimulation with ATP and lipopolysaccharides (LPS) in the presence or absence of the P2X7R inhibitor, A438079. MEASUREMENTS AND MAIN RESULTS P2X7R is overexpressed in CF monocytes and receptor inhibition decreased NLRP3 expression, caspase-1 activation, and IL-1β secretion. In vitro and in vivo, P2X7R expression is regulated by CFTR function and intracellular chloride (Cl-) levels. Trikafta therapy restored CFTR expression yet decreased P2X7R in CF monocytes, resulting in normalized Cl- and potassium efflux, and reduced intracellular calcium levels. CFTR modulator therapy decreased circulating levels of ATP and LPS and reduced inflammasome activation and IL-1β secretion. CONCLUSIONS P2X7R expression is regulated by intracellular Cl- levels, and in CF monocytes promotes inflammasome activation. Trikafta therapy significantly increased CFTR protein expression and reduced ATP/P2X7R -induced inflammasome activation. P2X7R may therefore be a promising target to reduce inflammation in PWCF non-eligible for Trikafta or other CFTR modulator therapy.
Collapse
Affiliation(s)
- Claudie Gabillard-Lefort
- Royal College of Surgeons in Ireland, Respiratory Research Division - Dept of Medicine, Dublin, Ireland
| | - Michelle Casey
- Royal College of Surgeons in Ireland, Respiratory Research Division - Dept of Medicine, Dublin, Ireland
| | - Arlene M A Glasgow
- Royal College of Surgeons in Ireland, Respiratory Research Division - Dept of Medicine, Dublin, Ireland
| | - Fiona Boland
- Royal College of Surgeons in Ireland, 8863, Population Health Sciences, Dublin, Ireland
| | - Orla Kerr
- Beaumont Hospital, 57978, Dublin, Ireland
| | | | | | - Cedric Gunaratnam
- Royal College of Surgeons in Ireland, Beaumont Hospital, Respiratory Research Division, Department of Medicine, Dublin, Ireland
| | | | - Emer P Reeves
- Royal College of Surgeons in Ireland, Respiratory Research Division - Dept of Medicine, Dublin, Ireland;
| |
Collapse
|
6
|
Averna M, Melotti P, Sorio C. Revisiting the Role of Leukocytes in Cystic Fibrosis. Cells 2021; 10:cells10123380. [PMID: 34943888 PMCID: PMC8699441 DOI: 10.3390/cells10123380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
Cystic fibrosis in characterized by pulmonary bacterial colonization and hyperinflammation. Lymphocytes, monocytes/macrophages, neutrophils, and dendritic cells of patients with CF express functional CFTR and are directly affected by altered CFTR expression/function, impairing their ability to resolve infections and inflammation. However, the mechanism behind and the contribution of leukocytes in the pathogenesis of CF are still poorly characterized. The recent clinical introduction of specific CFTR modulators added an important tool not only for the clinical management of the disease but also to the investigation of the pathophysiological mechanisms related to CFTR dysfunction and dysregulated immunity. These drugs treat the basic defect in cystic fibrosis (CF) by increasing CFTR function with improvement of lung function and quality of life, and may improve clinical outcomes also by correcting the dysregulated immune function that characterizes CF. Measure of CFTR function, protein expression profiling and several omics methods were used to identify molecular changes in freshly isolated leukocytes of CF patients, highlighting two roles of leukocytes in CF: one more generally related to the mechanism(s) causing immune dysregulation in CF and unresolved inflammation, and another more applicative role, which identifies in myeloid cells, an important tool predictive of the therapeutic response of CF patients. In this review we will summarize available data on CFTR expression and function in leukocyte populations and will discuss potential clinical applications based on available data.
