1
|
Cinek O, Furstova E, Novotna S, Hubackova K, Dousova T, Borek-Dohalska L, Drevinek P. Gene expression profile of intestinal organoids from people with cystic fibrosis upon exposure to elexacaftor/tezacaftor/ivacaftor. J Cyst Fibros 2025; 24:157-163. [PMID: 39278758 DOI: 10.1016/j.jcf.2024.09.005] [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: 04/01/2024] [Revised: 08/15/2024] [Accepted: 09/04/2024] [Indexed: 09/18/2024]
Abstract
The forskolin-induced swelling assay (FIS) in patient-derived intestinal organoids (PDIOs), used to determine in vitro responsiveness to elexacaftor/tezacaftor/ivacaftor (ETI), showed variability in swelling among PDIOs obtained from people with CF (pwCF) carrying the same F508del/F508del CFTR genotype. We aimed to characterise the effect of ETI on the transcriptional activity of PDIOs-derived cells to understand the intracellular processes triggered by ETI and the differences in treatment response. Six high- and six low-responding PDIOs to ETI, derived from F508del/F508del pwCF, were incubated with or without ETI for 2 to 6 h. Gene expression was assessed using 3'-mRNA sequencing and modelled using negative binomial models. Incubation with ETI resulted in a significant upregulation of several biological processes: mostly related to chemokines and signalling, chemotaxis, and tissue development processes. No changes were observed in abundance of the CFTR transcripts or in CFTR-related gene sets and pathways. The genes and pathways associated with ETI did not overlap with those whose expression changed with time only. PDIOs with a high FIS response did not significantly differ in any interpretable gene from the FIS-low organoids. The changes in the PDIOs gene expression upon the exposure to ETI cannot explain differences in the magnitude of PDIOs FIS-measured response to ETI. In conclusion, on incubation with ETI, genes of the CFTR-related pathways do not change their transcriptional activity; instead, overexpression was observed in genes of inflammatory-like cytokine response and receptor activation pathways.
Collapse
Affiliation(s)
- Ondrej Cinek
- Department of Medical Microbiology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia; Department of Paediatrics, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czechia.
| | - Eva Furstova
- Department of Paediatrics, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czechia
| | - Stepanka Novotna
- Department of Medical Microbiology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Klara Hubackova
- Department of Medical Microbiology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Tereza Dousova
- Department of Paediatrics, Second Faculty of Medicine Charles University and Motol University Hospital, Prague, Czechia
| | - Lucie Borek-Dohalska
- Department of Medical Microbiology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| | - Pavel Drevinek
- Department of Medical Microbiology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czechia
| |
Collapse
|
2
|
Caohuy H, Eidelman O, Chen T, Mungunsukh O, Yang Q, Walton NI, Pollard BS, Khanal S, Hentschel S, Florez C, Herbert AS, Pollard HB. Inflammation in the COVID-19 airway is due to inhibition of CFTR signaling by the SARS-CoV-2 spike protein. Sci Rep 2024; 14:16895. [PMID: 39043712 PMCID: PMC11266487 DOI: 10.1038/s41598-024-66473-4] [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: 12/15/2023] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
SARS-CoV-2-contributes to sickness and death in COVID-19 patients partly by inducing a hyper-proinflammatory immune response in the host airway. This hyper-proinflammatory state involves activation of signaling by NFκB, and unexpectedly, ENaC, the epithelial sodium channel. Post-infection inflammation may also contribute to "Long COVID"/PASC. Enhanced signaling by NFκB and ENaC also marks the airway of patients suffering from cystic fibrosis, a life-limiting proinflammatory genetic disease due to inactivating mutations in the CFTR gene. We therefore hypothesized that inflammation in the COVID-19 airway might similarly be due to inhibition of CFTR signaling by SARS-CoV-2 spike protein, and therefore activation of both NFκB and ENaC signaling. We used western blot and electrophysiological techniques, and an organoid model of normal airway epithelia, differentiated on an air-liquid-interface (ALI). We found that CFTR protein expression and CFTR cAMP-activated chloride channel activity were lost when the model epithelium was exposed to SARS-CoV-2 spike proteins. As hypothesized, the absence of CFTR led to activation of both TNFα/NFκB signaling and α and γ ENaC. We had previously shown that the cardiac glycoside drugs digoxin, digitoxin and ouabain blocked interaction of spike protein and ACE2. Consistently, addition of 30 nM concentrations of the cardiac glycoside drugs, prevented loss of both CFTR protein and CFTR channel activity. ACE2 and CFTR were found to co-immunoprecipitate in both basal cells and differentiated epithelia. Thus spike-dependent CFTR loss might involve ACE2 as a bridge between Spike and CFTR. In addition, spike exposure to the epithelia resulted in failure of endosomal recycling to return CFTR to the plasma membrane. Thus, failure of CFTR recovery from endosomal recycling might be a mechanism for spike-dependent loss of CFTR. Finally, we found that authentic SARS-CoV-2 virus infection induced loss of CFTR protein, which was rescued by the cardiac glycoside drugs digitoxin and ouabain. Based on experiments with this organoid model of small airway epithelia, and comparisons with 16HBE14o- and other cell types expressing normal CFTR, we predict that inflammation in the COVID-19 airway may be mediated by inhibition of CFTR signaling by the SARS-CoV-2 spike protein, thus inducing a cystic fibrosis-like clinical phenotype. To our knowledge this is the first time COVID-19 airway inflammation has been experimentally traced in normal subjects to a contribution from SARS-CoV-2 spike-dependent inhibition of CFTR signaling.
Collapse
Affiliation(s)
- Hung Caohuy
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Consortium for Health and Military Performance (CHAMP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Ofer Eidelman
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Tinghua Chen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Consortium for Health and Military Performance (CHAMP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Ognoon Mungunsukh
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Consortium for Health and Military Performance (CHAMP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Qingfeng Yang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Center for the Study of Traumatic Stress (CSTS), and Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Nathan I Walton
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Consortium for Health and Military Performance (CHAMP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | | | - Sara Khanal
- Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, 21702, USA
- The Geneva Foundation, Tacoma, WA, 98402, USA
| | - Shannon Hentschel
- Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, 21702, USA
- Cherokee Nation Assurance, Catoosa, OK, 74015, USA
| | - Catalina Florez
- Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, 21702, USA
- The Geneva Foundation, Tacoma, WA, 98402, USA
| | - Andrew S Herbert
- Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD, 21702, USA
| | - Harvey B Pollard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
- Collaborative Health Initiative Research Program (CHIRP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
- Consortium for Health and Military Performance (CHAMP), Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
3
|
Carbone A, Vitullo P, Di Gioia S, Conese M. Lung Inflammatory Genes in Cystic Fibrosis and Their Relevance to Cystic Fibrosis Transmembrane Conductance Regulator Modulator Therapies. Genes (Basel) 2023; 14:1966. [PMID: 37895314 PMCID: PMC10606852 DOI: 10.3390/genes14101966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cystic fibrosis (CF) is a monogenic syndrome determined by over 2000 mutations in the CF Transmembrane Conductance Regulator (CFTR) gene harbored on chromosome 7. In people with CF (PWCF), lung disease is the major determinant of morbidity and mortality and is characterized by a clinical phenotype which differs in the presence of equal mutational assets, indicating that genetic and environmental modifiers play an important role in this variability. Airway inflammation determines the pathophysiology of CF lung disease (CFLD) both at its onset and progression. In this narrative review, we aim to depict the inflammatory process in CF lung, with a particular emphasis on those genetic polymorphisms that could modify the clinical outcome of the respiratory disease in PWCF. The natural history of CF has been changed since the introduction of CFTR modulator therapies in the clinical arena. However, also in this case, there is a patient-to-patient variable response. We provide an overview on inflammatory/immunity gene variants that affect CFLD severity and an appraisal of the effects of CFTR modulator therapies on the inflammatory process in lung disease and how this knowledge may advance the optimization of the management of PWCF.
Collapse
Affiliation(s)
- Annalucia Carbone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Pamela Vitullo
- Cystic Fibrosis Support Center, Ospedale “G. Tatarella”, 71042 Cerignola, Italy;
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| |
Collapse
|
4
|
Mottais A, Riberi L, Falco A, Soccal S, Gohy S, De Rose V. Epithelial-Mesenchymal Transition Mechanisms in Chronic Airway Diseases: A Common Process to Target? Int J Mol Sci 2023; 24:12412. [PMID: 37569787 PMCID: PMC10418908 DOI: 10.3390/ijms241512412] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a reversible process, in which epithelial cells lose their epithelial traits and acquire a mesenchymal phenotype. This transformation has been described in different lung diseases, such as lung cancer, interstitial lung diseases, asthma, chronic obstructive pulmonary disease and other muco-obstructive lung diseases, such as cystic fibrosis and non-cystic fibrosis bronchiectasis. The exaggerated chronic inflammation typical of these pulmonary diseases can induce molecular reprogramming with subsequent self-sustaining aberrant and excessive profibrotic tissue repair. Over time this process leads to structural changes with progressive organ dysfunction and lung function impairment. Although having common signalling pathways, specific triggers and regulation mechanisms might be present in each disease. This review aims to describe the various mechanisms associated with fibrotic changes and airway remodelling involved in chronic airway diseases. Having better knowledge of the mechanisms underlying the EMT process may help us to identify specific targets and thus lead to the development of novel therapeutic strategies to prevent or limit the onset of irreversible structural changes.
Collapse
Affiliation(s)
- Angélique Mottais
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
| | - Luca Riberi
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Andrea Falco
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Simone Soccal
- Postgraduate School in Respiratory Medicine, University of Torino, 10124 Torino, Italy; (L.R.); (A.F.); (S.S.)
| | - Sophie Gohy
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium; (A.M.); (S.G.)
- Department of Pneumology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Cystic Fibrosis Reference Centre, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Virginia De Rose
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| |
Collapse
|
5
|
Santos L, Nascimento R, Duarte A, Railean V, Amaral MD, Harrison PT, Gama-Carvalho M, Farinha CM. Mutation-class dependent signatures outweigh disease-associated processes in cystic fibrosis cells. Cell Biosci 2023; 13:26. [PMID: 36759923 PMCID: PMC9912517 DOI: 10.1186/s13578-023-00975-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/28/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND The phenotypic heterogeneity observed in Cystic Fibrosis (CF) patients suggests the involvement of other genes, besides CFTR. Here, we combined transcriptome and proteome analysis to understand the global gene expression patterns associated with five prototypical CFTR mutations. RESULTS Evaluation of differentially expressed genes and proteins unveiled common and mutation-specific changes revealing functional signatures that are much more associated with the specific molecular defects associated with each mutation than to the CFTR loss-of-function phenotype. The combination of both datasets revealed that mutation-specific detected translated-transcripts (Dtt) have a high level of consistency. CONCLUSIONS This is the first combined transcriptomic and proteomic study focusing on prototypical CFTR mutations. Analysis of Dtt provides novel insight into the pathophysiology of CF, and the mechanisms through which each mutation class causes disease and will likely contribute to the identification of new therapeutic targets and/or biomarkers for CF.
