201
|
Muilwijk D, de Poel E, van Mourik P, Suen SWF, Vonk AM, Brunsveld JE, Kruisselbrink E, Oppelaar H, Hagemeijer MC, Berkers G, de Winter-de Groot KM, Heida-Michel S, Jans SR, van Panhuis H, van der Eerden MM, van der Meer R, Roukema J, Dompeling E, Weersink EJM, Koppelman GH, Vries R, Zomer-van Ommen DD, Eijkemans MJC, van der Ent CK, Beekman JM. Forskolin-induced Organoid Swelling is Associated with Long-term CF Disease Progression. Eur Respir J 2022; 60:13993003.00508-2021. [PMID: 35086832 PMCID: PMC9386333 DOI: 10.1183/13993003.00508-2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022]
Abstract
Rationale Cystic fibrosis (CF) is a monogenic life-shortening disease associated with highly variable individual disease progression which is difficult to predict. Here we assessed the association of forskolin-induced swelling (FIS) of patient-derived organoids with long-term CF disease progression in multiple organs and compared FIS with the golden standard biomarker sweat chloride concentration (SCC). Methods We retrieved 9-year longitudinal clinical data from the Dutch CF Registry of 173 people with mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Individual CFTR function was defined by FIS, measured as the relative size increase of intestinal organoids after stimulation with 0.8 µM forskolin, quantified as area under the curve (AUC). We used linear mixed-effect models and multivariable logistic regression to estimate the association of FIS with long-term forced expiratory volume in 1 s % predicted (FEV1pp) decline and development of pancreatic insufficiency, CF-related liver disease and diabetes. Within these models, FIS was compared with SCC. Results FIS was strongly associated with longitudinal changes of lung function, with an estimated difference in annual FEV1pp decline of 0.32% (95% CI 0.11–0.54%; p=0.004) per 1000-point change in AUC. Moreover, increasing FIS levels were associated with lower odds of developing pancreatic insufficiency (adjusted OR 0.18, 95% CI 0.07–0.46; p<0.001), CF-related liver disease (adjusted OR 0.18, 95% CI 0.06–0.54; p=0.002) and diabetes (adjusted OR 0.34, 95% CI 0.12–0.97; p=0.044). These associations were absent for SCC. Conclusion This study exemplifies the prognostic value of a patient-derived organoid-based biomarker within a clinical setting, which is especially important for people carrying rare CFTR mutations with unclear clinical consequences. Forskolin-induced swelling of patient-derived intestinal organoids is associated with long-term cystic fibrosis disease progression, expressed as FEV1pp decline and development of pancreatic insufficiency, CF-related liver disease and CF-related diabeteshttps://bit.ly/3tjjJzU
Collapse
Affiliation(s)
- Danya Muilwijk
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,These authors contributed equally to this work
| | - Eyleen de Poel
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands.,These authors contributed equally to this work
| | - Peter van Mourik
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Sylvia W F Suen
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Annelotte M Vonk
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Jesse E Brunsveld
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Evelien Kruisselbrink
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Hugo Oppelaar
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Marne C Hagemeijer
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands.,Current affiliation: Center for Lysosomal and Metabolic Diseases, Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gitte Berkers
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Karin M de Winter-de Groot
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Sabine Heida-Michel
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Stephan R Jans
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Hannah van Panhuis
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Menno M van der Eerden
- Department of Pulmonology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | - Jolt Roukema
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Edward Dompeling
- Maastricht University Medical Center, Maastricht, The Netherlands
| | - Els J M Weersink
- Amsterdam University Medical Center, location AMC, Amsterdam, The Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Robert Vries
- Hubrecht Organoid Technology (HUB), Utrecht, The Netherlands
| | | | - Marinus J C Eijkemans
- Department of Data Science and Biostatistics, Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Cornelis K van der Ent
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands.,These authors contributed equally to this work and are both corresponding authors
| | - Jeffrey M Beekman
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, Utrecht, The Netherlands .,Regenerative Medicine Utrecht, University Medical Center, Utrecht University, Utrecht, The Netherlands.,These authors contributed equally to this work and are both corresponding authors
| |
Collapse
|
202
|
Tam RY, van Dorst JM, McKay I, Coffey M, Ooi CY. Intestinal Inflammation and Alterations in the Gut Microbiota in Cystic Fibrosis: A Review of the Current Evidence, Pathophysiology and Future Directions. J Clin Med 2022; 11:649. [PMID: 35160099 PMCID: PMC8836727 DOI: 10.3390/jcm11030649] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-limiting autosomal recessive multisystem disease. While its burden of morbidity and mortality is classically associated with pulmonary disease, CF also profoundly affects the gastrointestinal (GI) tract. Chronic low-grade inflammation and alterations to the gut microbiota are hallmarks of the CF intestine. The etiology of these manifestations is likely multifactorial, resulting from cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction, a high-fat CF diet, and the use of antibiotics. There may also be a bidirectional pathophysiological link between intestinal inflammation and changes to the gut microbiome. Additionally, a growing body of evidence suggests that these GI manifestations may have significant clinical associations with growth and nutrition, quality of life, and respiratory function in CF. As such, the potential utility of GI therapies and long-term GI outcomes are areas of interest in CF. Further research involving microbial modulation and multi-omics techniques may reveal novel insights. This article provides an overview of the current evidence, pathophysiology, and future research and therapeutic considerations pertaining to intestinal inflammation and alterations in the gut microbiota in CF.
Collapse
Affiliation(s)
- Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Isabelle McKay
- Wagga Wagga Base Hospital, Wagga Wagga, NSW 2650, Australia;
| | - Michael Coffey
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| | - Chee Y. Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| |
Collapse
|
203
|
A Hyperglycemic Microenvironment Inhibits Tendon-to-Bone Healing through the let-7b-5p/CFTR Pathway. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8268067. [PMID: 35126637 PMCID: PMC8813224 DOI: 10.1155/2022/8268067] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022]
Abstract
Background Tendon-to-bone healing is a difficult process in treatment of rotator cuff tear (RCT). In addition, diabetes is an important risk factor for poor tendon-to-bone healing. Therefore, we investigated the specific mechanisms through which diabetes affects tendon-to-bone healing by regulating the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). Methods Tendon-derived stem cells (TDSCs) were extracted from rats after which their proliferative capacities were evaluated by the MTT assay. The expression levels of CFTR and tendon-related markers were determined by qRT-PCR. Then, bioinformatics analyses and dual luciferase reporter gene assays were used to identify miRNAs with the ability to bind CFTR mRNA. Finally, CFTR was overexpressed in TDSCs to validate the specific mechanisms through which the high glucose microenvironment inhibits tendon-to-bone healing. Results The high glucose microenvironment downregulated mRNA expression levels of tendon-related markers and CFTR in TDSCs cultured with different glucose concentrations. Additionally, bioinformatics analyses revealed that let-7b-5p may be regulated by the high glucose microenvironment and can regulate CFTR levels. Moreover, a dual luciferase reporter gene assay was used to confirm that let-7b-5p targets and binds CFTR mRNA. Additional experiments also confirmed that overexpressed CFTR effectively reversed the negative effects of the hyperglycaemic microenvironment and upregulation of let-7b-5p on TDSC proliferation and differentiation. These findings imply that the hyperglycemic microenvironment inhibits CFTR transcription and, consequently, proliferation and differentiation of TDSCs in vitro by upregulating let-7b-5p. Conclusions A hyperglycemic microenvironment inhibits TDSC proliferation in vitro via the let-7b-5p/CFTR pathway, and this is a potential mechanism in diabetes-induced poor tendon-to-bone healing.
Collapse
|
204
|
Shah VS, Chivukula RR, Lin B, Waghray A, Rajagopal J. Cystic Fibrosis and the Cells of the Airway Epithelium: What Are Ionocytes and What Do They Do? ANNUAL REVIEW OF PATHOLOGY 2022; 17:23-46. [PMID: 34437820 PMCID: PMC10837786 DOI: 10.1146/annurev-pathol-042420-094031] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cystic fibrosis (CF) is caused by defects in an anion channel, the cystic fibrosis transmembrane conductance regulator (CFTR). Recently, a new airway epithelial cell type has been discovered and dubbed the pulmonary ionocyte. Unexpectedly, these ionocytes express higher levels of CFTR than any other airway epithelial cell type. However, ionocytes are not the sole CFTR-expressing airway epithelial cells, and CF-associated disease genes are in fact expressed in multiple airway epithelial cell types. The experimental depletion of ionocytes perturbs epithelial physiology in the mouse trachea, but the role of these rare cells in the pathogenesis of human CF remains mysterious. Ionocytes have been described in diverse tissues(kidney and inner ear) and species (frog and fish). We draw on these prior studies to suggest potential roles of airway ionocytes in health and disease. A complete understanding of ionocytes in the mammalian airway will ultimately depend on cell type-specific genetic manipulation.
Collapse
Affiliation(s)
- Viral S Shah
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Raghu R Chivukula
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | - Brian Lin
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| | - Avinash Waghray
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| | - Jayaraj Rajagopal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; , , , ,
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Klarman Cell Observatory, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
205
|
Chimenti I, Sattler S, del Monte-Nieto G, Forte E. Editorial: Fibrosis and Inflammation in Tissue Pathophysiology. Front Physiol 2022; 12:830683. [PMID: 35126187 PMCID: PMC8814660 DOI: 10.3389/fphys.2021.830683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022] Open
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Naples, Italy
- *Correspondence: Isotta Chimenti
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Elvira Forte
- The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
206
|
Kim YJ, Sivetz N, Layne J, Voss DM, Yang L, Zhang Q, Krainer AR. Exon-skipping antisense oligonucleotides for cystic fibrosis therapy. Proc Natl Acad Sci U S A 2022; 119:e2114858118. [PMID: 35017301 PMCID: PMC8784140 DOI: 10.1073/pnas.2114858118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF), and the CFTR-W1282X nonsense mutation causes a severe form of CF. Although Trikafta and other CFTR-modulation therapies benefit most CF patients, targeted therapy for patients with the W1282X mutation is lacking. The CFTR-W1282X protein has residual activity but is expressed at a very low level due to nonsense-mediated messenger RNA (mRNA) decay (NMD). NMD-suppression therapy and read-through therapy are actively being researched for CFTR nonsense mutants. NMD suppression could increase the mutant CFTR mRNA, and read-through therapies may increase the levels of full-length CFTR protein. However, these approaches have limitations and potential side effects: because the NMD machinery also regulates the expression of many normal mRNAs, broad inhibition of the pathway is not desirable, and read-through drugs are inefficient partly because the mutant mRNA template is subject to NMD. To bypass these issues, we pursued an exon-skipping antisense oligonucleotide (ASO) strategy to achieve gene-specific NMD evasion. A cocktail of two splice-site-targeting ASOs induced the expression of CFTR mRNA without the premature-termination-codon-containing exon 23 (CFTR-Δex23), which is an in-frame exon. Treatment of human bronchial epithelial cells with this cocktail of ASOs that target the splice sites flanking exon 23 results in efficient skipping of exon 23 and an increase in CFTR-Δex23 protein. The splice-switching ASO cocktail increases the CFTR-mediated chloride current in human bronchial epithelial cells. Our results set the stage for developing an allele-specific therapy for CF caused by the W1282X mutation.
