1
|
Caballero I, Mbouamboua Y, Weise S, López-Gálvez R, Couralet M, Fleurot I, Pons N, Barrera-Conde M, Quílez-Playán N, Keller M, Klymiuk N, Robledo P, Hummel T, Barbry P, Chamero P. Cystic fibrosis alters the structure of the olfactory epithelium and the expression of olfactory receptors affecting odor perception. SCIENCE ADVANCES 2025; 11:eads1568. [PMID: 40020072 PMCID: PMC11870070 DOI: 10.1126/sciadv.ads1568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/27/2025] [Indexed: 03/03/2025]
Abstract
A reduced sense of smell is a common condition in people with cystic fibrosis (CF) that negatively affects their quality of life. While often attributed to nasal mucosa inflammation, the underlying causes of the olfactory loss remain unknown. Here, we characterized gene expression in olfactory epithelium cells from patients with CF using single-nuclei RNA sequencing and found altered expression of olfactory receptors (ORs) and genes related to progenitor cell proliferation. We confirmed these findings in newborn, inflammation-free samples of a CF animal model and further identified ultrastructural alterations in the olfactory epithelium and bulbs of these animals. We established that CFTR, the anion channel whose dysfunction causes CF, is dispensable for odor-evoked signaling in sensory neurons, yet CF animals displayed defective odor-guided behaviors consistent with the morphological and molecular alterations. Our study highlights CF's major role in modulating epithelial structure and OR expression, shedding light on the mechanisms contributing to olfactory loss in CF.
Collapse
Affiliation(s)
- Ignacio Caballero
- Infectiologie et Santé Publique, UMR1282, INRAE, University of Tours, 37380 Nouzilly, France
| | - Yvon Mbouamboua
- Université Côte d’Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Susanne Weise
- Department of Otorhinolaryngology, Smell & Taste Clinic, Carl Gustav Carus University Hospital, Technische Universität Dresden, 01307 Dresden, Germany
| | - Raquel López-Gálvez
- Infectiologie et Santé Publique, UMR1282, INRAE, University of Tours, 37380 Nouzilly, France
- Laboratoire de Physiologie de la Reproduction et des Comportements, CNRS, INRAE, University of Tours, 37380 Nouzilly, France
| | - Marie Couralet
- Université Côte d’Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Isabelle Fleurot
- Infectiologie et Santé Publique, UMR1282, INRAE, University of Tours, 37380 Nouzilly, France
| | - Nicolas Pons
- Université Côte d’Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Marta Barrera-Conde
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, 08003 Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Nayima Quílez-Playán
- Laboratoire de Physiologie de la Reproduction et des Comportements, CNRS, INRAE, University of Tours, 37380 Nouzilly, France
| | - Matthieu Keller
- Laboratoire de Physiologie de la Reproduction et des Comportements, CNRS, INRAE, University of Tours, 37380 Nouzilly, France
| | - Nikolai Klymiuk
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Center for Innovative Medical Models, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Patricia Robledo
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, IMIM-Hospital del Mar Research Institute, 08003 Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Thomas Hummel
- Department of Otorhinolaryngology, Smell & Taste Clinic, Carl Gustav Carus University Hospital, Technische Universität Dresden, 01307 Dresden, Germany
| | - Pascal Barbry
- Université Côte d’Azur, CNRS, INSERM, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
- 3IA-Côte d’Azur, 06560 Sophia Antipolis, France
- IHU Respirera, 06560 Sophia Antipolis, France
| | - Pablo Chamero
- Laboratoire de Physiologie de la Reproduction et des Comportements, CNRS, INRAE, University of Tours, 37380 Nouzilly, France
| |
Collapse
|
2
|
Li Y. The expression of MUC5AC in patients with rhinosinusitis: A systematic review and meta-analysis. Clin Transl Allergy 2024; 14:e70003. [PMID: 39482799 PMCID: PMC11527733 DOI: 10.1002/clt2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND To understand the connection between Muc5AC expression and the likelihood of rhinosinusitis, with the goal of providing insights into its prospective use as a biomarker. METHODS We searched PubMed, Embase, Cochrane Library, China National Knowledge Infrastructure, and Wanfang databases for studies up to November 2023 to conduct a literature review. After screening and quality assessment, eligible studies meeting the criteria were included. Muc5AC expression and rhinosinusitis association was analyzed by STATA 14.0. RESULTS Including weighted mean difference and 95% confidence interval, were reported. The meta-analysis included 16 studies with 1448 rhinosinusitis patients. MUC5AC expression was significantly up-regulated in both chronic rhinosinusitis with nasal polyps (CRSwNP; WMD: 0.52; 95% CI: 0.41-0.63) and chronic rhinosinusitis without nasal polyps (CRSsNP; WMD: 0.42; 95% CI: 0.28-0.56) patients compared to controls. IHC positive area analysis corroborated these findings, with elevated MUC5AC levels in CRSwNP (WMD: 25.61; 95% CI: 22.41-28.81) and CRSsNP (WMD: 39.74; 95% CI: 25.6-53.88) patients. Subgroup analysis based on tissue type (nasal tissue fluid and sinus mucosa) consistently supported the overall results. CONCLUSION Our meta-analysis robustly demonstrates a significant association between elevated MUC5AC expression and rhinosinusitis risk. This finding underscores the potential of MUC5AC as a molecular marker, providing valuable insights for future research and potential therapeutic interventions in rhinosinusitis management. SYSTEMATIC REVIEW REGISTRATION CRD42024518932.
Collapse
Affiliation(s)
- Yitao Li
- Department of Otolaryngology Head and Neck SurgeryFirst Hospital of Hainan Medical CollegeHaikouChina
| |
Collapse
|
3
|
Abou Alaiwa MA, Hilkin BM, Price MP, Gansemer ND, Rector MR, Stroik MR, Powers LS, Whitworth KM, Samuel MS, Jain A, Ostedgaard LS, Ernst SE, Philibert W, Boyken LD, Moninger TO, Karp PH, Hornick DB, Sinn PL, Fischer AJ, Pezzulo AA, McCray PB, Meyerholz DK, Zabner J, Prather RS, Welsh MJ, Stoltz DA. Development and Initial Characterization of Pigs with DNAI1 Mutations and Primary Ciliary Dyskinesia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.594822. [PMID: 39229081 PMCID: PMC11370470 DOI: 10.1101/2024.05.22.594822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Mutations in more than 50 different genes cause primary ciliary dyskinesia (PCD) by disrupting the activity of motile cilia that facilitate mucociliary transport (MCT). Knowledge of PCD has come from studies identifying disease-causing mutations, characterizing structural cilia abnormalities, finding genotype-phenotype relationships, and studying the cell biology of cilia. Despite these important findings, we still lack effective treatments and people with PCD have significant pulmonary impairment. As with many other diseases, a better understanding of pathogenic mechanisms may lead to effective treatments. To pursue disease mechanisms, we used CRISPR-Cas9 to develop a PCD pig with a disrupted DNAI1 gene. PCD pig airway cilia lacked the outer dynein arm and had impaired beating. MCT was impaired under both baseline conditions and after cholinergic stimulation in PCD pigs. Neonatal PCD pigs developed neonatal respiratory distress with evidence of atelectasis, air trapping, and airway mucus obstruction. Despite airway mucus accumulation, lung bacterial counts were similar between neonatal wild-type and PCD pigs. Sinonasal disease was present in all neonatal PCD pigs. Older PCD pigs developed worsening airway mucus obstruction, inflammation, and bacterial infection. This pig model closely mimics the disease phenotype seen in people with PCD and can be used to better understand the pathophysiology of PCD airway disease.
Collapse
Affiliation(s)
- Mahmoud A. Abou Alaiwa
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242
| | - Brie M. Hilkin
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Margaret P. Price
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Nicholas D. Gansemer
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Michael R. Rector
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Mal R. Stroik
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Linda S. Powers
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | | | - Melissa S. Samuel
- Division of Animal Sciences, University of Missouri, Columbia, Missouri 65211
| | - Akansha Jain
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Lynda S. Ostedgaard
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Sarah E. Ernst
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Winter Philibert
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242
| | - Linda D. Boyken
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Thomas O. Moninger
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Phillip H. Karp
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Douglas B. Hornick
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Patrick L. Sinn
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Anthony J. Fischer
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Alejandro A. Pezzulo
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Paul B. McCray
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - David K. Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Joseph Zabner
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| | - Randy S. Prather
- Division of Animal Sciences, University of Missouri, Columbia, Missouri 65211
| | - Michael J. Welsh
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa 52242
| | - David A. Stoltz
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
4
|
Meyerholz DK, Burrough ER, Kirchhof N, Anderson DJ, Helke KL. Swine models in translational research and medicine. Vet Pathol 2024; 61:512-523. [PMID: 38197394 DOI: 10.1177/03009858231222235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Swine are increasingly studied as animal models of human disease. The anatomy, size, longevity, physiology, immune system, and metabolism of swine are more like humans than traditional rodent models. In addition, the size of swine is preferred for surgical placement and testing of medical devices destined for humans. These features make swine useful for biomedical, pharmacological, and toxicological research. With recent advances in gene-editing technologies, genetic modifications can readily and efficiently be made in swine to study genetic disorders. In addition, gene-edited swine tissues are necessary for studies testing and validating xenotransplantation into humans to meet the critical shortfall of viable organs versus need. Underlying all of these biomedical applications, the knowledge of husbandry, background diseases and lesions, and biosecurity needs are important for productive, efficient, and reproducible research when using swine as a human disease model for basic research, preclinical testing, and translational studies.
Collapse
|
5
|
Kaluzna-Mlynarczyk A, Pucher B, Sroczynski J, Kotowski M, Jonczyk-Potoczna K, Szydlowski J. The development of paranasal sinuses in patients with cystic fibrosis: sinuses volume analysis. Eur Arch Otorhinolaryngol 2024; 281:795-803. [PMID: 37882847 PMCID: PMC10796702 DOI: 10.1007/s00405-023-08236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 09/08/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) is a severe systemic disease that affects many aspects of patients' lives. It is known that the progression of the disease adversely affects lower and upper airways including the paranasal sinuses. However, its impact on sinus development in the pediatric population is not fully examined. The purpose of this study was to evaluate the development of the paranasal sinuses in a pediatric population with CF and compare it to a control group consisting of healthy children. METHODS The results of computed tomography (CT) scans of children with the disease and the control group were evaluated. The study included 114 CT images of children in the study group and 126 images of healthy children aged 0-18 years. The volumes of maxillary, frontal, and sphenoid sinuses were analyzed. The obtained results were compared with those of the control group and analyzed statistically. RESULTS The volume and the development of the paranasal sinuses in both groups increased with age, but statistically significant differences were found between the study and the control group. CONCLUSIONS The obtained results provide valuable knowledge regarding the impact of the CF on sinuses development. Also, they may be important in understanding the progression of the disease and its influence on the quality and length of life of patients. The results may contribute to enhanced diagnostics and have implications for improving therapy for patients with chronic sinusitis associated with CF.
Collapse
Affiliation(s)
- Agata Kaluzna-Mlynarczyk
- Department of Pediatric Otolaryngology, Institute of Pediatrics, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572, Poznań, Poland.