Collapse
Affiliation(s)
- Lúcia Santos
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal ,grid.7872.a0000000123318773Department of Physiology, University College Cork, Cork, T12 K8AF Ireland
| | - Rui Nascimento
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Aires Duarte
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Violeta Railean
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Margarida D. Amaral
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Patrick T. Harrison
- grid.7872.a0000000123318773Department of Physiology, University College Cork, Cork, T12 K8AF Ireland
| | - Margarida Gama-Carvalho
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Carlos M. Farinha
- grid.9983.b0000 0001 2181 4263BioISI – Instituto de Biossistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| |
Collapse
|
6
|
Kingdom R, Wright CF. Incomplete Penetrance and Variable Expressivity: From Clinical Studies to Population Cohorts. Front Genet 2022; 13:920390. [PMID: 35983412 PMCID: PMC9380816 DOI: 10.3389/fgene.2022.920390] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/09/2022] [Indexed: 12/20/2022] Open
Abstract
The same genetic variant found in different individuals can cause a range of diverse phenotypes, from no discernible clinical phenotype to severe disease, even among related individuals. Such variants can be said to display incomplete penetrance, a binary phenomenon where the genotype either causes the expected clinical phenotype or it does not, or they can be said to display variable expressivity, in which the same genotype can cause a wide range of clinical symptoms across a spectrum. Both incomplete penetrance and variable expressivity are thought to be caused by a range of factors, including common variants, variants in regulatory regions, epigenetics, environmental factors, and lifestyle. Many thousands of genetic variants have been identified as the cause of monogenic disorders, mostly determined through small clinical studies, and thus, the penetrance and expressivity of these variants may be overestimated when compared to their effect on the general population. With the wealth of population cohort data currently available, the penetrance and expressivity of such genetic variants can be investigated across a much wider contingent, potentially helping to reclassify variants that were previously thought to be completely penetrant. Research into the penetrance and expressivity of such genetic variants is important for clinical classification, both for determining causative mechanisms of disease in the affected population and for providing accurate risk information through genetic counseling. A genotype-based definition of the causes of rare diseases incorporating information from population cohorts and clinical studies is critical for our understanding of incomplete penetrance and variable expressivity. This review examines our current knowledge of the penetrance and expressivity of genetic variants in rare disease and across populations, as well as looking into the potential causes of the variation seen, including genetic modifiers, mosaicism, and polygenic factors, among others. We also considered the challenges that come with investigating penetrance and expressivity.
Collapse
Affiliation(s)
| | - Caroline F. Wright
- Institute of Biomedical and Clinical Science, Royal Devon & Exeter Hospital, University of Exeter Medical School, Exeter, United Kingdom
| |
Collapse
|
7
|
Zhou W, Yu T, Hua Y, Hou Y, Ding Y, Nie H. Effects of Hypoxia on Respiratory Diseases: Perspective View of Epithelial Ion Transport. Am J Physiol Lung Cell Mol Physiol 2022; 323:L240-L250. [PMID: 35819839 DOI: 10.1152/ajplung.00065.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The balance of gas exchange and lung ventilation is essential for the maintenance of body homeostasis. There are many ion channels and transporters in respiratory epithelial cells, including epithelial sodium channel, Na,K-ATPase, cystic fibrosis transmembrane conductance regulator, and some transporters. These ion channels/transporters maintain the capacity of liquid layer on the surface of respiratory epithelial cells, and provide an immune barrier for the respiratory system to clear off foreign pathogens. However, in some harmful external environment and/or pathological conditions, the respiratory epithelium is prone to hypoxia, which would destroy the ion transport function of the epithelium and unbalance the homeostasis of internal environment, triggering a series of pathological reactions. Many respiratory diseases associated with hypoxia manifest an increased expression of hypoxia-inducible factor-1, which mediates the integrity of the epithelial barrier and affects epithelial ion transport function. It is important to study the relationship between hypoxia and ion transport function, whereas the mechanism of hypoxia-induced ion transport dysfunction in respiratory diseases is not clear. This review focuses on the relationship of hypoxia and respiratory diseases, as well as dysfunction of ion transport and tight junctions in respiratory epithelial cells under hypoxia.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yu Hua
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
8
|
Bojanowski CM, Lu S, Kolls JK. Mucosal Immunity in Cystic Fibrosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2901-2912. [PMID: 35802761 PMCID: PMC9270582 DOI: 10.4049/jimmunol.2100424] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/21/2021] [Indexed: 05/27/2023]
Abstract
The highly complex and variable genotype-phenotype relationships observed in cystic fibrosis (CF) have been an area of growing interest since the discovery of the CF transmembrane conductance regulator (CFTR) gene >30 y ago. The consistently observed excessive, yet ineffective, activation of both the innate and adaptive host immune systems and the establishment of chronic infections within the lung, leading to destruction and functional decline, remain the primary causes of morbidity and mortality in CF. The fact that both inflammation and pathogenic bacteria persist despite the introduction of modulator therapies targeting the defective protein, CFTR, highlights that we still have much to discover regarding mucosal immunity determinants in CF. Gene modifier studies have overwhelmingly implicated immune genes in the pulmonary phenotype of the disease. In this context, we aim to review recent advances in our understanding of the innate and adaptive immune systems in CF lung disease.
Collapse
Affiliation(s)
- Christine M Bojanowski
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, LA;
| | - Shiping Lu
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA; and
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Department of Medicine, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
9
|
Butnariu LI, Țarcă E, Cojocaru E, Rusu C, Moisă ȘM, Leon Constantin MM, Gorduza EV, Trandafir LM. Genetic Modifying Factors of Cystic Fibrosis Phenotype: A Challenge for Modern Medicine. J Clin Med 2021; 10:5821. [PMID: 34945117 PMCID: PMC8707808 DOI: 10.3390/jcm10245821] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Cystic fibrosis (CF) is a monogenic autosomal recessive disease caused by cystic fibrosis transmembrane conductance regulator (CFTR) gene mutations. CF is characterized by a high phenotypic variability present even in patients with the same genotype. This is due to the intervention of modifier genes that interact with both the CFTR gene and environmental factors. The purpose of this review is to highlight the role of non-CFTR genetic factors (modifier genes) that contribute to phenotypic variability in CF. We analyzed literature data starting with candidate gene studies and continuing with extensive studies, such as genome-wide association studies (GWAS) and whole exome sequencing (WES). The results of both types of studies revealed that the number of modifier genes in CF patients is impressive. Their identification offers a new perspective on the pathophysiological mechanisms of the disease, paving the way for the understanding of other genetic disorders. In conclusion, in the future, genetic analysis, such as GWAS and WES, should be performed routinely. A challenge for future research is to integrate their results in the process of developing new classes of drugs, with a goal to improve the prognosis, increase life expectancy, and enhance quality of life among CF patients.
Collapse
Affiliation(s)
- Lăcrămioara Ionela Butnariu
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.I.B.); (C.R.); (E.V.G.)
| | - Elena Țarcă
- Department of Surgery II—Pediatric Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iaşi, Romania
| | - Cristina Rusu
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.I.B.); (C.R.); (E.V.G.)
| | - Ștefana Maria Moisă
- Department of Mother and Child, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (Ș.M.M.); (L.M.T.)
| | | | - Eusebiu Vlad Gorduza
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.I.B.); (C.R.); (E.V.G.)
| | - Laura Mihaela Trandafir
- Department of Mother and Child, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (Ș.M.M.); (L.M.T.)
| |
Collapse
|
10
|
Giorgetti M, Klymiuk N, Bähr A, Hemmerling M, Jinton L, Tarran R, Malmgren A, Åstrand A, Hansson GC, Ermund A. New generation ENaC inhibitors detach cystic fibrosis airway mucus bundles via sodium/hydrogen exchanger inhibition. Eur J Pharmacol 2021; 904:174123. [PMID: 33974881 PMCID: PMC8477379 DOI: 10.1016/j.ejphar.2021.174123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 11/18/2022]
Abstract
Cystic fibrosis (CF) is a recessive inherited disease caused by mutations affecting anion transport by the epithelial ion channel cystic fibrosis transmembrane conductance regulator (CFTR). The disease is characterized by mucus accumulation in the airways and intestine, but the major cause of mortality in CF is airway mucus accumulation, leading to bacterial colonization, inflammation and respiratory failure. Several drug targets are under evaluation to alleviate airway mucus obstruction in CF and one of these targets is the epithelial sodium channel ENaC. To explore effects of ENaC inhibitors on mucus properties, we used two model systems to investigate mucus characteristics, mucus attachment in mouse ileum and mucus bundle transport in piglet airways. We quantified mucus attachment in explants from CFTR null (CF) mice and tracheobronchial explants from newborn CFTR null (CF) piglets to evaluate effects of ENaC or sodium/hydrogen exchanger (NHE) inhibitors on mucus attachment. ENaC inhibitors detached mucus in the CF mouse ileum, although the ileum lacks ENaC expression. This effect was mimicked by two NHE inhibitors. Airway mucus bundles were immobile in untreated newborn CF piglets but were detached by the therapeutic drug candidate AZD5634 (patent WO, 2015140527). These results suggest that the ENaC inhibitor AZD5634 causes detachment of CF mucus in the ileum and airway via NHE inhibition and that drug design should focus on NHE instead of ENaC inhibition.
Collapse
Affiliation(s)
- Melania Giorgetti
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Sweden.
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Germany.
| | - Andrea Bähr
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Germany.
| | - Martin Hemmerling
- Research and Early Development, Respiratory, Inflammation and Autoimmunity (RIA), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Lisa Jinton
- Research and Early Development, Respiratory, Inflammation and Autoimmunity (RIA), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Robert Tarran
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, North Carolina, United States.
| | - Anna Malmgren
- Research and Early Development, Respiratory, Inflammation and Autoimmunity (RIA), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Annika Åstrand
- Research and Early Development, Respiratory, Inflammation and Autoimmunity (RIA), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Gunnar C Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Sweden.
| | - Anna Ermund
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Sweden.
| |
Collapse
|
11
|
Sepahzad A, Morris-Rosendahl DJ, Davies JC. Cystic Fibrosis Lung Disease Modifiers and Their Relevance in the New Era of Precision Medicine. Genes (Basel) 2021; 12:genes12040562. [PMID: 33924524 PMCID: PMC8069009 DOI: 10.3390/genes12040562] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
Our understanding of cystic fibrosis (CF) has grown exponentially since the discovery of the cystic fibrosis transmembrane conductance regulator (CFTR) gene in 1989. With evolving genetic and genomic tools, we have come to better understand the role of CFTR genotypes in the pathophysiology of the disease. This, in turn, has paved the way for the development of modulator therapies targeted at mutations in the CFTR, which are arguably one of the greatest advances in the treatment of CF. These modulator therapies, however, do not target all the mutations in CFTR that are seen in patients with CF and, furthermore, a variation in response is seen in patients with the same genotype who are taking modulator therapies. There is growing evidence to support the role of non-CFTR modifiers, both genetic and environmental, in determining the variation seen in CF morbidity and mortality and also in the response to existing therapies. This review focusses on key findings from studies using candidate gene and genome-wide approaches to identify CF modifier genes of lung disease in cystic fibrosis and considers the interaction between modifiers and the response to modulator therapies. As the use of modulator therapies expands and we gain data around outcomes, it will be of great interest to investigate this interaction further. Going forward, it will also be crucial to better understand the relative influence of genomic versus environmental factors. With this understanding, we can truly begin to deliver personalised care by better profiling the likely disease phenotype for each patient and their response to treatment.
Collapse
Affiliation(s)
- Afsoon Sepahzad
- Department of Paediatric Respiratory Medicine, Royal Brompton and Harefield Hospitals, London SW3 6NP, UK;
| | | | - Jane C. Davies
- Department of Paediatric Respiratory Medicine, Royal Brompton and Harefield Hospitals, London SW3 6NP, UK;
- National Heart & Lung Institute, Imperial College London, Emmanuel Kay Building, 1b Manresa Rd, London SW3 6LR, UK
- Correspondence:
| |
Collapse
|
12
|
Jurado-Martín I, Sainz-Mejías M, McClean S. Pseudomonas aeruginosa: An Audacious Pathogen with an Adaptable Arsenal of Virulence Factors. Int J Mol Sci 2021; 22:3128. [PMID: 33803907 PMCID: PMC8003266 DOI: 10.3390/ijms22063128] [Citation(s) in RCA: 331] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa is a dominant pathogen in people with cystic fibrosis (CF) contributing to morbidity and mortality. Its tremendous ability to adapt greatly facilitates its capacity to cause chronic infections. The adaptability and flexibility of the pathogen are afforded by the extensive number of virulence factors it has at its disposal, providing P. aeruginosa with the facility to tailor its response against the different stressors in the environment. A deep understanding of these virulence mechanisms is crucial for the design of therapeutic strategies and vaccines against this multi-resistant pathogen. Therefore, this review describes the main virulence factors of P. aeruginosa and the adaptations it undergoes to persist in hostile environments such as the CF respiratory tract. The very large P. aeruginosa genome (5 to 7 MB) contributes considerably to its adaptive capacity; consequently, genomic studies have provided significant insights into elucidating P. aeruginosa evolution and its interactions with the host throughout the course of infection.