Collapse
Affiliation(s)
- Young Jin Kim
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794
- Medical Scientist Training Program, Stony Brook University School of Medicine, Stony Brook, NY 11794
| | - Nicole Sivetz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Jessica Layne
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Dillon M Voss
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794
- Medical Scientist Training Program, Stony Brook University School of Medicine, Stony Brook, NY 11794
| | - Lucia Yang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY 11794
- Medical Scientist Training Program, Stony Brook University School of Medicine, Stony Brook, NY 11794
| | - Qian Zhang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Graduate Program in Molecular and Cell Biology, Stony Brook University, Stony Brook, NY 11794
| | | |
Collapse
|
207
|
Fischer S, Klockgether J, Gonzalez Sorribes M, Dorda M, Wiehlmann L, Tümmler B. Sequence diversity of the Pseudomonas aeruginosa population in loci that undergo microevolution in cystic fibrosis airways. Access Microbiol 2022; 3:000286. [PMID: 35024551 PMCID: PMC8749138 DOI: 10.1099/acmi.0.000286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022] Open
Abstract
Five hundred and thirty-four unrelated Pseudomonas aeruginosa isolates from inanimate habitats, patients with cystic fibrosis (CF) and other human infections were sequenced in 19 genes that had been identified previously as the hot spots of genomic within-host evolution in serial isolates from 12 CF lungs. Amplicon sequencing confirmed a significantly higher sequence diversity of the 19 loci in P. aeruginosa isolates from CF patients compared to those from other habitats, but this overrepresentation was mainly due to the larger share of synonymous substitutions. Correspondingly, non-synonymous substitutions were either rare (gltT, lepA, ptsP) or benign (nuoL, fleR, pelF) in some loci. Other loci, however, showed an accumulation of non-neutral coding variants. Strains from the CF habitat were often mutated at evolutionarily conserved positions in the elements of stringent response (RelA, SpoT), LPS (PagL), polyamine transport (SpuE, SpuF) and alginate biosynthesis (AlgG, AlgU). The strongest skew towards the CF lung habitat was seen for amino acid sequence variants in AlgG that clustered in the carbohydrate-binding/sugar hydrolysis domain. The master regulators of quorum sensing lasR and rhlR were frequent targets for coding variants in isolates from chronic and acute human infections. Unique variants in lasR showed strong evidence of positive selection indicated by d N/d S values of ~4. The pelA gene that encodes a multidomain enzyme involved in both the formation and dispersion of Pel biofilms carried the highest number of single-nucleotide variants among the 19 genes and was the only gene with a higher frequency of missense mutations in P. aeruginosa strains from non-CF habitats than in isolates from CF airways. PelA protein variants are widely distributed in the P. aeruginosa population. In conclusion, coding variants in a subset of the examined loci are indeed characteristic for the adaptation of P. aeruginosa to the CF airways, but for other loci the elevated mutation rate is more indicative of infections in human habitats (lasR, rhlR) or global diversifying selection (pelA).
Collapse
Affiliation(s)
- Sebastian Fischer
- Clinical Research Group 'Molecular Pathology of Cystic Fibrosis', Department for Paediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Jens Klockgether
- Clinical Research Group 'Molecular Pathology of Cystic Fibrosis', Department for Paediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Marina Gonzalez Sorribes
- Clinical Research Group 'Molecular Pathology of Cystic Fibrosis', Department for Paediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Marie Dorda
- Clinical Research Group 'Molecular Pathology of Cystic Fibrosis', Department for Paediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Lutz Wiehlmann
- Clinical Research Group 'Molecular Pathology of Cystic Fibrosis', Department for Paediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Burkhard Tümmler
- Clinical Research Group 'Molecular Pathology of Cystic Fibrosis', Department for Paediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Germany
| |
Collapse
|
208
|
Fiedorczuk K, Chen J. Mechanism of CFTR correction by type I folding correctors. Cell 2022; 185:158-168.e11. [PMID: 34995514 DOI: 10.1016/j.cell.2021.12.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/02/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023]
Abstract
Small molecule chaperones have been exploited as therapeutics for the hundreds of diseases caused by protein misfolding. The most successful examples are the CFTR correctors, which transformed cystic fibrosis therapy. These molecules revert folding defects of the ΔF508 mutant and are widely used to treat patients. To investigate the molecular mechanism of their action, we determined cryo-electron microscopy structures of CFTR in complex with the FDA-approved correctors lumacaftor or tezacaftor. Both drugs insert into a hydrophobic pocket in the first transmembrane domain (TMD1), linking together four helices that are thermodynamically unstable. Mutating residues at the binding site rendered ΔF508-CFTR insensitive to lumacaftor and tezacaftor, underscoring the functional significance of the structural discovery. These results support a mechanism in which the correctors stabilize TMD1 at an early stage of biogenesis, prevent its premature degradation, and thereby allosterically rescuing many disease-causing mutations.
Collapse
Affiliation(s)
- Karol Fiedorczuk
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Jue Chen
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
209
|
Krajewska J, Zub K, Słowikowski A, Zatoński T. Chronic rhinosinusitis in cystic fibrosis: a review of therapeutic options. Eur Arch Otorhinolaryngol 2022; 279:1-24. [PMID: 34296343 PMCID: PMC8739462 DOI: 10.1007/s00405-021-06875-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/06/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE Chronic rhinosinusitis (CRS) is observed in almost 100% of patients with cystic fibrosis (CF). CF-related CRS treatment is extremely challenging because of the underlying genetic defect leading to its development. CRS in CF is often refractory to standard therapy, while recurrences after surgical treatment are inevitable in the majority of patients. This study provides a precise review of the current knowledge regarding possible therapeutic options for CF-related CRS. METHODS The Medline and Web of Science databases were searched without a time limit using the terms "cystic fibrosis" in conjunction with "otorhinolaryngological manifestation", "rhinology" and "sinusitis". RESULTS Precise guidelines for CF-induced CRS therapy are lacking due to the lack of large cohort randomized controlled trials. None of the existing therapeutic agents has already been recommended for CRS in CF. Therapy targeting the underlying genetic defect, intranasal dornase alfa administration, and topical delivery of colistin and tobramycin showed promising results in CF-related CRS therapy. Besides the potential effectiveness of nasal steroids, strong recommendations for their usage in CF have not been provided yet. Systemic corticosteroid usage is controversial due to its potential negative influence on pulmonary disease. Ibuprofen revealed some positive effects on CF-related CRS in molecular and small cohort studies. Intranasal irrigation with saline solutions could relieve sinonasal symptoms. Nasal decongestants are not recommended. Endoscopic sinus surgery is the first-line surgical option for refractory CRS. Extensive surgical approaches should be considered as they could improve long-term outcomes in CRS. CONCLUSION Further studies are warranted to establish consensus for CF-related CRS therapy.
Collapse
Affiliation(s)
- Joanna Krajewska
- Department of Otolaryngology Head and Neck Surgery, Wroclaw Medical University, Wrocław, Poland
| | - Krzysztof Zub
- Department of Otolaryngology Head and Neck Surgery, Wroclaw Medical University, Wrocław, Poland
| | - Adam Słowikowski
- Department of Pediatric Pulmonology, Medical Center Karpacz, Karpacz, Poland
| | - Tomasz Zatoński
- Department of Otolaryngology Head and Neck Surgery, Wroclaw Medical University, Wrocław, Poland.
| |
Collapse
|
210
|
Esgueira VLR, Lopes CPA, dos Santos ACA, Pinto F, Sousa SA, de Barros DPC, Leitão JH, Fonseca LP. LipNanoCar Technology – A Versatile and Scalable Technology for the Production of Lipid Nanoparticles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1357:43-82. [DOI: 10.1007/978-3-030-88071-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
211
|
Catellani C, Cirillo F, Graziano S, Montanini L, Marmiroli N, Gullì M, Street ME. MicroRNA global profiling in cystic fibrosis cell lines reveals dysregulated pathways related with inflammation, cancer, growth, glucose and lipid metabolism, and fertility: an exploratory study. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 93:e2022133. [PMID: 35775757 PMCID: PMC9335447 DOI: 10.23750/abm.v93i3.12842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND AIM Cystic fibrosis (CF), is due to CF transmembrane conductance regulator (CFTR) loss of function, and is associated with comorbidities. The increasing longevity of CF patients has been associated with increased cancer risk besides the other known comorbidities. The significant heterogeneity among patients, suggests potential epigenetic regulation. Little attention has been given to how CFTR influences microRNA (miRNA) expression and how this may impact on biological processes and pathways. METHODS We assessed the changes in miRNAs and subsequently identified the affected molecular pathways using CFBE41o-, and IB3 human immortalized cell lines since they reflect the most common genetic mutations in CF patients, and 16HBE14o- cells were used as controls. RESULTS In the CF cell lines, 41 miRNAs showed significant changes (FC (log2) ≥ +2 or FC (log2) ≤ -2 and p-value≤0.05). Gene target analysis evidenced 511 validated miRNA target genes. Gene Ontology analysis evidenced cancer, inflammation, body growth, glucose, and lipid metabolism as the biological processes most impacted by these miRNAs. Protein-protein interaction and pathway analysis highlighted 50 significantly enriched pathways among which RAS, TGF beta, JAK/STAT and insulin signaling. CONCLUSIONS CFTR loss of function is associated with changes in the miRNA network, which regulates genes involved in the major comorbidities that affect CF patients suggesting that further research is warranted.
Collapse
Affiliation(s)
- Cecilia Catellani
- Department of Mother and Child, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy, PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy, These authors contributed equally to this work
| | - Francesca Cirillo
- Department of Mother and Child, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy, These authors contributed equally to this work
| | - Sara Graziano
- Interdepartmental Center SITEIA.PARMA, University of Parma, Parma, Italy
| | - Luisa Montanini
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Nelson Marmiroli
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Mariolina Gullì
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Maria E. Street
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
212
|
Huang L, Zhang L, Li W, Li S, Wen J, Li H, Liu Z. Advances in Development of mRNA-Based Therapeutics. Curr Top Microbiol Immunol 2022; 440:147-166. [PMID: 32683507 DOI: 10.1007/82_2020_222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recently, mRNA-based therapeutics have been greatly boosted since the development of novel technologies of both mRNA synthesis and delivery system. Promising results were showed in both preclinical and clinical studies in the field of cancer vaccine, tumor immunotherapy, infectious disease prevention and protein replacement therapy. Recent advancements in clinical trials also encouraged scientists to attempt new applications of mRNA therapy such as gene editing and cell programming. These studies bring mRNA therapeutics closer to real-world application. Herein, we provide an overview of recent advances in mRNA-based therapeutics.
Collapse
Affiliation(s)
- Lei Huang
- Stemirna Therapeutics Inc, Shanghai, 201206, China
| | - Luyao Zhang
- Stemirna Therapeutics Inc, Shanghai, 201206, China
| | - Weiwei Li
- Stemirna Therapeutics Inc, Shanghai, 201206, China
| | - Shiqiang Li
- Stemirna Therapeutics Inc, Shanghai, 201206, China
| | - Jianguo Wen
- Stemirna Therapeutics Inc, Shanghai, 201206, China
| | - Hangwen Li
- Stemirna Therapeutics Inc, Shanghai, 201206, China.
| | | |
Collapse
|
213
|
Elkhalifa D, Rayan M, Negmeldin AT, Elhissi A, Khalil A. Chemically modified mRNA beyond COVID-19: Potential preventive and therapeutic applications for targeting chronic diseases. Biomed Pharmacother 2022; 145:112385. [PMID: 34915673 PMCID: PMC8552589 DOI: 10.1016/j.biopha.2021.112385] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Chemically modified mRNA represents a unique, efficient, and straightforward approach to produce a class of biopharmaceutical agents. It has been already approved as a vaccination-based method for targeting SARS-CoV-2 virus. The COVID-19 pandemic has highlighted the prospect of synthetic modified mRNA to efficiently and safely combat various diseases. Recently, various optimization advances have been adopted to overcome the limitations associated with conventional gene therapeutics leading to wide-ranging applications in different disease conditions. This review sheds light on emerging directions of chemically modified mRNAs to prevent and treat widespread chronic diseases, including metabolic disorders, cancer vaccination and immunotherapy, musculoskeletal disorders, respiratory conditions, cardiovascular diseases, and liver diseases.