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812, Poznań, Poland.
| | - Beata Pucher
- Department of Pediatric Otolaryngology, Institute of Pediatrics, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572, Poznań, Poland
| | - Jakub Sroczynski
- Department of Pediatric Otolaryngology, Institute of Pediatrics, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572, Poznań, Poland
| | - Michal Kotowski
- Department of Pediatric Otolaryngology, Institute of Pediatrics, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572, Poznań, Poland
| | - Katarzyna Jonczyk-Potoczna
- Department of Pediatric Radiology, Institute of Pediatrics, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572, Poznań, Poland
| | - Jarosław Szydlowski
- Department of Pediatric Otolaryngology, Institute of Pediatrics, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572, Poznań, Poland
| |
Collapse
|
6
|
Meyerholz DK, Leidinger MR, Adam Goeken J, Businga TR, Vizuett S, Akers A, Evans I, Zhang Y, Engelhardt JF. Immunohistochemical detection of MUC5AC and MUC5B mucins in ferrets. BMC Res Notes 2023; 16:111. [PMID: 37349833 PMCID: PMC10286488 DOI: 10.1186/s13104-023-06388-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVE Cystic fibrosis (CF) is a genetic condition that causes abnormal mucus secretions in affected organs. MUC5AC and MUC5B are gel-forming mucins and frequent targets for investigations in CF tissues. Our objective was to qualify MUC5AC and MUC5B immunohistochemical techniques to provide a useful tool to identify, localize and interpret mucin expression in ferret tissues. RESULTS MUC5AC and MUC5B mucins were detected most commonly in large airways and least in small airways, consistent with reported goblet cell density in airway surface epithelia. We evaluated whether staining method affected the detection of goblet cell mucins in serial sections of bronchial surface epithelia. Significant differences between stains were not observed suggesting common co-expression MUC5AC and MUC5B proteins in goblet cells of airway surface epithelia. Gallbladder and stomach tissues are reported to have differential mucin enrichment, so we tested these tissues in wildtype ferrets. Stomach tissues were enriched in MUC5AC and gallbladder tissues enriched in MUC5B, mucin enrichment similar to human tissues. Mucin immunostaining techniques were further qualified for specificity using lung tissue from recently generated MUC5AC-/- and MUC5B-/- ferrets. Qualified techniques for MUC5AC and MUC5B immunohistochemistry will be useful tools for mucin tissue studies in CF and other ferret models.
Collapse
Affiliation(s)
- David K. Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Mariah R. Leidinger
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - J. Adam Goeken
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Thomas R. Businga
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Sebastian Vizuett
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Allison Akers
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Idil Evans
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - Yan Zhang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242 USA
| |
Collapse
|
7
|
Darwish T, Al-Khulaifi A, Ali M, Mowafy R, Arredouani A, Doi SA, Emara MM. Assessing the consistency of iPSC and animal models in cystic fibrosis modelling: A meta-analysis. PLoS One 2022; 17:e0272091. [PMID: 35944004 PMCID: PMC9362911 DOI: 10.1371/journal.pone.0272091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 07/12/2022] [Indexed: 12/09/2022] Open
Abstract
INTRODUCTION Cystic fibrosis (CF) is a hereditary autosomal recessive disorder caused by a range of mutations in the CF Transmembrane Conductance Regulator (CFTR) gene. This gene encodes the CFTR protein, which acts as a chloride channel activated by cyclic AMP (cAMP). This meta-analysis aimed to compare the responsiveness of induced pluripotent stem cells (iPSCs) to cAMP analogues to that of commonly used animal models. METHODS Databases searched included PubMed, Scopus, and Medline from inception to January 2020. A total of 8 and 3 studies, respectively, for animal models and iPSCs, were analyzed. Studies were extracted for investigating cAMP-stimulated anion transport by measuring the short circuit current (Isc) of chloride channels in different animal models and iPSC systems We utilized an inverse variance heterogeneity model for synthesis. RESULTS Our analysis showed considerable heterogeneity in the mean Isc value in both animal models and iPSCs studies (compared to their WT counterparts), and both suffer from variable responsiveness based on the nature of the underlying model. There was no clear advantage of one over the other. CONCLUSIONS Studies on both animal and iPSCs models generated considerable heterogeneity. Given the potential of iPSC-derived models to study different diseases, we recommend paying more attention to developing reproducible models of iPSC as it has potential if adequately developed.
Collapse
Affiliation(s)
- Toqa Darwish
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Azhar Al-Khulaifi
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Menatalla Ali
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Rana Mowafy
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Abdelilah Arredouani
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, Doha, Qatar
| | - Suhail A. Doi
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mohamed M. Emara
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| |
Collapse
|
8
|
Grubb BR, Livraghi-Butrico A. Animal models of cystic fibrosis in the era of highly effective modulator therapies. Curr Opin Pharmacol 2022; 64:102235. [DOI: 10.1016/j.coph.2022.102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022]
|
9
|
Lunney JK, Van Goor A, Walker KE, Hailstock T, Franklin J, Dai C. Importance of the pig as a human biomedical model. Sci Transl Med 2021; 13:eabd5758. [PMID: 34818055 DOI: 10.1126/scitranslmed.abd5758] [Citation(s) in RCA: 329] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Angelica Van Goor
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Kristen E Walker
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Taylor Hailstock
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Jasmine Franklin
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Chaohui Dai
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA.,College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
10
|
Bouzek DC, Abou Alaiwa MH, Adam RJ, Pezzulo AA, Reznikov LR, Cook DP, Aguilar Pescozo MI, Ten Eyck P, Wu C, Gross TJ, Hornick DB, Hoffman EA, Meyerholz DK, Stoltz DA. Early Lung Disease Exhibits Bacterial-Dependent and -Independent Abnormalities in Cystic Fibrosis Pigs. Am J Respir Crit Care Med 2021; 204:692-702. [PMID: 34170795 DOI: 10.1164/rccm.202102-0451oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE While it is clear that cystic fibrosis airway disease begins at a very young age, the early and subsequent steps in disease pathogenesis and the relative contribution of infection, mucus, and inflammation are not well understood. OBJECTIVES As one approach to assessing the early contribution of infection, we tested the hypothesis that early and continuous antibiotics would decrease the airway bacterial burden. We thought that, if it does, it might reveal aspects of the disease that are more or less sensitive to decreasing infection. METHODS Three groups of pigs were studied from birth until ~3 weeks of age: 1) wild-type, 2) cystic fibrosis, and 3) cystic fibrosis pigs treated continuously with broad-spectrum antibiotics from birth until study completion. Disease was assessed with chest computed tomography, histopathology, microbiology, and bronchoalveolar lavage. MEASUREMENTS AND MAIN RESULTS Disease was present by 3 weeks of age in cystic fibrosis pigs. Continuous antibiotics from birth improved chest computed tomography imaging abnormalities and airway mucus accumulation, but not airway inflammation in the cystic fibrosis pig model. However, reducing bacterial infection did not improve two disease features already present at birth in cystic fibrosis pigs, air trapping and submucosal gland duct plugging. In the cystic fibrosis sinuses, antibiotics did not prevent the development of infection, disease, or the number of bacteria but did alter the bacterial species. CONCLUSIONS These findings suggest that cystic fibrosis airway disease begins immediately following birth, and that early and continuous antibiotics impact some, but not all, aspects of CF lung disease development.
Collapse
Affiliation(s)
- Drake C Bouzek
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Mahmoud H Abou Alaiwa
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Internal Medicine, Iowa City, Iowa, United States
| | - Ryan J Adam
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Alejandro A Pezzulo
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Internal Medicine, Iowa City, Iowa, United States
| | - Leah R Reznikov
- University of Florida, 3463, Physiological Sciences, Gainesville, Florida, United States
| | - Daniel P Cook
- Vanderbilt University Medical Center, 12328, Department of Medicine, Nashville, Tennessee, United States
| | - Maria I Aguilar Pescozo
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Patrick Ten Eyck
- The University of Iowa, 4083, Institute for Clinical and Translational Science, Iowa City, Iowa, United States
| | - Chaorong Wu
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Thomas J Gross
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Douglas B Hornick
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States
| | - Eric A Hoffman
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Radiology, Iowa City, Iowa, United States
| | - David K Meyerholz
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Pathology, Iowa City, Iowa, United States
| | - David A Stoltz
- The University of Iowa Roy J and Lucille A Carver College of Medicine, 12243, Iowa City, Iowa, United States;
| |
Collapse
|
11
|
Helke KL, Meyerholz DK, Beck AP, Burrough ER, Derscheid RJ, Löhr C, McInnes EF, Scudamore CL, Brayton CF. Research Relevant Background Lesions and Conditions: Ferrets, Dogs, Swine, Sheep, and Goats. ILAR J 2021; 62:133-168. [PMID: 33712827 DOI: 10.1093/ilar/ilab005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/17/2020] [Accepted: 01/06/2021] [Indexed: 01/01/2023] Open
Abstract
Animal models provide a valuable tool and resource for biomedical researchers as they investigate biological processes, disease pathogenesis, novel therapies, and toxicologic studies. Interpretation of animal model data requires knowledge not only of the processes/diseases being studied but also awareness of spontaneous conditions and background lesions in the model that can influence or even confound the study results. Species, breed/stock, sex, age, anatomy, physiology, diseases (noninfectious and infectious), and neoplastic processes are model features that can impact the results as well as study interpretation. Here, we review these features in several common laboratory animal species, including ferret, dog (beagle), pig, sheep, and goats.
Collapse
Affiliation(s)
- Kristi L Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Amanda P Beck
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eric R Burrough
- Veterinary Diagnostic and Production Animal Medicine Department, Iowa State University, Ames, Iowa, USA
| | - Rachel J Derscheid
- Veterinary Diagnostic and Production Animal Medicine Department, Iowa State University, Ames, Iowa, USA
| | - Christiane Löhr
- Department of Biomedical Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Elizabeth F McInnes
- Toxicologic Pathology, Toxicology Section, Human Safety at Syngenta, in Jealott's Hill, Bracknell, United Kingdom
| | - Cheryl L Scudamore
- ExePathology, Pathologist at ExePathology, Exmouth, Devon, United Kingdom
| | - Cory F Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Maggiore G, Pietragalla M, De Amicis C, Nardi C, Bruno C, Gallo O, Bonasera L, Perrone A, Cavallo A, Colagrande S, Taccetti G, Locatello LG. The Risks of Complications During Endoscopic Sinus Surgery in Cystic Fibrosis Patients: An Anatomical and Endoscopic Study. Laryngoscope 2021; 131:E2481-E2489. [PMID: 33464574 DOI: 10.1002/lary.29404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE/HYPOTHESIS An increasing proportion of adult cystic fibrosis (CF) patients is being referred to endoscopic sinus surgery (ESS) in order to relieve the symptoms of chronic rhinosinusitis (CRS). Given that CFTR mutations profoundly alter sinonasal development, we want to explore the relationship between their peculiar surgical anatomy and the risk of postoperative complications. STUDY DESIGN Retrospective case-control study. METHODS Paranasal sinuses CT scans of 103 CF adult patients with CRS were compared to those belonging to a cohort of 100 non-CF adult patients to explore their anatomical differences. Secondly, CF and non-CF patients who received primary/revision ESS were analyzed in order to assess their preoperative CT scan in terms of surgically relevant variants, and according to the CLOSE checklist. Surgical outcomes were statistically compared in order to explore the differences between groups. RESULTS CF group presented more frequently with smaller and less pneumatized paranasal sinuses and a higher Lund-Mckay score compared with controls. No anatomical differences emerged in terms of genotype stratification. Non-CF CRS patients undergoing ESS showed a significantly deeper olfactory fossa and a more frequent supraorbital pneumatization compared to CF patients (P < .001 and P = .031, respectively). Whereas this latter group underwent more often aggressive surgical procedures (P = .001), no difference in terms of postoperative adverse events was found (P = .620). CONCLUSIONS Despite receiving more often aggressive ESS procedures, adult CF patients do not show an increased risk of postoperative complication and this may be linked to a different proportion of anatomical and surgically-relevant variants. LEVEL OF EVIDENCE 4 Laryngoscope, 131:E2481-E2489, 2021.