Collapse
Affiliation(s)
| | | | - Siobhán McClean
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin 4 D04 V1W8, Ireland; (I.J.-M.); (M.S.-M.)
| |
Collapse
|
13
|
Two Siblings Homozygous for F508del-CFTR Have Varied Disease Phenotypes and Protein Biomarkers. Int J Mol Sci 2021; 22:ijms22052631. [PMID: 33807880 PMCID: PMC7961721 DOI: 10.3390/ijms22052631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/20/2021] [Accepted: 03/01/2021] [Indexed: 12/01/2022] Open
Abstract
Two siblings with CF are homozygous for F508del (referred to as Subject A and Subject B). Despite having the same CFTR genotype and similar environment, these two subjects exhibited different disease phenotypes. We analyzed their medical records and CF Foundation Registry data and measured inflammatory protein mediators in their sputum samples. Then, we examined the longitudinal relationships between inflammatory markers and disease severity for each subject and compared between them. Subject A presented a more severe disease than Subject B. During the study period, Subject A had two pulmonary exacerbations (PEs) whereas Subject B had one mild PE. The forced expiratory volume in 1 s (FEV1, % predicted) values for Subject A were between 34–45% whereas for Subject B varied between 48–90%. Inflammatory protein mediators associated with neutrophils, Th1, Th2, and Th17 responses were elevated in sputum of Subject A compared with Subject B, and also in samples collected prior to and during PEs for both subjects. Neutrophilic elastase (NE) seemed to be the most informative biomarkers. The infectious burden between these two subjects was different.
Collapse
|
14
|
Abstract
Because CFTR gene studies now represent one of the most frequent genetic analyses routinely performed worldwide, the number of rare CFTR variants identified in various clinical situations, regularly increases. To provide appropriate diagnosis and prognosis to CF patients as well as appropriate genetic counseling to families, the clinical impact and the phenotypic spectrum of variants identified by diagnostic techniques need to be characterized. Three complementary locus specific databases, called CFTR1, CFTR2 and CFTR-France were developed to address these issues. Besides, the growing knowledge of the CF pathophysiology and the technical evolution in molecular biology allowed to identify candidate modifier genes, regulatory loci, epigenetic profiles and trans-regulators that could help to refine genotype-phenotype correlations at the individual level. These different factors may contribute to the large phenotypic variability between patients with CF, even when they carry identical CFTR variants, regarding lung function, meconium ileus susceptibility or the risk for developing CFTR-related diabetes and liver disease. Finally, the availability of new therapies that target the CFTR protein for numbers of CF patients led to the identification of 'good' and 'poor' responders, thus raising questions of pharmacogenetics factors that may influence treatment efficiency as a novel feature of the complexity of CF patients' management. © 2020 French Society of Pediatrics. Published by Elsevier Masson SAS. All rights reserved.
Collapse
|
15
|
Ruffin M, Mercier J, Calmel C, Mésinèle J, Bigot J, Sutanto EN, Kicic A, Corvol H, Guillot L. Update on SLC6A14 in lung and gastrointestinal physiology and physiopathology: focus on cystic fibrosis. Cell Mol Life Sci 2020; 77:3311-3323. [PMID: 32166393 PMCID: PMC7426304 DOI: 10.1007/s00018-020-03487-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/24/2020] [Accepted: 02/17/2020] [Indexed: 12/17/2022]
Abstract
The solute carrier family 6 member 14 (SLC6A14) protein imports and concentrates all neutral amino acids as well as the two cationic acids lysine and arginine into the cytoplasm of different cell types. Primarily described as involved in several cancer and colonic diseases physiopathological mechanisms, the SLC6A14 gene has been more recently identified as a genetic modifier of cystic fibrosis (CF) disease severity. It was indeed shown to have a pleiotropic effect, modulating meconium ileus occurrence, lung disease severity, and precocity of P. aeruginosa airway infection. The biological mechanisms explaining the impact of SLC6A14 on intestinal and lung phenotypes of CF patients are starting to be elucidated. This review focuses on SLC6A14 in lung and gastrointestinal physiology and physiopathology, especially its involvement in the pathophysiology of CF disease.
Collapse
Affiliation(s)
- Manon Ruffin
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint‑Antoine (CRSA), Paris, France
| | - Julia Mercier
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint‑Antoine (CRSA), Paris, France
| | - Claire Calmel
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint‑Antoine (CRSA), Paris, France
| | - Julie Mésinèle
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint‑Antoine (CRSA), Paris, France
| | - Jeanne Bigot
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint‑Antoine (CRSA), Paris, France
| | - Erika N Sutanto
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- School of Public Health, Curtin University, Bentley, WA, Australia
| | - Anthony Kicic
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- School of Public Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia
| | - Harriet Corvol
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint‑Antoine (CRSA), Paris, France.
- Pneumologie Pédiatrique, APHP, Hôpital Trousseau, Paris, France.
| | - Loic Guillot
- Sorbonne Université, INSERM UMR S 938, Centre de Recherche Saint‑Antoine (CRSA), Paris, France
| |
Collapse
|
16
|
Cui X, Wu X, Li Q, Jing X. Mutations of the cystic fibrosis transmembrane conductance regulator gene in males with congenital bilateral absence of the vas deferens: Reproductive implications and genetic counseling (Review). Mol Med Rep 2020; 22:3587-3596. [PMID: 33000223 PMCID: PMC7533508 DOI: 10.3892/mmr.2020.11456] [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: 04/13/2020] [Accepted: 07/21/2020] [Indexed: 11/05/2022] Open
Abstract
Congenital bilateral absence of the vas deferens (CBAVD) is predominantly caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CBAVD accounts for 2–6% of male infertility cases and up to 25% of cases of obstructive azoospermia. With the use of pre-implantation genetic diagnosis, testicular or epididymal sperm aspiration, intracytoplasmic sperm injection and in vitro fertilization, patients affected by CBAVD are able to have children who do not carry CFTR gene mutations, thereby preventing disease. Therefore, genetic counseling should be provided to couples receiving assisted reproductive techniques to discuss the impact of CFTR gene mutations on reproductive health. In the present article, the current literature concerning the CFTR gene and its association with CBAVD is reviewed.
Collapse
Affiliation(s)
- Xiangrong Cui
- Reproductive Medicine Center, Children's Hospital of Shanxi and Women's Health Center of Shanxi, Taiyuan, Shanxi 030001, P.R. China
| | - Xueqing Wu
- Reproductive Medicine Center, Children's Hospital of Shanxi and Women's Health Center of Shanxi, Taiyuan, Shanxi 030001, P.R. China
| | - Qiang Li
- Reproductive Medicine Center, Children's Hospital of Shanxi and Women's Health Center of Shanxi, Taiyuan, Shanxi 030001, P.R. China
| | - Xuan Jing
- Clinical Laboratory, Shanxi Province People's Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
17
|
Paranjapye A, Ruffin M, Harris A, Corvol H. Genetic variation in CFTR and modifier loci may modulate cystic fibrosis disease severity. J Cyst Fibros 2020; 19 Suppl 1:S10-S14. [PMID: 31734115 PMCID: PMC7036019 DOI: 10.1016/j.jcf.2019.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022]
Abstract
In patients with cystic fibrosis (CF), genetic variants within and outside the CFTR locus contribute to the variability of the disease severity. CFTR transcription is tightly regulated by cis-regulatory elements (CREs) that control the three-dimensional structure of the locus, chromatin accessibility and transcription factor recruitment. Variants within these CREs may contribute to the pathophysiology and to the phenotypic heterogeneity by altering CFTR transcript abundance. In addition to the CREs, variants outside the CFTR locus, namely "modifiers genes", may also be associated with the clinical variability. This review addresses variants at the CFTR locus itself and CFTR CREs, together with the outcomes of the latest modifier gene studies with respect to the different CF phenotypes.
Collapse
Affiliation(s)
- Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University Medical School, 10900 Euclid Avenue, Cleveland, OH, USA
| | - Manon Ruffin
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University Medical School, 10900 Euclid Avenue, Cleveland, OH, USA.
| | - Harriet Corvol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France; AP-HP, Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France.
| |
Collapse
|
18
|
Pasterkamp H, Menzies KJ, Bayomi DJ. Cystic fibrosis in Canadian Hutterites. Pediatr Pulmonol 2020; 55:526-532. [PMID: 31782915 DOI: 10.1002/ppul.24590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/18/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND The Hutterite Brethren are a communal group of Anabaptists who live in the Western regions of North America, predominantly in the Western Canadian provinces. Due to a founder effect, Hutterites have a high rate of cystic fibrosis (CF) with genotypes limited to only two CFTR mutations. One-third of Hutterite patients with CF are pancreatic sufficient. Previously we found an unexplained younger age at death in Hutterites compared with nonHutterites homozygous for the common F508del mutation. The present study expanded the data collection and analysis for confirmation and further exploration. METHODS Anonymized information, based on Hutterite surnames, was extracted from the Canadian CF Registry. Summary data on nonHutterite patients with CF homozygous for F508del served as control. Statistical analyses explored the effects of genotype within Hutterites and compared nutritional status, lung function, and microbiologic findings between the groups. RESULTS The younger average age at death in Hutterites compared with controls was confirmed, but there was no suggestion of a generally shortened life expectancy. While the nutritional status in Hutterite children was better than that of controls, their lung function was slightly but significantly lower. Staphylococcus aureus was more frequent in Hutterites while there was no difference between the groups regarding Pseudomonas aeruginosa. CONCLUSIONS Despite less pancreatic insufficiency, better nutrition, communal life in socioeconomic stability, and without exposure to environmental tobacco smoke, the clinical course of CF appears to be more severe in a significant number of Hutterites. Investigations of gene-environment interactions and of CF disease gene modifiers may help to explain this conundrum.
Collapse
Affiliation(s)
- Hans Pasterkamp
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - Kathryn J Menzies
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - Dennis J Bayomi
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
19
|
Shanthikumar S, Neeland MN, Saffery R, Ranganathan S. Gene modifiers of cystic fibrosis lung disease: A systematic review. Pediatr Pulmonol 2019; 54:1356-1366. [PMID: 31140758 DOI: 10.1002/ppul.24366] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Lung disease is the major source of morbidity and mortality in cystic fibrosis (CF), with large variability in severity between patients. Although accurate prediction of lung disease severity would be extremely useful, no robust methods exist. Twin and sibling studies have highlighted the importance of non-cystic fibrosis transmembrane conductance regulator (CFTR) genes in determining lung disease severity but how these impact on the severity in CF remains unclear. METHODS A systematic review was undertaken to answer the question "In patients with CF which non-CFTR genes modify the severity of lung disease?" The method for this systematic review was based upon the "Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)" statement, with a narrative synthesis of results planned. RESULTS A total of 1168 articles were screened for inclusion, with 275 articles undergoing detailed assessment for inclusion. One hundred and forty articles were included. Early studies focused on candidate genes, whereas more recent studies utilized genome-wide approaches and also examined epigenetic mechanisms, gene expression, and therapeutic response. DISCUSSION A large body of evidence regarding non-CFTR gene modifiers of lung disease severity has been generated, examining a wide array of genes. Limitations to existing studies include heterogeneity in outcome measures used, limited replication, and relative lack of clinical impact. Future work examining non-CFTR gene modifiers will have to overcome these limitations if gene modifiers are to have a meaningful role in the care of patients with CF.