Collapse
Affiliation(s)
- Dana Elkhalifa
- Department of Pharmacy, Aspetar Orthopedic and Sports Medicine Hospital, Doha, Qatar
| | - Menatallah Rayan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Ahmed T Negmeldin
- Department of Pharmaceutical Sciences, College of Pharmacy and Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Abdelbary Elhissi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar; Office of the Vice President for Research and Graduate Studies, Qatar University, Doha, Qatar
| | - Ashraf Khalil
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
214
|
Apgar TL, Sanders CR. Compendium of causative genes and their encoded proteins for common monogenic disorders. Protein Sci 2022; 31:75-91. [PMID: 34515378 PMCID: PMC8740837 DOI: 10.1002/pro.4183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 01/19/2023]
Abstract
A compendium is presented of inherited monogenic disorders that have a prevalence of >1:20,000 in the human population, along with their causative genes and encoded proteins. "Simple" monogenic diseases are those for which the clinical features are caused by mutations impacting a single gene, usually in a manner that alters the sequence of the encoded protein. Of course, for a given "monogenic disorder", there is sometimes more than one potential disease gene, mutations in any one of which is sufficient to cause phenotypes of that disorder. Disease-causing mutations for monogenic disorders are usually passed on from generation to generation in a Mendelian fashion, and originate from spontaneous (de novo) germline founder mutations. In the past monogenic disorders have often been written off as targets for drug discovery because they sometimes are assumed to be rare disorders, for which the meager projected financial payoff of drug discovery and development has discouraged investment. However, not all monogenic diseases are rare. Here, we report that that currently available data identifies 72 disorders with a prevalence of at least 1 in 20,000 humans. For each, we tabulate the gene(s) for which mutations cause the spectrum of phenotypes associated with that disorder. We also identify the gene and protein that most commonly causes each disease. 34 of these disorders are caused exclusively by mutations in only a single gene and encoded protein.
Collapse
Affiliation(s)
- Tucker L. Apgar
- Department of Biochemistry and Center for Structural BiologyVanderbilt University School of Medicine Basic SciencesNashvilleTennesseeUSA
| | - Charles R. Sanders
- Department of Biochemistry and Center for Structural BiologyVanderbilt University School of Medicine Basic SciencesNashvilleTennesseeUSA
| |
Collapse
|
215
|
Middour-Oxler B, Bergman S, Blair S, Pendley S, Stecenko A, Hunt WR. Formal vs. informal transition in adolescents with cystic fibrosis: A retrospective comparison of outcomes. J Pediatr Nurs 2022; 62:177-183. [PMID: 34172371 DOI: 10.1016/j.pedn.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND The aim of this study was to survey young adults who participated in either a formal or semi-formal transition program at one cystic fibrosis (CF) care center to compare their self-perceived transition related anxiety, transition readiness and satisfaction with transition teaching and timing. METHODS This retrospective cohort study was conducted from 3/1/2015 to 9/30/2016. Study participants met inclusion criteria if they had a diagnosis of CF, received pediatric care from the care center, transitioned to adult care between 1/1/2009 and 3/1/2016 and had at least six months experience in adult care. Participants completed a 43 question Likert-type survey rating their pre-transfer transition related anxiety, transition readiness, and satisfaction with the transition preparation and process. FINDINGS Participation in a structured transition program was associated with decreased anxiety at transition time (p < .05), increased transition readiness (p < .01) and increased self-perceived healthcare independence (p < .01). Only 48% of participants were satisfied with their chosen transition time, with 18% wishing they had moved to adult care sooner and 34% wishing they could have delayed their transfer to adult care longer. DISCUSSION This study supports that participation in a formalized transition program was associated with significantly lower pre-transfer anxiety and higher post-transition satisfaction in individuals with CF. Age at transfer initiation was not associated with satisfaction or perceived readiness to transfer. PRACTICE IMPLICATIONS Disease-specific knowledge acquisition in transition curriculum does not necessarily correlate to task-completion skills. Teams should partner with young adults to choose the right transition time.
Collapse
Affiliation(s)
- Brandi Middour-Oxler
- Children's Healthcare of Atlanta + Emory University Cystic Fibrosis Care Center, Atlanta, GA, USA; Division of Pulmonary Allergy/Immunology, Cystic Fibrosis & Sleep, Department of Pediatrics, Emory University, Atlanta, GA, USA.
| | | | - Shaina Blair
- Children's Healthcare of Atlanta + Emory University Cystic Fibrosis Care Center, Atlanta, GA, USA
| | - Stephanie Pendley
- Children's Healthcare of Atlanta + Emory University Cystic Fibrosis Care Center, Atlanta, GA, USA
| | - Arlene Stecenko
- Children's Healthcare of Atlanta + Emory University Cystic Fibrosis Care Center, Atlanta, GA, USA; Division of Pulmonary Allergy/Immunology, Cystic Fibrosis & Sleep, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - William R Hunt
- Children's Healthcare of Atlanta + Emory University Cystic Fibrosis Care Center, Atlanta, GA, USA; Divison of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
216
|
Moreira A, Müller M, Costa PF, Kohl Y. Advanced In Vitro Lung Models for Drug and Toxicity Screening: The Promising Role of Induced Pluripotent Stem Cells. Adv Biol (Weinh) 2021; 6:e2101139. [PMID: 34962104 DOI: 10.1002/adbi.202101139] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/25/2021] [Indexed: 12/24/2022]
Abstract
The substantial socioeconomic burden of lung diseases, recently highlighted by the disastrous impact of the coronavirus disease 2019 (COVID-19) pandemic, accentuates the need for interventive treatments capable of decelerating disease progression, limiting organ damage, and contributing to a functional tissue recovery. However, this is hampered by the lack of accurate human lung research models, which currently fail to reproduce the human pulmonary architecture and biochemical environment. Induced pluripotent stem cells (iPSCs) and organ-on-chip (OOC) technologies possess suitable characteristics for the generation of physiologically relevant in vitro lung models, allowing for developmental studies, disease modeling, and toxicological screening. Importantly, these platforms represent potential alternatives for animal testing, according to the 3Rs (replace, reduce, refine) principle, and hold promise for the identification and approval of new chemicals under the European REACH (registration, evaluation, authorization and restriction of chemicals) framework. As such, this review aims to summarize recent progress made in human iPSC- and OOC-based in vitro lung models. A general overview of the present applications of in vitro lung models is presented, followed by a summary of currently used protocols to generate different lung cell types from iPSCs. Lastly, recently developed iPSC-based lung models are discussed.
Collapse
Affiliation(s)
| | - Michelle Müller
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Pedro F Costa
- BIOFABICS, Rua Alfredo Allen 455, Porto, 4200-135, Portugal
| | - Yvonne Kohl
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany.,Postgraduate Course for Toxicology and Environmental Toxicology, Medical Faculty, University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany
| |
Collapse
|
217
|
Aerosol-Mediated Non-Viral Lung Gene Therapy: The Potential of Aminoglycoside-Based Cationic Liposomes. Pharmaceutics 2021; 14:pharmaceutics14010025. [PMID: 35056921 PMCID: PMC8778791 DOI: 10.3390/pharmaceutics14010025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022] Open
Abstract
Aerosol lung gene therapy using non-viral delivery systems represents a credible therapeutic strategy for chronic respiratory diseases, such as cystic fibrosis (CF). Progress in CF clinical setting using the lipidic formulation GL67A has demonstrated the relevance of such a strategy while emphasizing the need for more potent gene transfer agents. In recent years, many novel non-viral gene delivery vehicles were proposed as potential alternatives to GL67 cationic lipid. However, they were usually evaluated using procedures difficult or even impossible to implement in clinical practice. In this study, a clinically-relevant administration protocol via aerosol in murine lungs was used to conduct a comparative study with GL67A. Diverse lipidic compounds were used to prepare a series of formulations inspired by the composition of GL67A. While some of these formulations were ineffective at transfecting murine lungs, others demonstrated modest-to-very-efficient activities and a series of structure-activity relationships were unveiled. Lipidic aminoglycoside derivative-based formulations were found to be at least as efficient as GL67A following aerosol delivery of a luciferase-encoding plasmid DNA. A single aerosol treatment with one such formulation was found to mediate long-term lung transgene expression, exceeding half the animal's lifetime. This study clearly supports the potential of aminoglycoside-based cationic lipids as potent GL67-alternative scaffolds for further enhanced aerosol non-viral lung gene therapy for diseases such as CF.
Collapse
|
218
|
Rare Trafficking CFTR Mutations Involve Distinct Cellular Retention Machineries and Require Different Rescuing Strategies. Int J Mol Sci 2021; 23:ijms23010024. [PMID: 35008443 PMCID: PMC8744605 DOI: 10.3390/ijms23010024] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 12/12/2022] Open
Abstract
Most of the ~2100 CFTR variants so far reported are very rare and still uncharacterized regarding their cystic fibrosis (CF) disease liability. Since some may respond to currently approved modulators, characterizing their defect and response to these drugs is essential. Here we aimed characterizing the defect associated with four rare missense (likely Class II) CFTR variants and assess their rescue by corrector drugs. We produced CFBE cell lines stably expressing CFTR with W57G, R560S, H1079P and Q1100P, assessed their effect upon CFTR expression and maturation and their rescue by VX-661/VX-445 correctors. Results were validated by forskolin-induced swelling assay (FIS) using intestinal organoids from individuals bearing these variants. Finally, knock-down (KD) of genes previously shown to rescue F508del-CFTR was assessed on these mutants. Results show that all the variants preclude the production of mature CFTR, confirming them as Class II mutations. None of the variants responded to VX-661 but the combination rescued H1079P- and Q1100P-CFTR. The KD of factors that correct F508del-CFTR retention only marginally rescued R560S- and H1079P-CFTR. Overall, data evidence that Class II mutations induce distinct molecular defects that are neither rescued by the same corrector compounds nor recognized by the same cellular machinery, thus requiring personalized drug discovery initiatives.
Collapse
|
219
|
Synergy in Cystic Fibrosis Therapies: Targeting SLC26A9. Int J Mol Sci 2021; 22:ijms222313064. [PMID: 34884866 PMCID: PMC8658147 DOI: 10.3390/ijms222313064] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
SLC26A9, a constitutively active Cl- transporter, has gained interest over the past years as a relevant disease modifier in several respiratory disorders including Cystic Fibrosis (CF), asthma, and non-CF bronchiectasis. SLC26A9 contributes to epithelial Cl- secretion, thus preventing mucus obstruction under inflammatory conditions. Additionally, SLC26A9 was identified as a CF gene modifier, and its polymorphisms were shown to correlate with the response to drugs modulating CFTR, the defective protein in CF. Here, we aimed to investigate the relationship between SLC26A9 and CFTR, and its role in CF pathogenesis. Our data show that SLC26A9 expression contributes to enhanced CFTR expression and function. While knocking-down SLC26A9 in human bronchial cells leads to lower wt- and F508del-CFTR expression, function, and response to CFTR correctors, the opposite occurs upon its overexpression, highlighting SLC26A9 relevance for CF. Accordingly, F508del-CFTR rescue by the most efficient correctors available is further enhanced by increasing SLC26A9 expression. Interestingly, SLC26A9 overexpression does not increase the PM expression of non-F508del CFTR traffic mutants, namely those unresponsive to corrector drugs. Altogether, our data indicate that SLC26A9 stabilizes CFTR at the ER level and that the efficacy of CFTR modulator drugs may be further enhanced by increasing its expression.