Collapse
Affiliation(s)
| | - Michele Pietragalla
- Department of Biomedical, Experimental and Clinical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Careggi University Hospital, Florence, Italy
| | - Christian De Amicis
- Department of Biomedical, Experimental and Clinical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Careggi University Hospital, Florence, Italy
| | - Cosimo Nardi
- Department of Biomedical, Experimental and Clinical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Careggi University Hospital, Florence, Italy
| | - Chiara Bruno
- Department of Otorhinolaryngology, Careggi University Hospital, Florence, Italy
| | - Oreste Gallo
- Department of Otorhinolaryngology, Careggi University Hospital, Florence, Italy
| | - Luigi Bonasera
- Department of Radiology, Careggi University Hospital, Florence, Italy
| | - Anna Perrone
- Department of Radiology, Anna Meyer Children's University Hospital, Florence, Italy
| | - Annalisa Cavallo
- Department of Infectious and Tropical Diseases, Careggi University Hospital, Florence, Italy
| | - Stefano Colagrande
- Department of Biomedical, Experimental and Clinical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Careggi University Hospital, Florence, Italy
| | - Giovanni Taccetti
- Cystic Fibrosis Center, Anna Meyer Children's University Hospital, University of Florence, Florence, Italy
| | | |
Collapse
|
13
|
Xu J, Livraghi-Butrico A, Hou X, Rajagopalan C, Zhang J, Song J, Jiang H, Wei HG, Wang H, Bouhamdan M, Ruan J, Yang D, Qiu Y, Xie Y, Barrett R, McClellan S, Mou H, Wu Q, Chen X, Rogers TD, Wilkinson KJ, Gilmore RC, Esther CR, Zaman K, Liang X, Sobolic M, Hazlett L, Zhang K, Frizzell RA, Gentzsch M, O'Neal WK, Grubb BR, Chen YE, Boucher RC, Sun F. Phenotypes of CF rabbits generated by CRISPR/Cas9-mediated disruption of the CFTR gene. JCI Insight 2021; 6:139813. [PMID: 33232302 PMCID: PMC7821608 DOI: 10.1172/jci.insight.139813] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Existing animal models of cystic fibrosis (CF) have provided key insights into CF pathogenesis but have been limited by short lifespans, absence of key phenotypes, and/or high maintenance costs. Here, we report the CRISPR/Cas9-mediated generation of CF rabbits, a model with a relatively long lifespan and affordable maintenance and care costs. CF rabbits supplemented solely with oral osmotic laxative had a median survival of approximately 40 days and died of gastrointestinal disease, but therapeutic regimens directed toward restoring gastrointestinal transit extended median survival to approximately 80 days. Surrogate markers of exocrine pancreas disorders were found in CF rabbits with declining health. CFTR expression patterns in WT rabbit airways mimicked humans, with widespread distribution in nasal respiratory and olfactory epithelia, as well as proximal and distal lower airways. CF rabbits exhibited human CF–like abnormalities in the bioelectric properties of the nasal and tracheal epithelia. No spontaneous respiratory disease was detected in young CF rabbits. However, abnormal phenotypes were observed in surviving 1-year-old CF rabbits as compared with WT littermates, and these were especially evident in the nasal respiratory and olfactory epithelium. The CF rabbit model may serve as a useful tool for understanding gut and lung CF pathogenesis and for the practical development of CF therapeutics.
Collapse
Affiliation(s)
- Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan (UM) Medical Center, Ann Arbor, Michigan, USA
| | | | | | | | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan (UM) Medical Center, Ann Arbor, Michigan, USA
| | - Jun Song
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan (UM) Medical Center, Ann Arbor, Michigan, USA
| | | | | | - Hui Wang
- Department of Oncology, Karmanos Cancer Institute
| | | | - Jinxue Ruan
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan (UM) Medical Center, Ann Arbor, Michigan, USA
| | - Dongshan Yang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan (UM) Medical Center, Ann Arbor, Michigan, USA
| | - Yining Qiu
- Center for Molecular Medicine and Genetics, and
| | - Youming Xie
- Department of Oncology, Karmanos Cancer Institute
| | - Ronald Barrett
- Department of Anatomy and Cell Biology, Wayne State University (WSU) School of Medicine, Detroit, Michigan, USA
| | - Sharon McClellan
- Department of Anatomy and Cell Biology, Wayne State University (WSU) School of Medicine, Detroit, Michigan, USA
| | - Hongmei Mou
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | - Troy D Rogers
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Kristen J Wilkinson
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Rodney C Gilmore
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Charles R Esther
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Khalequz Zaman
- Department of Pediatrics, Case Western Research University School of Medicine, Cleveland, Ohio, USA
| | - Xiubin Liang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan (UM) Medical Center, Ann Arbor, Michigan, USA
| | | | - Linda Hazlett
- Department of Anatomy and Cell Biology, Wayne State University (WSU) School of Medicine, Detroit, Michigan, USA
| | | | - Raymond A Frizzell
- Department of Pediatrics and Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvnia, USA
| | - Martina Gentzsch
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Wanda K O'Neal
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Barbara R Grubb
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan (UM) Medical Center, Ann Arbor, Michigan, USA
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
14
|
Abstract
Background: Paranasal sinus pneumatization is a complex process, and numerous computed tomography (CT) studies document developmental variations in the setting of underlying sinus disease. The purpose of this study was to investigate variation in paranasal sinus pneumatization in a population of nondiseased subjects using a metric validated for tracking individual anatomic variants as well as total sinus volume. Methods: A total of 591 consecutive sinus and maxillofacial CT scans were considered for study inclusion. After patients with inflammatory sinus or respiratory disease were excluded, 323 CT scans were scored using the Assessment of Pneumatization of the Paranasal Sinuses (APPS) instrument, and relevant demographic data were recorded for each scan. APPS findings were compared according to demographic characteristics and laterality. Results: Laterality and sex were associated with differences in paranasal sinus pneumatization in a nondiseased population. Based on APPS score, the left side (4.95) was more pneumatized than the right (4.74, P=0.006), and males (10.16) were more extensively pneumatized than females (9.18, P=0.005). We found no correlation of age with sinus pneumatization (ρ=0.025). The probability of perceptible asymmetry in any given individual's paranasal sinus pneumatization was 69%, and the probability of left-sided dominance was 53%. Conclusion: Substantial anatomic variation exists in paranasal sinus anatomy, even among patients without sinus disease. Significant differences are found between males and females and between the left and right sides. Continued systematic research of paranasal sinus anatomy may facilitate a standard for CT sinus assessment that will aid clinician evaluation of anatomic variation and surgical decision making.
Collapse
|
15
|
Mader BJ, Hammer RB. On the true nature of the sphenoidal “pits” in the common warthog, Phacochoerus africanus (Artiodactyla, Suidae), and a description of the cranial sinuses. J Mammal 2020. [DOI: 10.1093/jmammal/gyaa035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abstract
The peculiar sphenoidal “pits” of the common warthog (Phacochoerus africanus) long have been recognized as a feature that distinguishes that species from the closely related desert warthog (P. aethiopicus). Authors seem to regard these structures as blind pockets that do not extend beyond the basicranium. However, these structures actually are openings that lead into a complex system of sinuses (the sphenoidal sinuses) located in the sphenoid and squamosal, and occasionally extending into the occipital and parietal bones. The openings appear to serve as a drain into the nasopharynx. The sphenoidal “pits” of P. africanus as currently defined by most authors are, therefore, not homologous with the sphenoidal pits of P. aethiopicus. We suggest that the term “sphenoidal apertures” be applied to these openings. The true homologue of the sphenoidal pits of P. aethiopicus is the bone that surrounds the sphenoidal apertures in P. africanus. This would include the thin bony sheet that forms the floor of the opening and that bears a shallow fossa. We recommend the term “sphenoidal shelf” be employed to describe this part of the true sphenoidal pit of P. africanus. In addition to the sphenoidal sinuses, the maxillary and frontal sinuses of P. africanus also are described. We report the possibility of mammalian cranial sinuses arising from nasopharyngeal diverticula rather than from diverticula originating from the nasal cavity proper. Previously, such nasopharyngeal pneumatization only has been known to occur in Reptilia. Possible sinus functions are discussed including shock absorption, lightening of the skull, improvement of olfaction, and enhancement of the immune system. In addition to P. africanus, sphenoidal apertures also occur in Babyrousa, and in an extinct perissodactyl, a brontothere (= titanothere).
Collapse
Affiliation(s)
- Bryn J Mader
- Department of Biological Sciences and Geology, Queensborough Community College, 222-05 56th Avenue, Bayside, NY, USA
| | - Rachel B Hammer
- Department of Biological Sciences and Geology, Queensborough Community College, 222-05 56th Avenue, Bayside, NY, USA
| |
Collapse
|
16
|
Pappa AK, Sullivan KM, Lopez EM, Adams KN, Zanation AM, Ebert CS, Thorp BD, Senior BA, Leigh MW, Knowles MR, Kimple AJ. Sinus Development and Pneumatization in a Primary Ciliary Dyskinesia Cohort. Am J Rhinol Allergy 2020; 35:72-76. [PMID: 32551925 PMCID: PMC7750665 DOI: 10.1177/1945892420933175] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background Primary ciliary dyskinesia (PCD) is a genetically diverse disease which
causes impaired mucociliary clearance, and results in pulmonary, otologic,
and rhinologic disease in affected patients. Genetic mutations in multiple
genes impair the ability of patients to clear mucous from the lungs, middle
ear, and sinonasal cavity and lead to chronic pulmonary and sinonasal
symptoms. Methods We identified 17 PCD patients who had available CT scans. Volumes for
bilateral maxillary, sphenoid, and frontal sinuses were calculated. A
control population of patients who had preoperative CT scans for endoscopic
endonasal resection of skull base pathology without sinonasal cavity
involvement was also identified. Results The mean age of PCD was 33 and ranged from 13 to 54 years. Patients were age-
and gender-matched to a control group that underwent resection of anterior
skull-base tumors and had a mean age of 35 that ranged between 17–53 years
old. The volumes for all thee sinus cavities were significantly smaller
(p < 0.007) compared to the control population. The average Lund-Mackay
score was 10.6 in the PCD cohort (range 6–16) in comparison to an average of
0.7 in the control cohort (range 0–2). Conclusions Overall sinus volumes were smaller in patients with PCD compared to our
control population. Future studies will be aimed at understanding defects in
sinus development as a function of specific genetic mutations in PCD
patients. Ultimately, a better understanding of the underlying
pathophysiology of PCD will allow us to identify the optimal treatment
practices for this unique patient group.
Collapse
Affiliation(s)
- Andrew K Pappa
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kelli M Sullivan
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Erin M Lopez
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Katherine N Adams
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Adam M Zanation
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles S Ebert
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brian D Thorp
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brent A Senior
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Margaret W Leigh
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mike R Knowles
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Adam J Kimple
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center at the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
17
|
Dalesio NM, Aksit MA, Ahn K, Raraigh KS, Collaco JM, McGrath-Morrow S, Zeitlin PL, An SS, Cutting GR. Cystic fibrosis transmembrane conductance regulator function, not TAS2R38 gene haplotypes, predict sinus surgery in children and young adults with cystic fibrosis. Int Forum Allergy Rhinol 2020; 10:748-754. [PMID: 32282124 DOI: 10.1002/alr.22548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND Chronic rhinosinusitis symptomatology begins in early childhood individuals with cystic fibrosis (CF). Cystic fibrosis transmembrane conductance regulator (CFTR) function contributes to sinus development and disease. Genetic variants of the bitter taste receptor TAS2R38 have been suggested to contribute to sinus disease severity in individuals without CF. Our objective was to explore whether functional TAS2R38 haplotypes and CFTR function are associated with sinus disease or the need for sinus surgery in individuals with CF. METHODS We conducted a retrospective study using prospectively collected data from the CF Twin-Sibling Study. The function of CFTR was assessed via chloride conductance. Genotyping of the TAS2R38 gene identified patients who were homozygous for the functional haplotype, heterozygous, or homozygous for nonfunctional haplotypes. Clustered multivariate logistic regression was performed, controlling for sex and family relationship. RESULTS A total of 1291 patients were evaluated. Patients with ≤1% CFTR function were 1.56 times more likely to require sinus surgery than those with >1% CFTR function (p = 0.049). CFTR function did not correlate significantly with the presence of sinus disease (p = 0.30). In addition, there were no statistically significant differences in diagnosis of sinus disease or need for sinus surgery between patients with functional and nonfunctional TAS2R38 haplotypes. CONCLUSION CFTR function correlates with need for sinus surgery, whereas TAS2R38 function does not appear to contribute to sinus disease or the need for sinus surgery in patients with CF.