Collapse
Affiliation(s)
- Shivanthan Shanthikumar
- Respiratory and Sleep Medicine Department, Royal Children's Hospital, Melbourne, Australia.,Respiratory Diseases Department, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Australia
| | - Melanie N Neeland
- Department of Paediatrics, The University of Melbourne, Australia.,Centre of Food and Allergy Research, Murdoch Children's Research Institute, Melbourne, Australia
| | - Richard Saffery
- Department of Paediatrics, The University of Melbourne, Australia.,Cancer & Disease Epigenetics, Murdoch Children's Research Institute, Melbourne, Australia
| | - Sarath Ranganathan
- Respiratory and Sleep Medicine Department, Royal Children's Hospital, Melbourne, Australia.,Respiratory Diseases Department, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Australia
| |
Collapse
|
20
|
Welsner M, Straßburg S, Taube C, Sutharsan S. Use of ivacaftor in late diagnosed cystic fibrosis monozygotic twins heterozygous for F508del and R117H-7T - a case report. BMC Pulm Med 2019; 19:76. [PMID: 30975115 PMCID: PMC6458608 DOI: 10.1186/s12890-019-0840-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND CFTR modulator therapy with ivacaftor is a treatment option for Cystic Fibrosis (CF) patients with at least one copy of a R117H-7T mutation in the CFTR gene. Desirable effects of this therapy are improvement of lung function, decrease in exacerbation rate, normalization or reduction of sweat chloride and weight gain. Monogenetic CF-twins carry identical genetic information, so therapy response and side effects are expected to be nearly identical under this specific therapy. CASE PRESENTATION In monozygotic twins, at the age of 55, two pathogenic variants in the CFTR gene (F508del and R117H-7T) were detected. Both patients presented with a borderline sweat test (30-59 mmol/L) and despite the same genetic information and similar life circumstances the disease proceeds completely different. While one patient has severe pulmonary involvement with chronic P. aeruginosa infection, her twin sister is almost unimpaired. Liver or pancreatic involvement was not seen in either patient. Due to the presence of one copy of a R117H-7T mutation, CFTR modulator therapy with ivacaftor was initiated in both. Response and side effects were significantly different. In the less affected patient, we observed an improvement in lung function and a normalization of sweat chloride. In the severely affected patient, no functional response to treatment was seen, but stabilization of the disease state with a decrease in exacerbation and hospitalization rate and weight gain as well as a normalization of sweat chloride. There was an increase in liver enzymes in the less affected patient, which normalized after halving the dose of ivacaftor, while the therapeutic effect was maintained. CONCLUSIONS Despite nearly identical genetic information, as in monogenetic twins, therapy response and onset of side effects of CFTR modulating therapy are very different. In patients with late diagnosis and severe pulmonary involvement, ivacaftor does not seem to improve lung function, whereas in patients with late diagnosis and low disease severity a relevant therapy response was obtained. In addition to lung function, additional clinical parameters such as reduction of exacerbation and hospitalization rate and weight gain should be used to assess therapy response, especially in severely affected patients.
Collapse
Affiliation(s)
- Matthias Welsner
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Adult Cystic Fibrosis Center, University of Duisburg-Essen, Tueschener Weg 40, 45329, Essen, Germany.
| | - Svenja Straßburg
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Adult Cystic Fibrosis Center, University of Duisburg-Essen, Tueschener Weg 40, 45329, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Adult Cystic Fibrosis Center, University of Duisburg-Essen, Tueschener Weg 40, 45329, Essen, Germany
| | - Sivagurunathan Sutharsan
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Adult Cystic Fibrosis Center, University of Duisburg-Essen, Tueschener Weg 40, 45329, Essen, Germany
| |
Collapse
|
21
|
Reynaud Q, Boudreau V, Touzet S, Desjardins K, Bourdy SP, Blond E, Berthiaume Y, Rabasa-Lhoret R, Durieu I. Glucose tolerance in Canadian and French cystic fibrosis adult patients. Sci Rep 2019; 9:4763. [PMID: 30894563 PMCID: PMC6427035 DOI: 10.1038/s41598-019-40592-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 02/04/2019] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF)-related diabetes is associated with increased mortality. We analysed the clinical and glycemic profiles of two cohorts of patients treated according to the same guidelines in France and Canada. To investigate incidence differences in phenotypic and glucose abnormalities and to explore the evolution over a 4-year follow-up period, two cohorts of 224 Canadian and 147 French adult CF patients (≥18 years) without treated CF-related diabetes (CFRD) were followed over a 4 year period. In each of these groups, we investigated the longitudinal relationship between glucose tolerance and pulmonary function. An annual 2-hour oral glucose tolerance test was performed: fasting blood glucose (G0) and 2-h blood glucose (G2) were measured. Patients were classified at inclusion according to their glucose tolerance status: Normal glucose tolerant, abnormal glucose tolerant or de novo CFRD. Age, sex ratio and proportion of F508del homozygous patients were not statistically different between both cohorts. Canadian patients had better pulmonary function (median %FEV1 (IQR): 71.0 (55.0–82.0) vs. 64.0 (40.0–78.0), p < 0.001) and greater body mass index (BMI; median BMI in kg/m2) (IQR) 21.1 (19.5–22.8) vs. 19.9 (18.4–21.4), p < 0.001). Glucose values: G0 (5.4 (5.0–5.9) vs. 4.8 (4.5–5.1) mmol/L, p < 0.001) and G2 (7.6 (5.8–9.7) vs. 6.5 (5.2–8.5) mmol/L, p = 0.001) were higher in the Canadian cohort translating into a higher incidence of de novo CFRD diagnosis (19.2 vs. 9.8%, p = 0.003). Decline in FEV1 over time was not different between patients according to glucose tolerance groups. Despite higher glucose levels and incidence of de novo CFRD, Canadian CF patients have a better lung function and a higher BMI than French patients. In spite of these differences between the cohorts, the decline in FEV1 in patients with abnormal glucose tolerance is similar between these groups.
Collapse
Affiliation(s)
- Quitterie Reynaud
- Centre de référence Adulte de la Mucoviscidose, Service de médecine interne, Hospices civils de Lyon, F-69495, Pierre Bénite, France. .,Université de Lyon, Équipe d'Accueil Health Services and Performance Research (HESPER) 7425, F-69003, Lyon, France.
| | - Valérie Boudreau
- Montreal Clinical Research Institute, Québec, Canada.,Université de Montréal, Département de nutrition et de Médecine, Montréal, Québec, Canada
| | - Sandrine Touzet
- Hospices Civils de Lyon, Pôle de Santé Publique, Lyon, F-69003, France.,Université de Lyon, Équipe d'Accueil Health Services and Performance Research (HESPER) 7425, Lyon, France
| | | | - Stéphanie Poupon Bourdy
- Hospices Civils de Lyon, Pôle de Santé Publique, Lyon, F-69003, France.,Université de Lyon, Équipe d'Accueil Health Services and Performance Research (HESPER) 7425, Lyon, France
| | - Emilie Blond
- Service de Biochimie et Biologie Moléculaire, Hospices Civils de Lyon, F-69495, Pierre Bénite, France.,Université de Lyon, INSERM U1060, Laboratoire CarMen, F-69003, Lyon, France
| | - Yves Berthiaume
- Montreal Clinical Research Institute, Québec, Canada.,Cystic fibrosis clinic, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Rémi Rabasa-Lhoret
- Montreal Clinical Research Institute, Québec, Canada.,Université de Montréal, Département de nutrition et de Médecine, Montréal, Québec, Canada.,Cystic fibrosis clinic, Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Isabelle Durieu
- Centre de référence Adulte de la Mucoviscidose, Service de médecine interne, Hospices civils de Lyon, F-69495, Pierre Bénite, France.,Université de Lyon, Équipe d'Accueil Health Services and Performance Research (HESPER) 7425, F-69003, Lyon, France.,Hospices Civils de Lyon, Pôle de Santé Publique, Lyon, F-69003, France.,Université de Lyon, Équipe d'Accueil Health Services and Performance Research (HESPER) 7425, Lyon, France
| |
Collapse
|
22
|
Boëlle PY, Debray D, Guillot L, Corvol H. SERPINA1 Z allele is associated with cystic fibrosis liver disease. Genet Med 2019; 21:2151-2155. [PMID: 30739910 DOI: 10.1038/s41436-019-0449-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The SERPINA1 Z allele is associated with cystic fibrosis (CF)-related liver disease (CFLD), a common manifestation in patients with CF. We estimated CFLD incidence based on the SERPINA1 genotype in 3328 CF patients with CFLD-phenotype information. METHODS The associations of SERPINA1 Z (rs28929474) and S (rs17580) alleles with age at CFLD onset and the development of CFLD-related complications (severe liver disease with cirrhosis, portal hypertension, esophageal varices) were analyzed. RESULTS Overall, 3% of patients carried the SERPINA1 Z allele and 13% carried the S allele. The cumulative incidence of CFLD increased more rapidly in patients carrying the Z allele (hazard ratio [HR] = 1.6; 95% confidence interval [CI] = 1.1-2.4, P = 0.019), reaching 47% by age 25 compared with 30% in noncarriers. Increased risk was similar for patients with severe CFLD (HR = 1.5, 95% CI = 0.7-3.2, P = 0.31) but failed to reach significance due to a limited sample size of Z-allele carriers. No significant effect was found for the S allele. CONCLUSION CF patients carrying the SERPINA1 Z allele had an increased risk of developing CFLD and related complications compared with noncarriers. Routine SERPINA1 Z genotyping upon CF diagnosis is warranted for identifying patients worthy of closer liver disease monitoring.
Collapse
Affiliation(s)
- Pierre-Yves Boëlle
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (iPLESP), AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Dominique Debray
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France.,AP-HP, Hôpital Necker Enfants Malades, Unité d'Hépatologie Pédiatrique, Paris, France
| | - Loic Guillot
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Harriet Corvol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France. .,AP-HP, Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France.
| | | |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW To compile data from the past 10 years regarding the role of modifying genes in cystic fibrosis (CF). RECENT FINDINGS CF is a model disease for understanding of the action of modifying genes. Although it is a monogenic (CFTR) autosomal recessive disease, CF presents with wide phenotypic variability. In CF, variability occurs with different intensity among patients by each organ, being organ-specific, resulting from the mutual interaction of environmental and genetic factors, including CFTR mutations and various other genes, most of which are associated with inflammatory processes. In individuals, using precision medicine, gene modification studies have revealed individualized responses to drugs depending on particular CFTR mutations and modifying genes, most of which are alternative ion channels. SUMMARY Studies of modifying genes in CF allow: understanding of clinical variability among patients with the same CFTR genotype; evaluation of precision medicine; understanding of environmental and genetic effects at the organ level; understanding the involvement of genetic variants in inflammatory responses; improvements in genetic counseling; understanding the involvement of genetic variants in inflammatory responses in lung diseases, such as asthma; and understanding the individuality of the person with the disease.
Collapse
|
24
|
Kmit A, Marson FAL, Pereira SVN, Vinagre AM, Leite GS, Servidoni MF, Ribeiro JD, Ribeiro AF, Bertuzzo CS, Amaral MD. Extent of rescue of F508del-CFTR function by VX-809 and VX-770 in human nasal epithelial cells correlates with SNP rs7512462 in SLC26A9 gene in F508del/F508del Cystic Fibrosis patients. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1323-1331. [PMID: 30716472 DOI: 10.1016/j.bbadis.2019.01.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/16/2019] [Accepted: 01/30/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND We analyzed the CFTR response to VX-809/VX-770 drugs in conditionally reprogrammed cells (CRC) of human nasal epithelium (HNE) from F508del/F508del patients based on SNP rs7512462 in the Solute Carrier Family 26, Member 9 (SLC26A9; MIM: 608481) gene. METHODS The Isc-eq measurements of primary nasal epithelial cells from F508del/F508del patients (n = 12) for CFTR function were performed in micro Ussing chambers and compared with non-CF controls (n = 2). Data were analyzed according to the rs7512462 genotype which were determined by real-time PCR. RESULTS The CRC-HNE cells from F508del/F508del patients evidenced high variability in the basal levels of CFTR function. Also, the rs7512462*C allele showed an increased basal CFTR function and higher responses to VX-809 + VX-770. The rs7512462*CC + CT genotypes together evidenced CFTR function levels of 14.89% relatively to wt/wt (rs7512462*CT alone-15.29%) i.e., almost double of rs7512462*TT (7.13%). Furthermore, sweat [Cl-] and body mass index of patients also evidenced an association with the rs7512462 genotype. CONCLUSION The CFTR function can be performed in F508del/F508del patient-derived CRC-HNEs and its function and responses to VX-809 + VX-770 combination as well as clinical data, are all associated with the rs7512462 variant, which partially sheds light on the generally inter-individual phenotypic variability and in personalized responses to CFTR modulator drugs.