Collapse
Affiliation(s)
- Monica Averna
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Paola Melotti
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy;
| | - Claudio Sorio
- Department of Medicine, General Pathology Division, University of Verona, 37134 Verona, Italy
- Correspondence: ; Tel.: +39-045-802-7688
| |
Collapse
|
7
|
Macrophages from gut-corrected CF mice express human CFTR and lack a pro-inflammatory phenotype. J Cyst Fibros 2021; 21:370-374. [PMID: 34799297 PMCID: PMC9097694 DOI: 10.1016/j.jcf.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 11/24/2022]
Abstract
The cftrtm1unc Tg(FABP-hCFTR) mouse is a commonly-used animal model of CF. This mouse expresses human CFTR in the gut to prevent fatal intestinal obstruction. Macrophages from this mouse fail to replicate immune dysfunction seen in patient cells. We show ectopic expression of human CFTR transgene in macrophages from this CF mouse. This may help to explain anomalies in the field related to use of this model.
Macrophages represent prominent immune orchestrators of cystic fibrosis (CF) inflammation and, as such, are an ever-increasing focus of CF research with several reports of intrinsic immune dysfunction related to loss of CFTR activity in macrophages themselves. Animal models of CF have contributed, in no small part, to a deepening of our understanding of the pathophysiology of the disease and towards therapeutic development. A commonly-used animal model in CF research is the Cftrtm1Unc Tg(FABP-hCFTR) mouse, which displays gut-specific expression of a human CFTR transgene in order to rescue the high rate of early mortality in Cftr-null mice associated with severe intestinal obstruction. We find significant variation in the response to inflammatory challenge of patient macrophages and cells derived from the Cftrtm1Unc Tg(FABP-hCFTR) mouse and show that macrophages derived from this mouse exhibit aberrant expression of human CFTR. This may contribute to the absence of inflammatory changes in this model.
Collapse
|
8
|
Gillan JL, Davidson DJ, Gray RD. Targeting cystic fibrosis inflammation in the age of CFTR modulators: focus on macrophages. Eur Respir J 2020; 57:13993003.03502-2020. [PMID: 33303535 DOI: 10.1183/13993003.03502-2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/18/2020] [Indexed: 11/05/2022]
Abstract
Cystic fibrosis (CF) is a life-shortening, multi-organ, autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The most prominent clinical manifestation in CF is the development of progressive lung disease characterised by an intense, chronic inflammatory airway response that culminates in respiratory failure and, ultimately, death. In recent years, a new class of therapeutics that have the potential to correct the underlying defect in CF, known as CFTR modulators, have revolutionised the field. Despite the exciting success of these drugs, their impact on airway inflammation, and its long-term consequences, remains undetermined. In addition, studies querying the absolute requirement for infection as a driver of CF inflammation have challenged the traditional consensus on CF pathogenesis, and also emphasise the need to prioritise complementary anti-inflammatory treatments in CF. Macrophages, often overlooked in CF research despite their integral role in other chronic inflammatory pathologies, have increasingly become recognised as key players in the initiation, perpetuation and resolution of CF lung inflammation, perhaps as a direct result of CFTR dysfunction. These findings suggest that macrophages may be an important target for novel anti-inflammatory interventional strategies to effectively treat CF lung function decline. This review will consider evidence for the efficacy of anti-inflammatory drugs in the treatment of CF, the potential role of macrophages, and the significance of targeting these pathways at a time when rectifying the basic defect in CF, through use of novel CFTR modulator therapies, is becoming increasingly viable.