Collapse
Affiliation(s)
- Nicholas M Dalesio
- Division of Pediatric Anesthesia/Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD.,Department of Otolaryngology/Head & Neck Surgery, Johns Hopkins University, Baltimore, MD
| | - Melis A Aksit
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| | - Kwangmi Ahn
- Genetic Epidemiology Research Branch, National Institute of Mental Health, Bethesda, MD
| | - Karen S Raraigh
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| | - Joseph M Collaco
- Department of Pediatric Pulmonology, Johns Hopkins University, Baltimore, MD
| | | | | | - Steven S An
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ.,Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ
| | - Garry R Cutting
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
18
|
Jung JA. Cystic fibrosis lung disease: Current perspectives. ALLERGY ASTHMA & RESPIRATORY DISEASE 2020. [DOI: 10.4168/aard.2020.8.1.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Jin-A Jung
- Department of Pediatrics, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
19
|
Marino MJ, Riley CA, Wu EL, Weinstein JE, McCoul ED. The Unified Airway: Does Asthma Influence Paranasal Sinus Pneumatization? EAR, NOSE & THROAT JOURNAL 2019; 99:89-93. [PMID: 31064242 DOI: 10.1177/0145561319848992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Asthma has been implicated as a driving force in lower airway remodeling; however, its effect on upper airway development has not been studied. Clinical disease, particularly cystic fibrosis (CF), has been associated with anatomical paranasal sinus variation, although the mechanism for these variations remains unclear. The purpose of this study was to determine whether asthma is associated with altered sinus pneumatization. Five hundred ninety-one computed tomography scans, including 303 adolescents (age 13-18) and 288 adults (age > 18), were evaluated using the Assessment of Pneumatization of the Paranasal Sinuses (APPS) instrument. The APPS score is validated for assessing anatomical variation and total sinus volume. A diagnosis of asthma was ascertained from the medical record, and patients with CF were included as a positive control group. Patients with asthma had mean APPS score of 9.66, compared to 9.85 for participants without asthma (P = .585). Subgroup analysis demonstrated similar findings among adults (P = .817) and adolescents (P = .585). Patients with a diagnosis of CF had significant sinus hypoplasia according to a mean APPS scores of 3.50 (P < .001). Sinus hypoplasia persisted in both adults (P < .001) and adolescents (P < .001) with CF. The presence of asthma is not associated with altered paranasal sinus pneumatization. In contrast, CF is associated with significantly reduced sinus pneumatization. These findings suggest that aberrant sinus pneumatization may not be a feature of asthma and that chronic mucosal respiratory disease is not a generalizable cause for altered paranasal sinus pneumatization.
Collapse
Affiliation(s)
- Michael J Marino
- Department of Otorhinolaryngology, Mayo Clinic, Phoenix, AZ, USA
| | - Charles A Riley
- Division of Otolaryngology, Fort Belvoir Community Hospital, Fort Belvoir, VA, USA
| | - Eric L Wu
- Department of Otolaryngology-Head and Neck Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jacqueline E Weinstein
- Benioff Children's Hospital, Division of Pediatric Otolaryngology-Head and Neck Surgery, University of California-San Francisco, San Francisco, CA, USA
| | - Edward D McCoul
- Department of Otolaryngology-Head and Neck Surgery, Tulane University School of Medicine, New Orleans, LA, USA.,Department of Otorhinolaryngology, Ochsner Clinic Foundation, New Orleans, LA, USA.,Ochsner Clinical School, University of Queensland School of Medicine, New Orleans, LA, USA
| |
Collapse
|
20
|
Halderman AA, Lee S, London NR, Day A, Jain R, Moore JA, Lin SY. Impact of high- versus low-risk genotype on sinonasal radiographic disease in cystic fibrosis. Laryngoscope 2018; 129:788-793. [PMID: 30549259 DOI: 10.1002/lary.27595] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Understanding of how specific mutations impact the cystic fibrosis transmembrane conductance regulator (CFTR) protein has given rise to the classification of CF patients into low-risk and high-risk genotypes. Few prior studies have investigated differences in sinonasal disease between low-risk and high-risk CF genotypes. This multi-institutional review aimed to evaluate radiographic sinus disease severity based on genotype. METHODS A review was conducted on adult patients with CF evaluated between 2005 to 2017 at three academic institutions. Data including age, gender, CFTR mutation, and presence of a maxillofacial/sinus computed tomography scan was collected. A modified Lund-Mackay score (MLMS) was assigned to each scan, and the presence of sinus aplasia or hypoplasia was determined. Patients were further grouped depending on genotype into low- or high-risk for comparison. RESULTS A total of 126 patients were included with 99 patients in the high-risk and 21 in the low-risk groups. The high-risk group had significantly higher MLMS than the low-risk group (mean 13.88 vs. 8.06, P < 0.0001, 95% CI -8.196 to -3.462) The rate of frontal (P < 0.01), maxillary (P = 0.04), and sphenoid (P < 0.001) hypoplasia/aplasia was significantly higher in high-risk patients compared to low-risk. CONCLUSION This is one of the largest studies to date evaluating the impact of CF genotype on paranasal sinus development and disease. Genotype appears to impact sinonasal disease severity and also potentially paranasal sinus cavity development to a degree, although the exact mechanism is unknown. LEVEL OF EVIDENCE 4 Laryngoscope, 129:788-793, 2019.
Collapse
Affiliation(s)
| | - Stella Lee
- Department of Otolaryngology Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Nyall R London
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins Hospital, Baltimore, Maryland, U.S.A
| | - Andrew Day
- Department of Otolaryngology Head and Neck Surgery, Dallas, Texas
| | - Raksha Jain
- Department of Internal Medicine, Division of Pulmonary and Critical Care, University of Texas Southwestern, Dallas, Texas
| | - John A Moore
- Department of Otolaryngology Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Sandra Y Lin
- Department of Otolaryngology Head and Neck Surgery, Johns Hopkins Hospital, Baltimore, Maryland, U.S.A
| |
Collapse
|
21
|
Cooney AL, Abou Alaiwa MH, Shah VS, Bouzek DC, Stroik MR, Powers LS, Gansemer ND, Meyerholz DK, Welsh MJ, Stoltz DA, Sinn PL, McCray PB. Lentiviral-mediated phenotypic correction of cystic fibrosis pigs. JCI Insight 2018; 1:88730. [PMID: 27656681 DOI: 10.1172/jci.insight.88730] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cystic Fibrosis (CF) is an autosomal recessive disease caused by mutations in CF transmembrane conductance regulator (CFTR), resulting in defective anion transport. Regardless of the disease-causing mutation, gene therapy is a strategy to restore anion transport to airway epithelia. Indeed, viral vector-delivered CFTR can complement the anion channel defect. In this proof-of-principle study, functional in vivo CFTR channel activity was restored in the airways of CF pigs using a feline immunodeficiency virus-based (FIV-based) lentiviral vector pseudotyped with the GP64 envelope. Three newborn CF pigs received aerosolized FIV-CFTR to the nose and lung. Two weeks after viral vector delivery, epithelial tissues were analyzed for functional correction. In freshly excised tracheal and bronchus tissues and cultured ethmoid sinus cells, we observed a significant increase in transepithelial cAMP-stimulated current, evidence of functional CFTR. In addition, we observed increases in tracheal airway surface liquid pH and bacterial killing in CFTR vector-treated animals. Together, these data provide the first evidence to our knowledge that lentiviral delivery of CFTR can partially correct the anion channel defect in a large-animal CF model and validate a translational strategy to treat or prevent CF lung disease.
Collapse
Affiliation(s)
- Ashley L Cooney
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Departments of Microbiology
| | - Mahmoud H Abou Alaiwa
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine
| | - Viral S Shah
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine.,Molecular Physiology and Biophysics
| | - Drake C Bouzek
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine
| | - Mallory R Stroik
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine
| | - Linda S Powers
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine
| | - Nick D Gansemer
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine
| | - David K Meyerholz
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Pathology
| | - Michael J Welsh
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine.,Howard Hughes Medical Institute.,Molecular Physiology and Biophysics
| | - David A Stoltz
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Internal Medicine
| | - Patrick L Sinn
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Pediatrics, The University of Iowa, Iowa City, Iowa, USA
| | - Paul B McCray
- Pappajohn Biomedical Institute.,Roy J. and Lucille A. Carver College of Medicine.,Departments of Microbiology.,Pediatrics, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
22
|
Meyerholz DK, Stoltz DA, Gansemer ND, Ernst SE, Cook DP, Strub MD, LeClair EN, Barker CK, Adam RJ, Leidinger MR, Gibson-Corley KN, Karp PH, Welsh MJ, McCray PB. Lack of cystic fibrosis transmembrane conductance regulator disrupts fetal airway development in pigs. J Transl Med 2018; 98:825-838. [PMID: 29467455 PMCID: PMC6019641 DOI: 10.1038/s41374-018-0026-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/16/2017] [Accepted: 01/10/2018] [Indexed: 11/15/2022] Open
Abstract
Loss of cystic fibrosis transmembrane conductance regulator (CFTR) function causes cystic fibrosis (CF), predisposing the lungs to chronic infection and inflammation. In young infants with CF, structural airway defects are increasingly recognized before the onset of significant lung disease, which suggests a developmental origin and a possible role in lung disease pathogenesis. The role(s) of CFTR in lung development is unclear and developmental studies in humans with CF are not feasible. Young CF pigs have structural airway changes and develop spontaneous postnatal lung disease similar to humans; therefore, we studied lung development in the pig model (non-CF and CF). CF trachea and proximal airways had structural lesions detectable as early as pseudoglandular development. At this early developmental stage, budding CF airways had smaller, hypo-distended lumens compared to non-CF airways. Non-CF lung explants exhibited airway lumen distension in response to forskolin/IBMX as well as to fibroblast growth factor (FGF)-10, consistent with CFTR-dependent anion transport/secretion, but this was lacking in CF airways. We studied primary pig airway epithelial cell cultures and found that FGF10 increased cellular proliferation (non-CF and CF) and CFTR expression/function (in non-CF only). In pseudoglandular stage lung tissue, CFTR protein was exclusively localized to the leading edges of budding airways in non-CF (but not CF) lungs. This discreet microanatomic localization of CFTR is consistent with the site, during branching morphogenesis, where airway epithelia are responsive to FGF10 regulation. In summary, our results suggest that the CF proximal airway defects originate during branching morphogenesis and that the lack of CFTR-dependent anion transport/liquid secretion likely contributes to these hypo-distended airways.