Collapse
Affiliation(s)
- Arthur Kmit
- Department of Medical Genetics and Genomic Medicine, Faculty of Medical Sciences, University of Campinas, Brazil; Department of Pediatrics, Faculty of Medical Sciences, University of Campinas, Brazil.
| | - Fernando Augusto Lima Marson
- Department of Medical Genetics and Genomic Medicine, Faculty of Medical Sciences, University of Campinas, Brazil; Department of Pediatrics, Faculty of Medical Sciences, University of Campinas, Brazil.
| | - Stéphanie Villa-Nova Pereira
- Department of Medical Genetics and Genomic Medicine, Faculty of Medical Sciences, University of Campinas, Brazil
| | | | - Gabriela Silva Leite
- Department of Pediatrics, Faculty of Medical Sciences, University of Campinas, Brazil
| | | | - José Dirceu Ribeiro
- Department of Pediatrics, Faculty of Medical Sciences, University of Campinas, Brazil
| | | | - Carmen Sílvia Bertuzzo
- Department of Medical Genetics and Genomic Medicine, Faculty of Medical Sciences, University of Campinas, Brazil.
| | - Margarida Duarte Amaral
- University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, Portugal.
| |
Collapse
|
25
|
Pollard BS, Pollard HB. Induced pluripotent stem cells for treating cystic fibrosis: State of the science. Pediatr Pulmonol 2018; 53:S12-S29. [PMID: 30062693 DOI: 10.1002/ppul.24118] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/31/2018] [Indexed: 12/20/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are a recently developed technology in which fully differentiated cells such as fibroblasts from individual CF patients can be repaired with [wildtype] CFTR, and reprogrammed to differentiate into fully differentiated cells characteristic of the proximal and distal airways. Here, we review properties of different epithelial cells in the airway, and the in vitro genetic roadmap which iPSCs follow as they are step-wise differentiated into either basal stem cells, for the proximal airway, or into Type II Alveolar cells for the distal airways. The central theme is that iPSC-derived basal stem cells, are penultimately dependent on NOTCH signaling for differentiation into club cells, goblet cells, ciliated cells, and neuroendocrine cells. Furthermore, given the proper matrix, these cellular progenies are also able to self-assemble into a fully functional pseudostratified squamous proximal airway epithelium. By contrast, club cells are reserve stem cells which are able to either differentiate into goblet or ciliated cells, but also to de-differentiate into basal stem cells. Variant club cells, located at the transition between airway and alveoli, may also be responsible for differentiation into Type II Alveolar cells, which then differentiate into Type I Alveolar cells for gas exchange in the distal airway. Using gene editing, the mutant CFTR gene in iPSCs from CF patients can be repaired, and fully functional epithelial cells can thus be generated through directed differentiation. However, there is a limitation in that the lung has other CFTR-dependent cells besides epithelial cells. Another limitation is that there are CFTR-dependent cells in other organs which would continue to contribute to CF disease. Furthermore, there are also bystander or modifier genes which affect disease outcome, not only in the lung, but specifically in other CF-affected organs. Finally, we discuss future personalized applications of the iPSC technology, many of which have already survived the "proof-of-principle" test. These include (i) patient-derived iPSCs used as a "lung-on-a-chip" tool for personalized drug discovery; (ii) replacement of mutant lung cells by wildtype lung cells in the living lung; and (iii) development of bio-artificial lungs. It is hoped that this review will give the reader a roadmap through the most complicated of the obstacles, and foster a guardedly optimistic view of how some of the remaining obstacles might one day be overcome.
Collapse
Affiliation(s)
| | - Harvey B Pollard
- Department of Cell Biology and Genetics, Uniformed Services University School of Medicine-America's Medical School, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
26
|
Lin Z, Thorenoor N, Wu R, DiAngelo SL, Ye M, Thomas NJ, Liao X, Lin TR, Warren S, Floros J. Genetic Association of Pulmonary Surfactant Protein Genes, SFTPA1, SFTPA2, SFTPB, SFTPC, and SFTPD With Cystic Fibrosis. Front Immunol 2018; 9:2256. [PMID: 30333828 PMCID: PMC6175982 DOI: 10.3389/fimmu.2018.02256] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/11/2018] [Indexed: 01/03/2023] Open
Abstract
Surfactant proteins (SP) are involved in surfactant function and innate immunity in the human lung. Both lung function and innate immunity are altered in CF, and altered SP levels and genetic association are observed in Cystic Fibrosis (CF). We hypothesized that single nucleotide polymorphisms (SNPs) within the SP genes associate with CF or severity subgroups, either through single SNP or via SNP-SNP interactions between two SNPs of a given gene (intragenic) and/or between two genes (intergenic). We genotyped a total of 17 SP SNPs from 72 case-trio pedigree (SFTPA1 (5), SFTPA2 (4), SFTPB (4), SFTPC (2), and SFTPD (2)), and identified SP SNP associations by applying quantitative genetic principles. The results showed (a) Two SNPs, SFTPB rs7316 (p = 0.0083) and SFTPC rs1124 (p = 0.0154), each associated with CF. (b) Three intragenic SNP-SNP interactions, SFTPB (rs2077079, rs3024798), and SFTPA1 (rs1136451, rs1059057 and rs4253527), associated with CF. (c) A total of 34 intergenic SNP-SNP interactions among the 4 SP genes to be associated with CF. (d) No SNP-SNP interaction was observed between SFTPA1 or SFTPA2 and SFTPD. (e) Equal number of SNP-SNP interactions were observed between SFTPB and SFTPA1/SFTPA2 (n = 7) and SP-B and SFTPD (n = 7). (f) SFTPC exhibited significant SNP-SNP interactions with SFTPA1/SFTPA2 (n = 11), SFTPB (n = 4) and SFTPD (n = 3). (g) A single SFTPB SNP was associated with mild CF after Bonferroni correction, and several intergenic interactions that are associated (p < 0.01) with either mild or moderate/severe CF were observed. These collectively indicate that complex SNP-SNP interactions of the SP genes may contribute to the pulmonary disease in CF patients. We speculate that SPs may serve as modifiers for the varied progression of pulmonary disease in CF and/or its severity.
Collapse
Affiliation(s)
- Zhenwu Lin
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Nithyananda Thorenoor
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Rongling Wu
- Public Health Science, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Susan L. DiAngelo
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Meixia Ye
- Public Health Science, College of Medicine, Pennsylvania State University, Hershey, PA, United States
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Neal J. Thomas
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Xiaojie Liao
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Tony R. Lin
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Stuart Warren
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
- Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
27
|
O'Neal WK, Knowles MR. Cystic Fibrosis Disease Modifiers: Complex Genetics Defines the Phenotypic Diversity in a Monogenic Disease. Annu Rev Genomics Hum Genet 2018; 19:201-222. [DOI: 10.1146/annurev-genom-083117-021329] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In many respects, genetic studies in cystic fibrosis (CF) serve as a paradigm for a human Mendelian genetic success story. From recognition of the condition as a heritable pathological entity to implementation of personalized treatments based on genetic findings, this multistep pathway of progress has focused on the genetic underpinnings of CF clinical disease. Along this path was the recognition that not all CFTR gene mutations produce the same disease and the recognition of the complex, multifactorial nature of CF genotype–phenotype relationships. The non- CFTR genetic components (gene modifiers) that contribute to variation in phenotype are the focus of this review. A multifaceted approach involving candidate gene studies, genome-wide association studies, and gene expression studies has revealed significant gene modifiers for multiple CF phenotypes. The bold challenges for the future are to integrate the findings into our understanding of CF pathogenesis and to use the knowledge to develop novel therapies.
Collapse
Affiliation(s)
- Wanda K. O'Neal
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;,
| | - Michael R. Knowles
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;,
| |
Collapse
|
28
|
FAM13A is a modifier gene of cystic fibrosis lung phenotype regulating rhoa activity, actin cytoskeleton dynamics and epithelial-mesenchymal transition. J Cyst Fibros 2017; 17:190-203. [PMID: 29239766 DOI: 10.1016/j.jcf.2017.11.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/04/2017] [Accepted: 11/10/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) lung disease severity is highly variable and dependent on several factors including genetic modifiers. Family with sequence similarity 13 member A (FAM13A) has been previously associated with lung function in the general population as well as in several chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), we examined whether FAM13A is a modifier gene of CF lung phenotype. We also studied how FAM13A may contribute to the physiopathological mechanisms associated with CF. METHODS We investigated the association of FAM13A with lung function in CF French patients (n=1222) by SNP-wise analysis and Versatile Gene Based Association Study. We also analyzed the consequences of FAM13A knockdown in A549 cells and primary bronchial epithelial cells from CF patients. RESULTS We found that FAM13A is associated with lung function in CF patients. Utilizing lung epithelial A549 cells and primary human bronchial epithelial cells from CF patients we observed that IL-1β and TGFβ reduced FAM13A expression. Knockdown of FAM13A was associated with increased RhoA activity, induction of F-actin stress fibers and regulation of epithelial-mesenchymal transition markers such as E-cadherin, α-smooth muscle actin and vimentin. CONCLUSION Our data show that FAM13A is a modifier gene of CF lung phenotype regulating RhoA activity, actin cytoskeleton dynamics and epithelial-mesenchymal transition.
Collapse
|
29
|
Krick S, Baumlin N, Aller SP, Aguiar C, Grabner A, Sailland J, Mendes E, Schmid A, Qi L, David NV, Geraghty P, King G, Birket SE, Rowe SM, Faul C, Salathe M. Klotho Inhibits Interleukin-8 Secretion from Cystic Fibrosis Airway Epithelia. Sci Rep 2017; 7:14388. [PMID: 29085059 PMCID: PMC5662572 DOI: 10.1038/s41598-017-14811-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/16/2017] [Indexed: 01/08/2023] Open
Abstract
Chronic inflammation is a hallmark of cystic fibrosis (CF) and associated with increased production of transforming growth factor (TGF) β and interleukin (IL)-8. α-klotho (KL), a transmembrane or soluble protein, functions as a co-receptor for Fibroblast Growth Factor (FGF) 23, a known pro-inflammatory, prognostic marker in chronic kidney disease. KL is downregulated in airways from COPD patients. We hypothesized that both KL and FGF23 signaling modulate TGF β-induced IL-8 secretion in CF bronchial epithelia. Thus, FGF23 and soluble KL levels were measured in plasma from 48 CF patients and in primary CF bronchial epithelial cells (CF-HBEC). CF patients showed increased FGF23 plasma levels, but KL levels were not different. In CF-HBEC, TGF-β increased KL secretion and upregulated FGF receptor (FGFR) 1. Despite increases in KL, TGF-β also increased IL-8 secretion via activation of FGFR1 and Smad 3 signaling. However, KL excess via overexpression or supplementation decreased IL-8 secretion by inhibiting Smad 3 phosphorylation. Here, we identify a novel signaling pathway contributing to IL-8 secretion in the CF bronchial epithelium with KL functioning as an endocrine and local anti-inflammatory mediator that antagonizes pro-inflammatory actions of FGF23 and TGF-β.