Collapse
Affiliation(s)
- Jonathan L Gillan
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, UK
| | - Donald J Davidson
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, UK
| | - Robert D Gray
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
9
|
Tosco A, Villella VR, Castaldo A, Kroemer G, Maiuri L, Raia V. Repurposing therapies for the personalised treatment of cystic fibrosis. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1483231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Antonella Tosco
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Valeria R. Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Alice Castaldo
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Guido Kroemer
- Equipe11 labellisée Ligue Nationale Contrele Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Paris, Sorbonne Paris Cité, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, HôpitalEuropéen Georges Pompidou, AP-HP, Paris, France
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
10
|
Guerra L, D'Oria S, Favia M, Castellani S, Santostasi T, Polizzi AM, Mariggiò MA, Gallo C, Casavola V, Montemurro P, Leonetti G, Manca A, Conese M. CFTR-dependent chloride efflux in cystic fibrosis mononuclear cells is increased by ivacaftor therapy. Pediatr Pulmonol 2017; 52:900-908. [PMID: 28445004 DOI: 10.1002/ppul.23712] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 03/31/2017] [Indexed: 02/05/2023]
Abstract
AIM The Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) potentiator ivacaftor (Kalydeco®) improves clinical outcome in G551D cystic fibrosis (CF) patients. Here, we have investigated whether ivacaftor has a clinical impact on non-G551D gating mutations and function of circulating leukocytes as well. METHODS Seven patients were treated with ivacaftor and evaluated at baseline, and at 1-3 and 6 months. Besides clinical and systemic inflammatory parameters, circulating mononuclear cells (MNC) were evaluated for CFTR-dependent chloride efflux by spectrofluorimetry, neutrophils for oxidative burst by cytofluorimetry and HVCN1 mRNA expression by real time PCR. RESULTS Ivacaftor determined a significant decrease in sweat chloride concentrations at all time points during treatment. Body mass index (BMI), FEV1 , and FVC showed an increasing trend. While C-reactive protein decreased significantly at 2 months, the opposite behavior was noticed for circulating monocytes. CFTR activity in MNC was found to increase significantly at 3 and 6 months. Neutrophil oxidative burst peaked at 2 months and then decreased to baseline. HVCN1 mRNA expression was significantly higher than baseline at 1-3 months and decreased after 6 months of treatment. The chloride efflux in MNC correlated positively with both FEV1 and FVC. On the other hand, sweat chloride correlated positively with CRP and WBC, and negatively with both respiratory function tests. A cluster analysis confirmed that sweat chloride, FEV1 , FVC, BMI, and MNC chloride efflux behaved as a single entity over time. DISCUSSION In patients with non-G551D mutations, ivacaftor improved both chloride transport in sweat ducts and chloride efflux in MNC, that is, functions directly imputed to CFTR.
Collapse
Affiliation(s)
- Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Susanna D'Oria
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Stefano Castellani
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Teresa Santostasi
- Department of Biomedical and Human Oncology, Pediatrics Section, Cystic Fibrosis Regional Center, U.O. "B. Trambusti," Policlinico, University of Bari, Bari, Italy
| | - Angela M Polizzi
- Department of Biomedical and Human Oncology, Pediatrics Section, Cystic Fibrosis Regional Center, U.O. "B. Trambusti," Policlinico, University of Bari, Bari, Italy
| | - Maria A Mariggiò
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Crescenzio Gallo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Valeria Casavola
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Pasqualina Montemurro
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Giuseppina Leonetti
- Department of Biomedical and Human Oncology, Pediatrics Section, Cystic Fibrosis Regional Center, U.O. "B. Trambusti," Policlinico, University of Bari, Bari, Italy
| | | | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
11
|