Collapse
|
220
|
Cu-catalyzed endo-selective asymmetric 1,3-dipolar cycloaddition of azomethine ylides with ethenesulfonyl fluorides: Efficient access to chiral pyrrolidine-3-sulfonyl fluorides. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.05.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
221
|
Kotsimbos T, Kaye D, Keating D. Pulmonary arterial hypertension and CFTR: the paradox of going forward by tacking sideways! Eur Respir J 2021; 58:58/5/2101839. [PMID: 34824129 DOI: 10.1183/13993003.01839-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Tom Kotsimbos
- Dept of Respiratory Medicine, Alfred Hospital, Central Clinical School, Monash University, Melbourne, Australia .,Dept of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - David Kaye
- Dept of Cardiology, Alfred Hospital, Central Clinical School, Monash University, Melbourne, Australia.,Dept of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Dominic Keating
- Dept of Respiratory Medicine, Alfred Hospital, Central Clinical School, Monash University, Melbourne, Australia.,Dept of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
222
|
Soni DK, Biswas R. Role of Non-Coding RNAs in Post-Transcriptional Regulation of Lung Diseases. Front Genet 2021; 12:767348. [PMID: 34819948 PMCID: PMC8606426 DOI: 10.3389/fgene.2021.767348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Non-coding RNAs (ncRNAs), notably microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), have recently gained increasing consideration because of their versatile role as key regulators of gene expression. They adopt diverse mechanisms to regulate transcription and translation, and thereby, the function of the protein, which is associated with several major biological processes. For example, proliferation, differentiation, apoptosis, and metabolic pathways demand fine-tuning for the precise development of a specific tissue or organ. The deregulation of ncRNA expression is concomitant with multiple diseases, including lung diseases. This review highlights recent advances in the post-transcriptional regulation of miRNAs and lncRNAs in lung diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, and idiopathic pulmonary fibrosis. Further, we also discuss the emerging role of ncRNAs as biomarkers as well as therapeutic targets for lung diseases. However, more investigations are required to explore miRNAs and lncRNAs interaction, and their function in the regulation of mRNA expression. Understanding these mechanisms might lead to early diagnosis and the development of novel therapeutics for lung diseases.
Collapse
Affiliation(s)
- Dharmendra Kumar Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
223
|
Laselva O, Guerra L, Castellani S, Favia M, Di Gioia S, Conese M. Small-molecule drugs for cystic fibrosis: Where are we now? Pulm Pharmacol Ther 2021; 72:102098. [PMID: 34793977 DOI: 10.1016/j.pupt.2021.102098] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/24/2021] [Accepted: 11/12/2021] [Indexed: 01/05/2023]
Abstract
The cystic fibrosis (CF) lung disease is due to the lack/dysfunction of the CF Transmembrane Conductance Regulator (CFTR), a chloride channel expressed by epithelial cells as the main regulator of ion and fluid homeostasis. More than 2000 genetic variation in the CFTR gene are known, among which those with identified pathomechanism have been divided into six VI mutation classes. A major advancement in the pharmacotherapy of CF has been the development of small-molecule drugs hitting the root of the disease, i.e. the altered ion and fluid transport through the airway epithelium. These drugs, called CFTR modulators, have been advanced to the clinics to treat nearly 90% of CF patients, including the CFTR potentiator ivacaftor, approved for residual function mutations (Classes III and IV), and combinations of correctors (lumacaftor, tezacaftor, elexacaftor) and ivacaftor for patients bearing at least one the F508del mutation, the most frequent mutation belonging to class II. To cover the 10% of CF patients without etiological therapies, other novel small-molecule CFTR modulators are in evaluation of their effectiveness in all the CFTR mutation classes: read-through agents for Class I, correctors, potentiators and amplifiers from different companies for Class II-V, stabilizers for Class VI. In alternative, other solute carriers, such as SLC26A9 and SLC6A14, are the focus of intensive investigation. Finally, other molecular targets are being evaluated for patients with no approved CFTR modulator therapy or as means of enhancing CFTR modulatory therapy, including small molecules forming ion channels, inhibitors of the ENaC sodium channel and potentiators of the calcium-activated chloride channel TMEM16A. This paper aims to give an up-to-date overview of old and novel CFTR modulators as well as of novel strategies based on small-molecule drugs. Further investigations in in-vivo and cell-based models as well as carrying out large prospective studies will be required to determine if novel CFTR modulators, stabilizers, amplifiers, and the ENaC inhibitors or TMEM16A potentiators will further improve the clinical outcomes in CF management.
Collapse
Affiliation(s)
- Onofrio Laselva
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Stefano Castellani
- Department of Medical Sciences and Human Oncology, University of Bari, Bari, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
224
|
Yu X, Liu S, Cheng Q, Lee SM, Wei T, Zhang D, Farbiak L, Johnson LT, Wang X, Siegwart DJ. Hydrophobic Optimization of Functional Poly(TPAE-co-suberoyl chloride) for Extrahepatic mRNA Delivery following Intravenous Administration. Pharmaceutics 2021; 13:pharmaceutics13111914. [PMID: 34834329 PMCID: PMC8624493 DOI: 10.3390/pharmaceutics13111914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023] Open
Abstract
Messenger RNA (mRNA) has generated great attention due to its broad potential therapeutic applications, including vaccines, protein replacement therapy, and immunotherapy. Compared to other nucleic acids (e.g., siRNA and pDNA), there are more opportunities to improve the delivery efficacy of mRNA through systematic optimization. In this report, we studied a high-throughput library of 1200 functional polyesters for systemic mRNA delivery. We focused on the chemical investigation of hydrophobic optimization as a method to adjust mRNA polyplex stability, diameter, pKa, and efficacy. Focusing on a region of the library heatmap (PE4K-A17), we further explored the delivery of luciferase mRNA to IGROV1 ovarian cancer cells in vitro and to C57BL/6 mice in vivo following intravenous administration. PE4K-A17-0.2C8 was identified as an efficacious carrier for delivering mRNA to mouse lungs. The delivery selectivity between organs (lungs versus spleen) was found to be tunable through chemical modification of polyesters (both alkyl chain length and molar ratio in the formulation). Cre recombinase mRNA was delivered to the Lox-stop-lox tdTomato mouse model to study potential application in gene editing. Overall, we identified a series of polymer-mRNA polyplexes stabilized with Pluronic F-127 for safe and effective delivery to mouse lungs and spleens. Structure–activity relationships between alkyl side chains and in vivo delivery were elucidated, which may be informative for the continued development of polymer-based mRNA delivery.
Collapse
|
225
|
Raraigh KS, Aksit MA, Hetrick K, Pace RG, Ling H, O'Neal W, Blue E, Zhou YH, Bamshad MJ, Blackman SM, Gibson RL, Knowles MR, Cutting GR. Complete CFTR gene sequencing in 5,058 individuals with cystic fibrosis informs variant-specific treatment. J Cyst Fibros 2021; 21:463-470. [PMID: 34782259 DOI: 10.1016/j.jcf.2021.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) is a recessive condition caused by variants in each CF transmembrane conductance regulator (CFTR) allele. Clinically affected individuals without two identified causal variants typically have no further interrogation of CFTR beyond examination of coding regions, but the development of variant-specific CFTR-targeted treatments necessitates complete understanding of CFTR genotype. METHODS Whole genome sequences were analyzed on 5,058 individuals with CF. We focused on the full CFTR gene sequence and identified disease-causing variants in three phases: screening for known and structural variants; discovery of novel loss-of-function variants; and investigation of remaining variants. RESULTS All variants identified in the first two phases and coding region variants found in the third phase were interpreted according to CFTR2 or ACMG criteria (n = 371; 16 [4.3%] previously unreported). Full gene sequencing enabled delineation of 18 structural variants (large insertions or deletions), of which two were novel. Additional CFTR variants of uncertain effect were found in 76 F508del homozygotes and in 21 individuals with other combinations of CF-causing variants. Both causative variants were identified in 98.1% (n = 4,960) of subjects, an increase of 2.3 percentage points from the 95.8% (n = 4,847) who had a registry- or chart-reported disease-causing CFTR genotype. Of the remaining 98 individuals, 78 carried one variant that has been associated with CF (CF-causing [n = 70] or resulting in varying clinical consequences n = 8]). CONCLUSIONS Complete CFTR gene sequencing in 5,058 individuals with CF identified at least one DNA variant in 99.6% of the cohort that is targetable by current molecular or emerging gene-based therapeutic technologies.
Collapse
Affiliation(s)
- Karen S Raraigh
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Melis A Aksit
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Kurt Hetrick
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Rhonda G Pace
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Hua Ling
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Wanda O'Neal
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Elizabeth Blue
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, United States
| | - Yi-Hui Zhou
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, United States; Department of Pediatrics, University of Washington, Seattle, WA 98195, United States; Brotman-Baty Institute, Seattle, WA 98195, United States
| | - Scott M Blackman
- Division of Pediatric Endocrinology and Diabetes, Johns Hopkins University, Baltimore, MD 21287, United States
| | - Ronald L Gibson
- Department of Pediatrics, University of Washington, Seattle, WA 98195, United States
| | - Michael R Knowles
- Marsico Lung Institute/UNC CF Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Garry R Cutting
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States.
| |
Collapse
|
226
|
Noel S, Servel N, Hatton A, Golec A, Rodrat M, Ng DRS, Li H, Pranke I, Hinzpeter A, Edelman A, Sheppard DN, Sermet-Gaudelus I. Correlating genotype with phenotype using CFTR-mediated whole-cell Cl - currents in human nasal epithelial cells. J Physiol 2021; 600:1515-1531. [PMID: 34761808 DOI: 10.1113/jp282143] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/01/2021] [Indexed: 12/20/2022] Open
Abstract
Dysfunction of the epithelial anion channel cystic fibrosis transmembrane conductance regulator (CFTR) causes a wide spectrum of disease, including cystic fibrosis (CF) and CFTR-related diseases (CFTR-RDs). Here, we investigate genotype-phenotype-CFTR function relationships using human nasal epithelial (hNE) cells from a small cohort of non-CF subjects and individuals with CF and CFTR-RDs and genotypes associated with either residual or minimal CFTR function using electrophysiological techniques. Collected hNE cells were either studied directly with the whole-cell patch-clamp technique or grown as primary cultures at an air-liquid interface after conditional reprogramming. The properties of cAMP-activated whole-cell Cl- currents in freshly isolated hNE cells identified them as CFTR-mediated. Their magnitude varied between hNE cells from individuals within the same genotype and decreased in the rank order: non-CF > CFTR residual function > CFTR minimal function. CFTR-mediated whole-cell Cl- currents in hNE cells isolated from fully differentiated primary cultures were identical to those in freshly isolated hNE cells in both magnitude and behaviour, demonstrating that conditional reprogramming culture is without effect on CFTR expression and function. For the cohort of subjects studied, CFTR-mediated whole-cell Cl- currents in hNE cells correlated well with CFTR-mediated transepithelial Cl- currents measured in vitro with the Ussing chamber technique, but not with those determined in vivo with the nasal potential difference assay. Nevertheless, they did correlate with the sweat Cl- concentration of study subjects. Thus, this study highlights the complexity of genotype-phenotype-CFTR function relationships, but emphasises the value of conditionally reprogrammed hNE cells in CFTR research and therapeutic testing. KEY POINTS: The genetic disease cystic fibrosis is caused by pathogenic variants in the cystic fibrosis transmembrane conductance regulator (CFTR), an ion channel, which controls anion flow across epithelia lining ducts and tubes in the body. This study investigated CFTR function in nasal epithelial cells from people with cystic fibrosis and CFTR variants with a range of disease severity. CFTR function varied widely in nasal epithelial cells depending on the identity of CFTR variants, but was unaffected by conditional reprogramming culture, a cell culture technique used to grow large numbers of patient-derived cells. Assessment of CFTR function in vitro in nasal epithelial cells and epithelia, and in vivo in the nasal epithelium and sweat gland highlights the complexity of genotype-phenotype-CFTR function relationships.
Collapse
Affiliation(s)
- Sabrina Noel
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France
| | - Nathalie Servel
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France
| | - Aurélie Hatton
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France
| | - Anita Golec
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France
| | - Mayuree Rodrat
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.,Center of Research and Development for Biomedical Instrumentation, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Demi R S Ng
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Hongyu Li
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Iwona Pranke
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France
| | - Alexandre Hinzpeter
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France
| | - Aleksander Edelman
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Isabelle Sermet-Gaudelus
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Université de Paris, Paris, France.,Centre de Référence Maladies Rares, Mucoviscidose et Maladies Apparentées, Hôpital Necker-Enfants Malades, Paris, France.,European Reference Network on rare respiratory diseases, Frankfurt, Germany
| |
Collapse
|
227
|
Dumas MP, Xia S, Bear CE, Ratjen F. Perspectives on the translation of in-vitro studies to precision medicine in Cystic Fibrosis. EBioMedicine 2021; 73:103660. [PMID: 34740114 PMCID: PMC8577330 DOI: 10.1016/j.ebiom.2021.103660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 11/22/2022] Open
Abstract
Recent strides towards precision medicine in Cystic Fibrosis (CF) have been made possible by patient-derived in-vitro assays with the potential to predict clinical response to small molecule-based therapies. Here, we discuss the status of primary and stem-cell derived tissues used to evaluate the preclinical efficacy of CFTR modulators highlighting both their potential and limitations. Validation of these assays requires correlation of in-vitro responses to in-vivo measures of clinical biomarkers of disease outcomes. While initial efforts have shown some success, this translation requires methodologies that are sensitive enough to capture treatment responses in a CF population that now predominantly has mild lung disease. Future development of in-vitro and in-vivo biomarkers will facilitate the generation of new therapeutics particularly for those patients with rare mutations where clinical trials are not feasible so that in the future every CF patient will have access to effective targeted therapies.