Collapse
Affiliation(s)
- Stefanie Krick
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Nathalie Baumlin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Sheyla Paredes Aller
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Carolina Aguiar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Juliette Sailland
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Eliana Mendes
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Andreas Schmid
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Lixin Qi
- Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nicolae V David
- Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Patrick Geraghty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Gwendalyn King
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Susan E Birket
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Steven M Rowe
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Christian Faul
- Division of Nephrology and Hypertension, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Matthias Salathe
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| |
Collapse
|
30
|
Abstract
Cystic fibrosis (CF) is the most frequent lethal genetic disorder among Caucasians, but is considered to be a very rare disease in Chinese population. Here, we present an 11-year-old Chinese CF patient with disseminated bronchiectasis and salty sweat, for whom exon sequencing followed by multiplex ligation-dependent probe amplification analysis of the CFTR gene was applied for mutation screening. A homozygous deletion involving exon 20 of CFTR was observed in the patient's genome. Molecular characterization of the breakpoints indicated that both alleles of this locus had an identical novel complex rearrangement (c.3140-454_c.3367+249del931ins13, p.R1048_G1123del), leading to an in-frame removal of 76 amino acid residues in the second transmembrane domains of the CFTR protein. Although a same haplotype containing this complex rearrangement was observed on both of the maternal and paternal alleles, the parents denied any blood relationship as far as they know. Genome-wide homozygosity mapping was performed through SNP microarray and only a single homozygous interval of ~14.1 Mb at chromosome 7 containing the CFTR gene was observed, indicating the possible origin of the deletion from a common ancestor many generations ago. This study expands the mutation spectrum of CFTR in patients of Chinese origin and further emphasizes the necessity of MLPA analysis in mutation screening for CF patients.
Collapse
|
31
|
van Leeuwen J, Pons C, Boone C, Andrews BJ. Mechanisms of suppression: The wiring of genetic resilience. Bioessays 2017; 39. [PMID: 28582599 DOI: 10.1002/bies.201700042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recent analysis of genome sequences has identified individuals that are healthy despite carrying severe disease-associated mutations. A possible explanation is that these individuals carry a second genomic perturbation that can compensate for the detrimental effects of the disease allele, a phenomenon referred to as suppression. In model organisms, suppression interactions are generally divided into two classes: genomic suppressors which are secondary mutations in the genome that bypass a mutant phenotype, and dosage suppression interactions in which overexpression of a suppressor gene rescues a mutant phenotype. Here, we describe the general properties of genomic and dosage suppression, with an emphasis on the budding yeast. We propose that suppression interactions between genetic variants are likely relevant for determining the penetrance of human traits. Consequently, an understanding of suppression mechanisms may guide the discovery of protective variants in healthy individuals that carry disease alleles, which could direct the rational design of new therapeutics.
Collapse
Affiliation(s)
- Jolanda van Leeuwen
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Carles Pons
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute for Science and Technology, Barcelona, Catalonia, Spain
| | - Charles Boone
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Brenda J Andrews
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
|
33
|
Lucarelli M. New era of cystic fibrosis: Full mutational analysis and personalized therapy. World J Med Genet 2017; 7:1-9. [DOI: 10.5496/wjmg.v7.i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/19/2016] [Accepted: 02/21/2017] [Indexed: 02/07/2023] Open
Abstract
Despite its apparently simple genetics, cystic fibrosis (CF) is a rather complex genetic disease. A lot of variability in the steps of the path from the cystic fibrosis transmembrane conductance regulator (CFTR) gene to the clinical manifestations originates an uncertain genotype - phenotype relationship. A major determinant of this uncertainty is the incomplete knowledge of the CFTR mutated genotypes, due to the high number of CFTR mutations and to the higher number of their combinations in trans and in cis. Also the very limited knowledge of functional effects of CFTR mutated alleles severely impairs our diagnostic and prognostic ability. The final phenotypic modulation exerted by CFTR modifier genes and interactome further complicates the framework. The next generation sequencing approach is a rapid, low-cost and high-throughput tool that allows a near complete structural characterization of CFTR mutated genotypes, as well as of genotypes of several other genes cooperating to the final CF clinical manifestations. This powerful method perfectly complements the new personalized therapeutic approach for CF. Drugs active on specific CFTR mutational classes are already available for CF patients or are in phase 3 trials. A complete genetic characterization has been becoming crucial for a correct personalized therapy. However, the need of a functional classification of each CFTR mutation potently arises. Future big efforts towards an ever more detailed knowledge of both structural and functional CFTR defects, coupled to parallel personalized therapeutic interventions decisive for CF cure can be foreseen.
Collapse
Affiliation(s)
- Marco Lucarelli
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00161 Rome, Italy
| |
Collapse
|
34
|
Magalhães M, Rivals I, Claustres M, Varilh J, Thomasset M, Bergougnoux A, Mely L, Leroy S, Corvol H, Guillot L, Murris M, Beyne E, Caimmi D, Vachier I, Chiron R, De Sario A. DNA methylation at modifier genes of lung disease severity is altered in cystic fibrosis. Clin Epigenetics 2017; 9:19. [PMID: 28289476 PMCID: PMC5310067 DOI: 10.1186/s13148-016-0300-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/08/2016] [Indexed: 12/11/2022] Open
Abstract
Background Lung disease progression is variable among cystic fibrosis (CF) patients and depends on DNA mutations in the CFTR gene, polymorphic variations in disease modifier genes, and environmental exposure. The contribution of genetic factors has been extensively investigated, whereas the mechanism whereby environmental factors modulate the lung disease is unknown. In this project, we hypothesized that (i) reiterative stress alters the epigenome in CF-affected tissues and (ii) DNA methylation variations at disease modifier genes modulate the lung function in CF patients. Results We profiled DNA methylation at CFTR, the disease-causing gene, and at 13 lung modifier genes in nasal epithelial cells and whole blood samples from 48 CF patients and 24 healthy controls. CF patients homozygous for the p.Phe508del mutation and ≥18-year-old were stratified according to the lung disease severity. DNA methylation was measured by bisulfite and next-generation sequencing. The DNA methylation profile allowed us to correctly classify 75% of the subjects, thus providing a CF-specific molecular signature. Moreover, in CF patients, DNA methylation at specific genes was highly correlated in the same tissue sample. We suggest that gene methylation in CF cells may be co-regulated by disease-specific trans-factors. Three genes were differentially methylated in CF patients compared with controls and/or in groups of pulmonary severity: HMOX1 and GSTM3 in nasal epithelial samples; HMOX1 and EDNRA in blood samples. The association between pulmonary severity and DNA methylation at EDNRA was confirmed in blood samples from an independent set of CF patients. Also, lower DNA methylation levels at GSTM3 were associated with the GSTM3*B allele, a polymorphic 3-bp deletion that has a protective effect in cystic fibrosis. Conclusions DNA methylation levels are altered in nasal epithelial and blood cell samples from CF patients. Analysis of CFTR and 13 lung disease modifier genes shows DNA methylation changes of small magnitude: some of them are a consequence of the disease; other changes may result in small expression variations that collectively modulate the lung disease severity. Electronic supplementary material The online version of this article (doi:10.1186/s13148-016-0300-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Milena Magalhães
- Laboratoire de Génétique de Maladies Rares, EA7402 Montpellier University, Montpellier, France
| | - Isabelle Rivals
- Equipe de Statistique Appliquée-ESPCI ParisTech, PSL Research University-UMRS1158, Paris, France
| | - Mireille Claustres
- Laboratoire de Génétique de Maladies Rares, EA7402 Montpellier University, Montpellier, France.,Laboratoire de Génétique Moléculaire-CHU Montpellier, Montpellier, France
| | - Jessica Varilh
- Laboratoire de Génétique de Maladies Rares, EA7402 Montpellier University, Montpellier, France.,Laboratoire de Génétique Moléculaire-CHU Montpellier, Montpellier, France
| | - Mélodie Thomasset
- Laboratoire de Génétique de Maladies Rares, EA7402 Montpellier University, Montpellier, France
| | - Anne Bergougnoux
- Laboratoire de Génétique de Maladies Rares, EA7402 Montpellier University, Montpellier, France.,Laboratoire de Génétique Moléculaire-CHU Montpellier, Montpellier, France
| | - Laurent Mely
- CRCM, Renée Sabran Hospital-CHU Lyon, Hyères, France
| | | | - Harriet Corvol
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U938-CRSA, Paris, France.,APHP, Trousseau Hospital, Paris, France
| | - Loïc Guillot
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,INSERM U938-CRSA, Paris, France
| | | | - Emmanuelle Beyne
- Laboratoire de Génétique de Maladies Rares, EA7402 Montpellier University, Montpellier, France.,Laboratoire de Génétique Moléculaire-CHU Montpellier, Montpellier, France
| | - Davide Caimmi
- CRCM, Arnaud de Villeneuve Hospital-CHU Montpellier, Montpellier, France
| | - Isabelle Vachier
- CRCM, Arnaud de Villeneuve Hospital-CHU Montpellier, Montpellier, France
| | - Raphaël Chiron
- CRCM, Arnaud de Villeneuve Hospital-CHU Montpellier, Montpellier, France
| | - Albertina De Sario
- Laboratoire de Génétique de Maladies Rares, EA7402 Montpellier University, Montpellier, France
| |
Collapse
|
35
|
Abstract
Infection is one of the leading causes of human mortality and morbidity. Exposure to microbial agents is obviously required. However, also non-microbial environmental and host factors play a key role in the onset, development and outcome of infectious disease, resulting in large of clinical variability between individuals in a population infected with the same microbe. Controlled and standardized investigations of the genetics of susceptibility to infectious disease are almost impossible to perform in humans whereas mouse models allow application of powerful genomic techniques to identify and validate causative genes underlying human diseases with complex etiologies. Most of current animal models used in complex traits diseases genetic mapping have limited genetic diversity. This limitation impedes the ability to create incorporated network using genetic interactions, epigenetics, environmental factors, microbiota, and other phenotypes. A novel mouse genetic reference population for high-resolution mapping and subsequently identifying genes underlying the QTL, namely the Collaborative Cross (CC) mouse genetic reference population (GRP) was recently developed. In this chapter, we discuss a variety of approaches using CC mice for mapping genes underlying quantitative trait loci (QTL) to dissect the host response to polygenic traits, including infectious disease caused by bacterial agents and its toxins.
Collapse
|
36
|
De Boeck K, Amaral MD. Progress in therapies for cystic fibrosis. THE LANCET RESPIRATORY MEDICINE 2016; 4:662-674. [DOI: 10.1016/s2213-2600(16)00023-0] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 12/19/2022]
|
37
|
Dauletbaev N, Herscovitch K, Das M, Chen H, Bernier J, Matouk E, Bérubé J, Rousseau S, Lands LC. Down-regulation of IL-8 by high-dose vitamin D is specific to hyperinflammatory macrophages and involves mechanisms beyond up-regulation of DUSP1. Br J Pharmacol 2016; 172:4757-71. [PMID: 26178144 DOI: 10.1111/bph.13249] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 06/17/2015] [Accepted: 06/29/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE There is current interest in vitamin D as a potential anti-inflammatory treatment for chronic inflammatory lung disease, including cystic fibrosis (CF). Vitamin D transcriptionally up-regulates the anti-inflammatory gene DUSP1, which partly controls production of the inflammatory chemokine IL-8. IL-8 is overabundant in CF airways, potentially due to hyperinflammatory responses of CF macrophages. We tested the ability of vitamin D metabolites to down-regulate IL-8 production in CF macrophages. EXPERIMENTAL APPROACH CF and healthy monocyte-derived macrophages (MDM) were treated with two vitamin D metabolites, 25-hydroxyvitamin D3 (25OHD3 ) and 1,25-dihydroxyvitamin D3 (1,25(OH)2 D3 ), or paricalcitol, synthetic analogue of 1,25(OH)2 D3 . 25OHD3 was tested at doses of 25-150 nM, whereas 1,25(OH)2 D3 and paricalcitol at doses of up to 100 nM. IL-8 was stimulated by bacterial virulence factors. As potential anti-inflammatory mechanism of vitamin D metabolites, we assessed up-regulation of DUSP1. KEY RESULTS MDM from patients with CF and some healthy donors showed excessive production of stimulated IL-8, highlighting their hyperinflammatory phenotype. Vitamin D metabolites down-regulated stimulated IL-8 only in those hyperinflammatory MDM, and only when used at high doses (>100 nM for 25OHD3 , or >1 nM for 1,25(OH)2 D3 and paricalcitol). The magnitude of IL-8 down-regulation by vitamin D metabolites or paricalcitol was moderate (∼30% vs. >70% by low-dose dexamethasone). Transcriptional up-regulation of DUSP1 by vitamin D metabolites was seen in all tested MDM, regardless of IL-8 down-regulation. CONCLUSIONS AND IMPLICATIONS Vitamin D metabolites and their analogues moderately down-regulate IL-8 in hyperinflammatory macrophages, including those from CF. This down-regulation appears to go through DUSP1-independent mechanisms.