Collapse
Affiliation(s)
- David K Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| | - David A Stoltz
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nick D Gansemer
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sarah E Ernst
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Howard Hughes Medical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Daniel P Cook
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Matthew D Strub
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Erica N LeClair
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Carrie K Barker
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ryan J Adam
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mariah R Leidinger
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Philip H Karp
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Howard Hughes Medical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Michael J Welsh
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Howard Hughes Medical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Paul B McCray
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
23
|
Mikkelsen MLG, Ambrus R, Rasmussen R, Miles JE, Poulsen HH, Moltke FB, Eriksen T. The influence of norepinephrine and phenylephrine on cerebral perfusion and oxygenation during propofol-remifentanil and propofol-remifentanil-dexmedetomidine anaesthesia in piglets. Acta Vet Scand 2018; 60:8. [PMID: 29422100 PMCID: PMC5806235 DOI: 10.1186/s13028-018-0362-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 01/30/2018] [Indexed: 11/15/2022] Open
Abstract
Background Vasopressors are frequently used to increase blood pressure in order to ensure sufficient cerebral perfusion and oxygenation (CPO) during hypotensive periods in anaesthetized patients. Efficacy depends both on the vasopressor and anaesthetic protocol used. Propofol–remifentanil total intravenous anaesthesia (TIVA) is common in human anaesthesia, and dexmedetomidine is increasingly used as adjuvant to facilitate better haemodynamic stability and analgesia. Little is known of its interaction with vasopressors and subsequent effects on CPO. This study investigates the CPO response to infusions of norepinephrine and phenylephrine in piglets during propofol–remifentanil and propofol–remifentanil–dexmedetomidine anaesthesia. Sixteen healthy female piglets (25–34 kg) were randomly allocated into a two-arm parallel group design with either normal blood pressure (NBP) or induced low blood pressure (LBP). Anaesthesia was induced with propofol without premedication and maintained with propofol–remifentanil TIVA, and finally supplemented with continuous infusion of dexmedetomidine. Norepinephrine and phenylephrine were infused in consecutive intervention periods before and after addition of dexmedetomidine. Cerebral perfusion measured by laser speckle contrast imaging was related to cerebral oxygenation as measured by an intracerebral Licox probe (partial pressure of oxygen) and transcranial near infrared spectroscopy technology (NIRS) (cerebral oxygen saturation). Results During propofol–remifentanil anaesthesia, increases in blood pressure by norepinephrine and phenylephrine did not change cerebral perfusion significantly, but cerebral partial pressure of oxygen (Licox) increased following vasopressors in both groups and increases following norepinephrine were significant (NBP: P = 0.04, LBP: P = 0.02). In contrast, cerebral oxygen saturation (NIRS) fell significantly in NBP following phenylephrine (P = 0.003), and following both norepinephrine (P = 0.02) and phenylephrine (P = 0.002) in LBP. Blood pressure increase by both norepinephrine and phenylephrine during propofol–remifentanil–dexmedetomidine anaesthesia was not followed by significant changes in cerebral perfusion. Licox measures increased significantly following both vasopressors in both groups, whereas the decreases in NIRS measures were only significant in the NBP group. Conclusions Cerebral partial pressure of oxygen measured by Licox increased significantly in concert with the vasopressor induced increases in blood pressure in healthy piglets with both normal and low blood pressure. Cerebral oxygenation assessed by intracerebral Licox and transcranial NIRS showed opposing results to vasopressor infusions. Electronic supplementary material The online version of this article (10.1186/s13028-018-0362-z) contains supplementary material, which is available to authorized users.
Collapse
|
24
|
Mueller SK, Scangas G, Amiji MM, Bleier BS. Prospective transfrontal sheep model of skull-base reconstruction using vascularized mucosa. Int Forum Allergy Rhinol 2017; 8:614-619. [PMID: 29227037 DOI: 10.1002/alr.22058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/24/2017] [Accepted: 11/10/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND No high-fidelity animal model exists to examine prospective wound healing following vascularized reconstruction of the skull base. Such a model would require the ability to study the prospective behavior of vascularized mucosal repairs of large dural and arachnoid defects within the intranasal environment. The objective of this study was to therefore develop and validate a novel, in vivo, transfrontal sheep model of cranial base repair using vascularized sinonasal mucosa. METHODS Twelve transfrontal craniotomy and 1.5-cm durotomy reconstructions were performed in 60-kg to 70-kg Dorset/Ovis Aries sheep using vascularized mucosa with or without an adjunctive Biodesign™ underlay graft (n = 6 per group). Histologic outcomes were graded (scale, 0 to 4) by a blinded veterinary histopathologist after 7, 14, and 28 days for a range of wound healing parameters. RESULTS All sheep tolerated the surgery, which required 148 ± 33 minutes. By day 7, the mucosa was fully adherent with complete partitioning of the sinus and intracranial compartments. Fibroblast infiltration and flap neovascularization scores significantly increased between day 7 (0.3 ± 0.5 and 0.0 ± 0.0) and day 28 (4.0 ± 0.0, p = 0.01 and 2.0 ± 0.8, p = 0.01; respectively), while hemorrhage scores significantly decreased from 2.5 ± 0.6 to 0.0 ± 0.0 (p = 0.01). The inflammatory scores were not significantly different between the heterologous graft and control sides. CONCLUSION The described sheep model accurately reflects prospective intranasal wound healing following vascularized mucosal reconstruction of dural defects. This model can be used in future studies to examine novel reconstructive materials, tissue glues, and transmucosal drug delivery to the central nervous system.
Collapse
Affiliation(s)
- Sarina K Mueller
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA.,Department of Otolaryngology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - George Scangas
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| | - Mansor M Amiji
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA
| | - Benjamin S Bleier
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| |
Collapse
|
25
|
Carlon MS, Vidović D, Birket S. Roadmap for an early gene therapy for cystic fibrosis airway disease. Prenat Diagn 2017; 37:1181-1190. [DOI: 10.1002/pd.5164] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/12/2017] [Accepted: 09/28/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Marianne S. Carlon
- Molecular Virology and Gene Therapy; Department of Pharmaceutical and Pharmacological Sciences; KU Leuven Flanders Belgium
| | - Dragana Vidović
- Molecular Virology and Gene Therapy; Department of Pharmaceutical and Pharmacological Sciences; KU Leuven Flanders Belgium
- Current affiliation: Cellular Protein Chemistry, Faculty of Science; Utrecht University; The Netherlands
| | - Susan Birket
- Department of Medicine; University of Alabama at Birmingham; Birmingham AL USA
| |
Collapse
|
26
|
Tipirneni KE, Cho DY, Skinner DF, Zhang S, Mackey C, Lim DJ, Woodworth BA. Characterization of primary rat nasal epithelial cultures in CFTR knockout rats as a model for CF sinus disease. Laryngoscope 2017; 127:E384-E391. [PMID: 28771736 DOI: 10.1002/lary.26720] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/28/2017] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The objectives of the current experiments were to develop and characterize primary rat nasal epithelial cultures and evaluate their usefulness as a model of cystic fibrosis (CF) sinonasal transepithelial transport and CF transmembrane conductance regulator (CFTR) function. STUDY DESIGN Laboratory in vitro and animal studies. METHODS CFTR+/+ and CFTR-/- rat nasal septal epithelia (RNSE) were cultured on semipermeable supports at an air-liquid interface to confluence and full differentiation. Monolayers were mounted in Ussing chambers for pharmacologic manipulation of ion transport and compared to similar filters containing murine (MNSE) and human (HSNE) epithelia. Histology and scanning electron microscopy (SEM) were completed. Real-time polymerase chain reaction of CFTR+/+ RNSE, MNSE, and HSNE was performed to evaluate relative CFTR gene expression. RESULTS Forskolin-stimulated anion transport (ΔIsc in μA/cm2 ) was significantly greater in epithelia derived from CFTR+/+ when compared to CFTR-/- animals (100.9 ± 3.7 vs. 10.5 ± 0.9; P < 0.0001). Amiloride-sensitive ISC was equivalent (-42.3 ± 2.8 vs. -46.1 ± 2.3; P = 0.524). No inhibition of CFTR-mediated chloride (Cl- ) secretion was exhibited in CFTR-/- epithelia with the addition of the specific CFTR inhibitor, CFTRInh -172. However, calcium-activated Cl- secretion (UTP) was significantly increased in CFTR-/- RNSE (CFTR-/- -106.8 ± 1.6 vs. CFTR+/+ -32.2 ± 3.1; P < 0.0001). All responses were larger in RNSE when compared to CFTR+/+ and CFTR-/- (or F508del/F508del) murine and human cells (P < 0.0001). Scanning electron microscopy demonstrated 80% to 90% ciliation in all RNSE cultures. There was no evidence of infection in CFTR-/- rats at 4 months. CFTR expression was similar among species. CONCLUSION The successful development of the CFTR-/- rat enables improved evaluation of CF sinus disease based on characteristic abnormalities of ion transport. LEVEL OF EVIDENCE NA. Laryngoscope, 127:E384-E391, 2017.
Collapse
Affiliation(s)
- Kiranya E Tipirneni
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Do-Yeon Cho
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Daniel F Skinner
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Shaoyan Zhang
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Calvin Mackey
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Dong-Jin Lim
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Bradford A Woodworth
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| |
Collapse
|
27
|
Grayson J, Tipirneni KE, Skinner DF, Fort M, Cho DY, Zhang S, Prince AC, Lim DJ, Mackey C, Woodworth BA. Sinus hypoplasia in the cystic fibrosis rat resolves in the absence of chronic infection. Int Forum Allergy Rhinol 2017; 7:904-909. [PMID: 28597597 DOI: 10.1002/alr.21973] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/04/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sinus hypoplasia is a hallmark characteristic in cystic fibrosis (CF). Chronic rhinosinusitis (CRS) is nearly universal from a young age, impaired sinus development could be secondary to loss of the cystic fibrosis transmembrane conductance regulator (CFTR) or consequences of chronic infection during maturation. The objective of this study was to assess sinus development relative to overall growth in a novel CF animal model. METHODS Sinus development was evaluated in CFTR-/- and CFTR+/+ rats at 3 stages of development: newborn; 3 weeks; and 16 weeks. Microcomputed tomography (microCT) scanning, cultures, and histology were performed. Three-dimensional sinus and skull volumes were quantified. RESULTS At birth, sinus volumes were decreased in CFTR-/- rats compared with wild-type rats (mean ± SEM: 11.3 ± 0.85 mm3 vs 14.5 ± 0.73 mm3 ; p < 0.05), despite similar weights (8.4 ± 0.46 gm vs 8.3 ± 0.51 gm; p = 0.86). CF rat weights declined by 16 weeks (378.4 ± 10.6 gm vs 447.4 ± 15.9 gm; p < 0.05), sinus volume increased similar to wild-type rats (201.1 ± 3.77 gm vs 203.4 ± 7.13 gm; p = 0.8). The ratio of sinus volume to body weight indicates hypoplasia present at birth (1.37 ± 0.12 vs 1.78 ± 0.11; p < 0.05) and showed an increase compared with CFTR+/+ animals by 16 weeks (0.53 ± 0.02 vs 0.46 ± 0.02; p < 0.05). Rats did not develop histologic evidence of chronic infection. CONCLUSION CF rat sinuses are smaller at birth, but develop volumes similar to wild-type rats with maturation. This suggests that loss of CFTR may confer sinus hypoplasia at birth, but normal development ensues without chronic sinus infection.