Collapse
Affiliation(s)
- Marie-Pier Dumas
- Respiratory Medicine, Hospital for Sick Children, Toronto, Canada; Translational Medicine, Hospital for Sick Children, Toronto, Canada
| | - Sunny Xia
- Molecular Medicine, Hospital for Sick Children, Toronto, Canada.; Department of Physiology, University of Toronto, Toronto, Canada
| | - Christine E Bear
- Molecular Medicine, Hospital for Sick Children, Toronto, Canada.; Department of Physiology, University of Toronto, Toronto, Canada; Department of Biochemistry University of Toronto, Toronto, Canada
| | - Felix Ratjen
- Respiratory Medicine, Hospital for Sick Children, Toronto, Canada; Translational Medicine, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
228
|
Kaffenberger TM, Eichar BW, McCoy JL, Yang TS, Shaffer AD, Dohar JE. Cystic fibrosis mutation classes in pediatric otitis media - Fickle or faulty? Am J Otolaryngol 2021; 42:103067. [PMID: 33957545 DOI: 10.1016/j.amjoto.2021.103067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 11/17/2022]
Abstract
PURPOSE Recent data have challenged the historical paradigm that cystic fibrosis (CF) protects against otitis media (OM). These findings raised questions about the pathogenesis of this ostensible change. In this study our aim is to characterize acute OM (AOM) risk based on CF genotype. METHODS A retrospective chart review was completed at a tertiary care pediatric hospital. Charts of 159 CF patients seen at our facility from 2010 to 2019 were reviewed. Data collected included demographics, AOM infections, cystic fibrosis transmembrane conductance regulator (CFTR) allele mutations, pulmonary exacerbations (PE), and pancreatic insufficiency (PI) status. Mutation alleles were divided into five classes based on CF guidelines, which were further classified as severe (classes I-III) or mild (classes IV-V). RESULTS 54% of patients had at least one episode of AOM with a mean of 1.5 episodes of AOM (standard deviation = 2.3). 86% of patients had severe/severe (S/S) alleles and 14% had severe/mild (S/M). S/S patients had significantly more PE (p = .004) and increased rates of PI (p < .001). Of the 131 patients with S/S mutations, 57% had an episode of AOM while only 46% the 22 S/M patients had an AOM episode (p = .357). CONCLUSIONS To our knowledge this is the first report showing a clinical trend towards increased middle ear disease in patients with severe CFTR mutations. Future prospective studies will be powered to demonstrate whether this trend is statistically significant. Patients with S/S mutations not only have more severe clinical phenotypes but may have additional unexpected complications such as middle ear disease.
Collapse
Affiliation(s)
- Thomas M Kaffenberger
- Department of Otolaryngology, University of Pittsburgh School of Medicine, 203 Lothrop St. Suite 500, Pittsburgh, PA 15213, USA
| | - Bradley W Eichar
- Department of Otolaryngology, University of Pittsburgh School of Medicine, 203 Lothrop St. Suite 500, Pittsburgh, PA 15213, USA.
| | - Jennifer L McCoy
- Division of Pediatric Otolaryngology, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Tiffany S Yang
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Amber D Shaffer
- Division of Pediatric Otolaryngology, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Joseph E Dohar
- Department of Otolaryngology, University of Pittsburgh School of Medicine, 203 Lothrop St. Suite 500, Pittsburgh, PA 15213, USA; Division of Pediatric Otolaryngology, UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Department of Communication Science and Disorders, University of Pittsburgh School of Health & Rehabilitation, Forbes Tower, 3600 Forbes Ave., Pittsburgh, PA 15260, USA.
| |
Collapse
|
229
|
TMEM16A/ANO1: Current Strategies and Novel Drug Approaches for Cystic Fibrosis. Cells 2021; 10:cells10112867. [PMID: 34831090 PMCID: PMC8616501 DOI: 10.3390/cells10112867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/19/2022] Open
Abstract
Cystic fibrosis (CF) is the most common of rare hereditary diseases in Caucasians, and it is estimated to affect 75,000 patients globally. CF is a complex disease due to the multiplicity of mutations found in the CF transmembrane conductance regulator (CFTR) gene causing the CFTR protein to become dysfunctional. Correctors and potentiators have demonstrated good clinical outcomes for patients with specific gene mutations; however, there are still patients for whom those treatments are not suitable and require alternative CFTR-independent strategies. Although CFTR is the main chloride channel in the lungs, others could, e.g., anoctamin-1 (ANO1 or TMEM16A), compensate for the deficiency of CFTR. This review summarizes the current knowledge on calcium-activated chloride channel (CaCC) ANO1 and presents ANO1 as an exciting target in CF.
Collapse
|
230
|
Krishnamurthy S, Traore S, Cooney AL, Brommel CM, Kulhankova K, Sinn P, Newby G, Liu D, McCray P. Functional correction of CFTR mutations in human airway epithelial cells using adenine base editors. Nucleic Acids Res 2021; 49:10558-10572. [PMID: 34520545 PMCID: PMC8501978 DOI: 10.1093/nar/gkab788] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 12/31/2022] Open
Abstract
Mutations in the CFTR gene that lead to premature stop codons or splicing defects cause cystic fibrosis (CF) and are not amenable to treatment by small-molecule modulators. Here, we investigate the use of adenine base editor (ABE) ribonucleoproteins (RNPs) that convert A•T to G•C base pairs as a therapeutic strategy for three CF-causing mutations. Using ABE RNPs, we corrected in human airway epithelial cells premature stop codon mutations (R553X and W1282X) and a splice-site mutation (3849 + 10 kb C > T). Following ABE delivery, DNA sequencing revealed correction of these pathogenic mutations at efficiencies that reached 38-82% with minimal bystander edits or indels. This range of editing was sufficient to attain functional correction of CFTR-dependent anion channel activity in primary epithelial cells from CF patients and in a CF patient-derived cell line. These results demonstrate the utility of base editor RNPs to repair CFTR mutations that are not currently treatable with approved therapeutics.
Collapse
Affiliation(s)
| | - Soumba Traore
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Ashley L Cooney
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Christian M Brommel
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Molecular Medicine Graduate Program, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| | | | - Patrick L Sinn
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Molecular Medicine Graduate Program, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Paul B McCray
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Molecular Medicine Graduate Program, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
231
|
Mésinèle J, Ruffin M, Kemgang A, Guillot L, Boëlle PY, Corvol H. Risk factors for Pseudomonas aeruginosa airway infection and lung function decline in children with cystic fibrosis. J Cyst Fibros 2021; 21:45-51. [PMID: 34629287 DOI: 10.1016/j.jcf.2021.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/31/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022]
Abstract
Background Cystic fibrosis (CF) lung disease is characterised by recurrent Pseudomonas aeruginosa (Pa) infections, leading to structural lung damage and decreased survival. The epidemiology of Pa infection and its impact on lung function in people with CF (pwCF), especially in recent birth cohorts, remain uncertain. Methods We included 1,231 French pwCF under 18 years of age. Age at initial acquisition (Pa-IA), chronic colonisation (Pa-CC), and duration from Pa-IA to Pa-CC were estimated using the Kaplan-Meier method. Demographic, clinical, and genetic characteristics were analysed as risk factors for Pa infection using Cox regression models. Lung function decline was assessed by modelling percent-predicted forced expiratory volume in 1 s (ppFEV1) before Pa infection, after Pa-IA, and after Pa-CC. Results Among the 1,231 pwCF, 50% had Pa-IA by the age of 5.1 years [95% confidence interval (CI) 3.8-6.2] and 25% had Pa-CC by the age of 14.7 years (95% CI 12.1 to ∞). We observed that CF-related diabetes and liver disease were risk factors for Pa, while gender, CFTR variants, and CF centre size were not. Genetic variants of TNF, DCTN4, SLC9A3, and CAV2 were confirmed to be associated with Pa. The annual rate of ppFEV1 decline before Pa was -0.38% predicted/year (95% CI -0.59 to -0.18), which decreased significantly after Pa-IA to -0.93% predicted/year (95% CI -1.14 to -0.71) and after Pa-CC to -1.51% predicted/year (95% CI -1.86 to -1.16). Conclusions We identified and replicated several risk factors associated with Pa infection and showed its deleterious impact on lung function in young pwCF. This large-scale study confirmed that Pa airway infection is a major determinant of lung disease severity.
Collapse
Affiliation(s)
- Julie Mésinèle
- Sorbonne Université, INSERM UMR S_938, Centre de Recherche Saint-Antoine (CRSA), Paris, France; Sorbonne Université, Inserm, Institut Pierre Louis d'épidémiologie et de Santé Publique, IPLESP, APHP, Hôpital Saint-Antoine, Paris, France
| | - Manon Ruffin
- Sorbonne Université, INSERM UMR S_938, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Astrid Kemgang
- Sorbonne Université, INSERM UMR S_938, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Loïc Guillot
- Sorbonne Université, INSERM UMR S_938, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Pierre-Yves Boëlle
- Sorbonne Université, Inserm, Institut Pierre Louis d'épidémiologie et de Santé Publique, IPLESP, APHP, Hôpital Saint-Antoine, Paris, France
| | - Harriet Corvol
- Sorbonne Université, INSERM UMR S_938, Centre de Recherche Saint-Antoine (CRSA), Paris, France; AP-HP, Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France.
| |
Collapse
|
232
|
Shaughnessy CA, Zeitlin PL, Bratcher PE. Elexacaftor is a CFTR potentiator and acts synergistically with ivacaftor during acute and chronic treatment. Sci Rep 2021; 11:19810. [PMID: 34615919 PMCID: PMC8494914 DOI: 10.1038/s41598-021-99184-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR), which lead to early death due to progressive lung disease. The development of small-molecule modulators that directly interact with CFTR to aid in protein folding (“correctors”) and/or increase channel function (“potentiators”) have proven to be highly effective in the therapeutic treatment of CF. Notably, incorporation of the next-generation CFTR corrector, elexacaftor, into a triple combination therapeutic (marketed as Trikafta) has shown tremendous clinical promise in treating CF caused by F508del-CFTR. Here, we report on a newly-described role of elexacaftor as a CFTR potentiator. We explore the acute and chronic actions, pharmacology, and efficacy of elexacaftor as a CFTR potentiator in restoring function to multiple classes of CFTR mutations. We demonstrate that the potentiating action of elexacaftor exhibits multiplicative synergy with the established CFTR potentiator ivacaftor in rescuing multiple CFTR class defects, indicating that a new combination therapeutic of ivacaftor and elexacaftor could have broad impact on CF therapies.
Collapse
Affiliation(s)
| | - Pamela L Zeitlin
- Department of Pediatrics, National Jewish Health, Denver, CO, USA.,Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Preston E Bratcher
- Department of Pediatrics, National Jewish Health, Denver, CO, USA.,Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| |
Collapse
|
233
|
Abstract
Cystic fibrosis (CF) is a heritable, multiorgan disease that impacts all tissues that normally express cystic fibrosis transmembrane conductance regulator (CFTR) protein. While the importance of the airway microbiota has long been recognized, the intestinal microbiota has only recently been recognized as an important player in both intestinal and lung health outcomes for persons with CF (pwCF). Here, we summarize current literature related to the gut-lung axis in CF, with a particular focus on three key ideas: (i) mechanisms through which microbes influence the gut-lung axis, (ii) drivers of microbiota alterations, and (iii) the potential for intestinal microbiota remediation.