Collapse
Affiliation(s)
- N Dauletbaev
- Translational Research in Respiratory Diseases, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - K Herscovitch
- Translational Research in Respiratory Diseases, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - M Das
- Translational Research in Respiratory Diseases, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - H Chen
- Translational Research in Respiratory Diseases, The Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - J Bernier
- Division of Respiratory Medicine, Montreal Chest Institute, Montreal, QC, Canada
| | - E Matouk
- Division of Respiratory Medicine, Montreal Chest Institute, Montreal, QC, Canada
| | - J Bérubé
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - S Rousseau
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - L C Lands
- Translational Research in Respiratory Diseases, The Research Institute of McGill University Health Centre, Montreal, QC, Canada.,Division of Respiratory Medicine, Montreal Children's Hospital, Montreal, QC, Canada
| |
Collapse
|
38
|
Ralhan A, Laval J, Lelis F, Ballbach M, Grund C, Hector A, Hartl D. Current Concepts and Controversies in Innate Immunity of Cystic Fibrosis Lung Disease. J Innate Immun 2016; 8:531-540. [PMID: 27362371 PMCID: PMC6738757 DOI: 10.1159/000446840] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/17/2016] [Accepted: 05/17/2016] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) lung disease is characterized by chronic infection and inflammation. The inflammatory response in CF is dominated by the activation of the innate immune system. Bacteria and fungi represent the key pathogens chronically colonizing the CF airways. In response, innate immune pattern recognition receptors, expressed by airway epithelial and myeloid cells, sense the microbial threat and release chemoattractants to recruit large numbers of neutrophils into CF airways. However, neutrophils fail to efficiently clear the invading pathogens, but instead release harmful proteases and oxidants and finally cause tissue injury. Here, we summarize and discuss current concepts and controversies in the field of innate immunity in CF lung disease, facing the ongoing questions of whether inflammation is good or bad in CF and how innate immune mechanisms could be harnessed therapeutically.
Collapse
Affiliation(s)
- Anjali Ralhan
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Julie Laval
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Felipe Lelis
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Marlene Ballbach
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Charlotte Grund
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Andreas Hector
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Dominik Hartl
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
- Immunology, Inflammation and Infectious Diseases (I3) Discovery and Translational Area, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
39
|
De Simone M, Spagnuolo L, Lorè NI, Cigana C, De Fino I, Broman KW, Iraqi FA, Bragonzi A. Mapping genetic determinants of host susceptibility to Pseudomonas aeruginosa lung infection in mice. BMC Genomics 2016; 17:351. [PMID: 27169516 PMCID: PMC4866434 DOI: 10.1186/s12864-016-2676-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/28/2016] [Indexed: 12/16/2022] Open
Abstract
Background P. aeruginosa is one of the top three causes of opportunistic human bacterial infections. The remarkable variability in the clinical outcomes of this infection is thought to be associated with genetic predisposition. However, the genes underlying host susceptibility to P. aeruginosa infection are still largely unknown. Results As a step towards mapping these genes, we applied a genome wide linkage analysis approach to a mouse model. A large F2 intercross population, obtained by mating P. aeruginosa-resistant C3H/HeOuJ, and susceptible A/J mice, was used for quantitative trait locus (QTL) mapping. The F2 progenies were challenged with a P. aeruginosa clinical strain and monitored for the survival time up to 7 days post-infection, as a disease phenotype associated trait. Selected phenotypic extremes of the F2 distribution were genotyped with high-density single nucleotide polymorphic (SNP) markers, and subsequently QTL analysis was performed. A significant locus was mapped on chromosome 6 and was named P. aeruginosa infection resistance locus 1 (Pairl1). The most promising candidate genes, including Dok1, Tacr1, Cd207, Clec4f, Gp9, Gata2, Foxp1, are related to pathogen sensing, neutrophils and macrophages recruitment and inflammatory processes. Conclusions We propose a set of genes involved in the pathogenesis of P. aeruginosa infection that may be explored to complement human studies. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2676-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maura De Simone
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenza Spagnuolo
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Ivan Lorè
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Cigana
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ida De Fino
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Karl W Broman
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Fuad A Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, 69978, Tel Aviv, Israel
| | - Alessandra Bragonzi
- Infection and Cystic Fibrosis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
40
|
Cigana C, Lorè NI, Riva C, De Fino I, Spagnuolo L, Sipione B, Rossi G, Nonis A, Cabrini G, Bragonzi A. Tracking the immunopathological response to Pseudomonas aeruginosa during respiratory infections. Sci Rep 2016; 6:21465. [PMID: 26883959 PMCID: PMC4756310 DOI: 10.1038/srep21465] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/19/2016] [Indexed: 12/18/2022] Open
Abstract
Repeated cycles of infections, caused mainly by Pseudomonas aeruginosa, combined with a robust host immune response and tissue injury, determine the course and outcome of cystic fibrosis (CF) lung disease. As the disease progresses, P. aeruginosa adapts to the host modifying dramatically its phenotype; however, it remains unclear whether and how bacterial adaptive variants and their persistence influence the pathogenesis and disease development. Using in vitro and murine models of infection, we showed that P. aeruginosa CF-adaptive variants shaped the innate immune response favoring their persistence. Next, we refined a murine model of chronic pneumonia extending P. aeruginosa infection up to three months. In this model, including CFTR-deficient mice, we unveil that the P. aeruginosa persistence lead to CF hallmarks of airway remodelling and fibrosis, including epithelial hyperplasia and structure degeneration, goblet cell metaplasia, collagen deposition, elastin degradation and several additional markers of tissue damage. This murine model of P. aeruginosa chronic infection, reproducing CF lung pathology, will be instrumental to identify novel molecular targets and test newly tailored molecules inhibiting chronic inflammation and tissue damage processes in pre-clinical studies.
Collapse
Affiliation(s)
- Cristina Cigana
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Nicola Ivan Lorè
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Camilla Riva
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Ida De Fino
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Lorenza Spagnuolo
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Barbara Sipione
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, Italy
| | - Alessandro Nonis
- University Center for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cabrini
- Department of Pathology and Diagnostics, University Hospital, Verona, Italy
| | - Alessandra Bragonzi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
41
|
Corvol H, Thompson KE, Tabary O, le Rouzic P, Guillot L. Translating the genetics of cystic fibrosis to personalized medicine. Transl Res 2016; 168:40-49. [PMID: 25940043 DOI: 10.1016/j.trsl.2015.04.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 01/06/2023]
Abstract
Cystic fibrosis (CF) is the most common life-threatening recessive genetic disease in the Caucasian population. This multiorgan disease is caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR) protein, a chloride channel recognized as regulating several apical ion channels. The gene mutations result either in the lack of the protein at the apical surface or in an improperly functioning protein. Morbidity and mortality because of the mutation of CFTR are mainly attributable to lung disease resulting from chronic infection and inflammation. Since its discovery as the causative gene in 1989, much progress has been achieved not only in clinical genetics but also in basic science studies. Recently, combinations of these efforts have been successfully translated into development and availability for patients of new therapies targeting specific CFTR mutations to correct the CFTR at the protein level. Current technologies such as next gene sequencing and novel genomic editing tools may offer new strategies to identify new CFTR variants and modifier genes, and to correct CFTR to pursue personalized medicine, which is already developed in some patient subsets. Personalized medicine or P4 medicine ("personalized," "predictive," "preventive," and "participatory") is currently booming for CF. The various current and future challenges of personalized medicine as they apply to the issues faced in CF are discussed in this review.
Collapse
Affiliation(s)
- Harriet Corvol
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France; Pneumologie pédiatrique, APHP, Hôpital Trousseau, Paris, France
| | - Kristin E Thompson
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - Olivier Tabary
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - Philippe le Rouzic
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - Loïc Guillot
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France; Sorbonne Universités, UPMC University Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France.
| |
Collapse
|
42
|
Dewulf J, Vermeulen F, Wanyama S, Thomas M, Proesmans M, Dupont L, De Boeck K. Treatment burden in patients with at least one class IV or V CFTR mutation. Pediatr Pulmonol 2015; 50:1230-6. [PMID: 26540286 DOI: 10.1002/ppul.23313] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/04/2015] [Accepted: 08/28/2015] [Indexed: 12/27/2022]
Abstract
CFTR mutations are grouped according to disease-causing mechanism. Several studies demonstrated that patients having at least one mutation of class IV/V, present with a milder phenotype, but little is known about their relative treatment burden. We compared treatment burden between patients with two class I, II, or III mutations and patients with at least one mutation of class IV/V in the 2010 database of the Belgian CF Registry. We calculated a "Treatment Burden Index" (TBI) by assigning long term therapies to categories low, medium and high intensity, for differential weighing in the total score. There were 779 patients with two known class I/II/III mutations and 94 patients with at least one class IV/V mutation. Compared to class I/II/III, class IV/V patients had a lower median number of clinic visits (4 vs. 5; P < 0.001), a lower risk of hospitalization (24.7% vs. 50.8%; P < 0.001) and intravenous antibiotic treatment (23.5% vs. 46.0%; P < 0.001) and a lower median TBI (6 vs. 9; P < 0.001). These differences remained significant when only class IV/V patients with pancreatic insufficiency (n = 31) were considered. This study clearly demonstrates the significantly lower treatment burden in patients with CF and at least one class IV/V mutation compared to patients with two class I/II/III mutations and contributes to providing better individual counseling at time of diagnosis.
Collapse
Affiliation(s)
- Jonas Dewulf
- Department of Pediatrics, University Hospitals of Leuven, Belgium
| | | | - Simeon Wanyama
- Scientific Institute of Public Health, on behalf of the Belgian CF Registry, Brussels, Belgium
| | - Muriel Thomas
- Scientific Institute of Public Health, on behalf of the Belgian CF Registry, Brussels, Belgium
| | | | - Lieven Dupont
- Department of Pulmonology, University Hospitals of Leuven, Belgium
| | - Kris De Boeck
- Department of Pediatrics, University Hospitals of Leuven, Belgium
| |
Collapse
|
43
|
Shteinberg M, Rivlin J, Gur M, Konopnicki M, Stein N, Tunney MM, Elborn JS, Downey DG, Johnston E, Shalom H, Levy A. Lack of Association Between Haptoglobin Phenotype and Cystic Fibrosis Outcomes. Lung 2015; 193:1017-21. [PMID: 26370551 DOI: 10.1007/s00408-015-9801-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/07/2015] [Indexed: 11/25/2022]
Abstract
Haptoglobin (Hp), a heme-Iron chelator, has different isoforms which are associated with variable tendency toward infections: Hp 1-1, Hp 2-1, and Hp 2-2. Cystic fibrosis (CF) outcomes are variable and influenced by genetic and environmental factors. The aim of this study was to determine whether Hp phenotype influenced disease severity in CF. One hundred forty-two CF patients from two centers were analyzed for Haptoglobin phenotype using gel electrophoresis of hemoglobin enriched serum. Clinical and microbiological data including bacterial colonization status, lung function, presence of CF-related diabetes and liver disease, rate of exacerbation, and mortality were compared between Hp phenotype groups. We found a trend toward less mucoid PA among Hp 2-2 (20.4 %) compared with Hp 1-1 and Hp 2-1 individuals (33.3 %), p = 0.317. Hp 2-2 individuals also had less antibiotic courses, and lower inflammatory markers without statistical significance. Haptoglobin phenotype is unlikely to be an important modifier of CF phenotype.