Collapse
Affiliation(s)
- Jessica Grayson
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL
| | | | - Daniel F Skinner
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - Matthew Fort
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL
| | - Do-Yeon Cho
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - Shaoyan Zhang
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - Andrew C Prince
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Dong-Jin Lim
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - Calvin Mackey
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| | - Bradford A Woodworth
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
28
|
Tipirneni KE, Woodworth BA. Medical and Surgical Advancements in the Management of Cystic Fibrosis Chronic Rhinosinusitis. CURRENT OTORHINOLARYNGOLOGY REPORTS 2017; 5:24-34. [PMID: 28989817 PMCID: PMC5626435 DOI: 10.1007/s40136-017-0139-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide otolaryngologists with the most up-to-date advancements in both the medical and surgical management of CF-related sinus disease. RECENT FINDINGS Recent studies have supported more aggressive CRS management, often with a combination of both medical and surgical therapies. Comprehensive treatment strategies have been shown to reduce hospital admissions secondary to pulmonary exacerbations in addition to improving CRS symptoms. Still, current management strategies are lacking in both high-level evidence and standardized guidelines. SUMMARY The unified airway model describes the bi-directional relationship between the upper and lower airways as a single functional unit and suggests that CRS may play a pivotal role in both the development and progression of lower airway disease. Current strategies for CF CRS focus primarily on amelioration of symptoms with antibiotics, nasal saline and/or topical medicated irrigations, and surgery. However, there are no definitive management guidelines and there remains a persistent need for additional studies. Nevertheless, otolaryngologists have a significant role in the overall management of CF, which requires a multi-disciplinary approach and a combination of both surgical and medical interventions for optimal outcomes of airway disease. Here we present a review of currently available literature and summarize medical and surgical therapies best suited for the management of CF-related sinus disease.
Collapse
|
29
|
Are Chronic Rhinosinusitis and Paranasal Sinus Pneumatization Related? SINUSITIS 2016. [DOI: 10.3390/sinusitis1010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
30
|
Calton JB, Koripella PC, Willis AL, Le CH, Chiu AG, Chang EH. Paranasal sinus size is decreased inCFTRheterozygotes with chronic rhinosinusitis. Int Forum Allergy Rhinol 2016; 7:256-260. [DOI: 10.1002/alr.21874] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/05/2016] [Accepted: 10/11/2016] [Indexed: 01/22/2023]
Affiliation(s)
| | | | | | | | | | - Eugene H. Chang
- Department of Otolaryngology; University of Arizona; Tucson AZ
| |
Collapse
|
31
|
Steines B, Dickey DD, Bergen J, Excoffon KJ, Weinstein JR, Li X, Yan Z, Abou Alaiwa MH, Shah VS, Bouzek DC, Powers LS, Gansemer ND, Ostedgaard LS, Engelhardt JF, Stoltz DA, Welsh MJ, Sinn PL, Schaffer DV, Zabner J. CFTR gene transfer with AAV improves early cystic fibrosis pig phenotypes. JCI Insight 2016; 1:e88728. [PMID: 27699238 DOI: 10.1172/jci.insight.88728] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The physiological components that contribute to cystic fibrosis (CF) lung disease are steadily being elucidated. Gene therapy could potentially correct these defects. CFTR-null pigs provide a relevant model to test gene therapy vectors. Using an in vivo selection strategy that amplifies successful capsids by replicating their genomes with helper adenovirus coinfection, we selected an adeno-associated virus (AAV) with tropism for pig airway epithelia. The evolved capsid, termed AAV2H22, is based on AAV2 with 5 point mutations that result in a 240-fold increased infection efficiency. In contrast to AAV2, AAV2H22 binds specifically to pig airway epithelia and is less reliant on heparan sulfate for transduction. We administer AAV2H22-CFTR expressing the CF transmembrane conductance regulator (CFTR) cDNA to the airways of CF pigs. The transduced airways expressed CFTR on ciliated and nonciliated cells, induced anion transport, and improved the airway surface liquid pH and bacterial killing. Most gene therapy studies to date focus solely on Cl- transport as the primary metric of phenotypic correction. Here, we describe a gene therapy experiment where we not only correct defective anion transport, but also restore bacterial killing in CFTR-null pig airways.
Collapse
Affiliation(s)
- Benjamin Steines
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - David D Dickey
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and
| | - Jamie Bergen
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, California, USA
| | | | - John R Weinstein
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, California, USA
| | - Xiaopeng Li
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | - Mahmoud H Abou Alaiwa
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Viral S Shah
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | | | | | - Lynda S Ostedgaard
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | - David A Stoltz
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Michael J Welsh
- Department of Internal Medicine.,Molecular and Cellular Biology Program, and.,Molecular Physiology and Biophysics
| | - Patrick L Sinn
- Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA.,Howard Hughes Medical Institute, and
| | - David V Schaffer
- Departments of Chemical and Biomolecular Engineering, Bioengineering, The Helen Wills Neuroscience Institute, Molecular and Cellular Biology, University of California, Berkeley, California, USA
| | - Joseph Zabner
- Department of Internal Medicine.,Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
32
|
Jeong JH, Hwang PH, Cho DY, Joo NS, Wine JJ. Secretion rates of human nasal submucosal glands from patients with chronic rhinosinusitis or cystic fibrosis. Am J Rhinol Allergy 2016; 29:334-8. [PMID: 26358343 DOI: 10.2500/ajra.2015.29.4213] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND A majority of patients with cystic fibrosis (CF) have chronic rhinosinusitis (CRS) and/or nasal polyps, both of which may be secondary to reduced fluid secretion from nasal submucosal glands. OBJECTIVE To determine whether decreased fluid secretion from nasal submucosal glands also occurs in patients without CF and with CRS. METHODS Inferior turbinates of the nasal cavity were harvested from controls, subjects with CRS, and subjects with CF (n = 5-7 per group). The secretion rates of the nasal submucosal glands of the three groups in response to carbachol and forskolin were measured by using time lapse digital imaging of mucus bubbles from single glands as they formed on the mucosal surface under oil. RESULTS Carbachol-stimulated secretion rates were the following: controls, 1670 ± 381 pl·min(-1)·gland(-1); CRS, 965 ± 440 pl·min(-1)·gland(-1); and CF, 933 ± 588 pl·min(-1)·gland(-1) (p = 0.23, Kruskal-Wallis test). Forskolin-stimulated secretion rates were the following: controls, 229 ± 14 pl·min(-1)·gland(-1); CRS, 154 ± 48 pl·min(-1)·gland(-1); and CF, 22 ± 15 pl·min(-1)·gland(-1) (p = 0.008, Kruskal-Wallis test). The ratio of the average secretion rate induced by forskolin to that induced by carbachol was 13.7% in the controls, and 15.9% in CRS and 2.3% in CF groups. CONCLUSION The only significant difference in this small study was decreased forskolin-stimulated secretion in subjects with CF relative to the other subjects. However, there was a trend toward reduced carbachol-stimulated secretion rates in subjects with CRS and with and without CF relative to controls. Additional studies are needed to determine if nasal submucosal gland hyposecretion occurs in CRS either as a contributor to or as a consequence of CRS pathogenesis.
Collapse
Affiliation(s)
- Jin Hyeok Jeong
- Cystic Fibrosis Research Laboratory, Stanford University, Stanford, California, USA
| | | | | | | | | |
Collapse
|
33
|
London NR, Lane AP. Innate immunity and chronic rhinosinusitis: What we have learned from animal models. Laryngoscope Investig Otolaryngol 2016; 1:49-56. [PMID: 28459101 PMCID: PMC5409101 DOI: 10.1002/lio2.21] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Chronic rhinosinusitis (CRS) is a heterogeneous and multifactorial disease characterized by dysregulated inflammation. Abnormalities in innate immune function including sinonasal epithelial cell barrier function, mucociliary clearance, response to pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs), and the contribution of innate immune cells will be highlighted in this review. DATA SOURCES PubMed literature review. REVIEW METHODS A review of the literature was conducted to determine what we have learned from animal models in relation to innate immunity and chronic rhinosinusitis. RESULTS Dysregulation of innate immune mechanisms including sinonasal barrier function, mucociliary clearance, PAMPs, and innate immune cells such as eosinophils, mast cells, and innate lymphoid cells may contribute to CRS pathogenesis. Sinonasal inflammation has been studied using mouse, rat, rabbit, pig, and sheep explant or in vivo models. Study using these models has allowed for analysis of experimental therapeutics and furthered our understanding of the aforementioned aspects of the innate immune mechanism as it relates to sinonasal inflammation. These include augmenting mucociliary clearance through activation of the cystic fibrosis transmembrane conductance regulator (CFTR) and study of drug toxicity on ciliary beat frequency. Knockout models of Toll-like receptors (TLR) have demonstrated the critical role these PRRs play in allergic inflammation as loss of TLR2 and TLR4 leads to decreased lower airway inflammation. Mast cell deficient mice are less susceptible to ovalbumin-induced sinonasal inflammation. CONCLUSION Animal models have shed light as to the potential contribution of dysregulated innate immunity in chronic sinonasal inflammation.
Collapse
Affiliation(s)
- Nyall R. London
- Department of OtolaryngologyHead and Neck Surgery, Johns Hopkins School of MedicineBaltimoreMarylandU.S.A.
| | - Andrew P. Lane
- Department of OtolaryngologyHead and Neck Surgery, Johns Hopkins School of MedicineBaltimoreMarylandU.S.A.
| |
Collapse
|
34
|
Marino MJ, Riley CA, Kessler RH, McCoul ED. Clinician assessment of paranasal sinus pneumatization is correlated with total sinus volume. Int Forum Allergy Rhinol 2016; 6:1088-1093. [DOI: 10.1002/alr.21779] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/18/2016] [Accepted: 03/01/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Michael J. Marino
- Department of Otolaryngology-Head and Neck Surgery; Tulane University School of Medicine; New Orleans LA
| | - Charles A. Riley
- Department of Otolaryngology-Head and Neck Surgery; Tulane University School of Medicine; New Orleans LA
| | - Raymond H. Kessler
- Department of Otolaryngology-Head and Neck Surgery; Tulane University School of Medicine; New Orleans LA
| | - Edward D. McCoul
- Department of Otolaryngology-Head and Neck Surgery; Tulane University School of Medicine; New Orleans LA
- Department of Otorhinolaryngology; Ochsner Clinic Foundation; New Orleans LA
- Ochsner Clinical School; University of Queensland School of Medicine; New Orleans LA
| |
Collapse
|
35
|
Relationships among CFTR expression, HCO3- secretion, and host defense may inform gene- and cell-based cystic fibrosis therapies. Proc Natl Acad Sci U S A 2016; 113:5382-7. [PMID: 27114540 DOI: 10.1073/pnas.1604905113] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel. Airway disease is the major source of morbidity and mortality. Successful implementation of gene- and cell-based therapies for CF airway disease requires knowledge of relationships among percentages of targeted cells, levels of CFTR expression, correction of electrolyte transport, and rescue of host defense defects. Previous studies suggested that, when ∼10-50% of airway epithelial cells expressed CFTR, they generated nearly wild-type levels of Cl(-) secretion; overexpressing CFTR offered no advantage compared with endogenous expression levels. However, recent discoveries focused attention on CFTR-mediated HCO3 (-) secretion and airway surface liquid (ASL) pH as critical for host defense and CF pathogenesis. Therefore, we generated porcine airway epithelia with varying ratios of CF and wild-type cells. Epithelia with a 50:50 mix secreted HCO3 (-) at half the rate of wild-type epithelia. Likewise, heterozygous epithelia (CFTR(+/-) or CFTR(+/∆F508)) expressed CFTR and secreted HCO3 (-) at ∼50% of wild-type values. ASL pH, antimicrobial activity, and viscosity showed similar relationships to the amount of CFTR. Overexpressing CFTR increased HCO3 (-) secretion to rates greater than wild type, but ASL pH did not exceed wild-type values. Thus, in contrast to Cl(-) secretion, the amount of CFTR is rate-limiting for HCO3 (-) secretion and for correcting host defense abnormalities. In addition, overexpressing CFTR might produce a greater benefit than expressing CFTR at wild-type levels when targeting small fractions of cells. These findings may also explain the risk of airway disease in CF carriers.