Collapse
Affiliation(s)
- Courtney E. Price
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover New Hampshire, USA
| | - George A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover New Hampshire, USA
| |
Collapse
|
234
|
Pan FC, Evans T, Chen S. Modeling endodermal organ development and diseases using human pluripotent stem cell-derived organoids. J Mol Cell Biol 2021; 12:580-592. [PMID: 32652003 PMCID: PMC7683020 DOI: 10.1093/jmcb/mjaa031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/24/2020] [Accepted: 03/23/2020] [Indexed: 01/13/2023] Open
Abstract
Recent advances in development of protocols for directed differentiation from human pluripotent stem cells (hPSCs) to defined lineages, in combination with 3D organoid technology, have facilitated the generation of various endoderm-derived organoids for in vitro modeling of human gastrointestinal development and associated diseases. In this review, we discuss current state-of-the-art strategies for generating hPSC-derived endodermal organoids including stomach, liver, pancreatic, small intestine, and colonic organoids. We also review the advantages of using this system to model various human diseases and evaluate the shortcomings of this technology. Finally, we emphasize how other technologies, such as genome editing and bioengineering, can be incorporated into the 3D hPSC-organoid models to generate even more robust and powerful platforms for understanding human organ development and disease modeling.
Collapse
Affiliation(s)
- Fong Cheng Pan
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
235
|
Schamschula E, Lahnsteiner A, Assenov Y, Hagmann W, Zaborsky N, Wiederstein M, Strobl A, Stanke F, Muley T, Plass C, Tümmler B, Risch A. Disease-related blood-based differential methylation in cystic fibrosis and its representation in lung cancer revealed a regulatory locus in PKP3 in lung epithelial cells. Epigenetics 2021; 17:837-860. [PMID: 34415821 PMCID: PMC9423854 DOI: 10.1080/15592294.2021.1959976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis (CF) is a monogenic disease, characterized by massive chronic lung inflammation. The observed variability in clinical phenotypes in monozygotic CF twins is likely associated with the extent of inflammation. This study sought to investigate inflammation-related aberrant DNA methylation in CF twins and to determine to what extent acquired methylation changes may be associated with lung cancer. Blood-based genome-wide DNA methylation analysis was performed to compare the DNA methylomes of monozygotic twins, from the European CF Twin and Sibling Study with various degrees of disease severity. Putatively inflammation-related and differentially methylated positions were selected from a large lung cancer case-control study and investigated in blood by targeted bisulphite next-generation-sequencing. An inflammation-related locus located in the Plakophilin-3 (PKP3) gene was functionally analysed regarding promoter and enhancer activity in presence and absence of methylation using luciferase reporter assays. We confirmed in a unique cohort that monozygotic twins, even if clinically discordant, have only minor differences in global DNA methylation patterns and blood cell composition. Further, we determined the most differentially methylated positions, a high proportion of which are blood cell-type-specific, whereas others may be acquired and thus have potential relevance in the context of inflammation as lung cancer risk factors. We identified a sequence in the gene body of PKP3 which is hypermethylated in blood from CF twins with severe phenotype and highly variably methylated in lung cancer patients and controls, independent of known clinical parameters, and showed that this region exhibits methylation-dependent promoter activity in lung epithelial cells.
Collapse
Affiliation(s)
| | | | - Yassen Assenov
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Hagmann
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadja Zaborsky
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| | | | - Anna Strobl
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Frauke Stanke
- Clinical Research Group, Clinic for Pediatric Pneumology, Allergology and NeonatologyClinic for Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik Heidelberg, University of Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Burkhard Tümmler
- Clinical Research Group, Clinic for Pediatric Pneumology, Allergology and NeonatologyClinic for Pediatric Pneumology, Allergology and Neonatology, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| | - Angela Risch
- Department of Biosciences, University of Salzburg, Salzburg, Austria.,Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Cancer Cluster Salzburg, Salzburg, Austria.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
236
|
Sette G, Lo Cicero S, Blaconà G, Pierandrei S, Bruno SM, Salvati V, Castelli G, Falchi M, Fabrizzi B, Cimino G, De Maria R, Biffoni M, Eramo A, Lucarelli M. Theratyping cystic fibrosis in vitro in ALI-culture and organoid models generated from patient-derived nasal epithelial Conditionally Reprogrammed Stem Cells. Eur Respir J 2021; 58:13993003.00908-2021. [PMID: 34413153 PMCID: PMC8675295 DOI: 10.1183/13993003.00908-2021] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/28/2021] [Indexed: 11/05/2022]
Abstract
QUESTION Cystic Fibrosis (CF) is due to pathogenic variants in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene. Recent improvement enabled pharmacologic therapy aiming at restoring mutated CFTR expression and function. CFTR "modulators" have revolutionised the CF therapeutic landscape, particularly the last approved Trikafta. This drug-combination is indicated by FDA and very recently by EMA for genotypes carrying at least one copy of CFTR with F508del pathogenic variant. However, several genotypes, are not eligible for Trikafta treatment, yet. MATERIALS/PATIENTS AND METHODS We exploited an innovative cellular approach allowing highly efficient in vitro-expansion of airway epithelial stem cells (AESC) through conditional reprogramming (CRC) from nasal brushing of CF patients. This approach, coupled to development of AESC-derived personalised disease models, as organoids and air liquid interface (ALI) cultures, revealed highly suitable for CFTR pharmacological-testing. RESULTS AND ANSWER TO THE QUESTION We fully validated the experimental models and implemented the CFTR functional assays and biochemical CFTR protein characterisation, that allowed to evaluate the efficacy of clinically available modulators in restoring CFTR maturation and function of each patient-derived "avatar" (theratyping). F508del homozygous genotypes, used as controls, confirmed the higher clinical activity of Trikafta in comparison with older modulators. Trikafta showed its efficacy also on three rare genotypes previously not eligible for modulators-treatment, opening the way to clinical translation. Finally, encouraging results for innovative drug combinations were also obtained.
Collapse
Affiliation(s)
- Giovanni Sette
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.,Co-first authors
| | - Stefania Lo Cicero
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.,Co-first authors
| | - Giovanna Blaconà
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvia Pierandrei
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Sabina Maria Bruno
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Valentina Salvati
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Benedetta Fabrizzi
- Cystic Fibrosis Care Center, Mother - Child Department, United Hospitals, Ancona, Italy
| | - Giuseppe Cimino
- Cystic Fibrosis Reference Center of Lazio Region, AOU Policlinico Umberto I, Rome, Italy
| | - Ruggero De Maria
- U.O.C. Medical Oncology, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Adriana Eramo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy .,Co-last authors
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.,Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy.,Co-last authors
| |
Collapse
|
237
|
Rehman T, Karp PH, Tan P, Goodell BJ, Pezzulo AA, Thurman AL, Thornell IM, Durfey SL, Duffey ME, Stoltz DA, McKone EF, Singh PK, Welsh MJ. Inflammatory cytokines TNF-α and IL-17 enhance the efficacy of cystic fibrosis transmembrane conductance regulator modulators. J Clin Invest 2021; 131:e150398. [PMID: 34166230 DOI: 10.1172/jci150398] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Without cystic fibrosis transmembrane conductance regulator-mediated (CFTR-mediated) HCO3- secretion, airway epithelia of newborns with cystic fibrosis (CF) produce an abnormally acidic airway surface liquid (ASL), and the decreased pH impairs respiratory host defenses. However, within a few months of birth, ASL pH increases to match that in non-CF airways. Although the physiological basis for the increase is unknown, this time course matches the development of inflammation in CF airways. To learn whether inflammation alters CF ASL pH, we treated CF epithelia with TNF-α and IL-17 (TNF-α+IL-17), 2 inflammatory cytokines that are elevated in CF airways. TNF-α+IL-17 markedly increased ASL pH by upregulating pendrin, an apical Cl-/HCO3- exchanger. Moreover, when CF epithelia were exposed to TNF-α+IL-17, clinically approved CFTR modulators further alkalinized ASL pH. As predicted by these results, in vivo data revealed a positive correlation between airway inflammation and CFTR modulator-induced improvement in lung function. These findings suggest that inflammation is a key regulator of HCO3- secretion in CF airways. Thus, they explain earlier observations that ASL pH increases after birth and indicate that, for similar levels of inflammation, the pH of CF ASL is abnormally acidic. These results also suggest that a non-cell-autonomous mechanism, airway inflammation, is an important determinant of the response to CFTR modulators.
Collapse
Affiliation(s)
- Tayyab Rehman
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Philip H Karp
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Ping Tan
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Brian J Goodell
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Alejandro A Pezzulo
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Andrew L Thurman
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Ian M Thornell
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Samantha L Durfey
- Departments of Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - Michael E Duffey
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - David A Stoltz
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Edward F McKone
- National Referral Centre for Adult Cystic Fibrosis, St. Vincent's University Hospital and University College Dublin School of Medicine, Dublin, Ireland
| | - Pradeep K Singh
- Departments of Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - Michael J Welsh
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
238
|
Heydari Z, Moeinvaziri F, Agarwal T, Pooyan P, Shpichka A, Maiti TK, Timashev P, Baharvand H, Vosough M. Organoids: a novel modality in disease modeling. Biodes Manuf 2021; 4:689-716. [PMID: 34395032 PMCID: PMC8349706 DOI: 10.1007/s42242-021-00150-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/12/2021] [Indexed: 12/17/2022]
Abstract
Limitations of monolayer culture conditions have motivated scientists to explore new models that can recapitulate the architecture and function of human organs more accurately. Recent advances in the improvement of protocols have resulted in establishing three-dimensional (3D) organ-like architectures called 'organoids' that can display the characteristics of their corresponding real organs, including morphological features, functional activities, and personalized responses to specific pathogens. We discuss different organoid-based 3D models herein, which are classified based on their original germinal layer. Studies of organoids simulating the complexity of real tissues could provide novel platforms and opportunities for generating practical knowledge along with preclinical studies, including drug screening, toxicology, and molecular pathophysiology of diseases. This paper also outlines the key challenges, advantages, and prospects of current organoid systems.
Collapse
Affiliation(s)
- Zahra Heydari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, 14155-4364 Iran
| | - Farideh Moeinvaziri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, 14155-4364 Iran
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302 India
| | - Paria Pooyan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
| | - Anastasia Shpichka
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 19991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Tapas K. Maiti
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302 India
| | - Peter Timashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 19991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
- Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, 14155-4364 Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 14155-4364 Iran
| |
Collapse
|
239
|
Rajasekar P, Patel J, Clifford RL. DNA Methylation of Fibroblast Phenotypes and Contributions to Lung Fibrosis. Cells 2021; 10:cells10081977. [PMID: 34440746 PMCID: PMC8391838 DOI: 10.3390/cells10081977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 11/22/2022] Open
Abstract
Fibroblasts are an integral part of connective tissue and play a crucial role in developing and modulating the structural framework of tissues by acting as the primary source of extracellular matrix (ECM). A precise definition of the fibroblast remains elusive. Lung fibroblasts orchestrate the assembly and turnover of ECM to facilitate gas exchange alongside performing immune functions including the secretion of bioactive molecules and antigen presentation. DNA methylation is the covalent attachment of a methyl group to primarily cytosines within DNA. DNA methylation contributes to diverse cellular phenotypes from the same underlying genetic sequence, with DNA methylation profiles providing a memory of cellular origin. The lung fibroblast population is increasingly viewed as heterogeneous with between 6 and 11 mesenchymal populations identified across health and lung disease to date. DNA methylation has been associated with different lung fibroblast populations in health and with alterations in lung disease, but to varying extents. In this review, we will discuss lung fibroblast heterogeneity and the evidence for a contribution from DNA methylation to defining cell populations and alterations in disease.