Collapse
Affiliation(s)
- Michal Shteinberg
- CF Center, Carmel Medical Center, Haifa, Israel. .,Queens University Belfast, Belfast, UK. .,The B. Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel. .,Pulnonary Institute, Carmel Medical Center, 7 Michal St., 3463209, Haifa, Israel.
| | | | - Michal Gur
- CF Center, Carmel Medical Center, Haifa, Israel
| | | | - Nili Stein
- CF Center, Carmel Medical Center, Haifa, Israel
| | | | | | | | | | - Hadar Shalom
- The B. Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Andrew Levy
- The B. Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
44
|
Manzanares D, Krick S, Baumlin N, Dennis JS, Tyrrell J, Tarran R, Salathe M. Airway Surface Dehydration by Transforming Growth Factor β (TGF-β) in Cystic Fibrosis Is Due to Decreased Function of a Voltage-dependent Potassium Channel and Can Be Rescued by the Drug Pirfenidone. J Biol Chem 2015; 290:25710-6. [PMID: 26338706 DOI: 10.1074/jbc.m115.670885] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor β1 (TGF-β1) is not only elevated in airways of cystic fibrosis (CF) patients, whose airways are characterized by abnormal ion transport and mucociliary clearance, but TGF-β1 is also associated with worse clinical outcomes. Effective mucociliary clearance depends on adequate airway hydration, governed by ion transport. Apically expressed, large-conductance, Ca(2+)- and voltage-dependent K(+) (BK) channels play an important role in this process. In this study, TGF-β1 decreased airway surface liquid volume, ciliary beat frequency, and BK activity in fully differentiated CF bronchial epithelial cells by reducing mRNA expression of the BK γ subunit leucine-rich repeat-containing protein 26 (LRRC26) and its function. Although LRRC26 knockdown itself reduced BK activity, LRRC26 overexpression partially reversed TGF-β1-induced BK dysfunction. TGF-β1-induced airway surface liquid volume hyper-absorption was reversed by the BK opener mallotoxin and the clinically useful TGF-β signaling inhibitor pirfenidone. The latter increased BK activity via rescue of LRRC26. Therefore, we propose that TGF-β1-induced mucociliary dysfunction in CF airways is associated with BK inactivation related to a LRRC26 decrease and is amenable to treatment with clinically useful TGF-β1 inhibitors.
Collapse
Affiliation(s)
- Dahis Manzanares
- From the Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami, Miami, Florida 33136 and
| | - Stefanie Krick
- From the Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami, Miami, Florida 33136 and
| | - Nathalie Baumlin
- From the Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami, Miami, Florida 33136 and
| | - John S Dennis
- From the Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami, Miami, Florida 33136 and
| | - Jean Tyrrell
- Cystic Fibrosis Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Robert Tarran
- Cystic Fibrosis Center, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Matthias Salathe
- From the Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami, Miami, Florida 33136 and
| |
Collapse
|
45
|
Oueslati S, Hadj Fredj S, Dakhlaoui B, Othmani R, Siala H, Messaoud T. Association of TGFB1 -509C/T polymorphism gene with clinical variability in cystic fibrosis patients: A case-control study. ACTA ACUST UNITED AC 2015; 63:175-8. [PMID: 26277914 DOI: 10.1016/j.patbio.2015.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/10/2015] [Indexed: 01/18/2023]
Abstract
PURPOSE In this work, we are interested to study the implication of -509C/T polymorphism, located in the promoter region of TGFB1 (transforming growth factor β1), in the phenotypic variability of CF patients. PATIENTS AND METHODS The present study enrolled 111 CF patients and 100 healthy control subjects. The study of the -509C/T polymorphism was performed using PCR-RFLP method. RESULTS We found that patients carried non-F508del homozygous mutation with TT genotype was associated to lung symptoms (P=0.04). This association was not found in the sub-groups of patients with F508del at homozygous state P=0.145. No association was found between this polymorphism and the variability of digestive, pancreatic and ileus meconial symptoms. CONCLUSION On the basis of our results, the -509C/T polymorphism of the TGFB1 gene seems to be a modulator factor of cystic fibrosis.
Collapse
Affiliation(s)
- S Oueslati
- Biochemistry Laboratory (Research Laboratory Haemoglobinopathies and Cystic Fibrosis), Children's Hospital, Bab Saadoun Square, 1007 Tunis, Tunisia.
| | - S Hadj Fredj
- Biochemistry Laboratory (Research Laboratory Haemoglobinopathies and Cystic Fibrosis), Children's Hospital, Bab Saadoun Square, 1007 Tunis, Tunisia
| | - B Dakhlaoui
- Biochemistry Laboratory (Research Laboratory Haemoglobinopathies and Cystic Fibrosis), Children's Hospital, Bab Saadoun Square, 1007 Tunis, Tunisia
| | - R Othmani
- Biochemistry Laboratory (Research Laboratory Haemoglobinopathies and Cystic Fibrosis), Children's Hospital, Bab Saadoun Square, 1007 Tunis, Tunisia
| | - H Siala
- Biochemistry Laboratory (Research Laboratory Haemoglobinopathies and Cystic Fibrosis), Children's Hospital, Bab Saadoun Square, 1007 Tunis, Tunisia
| | - T Messaoud
- Biochemistry Laboratory (Research Laboratory Haemoglobinopathies and Cystic Fibrosis), Children's Hospital, Bab Saadoun Square, 1007 Tunis, Tunisia
| |
Collapse
|
46
|
Abstract
Despite decades of scientific and clinical research, pancreatic ductal adenocarcinoma (PDAC) remains a lethal malignancy. The clinical and pathologic features of PDAC, specifically the known environmental and genetic risk factors, are reviewed here with special emphasis on the hereditary pancreatic cancer (HPC) syndromes. For these latter conditions, strategies are described for their identification, for primary and secondary prevention in unaffected carriers, and for disease management in affected carriers. Nascent steps have been made toward personalized medicine based on the rational use of screening, tumor subtyping, and targeted therapies; these have been guided by growing knowledge of HPC syndromes in PDAC.
Collapse
Affiliation(s)
- Ashton A Connor
- Division of General Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Steven Gallinger
- Division of General Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Corvol H, Taytard J, Tabary O, Le Rouzic P, Guillot L, Clement A. Les enjeux de la médecine personnalisée appliquée à la mucoviscidose. Arch Pediatr 2015; 22:778-86. [DOI: 10.1016/j.arcped.2015.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/04/2015] [Accepted: 04/24/2015] [Indexed: 11/26/2022]
|
48
|
Esmaeili Dooki MR, Tabaripour R, Rahimi R, Akhavan-Niaki H. Mutation and new polymorphisms insight in introns 11 to 14a of CFTR gene of northern Iranian cystic fibrosis patients. Gene 2015; 564:193-196. [PMID: 25824381 DOI: 10.1016/j.gene.2015.03.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/18/2015] [Accepted: 03/23/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cystic fibrosis (CF) is the most common autosomal recessive disease in Caucasians, caused by mutation in cystic fibrosis transmembrane conductance regulator (CFTR). The type and distribution of mutations vary widely between different countries and ethnic groups. We therefore aimed to perform a comprehensive analysis of the CFTR gene in northern Iranian CF patients. METHODS Forty northern Iranian CF patients were analyzed for mutations in introns 11 to 14a of their CFTR genes, using sequencing and reverse dot blot methods. Five normal subjects were also analyzed as normal control. RESULTS One mutation and seven polymorphisms were identified. Of the eighty alleles studied, c.2043delG in exon 13 represented 12.5% of mutant alleles and was associated with two distinct haplotypes. rs1042077T>G, rs4148712delAT, rs4148711T>A and rs3808183 T>C with frequencies varying between 29.2% and 6.9% for the least common allele, as well as three new polymorphisms c.1680-224C>A (11.1%), c.2491-275T>G (14.1%) and c.2491-274C>G (35.9%) were detected. CONCLUSION These findings suggest a founder effect for c.2043delG in the Middle East and will assist in genetic counseling, prenatal diagnosis and future screening of CF in Iran.
Collapse
Affiliation(s)
| | | | | | - Haleh Akhavan-Niaki
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
49
|
Veit G, Avramescu RG, Perdomo D, Phuan PW, Bagdany M, Apaja PM, Borot F, Szollosi D, Wu YS, Finkbeiner WE, Hegedus T, Verkman AS, Lukacs GL. Some gating potentiators, including VX-770, diminish ΔF508-CFTR functional expression. Sci Transl Med 2015; 6:246ra97. [PMID: 25101887 DOI: 10.1126/scitranslmed.3008889] [Citation(s) in RCA: 255] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cystic fibrosis (CF) is caused by mutations in the CF transmembrane regulator (CFTR) that result in reduced anion conductance at the apical membrane of secretory epithelia. Treatment of CF patients carrying the G551D gating mutation with the potentiator VX-770 (ivacaftor) largely restores channel activity and has shown substantial clinical benefit. However, most CF patients carry the ΔF508 mutation, which impairs CFTR folding, processing, function, and stability. Studies in homozygous ΔF508 CF patients indicated little clinical benefit of monotherapy with the investigational corrector VX-809 (lumacaftor) or VX-770, whereas combination clinical trials show limited but significant improvements in lung function. We show that VX-770, as well as most other potentiators, reduces the correction efficacy of VX-809 and another investigational corrector, VX-661. To mimic the administration of VX-770 alone or in combination with VX-809, we examined its long-term effect in immortalized and primary human respiratory epithelia. VX-770 diminished the folding efficiency and the metabolic stability of ΔF508-CFTR at the endoplasmic reticulum (ER) and post-ER compartments, respectively, causing reduced cell surface ΔF508-CFTR density and function. VX-770-induced destabilization of ΔF508-CFTR was influenced by second-site suppressor mutations of the folding defect and was prevented by stabilization of the nucleotide-binding domain 1 (NBD1)-NBD2 interface. The reduced correction efficiency of ΔF508-CFTR, as well as of two other processing mutations in the presence of VX-770, suggests the need for further optimization of potentiators to maximize the clinical benefit of corrector-potentiator combination therapy in CF.
Collapse
Affiliation(s)
- Guido Veit
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Radu G Avramescu
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Doranda Perdomo
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Puay-Wah Phuan
- Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, CA 94143-0521, USA
| | - Miklos Bagdany
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Pirjo M Apaja
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Florence Borot
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Daniel Szollosi
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences, 1444 Budapest, Hungary. Department of Biophysics and Radiation Biology, Semmelweis University, 1444 Budapest P.O. Box 263, Hungary
| | - Yu-Sheng Wu
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Walter E Finkbeiner
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143-0511, USA
| | - Tamas Hegedus
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences, 1444 Budapest, Hungary. Department of Biophysics and Radiation Biology, Semmelweis University, 1444 Budapest P.O. Box 263, Hungary
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, CA 94143-0521, USA
| | - Gergely L Lukacs
- Department of Physiology, McGill University, Montréal, Quebec H3G 1Y6, Canada. Department of Biochemistry, McGill University, Montréal, Quebec H3G 1Y6, Canada. Groupe de Recherche Axé sur la Structure des Protéines (GRASP), McGill University, Montréal, Quebec H3G 1Y6, Canada.
| |
Collapse
|
50
|
Bell SC, De Boeck K, Amaral MD. New pharmacological approaches for cystic fibrosis: Promises, progress, pitfalls. Pharmacol Ther 2015; 145:19-34. [DOI: 10.1016/j.pharmthera.2014.06.005] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 12/17/2022]
|