Collapse
|
36
|
Meyerholz DK. Lessons learned from the cystic fibrosis pig. Theriogenology 2016; 86:427-32. [PMID: 27142487 DOI: 10.1016/j.theriogenology.2016.04.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/14/2015] [Accepted: 03/14/2016] [Indexed: 12/01/2022]
Abstract
Deficient function in the anion channel cystic fibrosis (CF) transmembrane conductance regulator is the fundamental cause for CF. This is a monogenic condition that causes lesions in several organs including the respiratory tract, pancreas, liver, intestines, and reproductive tract. Lung disease is most notable, given it is the leading cause of morbidity and mortality in people with CF. Shortly after the identification of CF transmembrane conductance regulator, CF mouse models were developed that did not show spontaneous lung disease as seen in humans, and this spurred development of additional CF animal models. Pig models were considered a leading choice for several reasons including their similarity to humans in respiratory anatomy, physiology, and in size for translational imaging. The first CF pig models were reported in 2008 and have been extremely valuable to help clarify persistent questions in the field and advance understanding of disease pathogenesis. Because CF pigs are susceptible to lung disease like humans, they have direct utility in translational research. In addition, CF pig models are useful to compare and contrast with current CF mouse models, human clinical studies, and even newer CF animal models being characterized. This "triangulation" strategy could help identify genetic differences that underlie phenotypic variations, so as to focus and accelerate translational research.
Collapse
Affiliation(s)
- David K Meyerholz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.
| |
Collapse
|
37
|
Tang XX, Ostedgaard LS, Hoegger MJ, Moninger TO, Karp PH, McMenimen JD, Choudhury B, Varki A, Stoltz DA, Welsh MJ. Acidic pH increases airway surface liquid viscosity in cystic fibrosis. J Clin Invest 2016; 126:879-91. [PMID: 26808501 DOI: 10.1172/jci83922] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022] Open
Abstract
Cystic fibrosis (CF) disrupts respiratory host defenses, allowing bacterial infection, inflammation, and mucus accumulation to progressively destroy the lungs. Our previous studies revealed that mucus with abnormal behavior impaired mucociliary transport in newborn CF piglets prior to the onset of secondary manifestations. To further investigate mucus abnormalities, here we studied airway surface liquid (ASL) collected from newborn piglets and ASL on cultured airway epithelia. Fluorescence recovery after photobleaching revealed that the viscosity of CF ASL was increased relative to that of non-CF ASL. CF ASL had a reduced pH, which was necessary and sufficient for genotype-dependent viscosity differences. The increased viscosity of CF ASL was not explained by pH-independent changes in HCO3- concentration, altered glycosylation, additional pH-induced disulfide bond formation, increased percentage of nonvolatile material, or increased sulfation. Treating acidic ASL with hypertonic saline or heparin largely reversed the increased viscosity, suggesting that acidic pH influences mucin electrostatic interactions. These findings link loss of cystic fibrosis transmembrane conductance regulator-dependent alkalinization to abnormal CF ASL. In addition, we found that increasing Ca2+ concentrations elevated ASL viscosity, in part, independently of pH. The results suggest that increasing pH, reducing Ca2+ concentration, and/or altering electrostatic interactions in ASL might benefit early CF.
Collapse
|
38
|
Klymiuk N, Seeliger F, Bohlooly-Y M, Blutke A, Rudmann DG, Wolf E. Tailored Pig Models for Preclinical Efficacy and Safety Testing of Targeted Therapies. Toxicol Pathol 2015; 44:346-57. [PMID: 26511847 DOI: 10.1177/0192623315609688] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite enormous advances in translational biomedical research, there remains a growing demand for improved animal models of human disease. This is particularly true for diseases where rodent models do not reflect the human disease phenotype. Compared to rodents, pig anatomy and physiology are more similar to humans in cardiovascular, immune, respiratory, skeletal muscle, and metabolic systems. Importantly, efficient and precise techniques for genetic engineering of pigs are now available, facilitating the creation of tailored large animal models that mimic human disease mechanisms at the molecular level. In this article, the benefits of genetically engineered pigs for basic and translational research are exemplified by a novel pig model of Duchenne muscular dystrophy and by porcine models of cystic fibrosis. Particular emphasis is given to potential advantages of using these models for efficacy and safety testing of targeted therapies, such as exon skipping and gene editing, for example, using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system. In general, genetically tailored pig models have the potential to bridge the gap between proof-of-concept studies in rodents and clinical trials in patients, thus supporting translational medicine.
Collapse
Affiliation(s)
- Nikolai Klymiuk
- Gene Center and Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Frank Seeliger
- Pathology Science, DSM, Transgenic, AstraZeneca RD, Mölndal, Sweden
| | | | - Andreas Blutke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel G Rudmann
- Pathology Science, DSM, Transgenic, AstraZeneca RD, Mölndal, Sweden
| | - Eckhard Wolf
- Gene Center and Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
39
|
Abstract
The successful application of precision genomic medicine requires an understanding of how a person's genome can influence his or her disease phenotype and how medical therapies can provide personalized therapy to one's genotype. In this review, we highlight advances in precision genomic medicine in cystic fibrosis (CF), a classic autosomal recessive genetic disorder. We discuss genotype-phenotype correlations in CF, genetic and environmental modifiers of disease, and pharmacogenetic therapies that target specific genetic mutations thereby addressing the primary defect of cystic fibrosis.
Collapse
Affiliation(s)
- Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Joseph Zabner
- Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
40
|
Diwakar A, Adam RJ, Michalski AS, Tamegnon MM, Fischer AJ, Launspach JL, Horan RA, Kao SC, Chaloner K, Meyerholz DK, Stoltz DA. Sonographic evidence of abnormal tracheal cartilage ring structure in cystic fibrosis. Laryngoscope 2015; 125:2398-404. [PMID: 25827636 DOI: 10.1002/lary.25255] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/13/2015] [Accepted: 02/18/2015] [Indexed: 11/10/2022]
Abstract
OBJECTIVES/HYPOTHESIS Tracheal cartilage ring structural abnormalities have been reported in cystic fibrosis (CF) mice and pigs. Whether similar findings are present in humans with CF is unknown. We hypothesized that tracheal cartilage ring shape and size would be different in people with CF. STUDY DESIGN Tracheal cartilage ring size and shape were measured in adults with (n = 21) and without CF (n = 18). METHODS Ultrasonography was used in human subjects to noninvasively assess tracheal cartilage ring structure in both the sagittal and the transverse planes. Tracheal cartilage ring thickness was also determined from histological sections obtained from newborn non-CF and CF pigs. These values were compared with human data. RESULTS Human CF tracheas had a greater width and were less circular in shape compared to non-CF subjects. CF tracheal cartilage rings had a greater midline cross-sectional area and were thicker compared to non-CF rings. Maximal tracheal cartilage ring thickness was also greater in both newborn CF pigs and human adults with CF, compared to non-CF controls. CONCLUSIONS Our findings demonstrate that structural differences exist in tracheal cartilage rings in adults with CF. Comparison with newborn CF pig data suggests that some of these changes may be congenital in nature. LEVEL OF EVIDENCE 3b
Collapse
Affiliation(s)
- Amit Diwakar
- Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa
| | - Ryan J Adam
- Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa.,Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa
| | - Andrew S Michalski
- Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa
| | - Monelle M Tamegnon
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, U.S.A
| | - Anthony J Fischer
- Department of Pediatrics, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa
| | - Jan L Launspach
- Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa
| | - Rebecca A Horan
- Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa
| | - Simon C Kao
- Department of Radiology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa
| | - Kathryn Chaloner
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, U.S.A
| | - David K Meyerholz
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa
| | - David A Stoltz
- Department of Internal Medicine, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa.,Department of Molecular Physiology and Biophysics, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa.,Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa
| |
Collapse
|
41
|
Affiliation(s)
- David A Stoltz
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Department of Molecular Physiology and Biophysics, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa 52242
| | - David K Meyerholz
- Department of Pathology, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Michael J Welsh
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Department of Molecular Physiology and Biophysics, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
- Howard Hughes Medical Institute, Roy J and Lucille A Carver College of Medicine University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Upper airway disease engenders significant morbidity for patients with cystic fibrosis and is increasingly recognized as having a much greater role in pulmonary outcomes and quality of life than originally believed. Widespread disparate therapeutic strategies for cystic fibrosis chronic rhinosinusitis underscore the absence of a standardized treatment paradigm. This review outlines the most recent evidence-based trends in the management of upper airway disease in cystic fibrosis. RECENT FINDINGS The unified airway theory proposes that the sinuses are a focus of initial bacterial colonization which seeds the lower airway and may play a large role in maintaining lung infections. Mounting evidence suggests more aggressive treatment of the sinuses may confer significant improvement in pulmonary disease and quality of life outcomes in cystic fibrosis patients. However, there is a lack of high-level evidence regarding medical and surgical management of cystic fibrosis chronic rhinosinusitis that makes generalizations difficult. SUMMARY Well designed clinical trials with long-term follow-up concerning medical and surgical interventions for cystic fibrosis sinus disease are required to establish standardized treatment protocols, but increased interest in the sinuses as a bacterial reservoir for pulmonary infections has generated considerable attention.
Collapse
Affiliation(s)
- Elisa A Illing
- Department of Surgery/Division of Otolaryngology and the Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
43
|
Chang EH, Tang XX, Shah VS, Launspach JL, Ernst SE, Hilkin B, Karp PH, Abou Alaiwa MH, Graham SM, Hornick DB, Welsh MJ, Stoltz DA, Zabner J. Medical reversal of chronic sinusitis in a cystic fibrosis patient with ivacaftor. Int Forum Allergy Rhinol 2014; 5:178-81. [PMID: 25363320 DOI: 10.1002/alr.21440] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 09/17/2014] [Accepted: 09/25/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Chronic sinusitis is universal in cystic fibrosis (CF) and our current treatments are ineffective in reversing sinus disease. The objective of this work was to determine if increasing CF transmembrane conductance regulator (CFTR) activity by ivacaftor could treat CF sinus disease and assess its effect on primary sinus epithelial cultures. METHODS Case report of 1 patient with long-standing chronic sinus disease and a new diagnosis of CF with a mild mutation (P205S) and a severe mutation (G551D). We discuss clinical changes in symptoms, radiographic findings, nasal potential difference testing, and nasal pH values before and after treatment with ivacaftor. We then developed primary sinonasal epithelial cell cultures from a biopsy of the patient to determine changes in airway surface liquid (ASL) pH and ASL viscosity after ivacaftor treatment. RESULTS Ivacaftor treatment reversed CT findings of CF sinus disease, increased nasal voltage and pH, and resolved sinus symptoms after 10 months of therapy. Ivacaftor significantly increased ASL pH and decreased ASL viscosity in primary airway cultures. CONCLUSION This report documents the reversal of CF sinus disease. Based on our in vivo and in vitro results, we speculate that ivacaftor may reverse CF sinusitis by increasing ASL pH and decreasing ASL viscosity. These studies suggest that CFTR modulation may be effective in treating CF and perhaps non-CF sinusitis.