Collapse
|
240
|
Leonhardt K, Autry EB, Kuhn RJ, Wurth MA. CFTR modulator drug desensitization: Preserving the hope of long term improvement. Pediatr Pulmonol 2021; 56:2546-2552. [PMID: 33913624 DOI: 10.1002/ppul.25437] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/26/2021] [Accepted: 04/13/2021] [Indexed: 11/07/2022]
Abstract
The development of modulator therapy has, for the first time, allowed direct targeting of the underlying cause of cystic fibrosis (CF), the cystic fibrosis transmembrane conductance regulator (CFTR). Patients treated with CFTR modulators have improvement in lung function and decreased rates of pulmonary exacerbations. In 2019, elexacaftor/tezacaftor/ivacaftor was approved for use in the United States, opening these therapies to 90% of patients with CF. Intolerable adverse drug reactions to CFTR modulators results in discontinuation of therapy, which can be devastating to our patients. We describe our approach to two cases, not previously reported, of rash to elexacaftor/tezacaftor/ivacaftor in patients with a previous history of cutaneous adverse reactions to dual modulator therapy that had been addressed by desensitization. Case 1 was able to tolerate elexacaftor/tezacaftor/ivacaftor after desensitization to the triple combination therapy, while in Case 2 tolerance was obtained by treating through the reaction. The loss of tolerance in these patients was unexpected, and may be a common finding in patients with history of cutaneous adverse reactions to these drugs. We hope reporting our experience, including our desensitization protocol, may benefit CF patients for whom these drug reactions may be limiting access to powerful disease altering therapies.
Collapse
Affiliation(s)
- Kelsey Leonhardt
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky, USA
| | - Elizabeth B Autry
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky, USA.,Department of Pharmacy Practice and Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Robert J Kuhn
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky, USA.,Department of Pharmacy Practice and Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Mark A Wurth
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
241
|
Sharma J, Du M, Wong E, Mutyam V, Li Y, Chen J, Wangen J, Thrasher K, Fu L, Peng N, Tang L, Liu K, Mathew B, Bostwick RJ, Augelli-Szafran CE, Bihler H, Liang F, Mahiou J, Saltz J, Rab A, Hong J, Sorscher EJ, Mendenhall EM, Coppola CJ, Keeling KM, Green R, Mense M, Suto MJ, Rowe SM, Bedwell DM. A small molecule that induces translational readthrough of CFTR nonsense mutations by eRF1 depletion. Nat Commun 2021; 12:4358. [PMID: 34272367 PMCID: PMC8285393 DOI: 10.1038/s41467-021-24575-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/22/2021] [Indexed: 11/09/2022] Open
Abstract
Premature termination codons (PTCs) prevent translation of a full-length protein and trigger nonsense-mediated mRNA decay (NMD). Nonsense suppression (also termed readthrough) therapy restores protein function by selectively suppressing translation termination at PTCs. Poor efficacy of current readthrough agents prompted us to search for better compounds. An NMD-sensitive NanoLuc readthrough reporter was used to screen 771,345 compounds. Among the 180 compounds identified with readthrough activity, SRI-37240 and its more potent derivative SRI-41315, induce a prolonged pause at stop codons and suppress PTCs associated with cystic fibrosis in immortalized and primary human bronchial epithelial cells, restoring CFTR expression and function. SRI-41315 suppresses PTCs by reducing the abundance of the termination factor eRF1. SRI-41315 also potentiates aminoglycoside-mediated readthrough, leading to synergistic increases in CFTR activity. Combining readthrough agents that target distinct components of the translation machinery is a promising treatment strategy for diseases caused by PTCs. Premature termination codons can cause early translation termination and lead to disease. Here the authors perform a screen to identify compounds with readthrough activity and show that these reduce eRF1 levels to suppress premature termination associated with cystic fibrosis.
Collapse
Affiliation(s)
- Jyoti Sharma
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Microbiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Ming Du
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Eric Wong
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | - Venkateshwar Mutyam
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Yao Li
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Jianguo Chen
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Jamie Wangen
- Department of Molecular Biology and Genetics and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kari Thrasher
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Lianwu Fu
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Ning Peng
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Liping Tang
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Kaimao Liu
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | | | | | | | - Hermann Bihler
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | - Feng Liang
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | - Jerome Mahiou
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | - Josef Saltz
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | - Andras Rab
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Jeong Hong
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Eric J Sorscher
- Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Eric M Mendenhall
- Department of Biological Sciences, The University of Alabama in Huntsville, Huntsville, AL, USA
| | - Candice J Coppola
- Department of Biological Sciences, The University of Alabama in Huntsville, Huntsville, AL, USA
| | - Kim M Keeling
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Rachel Green
- Department of Molecular Biology and Genetics and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin Mense
- CFFT Lab, Cystic Fibrosis Foundation, Lexington, MA, USA
| | | | - Steven M Rowe
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Pediatrics, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - David M Bedwell
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA. .,Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| |
Collapse
|
242
|
Dang Y, van Heusden C, Nickerson V, Chung F, Wang Y, Quinney NL, Gentzsch M, Randell SH, Moulton HM, Kole R, Ni A, Juliano RL, Kreda SM. Enhanced delivery of peptide-morpholino oligonucleotides with a small molecule to correct splicing defects in the lung. Nucleic Acids Res 2021; 49:6100-6113. [PMID: 34107015 PMCID: PMC8216463 DOI: 10.1093/nar/gkab488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary diseases offer many targets for oligonucleotide therapeutics. However, effective delivery of oligonucleotides to the lung is challenging. For example, splicing mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) affect a significant cohort of Cystic Fibrosis (CF) patients. These individuals could potentially benefit from treatment with splice switching oligonucleotides (SSOs) that can modulate splicing of CFTR and restore its activity. However, previous studies in cell culture used oligonucleotide transfection methods that cannot be safely translated in vivo. In this report, we demonstrate effective correction of a splicing mutation in the lung of a mouse model using SSOs. Moreover, we also demonstrate effective correction of a CFTR splicing mutation in a pre-clinical CF patient-derived cell model. We utilized a highly effective delivery strategy for oligonucleotides by combining peptide-morpholino (PPMO) SSOs with small molecules termed OECs. PPMOs distribute broadly into the lung and other tissues while OECs potentiate the effects of oligonucleotides by releasing them from endosomal entrapment. The combined PPMO plus OEC approach proved to be effective both in CF patient cells and in vivo in the mouse lung and thus may offer a path to the development of novel therapeutics for splicing mutations in CF and other lung diseases.
Collapse
Affiliation(s)
- Yan Dang
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Catharina van Heusden
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Veronica Nickerson
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Felicity Chung
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Yang Wang
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Nancy L Quinney
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Martina Gentzsch
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Hong M Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Ryszard Kole
- Department of Pharmacology, The University of North Carolina at Chapel Hill, 4010 Genetic Medicine Bldg, Chapel Hill, NC 27599, USA
| | - Aiguo Ni
- Initos Pharmaceuticals, LLC, Chapel Hill, NC 27514, USA
| | | | - Silvia M Kreda
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| |
Collapse
|
243
|
Budai-Szűcs M, Berkó S, Kovács A, Jaikumpun P, Ambrus R, Halász A, Szabó-Révész P, Csányi E, Zsembery Á. Rheological effects of hypertonic saline and sodium bicarbonate solutions on cystic fibrosis sputum in vitro. BMC Pulm Med 2021; 21:225. [PMID: 34253193 PMCID: PMC8276516 DOI: 10.1186/s12890-021-01599-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is a life-threatening multiorgan genetic disease, particularly affecting the lungs, where recurrent infections are the main cause of reduced life expectancy. In CF, mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) protein impair transepithelial electrolyte and water transport, resulting in airway dehydration, and a thickening of the mucus associated with abnormal viscoelastic properties. Our aim was to develop a rheological method to assess the effects of hypertonic saline (NaCl) and NaHCO3 on CF sputum viscoelasticity in vitro, and to identify the critical steps in sample preparation and in the rheological measurements. METHODS Sputum samples were mixed with hypertonic salt solutions in vitro in a ratio of either 10:4 or 10:1. Distilled water was applied as a reference treatment. The rheological properties of sputum from CF patients, and the effects of these in vitro treatments, were studied with a rheometer at constant frequency and strain, followed by frequency sweep tests, where storage modulus (G'), loss modulus (G″) and loss factor were determined. RESULTS We identified three distinct categories of sputum: (i) highly elastic (G' > 100,000 Pa), (ii) elastic (100,000 Pa > G' > 1000 Pa), and (iii) viscoelastic (G' < 1000). At the higher additive ratio (10:4), all of the added solutions were found to significantly reduce the gel strength of the sputum, but the most pronounced changes were observed with NaHCO3 (p < 0.001). Samples with high elasticity exhibited the greatest changes while, for less elastic samples, a weakening of the gel structure was observed when they were treated with water or NaHCO3, but not with NaCl. For the viscoelastic samples, the additives did not cause significant changes in the parameters. When the lower additive ratio (10:1) was used, the mean values of the rheological parameters usually decreased, but the changes were not statistically significant. CONCLUSION Based on the rheological properties of the initial sputum samples, we can predict with some confidence the treatment efficacy of each of the alternative additives. The marked differences between the three categories suggest that it is advisable to evaluate each sample individually using a rheological approach such as that described here.
Collapse
Affiliation(s)
- Mária Budai-Szűcs
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary.
| | - Szilvia Berkó
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Anita Kovács
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | | | - Rita Ambrus
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Adrien Halász
- National Korányi Institute for Pulmonology, Budapest, Hungary
| | - Piroska Szabó-Révész
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Erzsébet Csányi
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Ákos Zsembery
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
244
|
A Precision Medicine Approach to Optimize Modulator Therapy for Rare CFTR Folding Mutants. J Pers Med 2021; 11:jpm11070643. [PMID: 34357110 PMCID: PMC8307171 DOI: 10.3390/jpm11070643] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/23/2022] Open
Abstract
Trikafta, a triple-combination drug, consisting of folding correctors VX-661 (tezacaftor), VX-445 (elexacaftor) and the gating potentiator VX-770 (ivacaftor) provided unprecedented clinical benefits for patients with the most common cystic fibrosis (CF) mutation, F508del. Trikafta indications were recently expanded to additional 177 mutations in the CF transmembrane conductance regulator (CFTR). To minimize life-long pharmacological and financial burden of drug administration, if possible, we determined the necessary and sufficient modulator combination that can achieve maximal benefit in preclinical setting for selected mutants. To this end, the biochemical and functional rescue of single corrector-responsive rare mutants were investigated in a bronchial epithelial cell line and patient-derived human primary nasal epithelia (HNE), respectively. The plasma membrane density of P67L-, L206W- or S549R-CFTR corrected by VX-661 or other type I correctors was moderately increased by VX-445. Short-circuit current measurements of HNE, however, uncovered that correction comparable to Trikafta was achieved for S549R-CFTR by VX-661 + VX-770 and for P67L- and L206W-CFTR by the VX-661 + VX-445 combination. Thus, introduction of a third modulator may not provide additional benefit for patients with a subset of rare CFTR missense mutations. These results also underscore that HNE, as a precision medicine model, enable the optimization of mutation-specific modulator combinations to maximize their efficacy and minimize life-long drug exposure of CF patients.
Collapse
|
245
|
Trapnell BC, Chen S, Khurmi R, Bodhani A, Kapoor M, Haupt M. Hospitalization rates among patients with cystic fibrosis using pancreatic enzyme replacement therapy. Chron Respir Dis 2021; 17:1479973119900612. [PMID: 31984768 PMCID: PMC6985972 DOI: 10.1177/1479973119900612] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We investigated the relationship between self-reported adherence to pancreatic enzyme replacement therapy (PERT), nutritional status, and all-cause hospitalization in cystic fibrosis (CF) patients with a record of PERT use. Association of self-reported annual PERT use rate (adherence) with annual hospital admission rate (HAR) and annual total hospital nights (THNs) were analyzed for 5301 children (2000–2012) and 13,989 adults (2000–2013) from the CF Foundation Patient Registry. Multivariate linear regression was used to determine the association of HAR and THN with mean annual PERT use rate, cumulative PERT use rate, mean body mass index (BMI) (adult) or BMI percentile (pediatric), age, and sex. The median annual PERT use rate was 87% in children and 80% in adults. Statistically, higher annual PERT use, longer cumulative PERT, and higher BMI percentile (children) or BMI (adults) were significantly (p < 0.0001) associated with lower annual HAR and fewer annual THN in children and adults. Female sex was associated with higher annual HAR and more annual THN in children and adults (p < 0.05). Results indicate self-reported adherence to PERT, increased BMI, and male sex were associated with fewer hospital admissions and annual hospital nights in CF patients.