Sorio C, Montresor A, Bolomini-Vittori M, Caldrer S, Rossi B, Dusi S, Angiari S, Johansson JE, Vezzalini M, Leal T, Calcaterra E, Assael BM, Melotti P, Laudanna C. Mutations of Cystic Fibrosis Transmembrane Conductance Regulator Gene Cause a Monocyte-Selective Adhesion Deficiency. Am J Respir Crit Care Med 2017; 193:1123-33. [PMID: 26694899 DOI: 10.1164/rccm.201510-1922oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE Cystic fibrosis (CF) is a common genetic disease caused by mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Persistent lung inflammation, characterized by increasing polymorphonuclear leukocyte recruitment, is a major cause of the decline in respiratory function in patients with CF and is a leading cause of morbidity and mortality. CFTR is expressed in various cell types, including leukocytes, but its involvement in the regulation of leukocyte recruitment is unknown. OBJECTIVES We evaluated whether CF leukocytes might present with alterations in cell adhesion and migration, a key process governing innate and acquired immune responses. METHODS We used ex vivo adhesion and chemotaxis assays, flow cytometry, immunofluorescence, and GTPase activity assays in this study. MEASUREMENTS AND MAIN RESULTS We found that chemoattractant-induced activation of β1 and β2 integrins and of chemotaxis is defective in mononuclear cells isolated from patients with CF. In contrast, polymorphonuclear leukocyte adhesion and chemotaxis were normal. The functionality of β1 and β2 integrins was restored by treatment of CF monocytes with the CFTR-correcting drugs VRT325 and VX809. Moreover, treatment of healthy monocytes with the CFTR inhibitor CFTR(inh)-172 blocked integrin activation by chemoattractants. In a murine model of lung inflammation, we found that integrin-independent migration of CF monocytes into the lung parenchyma was normal, whereas, in contrast, integrin-dependent transmigration into the alveolar space was impaired. Finally, signal transduction analysis showed that, in CF monocytes, chemoattractant-triggered activation of RhoA and CDC42 Rho small GTPases (controlling integrin activation and chemotaxis, respectively) was strongly deficient. CONCLUSIONS Altogether, these data highlight the critical regulatory role of CFTR in integrin activation by chemoattractants in monocytes and identify CF as a new, cell type-selective leukocyte adhesion deficiency disease, providing new insights into CF pathogenesis.
Collapse
Affiliation(s)
- Claudio Sorio
- 1 Division of General Pathology, Department of Medicine, and.,2 Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | | | - Matteo Bolomini-Vittori
- 1 Division of General Pathology, Department of Medicine, and.,2 Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," Department of Medicine, University of Verona School of Medicine, Verona, Italy.,3 Department of Tumor Immunology (278), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sara Caldrer
- 1 Division of General Pathology, Department of Medicine, and.,2 Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Barbara Rossi
- 1 Division of General Pathology, Department of Medicine, and
| | - Silvia Dusi
- 1 Division of General Pathology, Department of Medicine, and
| | - Stefano Angiari
- 1 Division of General Pathology, Department of Medicine, and
| | - Jan E Johansson
- 1 Division of General Pathology, Department of Medicine, and.,2 Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Marzia Vezzalini
- 1 Division of General Pathology, Department of Medicine, and.,2 Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Teresinha Leal
- 4 Faculté de Pharmacie et des Sciences Biomédicales, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; and
| | - Elisa Calcaterra
- 1 Division of General Pathology, Department of Medicine, and.,2 Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Baroukh M Assael
- 5 Centro Fibrosi Cistica, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Paola Melotti
- 5 Centro Fibrosi Cistica, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Carlo Laudanna
- 1 Division of General Pathology, Department of Medicine, and
| |
Collapse
|
12
|
Vardenafil reduces macrophage pro-inflammatory overresponses in cystic fibrosis through PDE5- and CFTR-dependent mechanisms. Clin Sci (Lond) 2017; 131:1107-1121. [PMID: 28196856 DOI: 10.1042/cs20160749] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 02/07/2023]
Abstract
Chronic inflammation that progressively disrupts the lung tissue is a hallmark of cystic fibrosis (CF). In mice, vardenafil, an inhibitor of phosphodiesterase type 5 (PDE5), restores transepithelial ion transport and corrects mislocalization of the most common CF mutation, F508del-CFTR. It also reduces lung pro-inflammatory responses in mice and in patients with CF. To test the hypothesis that macrophages are target effector cells of the immunomo-dulatory effect of vardenafil, we isolated lung macrophages from mice homozygous for the F508del mutation or invalidated for the cftr gene and from their corresponding wild-type (WT) littermates. We then evaluated the effect of vardenafil on the classical M1 polarization, mirroring release of pro-inflammatory cytokines. We confirmed that macrophages from different body compartments express CF transmembrane conductance regulator (CFTR) and showed that vardenafil targets the cells through PDE5- and CFTR-dependent mechanisms. In the presence of the F508del mutation, vardenafil down-regulated overresponses of the M1 markers, tumour necrosis factor (TNF)-α and inducible nitric oxide synthase (NOS)-2. Our study identifies lung macrophages as target cells of the anti-inflammatory effect of vardenafil in CF and supports the view that the drug is potentially beneficial for treating CF as it combines rescue of CFTR protein and anti-inflammatory properties.