Collapse
Affiliation(s)
- Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ferril GR, Nick JA, Getz AE, Barham HP, Saavedra MT, Taylor-Cousar JL, Nichols DP, Curran-Everett D, Kingdom TT, Ramakrishnan VR. Comparison of radiographic and clinical characteristics of low-risk and high-risk cystic fibrosis genotypes. Int Forum Allergy Rhinol 2014; 4:915-20. [PMID: 25224556 DOI: 10.1002/alr.21412] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/13/2014] [Accepted: 08/05/2014] [Indexed: 11/11/2022]
Abstract
BACKGROUND Patients with cystic fibrosis (CF) exhibit a wide range of disease severity, and can be broadly stratified into high-risk and low-risk groups based on cystic fibrosis transmembrane conductance regulator (CFTR) mutation class. Patients with a low-risk genotype are often diagnosed as adults, with milder disease and lower sweat chloride values. The aim of the current study was to better understand radiographic and clinical characteristics of sinus disease in adult CF patients within this risk category. METHODS Adult CF patients were retrospectively compared to a control group of patients with chronic rhinosinusitis. CF diagnostic testing and pulmonary characteristics were compared between high-risk and low-risk CF groups, and sinus CT findings were compared among all 3 groups. RESULTS When comparing CF cohorts (n = 25 and 30, respectively), earlier age at diagnosis (p < 0.001), higher sweat chloride values (p < 0.001), lower forced expiratory volume in 1 second (FEV1 ) values (p < 0.001), and a higher prevalence of pulmonary infection with Pseudomonas aeruginosa (p = 0.001) were found in the high-risk genotype group. A significantly increased incidence of sinus hypoplasia/aplasia and bony sclerosis was seen when comparing both CF groups to the control cohort (n = 30), as well as when comparing the high-risk and low-risk CF genotype cohorts. CONCLUSION The current study describes clinicopathologic findings of sinus disease in adult CF patients in the context of genotype severity. Our data demonstrate that while patients within a low-risk genotype cohort have generally milder lung disease, they retain classic radiographic findings of CF sinus disease that can help raise the index of suspicion for undiagnosed CF.
Collapse
Affiliation(s)
- Geoffrey R Ferril
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado, Aurora, CO
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sinonasal manifestations of cystic fibrosis: A correlation between genotype and phenotype? J Cyst Fibros 2014; 13:442-8. [DOI: 10.1016/j.jcf.2013.10.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/11/2013] [Accepted: 10/21/2013] [Indexed: 12/26/2022]
|
46
|
Dean N, Ranganath NK, Jones B, Zhang S, Skinner D, Rowe SM, Sorscher EJ, Woodworth BA. Porcine nasal epithelial cultures for studies of cystic fibrosis sinusitis. Int Forum Allergy Rhinol 2014; 4:565-70. [PMID: 24733748 DOI: 10.1002/alr.21335] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 03/04/2014] [Accepted: 03/11/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Transgenic cystic fibrosis (CF) murine models do not develop spontaneous lung or sinus disease, 2 major causes of morbidity in human CF patients. Because of these limitations, transgenic cystic fibrosis transmembrane conductance regulator (CFTR)(-/-) pigs have been developed and are currently being characterized. These CF animal models have phenotypes closely resembling that of human CF subjects. The objectives of the current study were to develop primary porcine nasal epithelial (PNE) cultures and evaluate their usefulness as a means to investigate sinonasal transepithelial transport and CFTR function. METHODS PNE derived from the septum or turbinates of CFTR(+/+) and CFTR(-/-) pigs were cultured at an air-liquid interface to confluence and full differentiation. Epithelial monolayers were mounted in Ussing chambers to investigate pharmacologic manipulation of ion transport. Ciliary beat frequency (CBF) and scanning electron microscopy of monolayers were used to indicate degree of ciliation and cell differentiation. RESULTS Stimulation of CFTR-mediated anion transport (ΔIsc in μA/cm(2) ) was significantly greater in epithelia derived from the septum when compared to turbinates (33.04 ± 1.17 vs 18.9 ± 0.73; p < 0.05). Cyclic adenosine monophosphate (cAMP)-activated Cl(-) secretion was absent in CFTR(-/-) and present in CFTR(+/+) epithelia. Calcium-activated Cl(-) (CaCC) secretion was increased in CF; however, overall Cl(-) transport through CaCCs was very low. Degree of ciliation (90%) and CBF were similar between groups. CONCLUSION Septal PNE exhibit a robust ion transport phenotype and indicate CFTR(-/-) sinus disease could be attributable to diminished alternative pathways for Cl(-) transport. Overall, PNE have similarities to human respiratory epithelia not demonstrated in murine cells and represent useful in vitro models for studying CF sinus disease.
Collapse
Affiliation(s)
- Nichole Dean
- Department of Surgery, Division of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Tuggle KL, Birket SE, Cui X, Hong J, Warren J, Reid L, Chambers A, Ji D, Gamber K, Chu KK, Tearney G, Tang LP, Fortenberry JA, Du M, Cadillac JM, Bedwell DM, Rowe SM, Sorscher EJ, Fanucchi MV. Characterization of defects in ion transport and tissue development in cystic fibrosis transmembrane conductance regulator (CFTR)-knockout rats. PLoS One 2014; 9:e91253. [PMID: 24608905 PMCID: PMC3946746 DOI: 10.1371/journal.pone.0091253] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/10/2014] [Indexed: 11/28/2022] Open
Abstract
Animal models for cystic fibrosis (CF) have contributed significantly to our understanding of disease pathogenesis. Here we describe development and characterization of the first cystic fibrosis rat, in which the cystic fibrosis transmembrane conductance regulator gene (CFTR) was knocked out using a pair of zinc finger endonucleases (ZFN). The disrupted Cftr gene carries a 16 base pair deletion in exon 3, resulting in loss of CFTR protein expression. Breeding of heterozygous (CFTR+/−) rats resulted in Mendelian distribution of wild-type, heterozygous, and homozygous (CFTR−/−) pups. Nasal potential difference and transepithelial short circuit current measurements established a robust CF bioelectric phenotype, similar in many respects to that seen in CF patients. Young CFTR−/− rats exhibited histological abnormalities in the ileum and increased intracellular mucus in the proximal nasal septa. By six weeks of age, CFTR−/− males lacked the vas deferens bilaterally. Airway surface liquid and periciliary liquid depth were reduced, and submucosal gland size was abnormal in CFTR−/− animals. Use of ZFN based gene disruption successfully generated a CF animal model that recapitulates many aspects of human disease, and may be useful for modeling other CF genotypes, including CFTR processing defects, premature truncation alleles, and channel gating abnormalities.
Collapse
Affiliation(s)
- Katherine L. Tuggle
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Susan E. Birket
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Xiaoxia Cui
- SAGE Labs, Inc., St. Louis, Missouri, United States of America
| | - Jeong Hong
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Joe Warren
- SAGE Labs, Inc., St. Louis, Missouri, United States of America
| | - Lara Reid
- SAGE Labs, Inc., St. Louis, Missouri, United States of America
| | - Andre Chambers
- SAGE Labs, Inc., St. Louis, Missouri, United States of America
| | - Diana Ji
- SAGE Labs, Inc., St. Louis, Missouri, United States of America
| | - Kevin Gamber
- SAGE Labs, Inc., St. Louis, Missouri, United States of America
| | - Kengyeh K. Chu
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Guillermo Tearney
- Wellman Center for Photomedicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Li Ping Tang
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - James A. Fortenberry
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ming Du
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Animal Resources Program, Office of the Vice President for Research, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Joan M. Cadillac
- Animal Resources Program, Office of the Vice President for Research, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David M. Bedwell
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Steven M. Rowe
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Eric J. Sorscher
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Michelle V. Fanucchi
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
48
|
Berkhout MC, van Rooden CJ, Aalbers RC, el Bouazzaoui LH, Fokkens WJ, Rijntjes E, Heijerman HGM. Temporal bone pneumatization in cystic fibrosis: a correlation with genotype? Laryngoscope 2014; 124:1682-6. [PMID: 24374715 DOI: 10.1002/lary.24575] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/05/2013] [Accepted: 12/23/2013] [Indexed: 11/10/2022]
Abstract
OBJECTIVES/HYPOTHESIS Paranasal sinus pneumatization in patients with cystic fibrosis (CF) is less extensive compared to the general population and seems to be correlated to CF genotype. Interestingly, in CF patients temporal bone pneumatization (TBP) is more extensive compared to the general population, and middle ear pathology is generally uncommon in CF. It is debated whether TBP is influenced environmentally or genetically. The aim of the present study was to investigate pneumatization of the temporal bone in patients with CF and to correlate this with genotype and paranasal sinus volume. STUDY DESIGN Prospective collection of data. METHODS In 104 adult CF patients, computed tomography of the temporal bone and the paranasal sinuses was performed. TBP was graded using a validated scoring system. Patients were divided into two groups, mild and severe CF, based on their mutations in the CF transmembrane conductance regulator gene. RESULTS Of the 31 patients with mild CF, 71% had extensive TBP, and of the 73 patients with severe CF, 82% had extensive pneumatization of the temporal bone. TBP did not differ significantly for CF genotype, and TBP was not correlated to paranasal sinus volume. CONCLUSIONS Whereas paranasal sinus pneumatization in CF patients seems to be related to CF genotype among other influencing factors, this study showed no correlation between TBP and CF genotype. TBP was not correlated to paranasal sinus volume. Hypothetically, in CF, pneumatization of the temporal bone is under a different influence than paranasal sinus pneumatization. LEVEL OF EVIDENCE 4.
Collapse
Affiliation(s)
- Maaike C Berkhout
- Departments of Pulmonology, Haga Teaching Hospital, the Hague, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
49
|
Lung inflammation in cystic fibrosis: pathogenesis and novel therapies. Clin Biochem 2013; 47:539-46. [PMID: 24380764 DOI: 10.1016/j.clinbiochem.2013.12.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 12/11/2013] [Accepted: 12/16/2013] [Indexed: 12/24/2022]
Abstract
Despite remarkable progress following the identification of the causing gene, the final outcome of cystic fibrosis (CF) remains determined mainly by the progressive reduction of lung function. Inflammation of the airways is one of the key elements of the pathogenesis of the disease: it is responsible for the destruction of lung architecture, resulting in progressive loss of respiratory function. Bronchial infection induces an intense inflammatory reaction characterized by a massive invasion of neutrophils, the properties of which seems altered in CF. Moreover, the inflammatory process is also marked by a profuse release of soluble pro-inflammatory mediators, such as interleukin (IL)-6, IL-1β and IL-8 cytokines. In contrast, release of the anti-inflammatory mediator IL-10 is reduced, thus reflecting a pro-/anti-inflammatory imbalance. The inflammation/infection pair seems hard to dissociate, and the origin of the baneful consequences of the persisting excessive inflammatory responses remains to be cleared up: does inflammation follow or rather precede infection? Recent data suggest that uncontrolled inflammation is constitutive in CF. Countering it at early stages of the disease in order to prevent irretrievable damages in lungs remains a major priority in treating patients with CF. In this review, we discuss the usefulness and limitations of mouse models of CF to study the pathogenesis of human lung inflammatory disease, and the development of new potential strategies to reduce the inflammatory burden in the airways.
Collapse
|
50
|
Chang EH. New insights into the pathogenesis of cystic fibrosis sinusitis. Int Forum Allergy Rhinol 2013; 4:132-7. [PMID: 24282147 DOI: 10.1002/alr.21252] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND People with cystic fibrosis (CF) sinus disease have developmental sinus abnormalities with airway bacterial infection, inflammation, impaired mucociliary clearance and thick obstructive mucus. The pathophysiology of airway disease in CF is not completely understood, and current treatments in CF sinus disease ameliorate symptoms but do not provide a cure. METHODS This manuscript reviews the history of CF, its manifestations in sinus disease, and the potential impact and relationship of CF on the upper and lower airway. RESULTS There is increasing evidence that CF sinus disease may affect CF lung disease, the most common cause of mortality in CF. We have been limited in treating the symptoms of advanced CF sinus disease with our current therapies. CONCLUSIONS Recent discoveries in the pathophysiology of CF using the CF porcine animal model and exciting treatments that address the primary gene defect that may translate into improved outcomes in CF and non-CF sinusitis in humans.
Collapse
Affiliation(s)
- Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|