Collapse
Affiliation(s)
- Bruce C Trapnell
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Su Chen
- AbbVie Inc., North Chicago, IL, USA
| | | | | | | | - Mark Haupt
- AbbVie Inc., North Chicago, IL, USA.,ARIEL Precision Medicine, Pittsburgh, PA, USA
| |
Collapse
|
246
|
Carpentieri C, Farrow N, Cmielewski P, Rout-Pitt N, McCarron A, Knight E, Parsons D, Donnelley M. The Effects of Conditioning and Lentiviral Vector Pseudotype on Short- and Long-Term Airway Reporter Gene Expression in Mice. Hum Gene Ther 2021; 32:817-827. [PMID: 33947249 DOI: 10.1089/hum.2021.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A gene addition therapy into the conducting airway epithelium is a potential cure for cystic fibrosis lung disease. Achieving sustained lung gene expression has proven difficult due to the natural barriers of the lung. The development of lentiviral (LV) vectors pseudotyped with viral envelopes that have a natural tropism to the airway has enabled persistent gene expression to be achieved in vivo. The aims of this study were to compare the yields of hemagglutinin (HA) and vesicular stomatitis virus-glycoprotein (VSV-G) pseudotyped HIV-1 vectors produced under the same conditions by our standard LV vector production method. We then sought to measure gene expression in mouse airways and to determine whether lysophosphatidylcholine (LPC) conditioning enhances short- and long-term gene expression. C57Bl/6 mouse airways were conditioned with 10 μL of 0.1% LPC or saline control, followed 1 h later by a 30 μL dose of an HA or VSV-G pseudotyped vector carrying either the LacZ or luciferase reporter genes. LacZ expression was assessed by X-gal staining after 7 days, while lung luminescence was quantified regularly for up to 18 months by bioluminescent imaging. The HA pseudotyped vectors had functional titers 25 to 60 times lower than the VSV-G pseudotyped vectors. Conditioning the lung with LPC significantly increased the total number of LacZ-transduced cells for both pseudotypes compared to saline control. Regardless of LPC conditioning, the VSV-G pseudotype produced higher initial levels of gene expression compared to HA. LPC conditioning did not increase the number of transduced basal cells for either pseudotype compared to saline, and was not required for long-term gene expression. Both pseudotyped vectors effectively transduced the upper conducting airways of wild-type mice. The use of LPC conditioning before vector delivery was not required in mouse lungs to produce long-term gene expression, but did improve short-term gene expression.
Collapse
Affiliation(s)
- Chantelle Carpentieri
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Nigel Farrow
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Patricia Cmielewski
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Nathan Rout-Pitt
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Alexandra McCarron
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Emma Knight
- South Australian Health and Medical Research Institute, Adelaide, Australia.,School of Public Health, University of Adelaide, Adelaide, Australia
| | - David Parsons
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Martin Donnelley
- Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
247
|
Both Pseudomonas aeruginosa and Candida albicans Accumulate Greater Biomass in Dual-Species Biofilms under Flow. mSphere 2021; 6:e0041621. [PMID: 34160236 PMCID: PMC8265656 DOI: 10.1128/msphere.00416-21] [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] [Indexed: 12/12/2022] Open
Abstract
Microbe-microbe interactions can strongly influence growth and biofilm formation kinetics. For Pseudomonas aeruginosa and Candida albicans, which are found together in diverse clinical sites, including urinary and intravenous catheters and the lungs of individuals with cystic fibrosis (CF), we compared the kinetics of biofilm formation by each species in dual-species and single-species biofilms. We engineered fluorescent protein constructs for P. aeruginosa (producing mKO-κ) and C. albicans (producing mKate2) that did not alter growth and enabled single-cell resolution imaging by live-sample microscopy. Using these strains in an optically clear derivative of synthetic CF sputum medium, we found that both P. aeruginosa and C. albicans displayed increased biovolume accumulation—by three- and sixfold, respectively—in dual-species biofilms relative to single-species biofilms. This result was specific to the biofilm environment, as enhanced growth was not observed in planktonic cocultures. Stimulation of C. albicans biofilm formation occurred regardless of whether P. aeruginosa was added at the time of fungal inoculation or 24 h after the initiation of biofilm development. P. aeruginosa biofilm increases in cocultures did not require the Pel extracellular polysaccharide, phenazines, and siderophores known to influence C. albicans. P. aeruginosa mutants lacking Anr, LasR, and BapA were not significantly stimulated by C. albicans, but they still promoted a significant enhancement of biofilm development of the fungus, suggesting a fungal response to the presence of bacteria. Last, we showed that a set of P. aeruginosa clinical isolates also prompted an increase of biovolume by C. albicans in coculture. IMPORTANCE There is an abundance of work on both P. aeruginosa and C. albicans in isolation, and quite some work as well on the way these two microbes interact. These studies do not, however, consider biofilm environments under flow, and our results here show that the expected outcome of interaction between these two pathogens can actually be reversed under flow, from pure antagonism to an increase in biomass on the part of both. Our work also highlights the importance of cellular-scale spatial structure in biofilms for understanding multispecies population dynamics.
Collapse
|
248
|
Walton NI, Zhang X, Soltis AR, Starr J, Dalgard CL, Wilkerson MD, Conrad D, Pollard HB. Tensin 1 (TNS1) is a modifier gene for low body mass index (BMI) in homozygous [F508del]CFTR patients. Physiol Rep 2021; 9:e14886. [PMID: 34086412 PMCID: PMC8176904 DOI: 10.14814/phy2.14886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/02/2021] [Indexed: 11/24/2022] Open
Abstract
Cystic fibrosis (CF) is a life‐limiting autosomal recessive genetic disease caused by variants in the CFTR gene, most commonly by the [F508del] variant. Although CF is a classical Mendelian disease, genetic variants in several modifier genes have been associated with variation of the clinical phenotype for pulmonary and gastrointestinal function and urogenital development. We hypothesized that whole genome sequencing of a well‐phenotyped CF populations might identify novel variants in known, or hitherto unknown, modifier genes. Whole genome sequencing was performed on the Illumina HiSeq X platform for 98 clinically diagnosed cystic fibrosis patient samples from the Adult CF Clinic at the University of California San Diego (UCSD). We compared protein‐coding, non‐silent variants genome wide between CFTR [F508del] homozygotes vs CFTR compound heterozygotes. Based on a single variant score test, we found 3 SNPs in common variants (MAF >5%) that occurred at significantly different rates between homozygous [F508del]CFTR and compound heterozygous [F508del]CFTR patients. The 3 SNPs were all located in one gene on chromosome 2: Tensin 1 (TNS1: rs3796028; rs2571445: and rs918949). We observed significantly lower BMIs in homozygous [F508del]CFTR patients who were also homozygous for Tensin 1 rs918949 (T/T) (p = 0.023) or rs2571445 (G/G) (p = 0.02) variants. The Tensin 1 gene is thus a potential modifier gene for low BMI in CF patients homozygous for the [F508del]CFTR variant.
Collapse
Affiliation(s)
- Nathan I Walton
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Consortium for Health and Military Performance, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Xijun Zhang
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Anthony R Soltis
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Joshua Starr
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Clifton L Dalgard
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Matthew D Wilkerson
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Douglas Conrad
- Department of Medicine, University of California, San Diego, CA, USA
| | - Harvey B Pollard
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Consortium for Health and Military Performance, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
249
|
Aslanhan U, Cakir E, Pur Ozyigit L, Kucuksezer UC, Gelmez YM, Yuksel M, Deniz G, Cetin Aktas E. Pseudomonas aeruginosa colonization in cystic fibrosis: Impact on neutrophil functions and cytokine secretion capacity. Pediatr Pulmonol 2021; 56:1504-1513. [PMID: 33512090 DOI: 10.1002/ppul.25294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 01/03/2021] [Accepted: 01/22/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Chronic colonization with Pseudomonas (P.) aeruginosa worsens the prognosis of cystic fibrosis (CF) patients. This study aims to analyze the functional properties of neutrophils in CF patients with P. aeruginosa colonization. METHODS Patients with CF (n = 16) were grouped by positivity of P. aeruginosa in sputum culture, as positive (P.+) or negative (P.-), then compared with age and sex matched healthy controls (n = 8). Adhesion molecules, apoptotic index, intracellular CAP-18, interleukin 8 (IL-8), and tumor necrosis factor α (TNF-α) levels of neutrophils, following P. aeruginosa and lipopolysaccharides (LPS) stimulation, were analyzed by flow cytometry. IL-1β, IL-6, TNF-α, and IL-17 plasma levels were determined by Luminex. RESULTS Patients with CF had increased phagocytosis of Escherichia coli and P. aeruginosa, upregulated oxidative burst and chemotaxis. Increased neutrophil apoptosis was noted in CF patients. In unstimulated conditions, higher levels of CD16+ TNF-α+ and CD16+ IL-8+ neutrophils were determined, whereas bacteria and LPS stimulation significantly decreased secretion of CAP-18 from CD16+ neutrophils of CF patients. Plasma levels of IL-1β, TNF-α and IL-17 in P.+ patients were higher than in P.- group. CONCLUSION Our findings confirm inadequate neutrophil defense towards pathogens in CF. A significant difference in migration, phagocytosis, oxidative burst, percentage of IL-8 producing neutrophils, IL-1β, TNF-α, and IL-17 secretions were noted among CF patients according to their colonization status, which might induce a further destructive effect on airways, resulting in an unfavorable prognosis for children with CF who also have colonization.
Collapse
Affiliation(s)
- Umit Aslanhan
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.,Department of Immunology, Institute of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Erkan Cakir
- Department of Pediatric Pulmonology, Bezmialem Vakif University Medical Faculty, Istanbul, Turkey
| | - Leyla Pur Ozyigit
- Department of Allergy and Immunology, University Hospitals of Leicester, Leicester, UK
| | - Umut Can Kucuksezer
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Yusuf Metin Gelmez
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Mine Yuksel
- Department of Pediatric Pulmonology, Bezmialem Vakif University Medical Faculty, Istanbul, Turkey
| | - Gunnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Esin Cetin Aktas
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
250
|
Quantification of Phenotypic Variability of Lung Disease in Children with Cystic Fibrosis. Genes (Basel) 2021; 12:genes12060803. [PMID: 34070354 PMCID: PMC8229033 DOI: 10.3390/genes12060803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/15/2021] [Accepted: 05/19/2021] [Indexed: 12/28/2022] Open
Abstract
Cystic fibrosis (CF) lung disease has the greatest impact on the morbidity and mortality of patients suffering from this autosomal-recessive multiorgan disorder. Although CF is a monogenic disorder, considerable phenotypic variability of lung disease is observed in patients with CF, even in those carrying the same mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene or CFTR mutations with comparable functional consequences. In most patients with CF, lung disease progresses from childhood to adulthood, but is already present in infants soon after birth. In addition to the CFTR genotype, the variability of early CF lung disease can be influenced by several factors, including modifier genes, age at diagnosis (following newborn screening vs. clinical symptoms) and environmental factors. The early onset of CF lung disease requires sensitive, noninvasive measures to detect and monitor changes in lung structure and function. In this context, we review recent progress with using multiple-breath washout (MBW) and lung magnetic resonance imaging (MRI) to detect and quantify CF lung disease from infancy to adulthood. Further, we discuss emerging data on the impact of variability of lung disease severity in the first years of life on long-term outcomes and the potential use of this information to improve personalized medicine for patients with CF.
Collapse
|