Collapse
|
13
|
Eickmeier O, Fussbroich D, Mueller K, Serve F, Smaczny C, Zielen S, Schubert R. Pro-resolving lipid mediator Resolvin D1 serves as a marker of lung disease in cystic fibrosis. PLoS One 2017; 12:e0171249. [PMID: 28158236 PMCID: PMC5291435 DOI: 10.1371/journal.pone.0171249] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/17/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is an autosomal recessive genetic disorder that affects multiple organs, including the lungs, pancreas, liver and intestine. Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) locus lead to defective proteins and reduced Cl- secretion and Na+ hyperabsorption in the affected organs. In addition, patients suffering from CF display chronic inflammation that contributes to the pathogenesis of CF. Recent work suggests that CF patients have a reduced capacity to biosynthesize specialized pro-resolving lipid mediators (SPMs), which contributes to the development and duration of the unwanted inflammation. Alterations in the metabolism of arachidonic acid (AA) and docosahexaenoic acid (DHA) to specialized pro-resolving mediators (SPMs), like lipoxins (LXs), maresins (MaRs), protectins (PDs) and resolvins (Rvs), may play a major role on clinical impact of airway inflammation in CF. METHODS In this study, our aims were to detect and quantitate Resolvin D1 (RvD1) in sputum and plasma from patients with CF and compare levels of RvD1 with biomarkers of inflammation and lung function. We studied 27 CF patients aged 6 to 55 years (median 16 years) in a prospective approach. RESULTS DHA can be found in the plasma of our CF patients in the milligram range and is decreased in comparison to a healthy control group. The DHA-derived pro-resolving mediator Resolvin D1 (RvD1) was also present in the plasma (286.4 ± 50 pg/ mL, mean ± SEM) and sputum (30.0 ± 2.6 pg/ mL, mean ± SEM) samples from our patients with CF and showed a positive correlation with sputum inflammatory markers. The plasma concentrations of RvD1 were ten times higher than sputum concentrations. Interestingly, sputum RvD1/ IL-8 levels showed a positive correlation with FEV1 (rs = 0.3962, p< 0.05). CONCLUSIONS SPMs, like RvD1, are well known to down-regulate inflammatory pathways. Our study shows that the bioactive lipid mediator RvD1, derived from DHA, was present in sputum and plasma of CF patients and may serve as a representative peripheral biomarker of the lung resolution program for CF patients.
Collapse
Affiliation(s)
- Olaf Eickmeier
- Department for Children and Adolescents, Division of Allergology, Pulmonology, and Cystic Fibrosis, Goethe-University, Frankfurt, Germany
| | - Daniela Fussbroich
- Department for Children and Adolescents, Division of Allergology, Pulmonology, and Cystic Fibrosis, Goethe-University, Frankfurt, Germany
- Department of Food Technology, University of Applied Sciences, Fulda, Germany
| | - Klaus Mueller
- Department for Children and Adolescents, Division of Allergology, Pulmonology, and Cystic Fibrosis, Goethe-University, Frankfurt, Germany
| | - Friederike Serve
- Department for Children and Adolescents, Division of Allergology, Pulmonology, and Cystic Fibrosis, Goethe-University, Frankfurt, Germany
| | - Christina Smaczny
- Department of Internal Medicine III, Goethe-University, Frankfurt, Germany
| | - Stefan Zielen
- Department for Children and Adolescents, Division of Allergology, Pulmonology, and Cystic Fibrosis, Goethe-University, Frankfurt, Germany
| | - Ralf Schubert
- Department for Children and Adolescents, Division of Allergology, Pulmonology, and Cystic Fibrosis, Goethe-University, Frankfurt, Germany
| |
Collapse
|
14
|
Wang H, Cebotaru L, Lee HW, Yang Q, Pollard BS, Pollard HB, Guggino WB. CFTR Controls the Activity of NF-κB by Enhancing the Degradation of TRADD. Cell Physiol Biochem 2016; 40:1063-1078. [PMID: 27960153 DOI: 10.1159/000453162] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND/AIMS Chronic lung infection in cystic fibrosis leads to an inflammatory response that persists because of the chronic presence of bacteria and ultimately leads to a catastrophic failure of lung function. METHODS We use a combination of biochemistry, cell and molecular biology to study the interaction of TRADD, a key adaptor molecule in TNFα signaling, with CFTR in the regulation of NFκB. RESULTS We show that Wt CFTR binds to and colocalizes with TRADD. TRADD is a key signaling intermediate connecting TNFα with activation of NFκB. By contrast, ΔF508 CFTR does not bind to TRADD. NF-κB activation is higher in CFBE expressing ΔF508 CFTR than in cells expressing Wt CFTR. However, this differential effect is abolished when TRADD levels are knocked down. Transfecting Wt CFTR into CFBE cells reduces NF-κB activity. However the reduction is abolished by the CFTR chloride transport inhibitor-172. Consistently, transfecting in the correctly trafficked CFTR conduction mutants G551D or S341A also fail to reduce NFκB activity. Thus CFTR must be functional if it is to regulate NF-κB activity. We also found that TNFα produced a greater increase in NF-κB activity in CFBE cells than in the same cell when Wt CFTR-corrected. Consistently, the effect is also abolished when TRADD is knocked down by shRNA. Thus, Wt CFTR control of TRADD modulates the physiological activation of NF-κB by TNFα. Based on studies with proteosomal and lysosomal inhibitors, the mechanism by which Wt CFTR, but not ΔF508 CFTR, suppresses TRADD is by lysosomal degradation. CONCLUSION We have uncovered a novel mechanism whereby Wt CFTR regulates TNFα signaling by enhancing TRADD degradation. Thus by reducing the levels of TRADD, Wt CFTR suppresses downstream proinflammatory NFκB signaling. By contrast, suppression of NF-κB activation fails in CF cells expressing ΔF508 CFTR.
Collapse
Affiliation(s)
- Hua Wang
- Department of Physiology, Medicine, School of Medicine, The Johns Hopkins University, Baltimore, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Molinski SV, Ahmadi S, Hung M, Bear CE. Facilitating Structure-Function Studies of CFTR Modulator Sites with Efficiencies in Mutagenesis and Functional Screening. ACTA ACUST UNITED AC 2015; 20:1204-17. [PMID: 26385858 DOI: 10.1177/1087057115605834] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/23/2015] [Indexed: 12/20/2022]
Abstract
There are nearly 2000 mutations in the CFTR gene associated with cystic fibrosis disease, and to date, the only approved drug, Kalydeco, has been effective in rescuing the functional expression of a small subset of these mutant proteins with defects in channel activation. However, there is currently an urgent need to assess other mutations for possible rescue by Kalydeco, and further, definition of the binding site of such modulators on CFTR would enhance our understanding of the mechanism of action of such therapeutics. Here, we describe a simple and rapid one-step PCR-based site-directed mutagenesis method to generate mutations in the CFTR gene. This method was used to generate CFTR mutants bearing deletions (p.Gln2_Trp846del, p.Ser700_Asp835del, p.Ile1234_Arg1239del) and truncation with polyhistidine tag insertion (p.Glu1172-3Gly-6-His*), which either recapitulate a disease phenotype or render tools for modulator binding site identification, with subsequent evaluation of drug responses using a high-throughput (384-well) membrane potential-sensitive fluorescence assay of CFTR channel activity within a 1 wk time frame. This proof-of-concept study shows that these methods enable rapid and quantitative comparison of multiple CFTR mutants to emerging drugs, facilitating future large-scale efforts to stratify mutants according to their "theratype" or most promising targeted therapy.
Collapse
Affiliation(s)
- Steven V Molinski
- Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Saumel Ahmadi
- Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Maurita Hung
- Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Christine E Bear
- Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|