1
|
Ran P, Jiang F, Pan L, Shu Y, Hu F, Wang Y, Zhao R, Wang W, Mu H, Wang J, Wei J, Fu G. Polysaccharide from Atractylodes macrocephala Koidz. alleviates pyrotinib-induced diarrhea through regulating cAMP/LKB1/AMPK/CFTR pathway and restoring gut microbiota and metabolites. Int J Biol Macromol 2025; 308:142512. [PMID: 40157659 DOI: 10.1016/j.ijbiomac.2025.142512] [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: 10/21/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Polysaccharide from Atractylodes macrocephala Koidz. (PAMK), a bioactive component of Atractylodes macrocephala Koidz. (AMK), demonstrates a wide range of pharmacological activities, including the enhancement of gastrointestinal function and regulation of internal homeostasis. This study explores the potential of PAMK in alleviating pyrotinib-induced diarrhea and modulating gut microbiota and its metabolites. Pyrotinib is a tyrosine kinase inhibitor used in cancer treatment, is known for its side effect of diarrhea, which significantly diminishes patients' quality of life. Our prior research suggests that pyrotinib-induced diarrhea may be linked to CFTR-mediated dysregulation of chloride secretion. The present findings indicate that PAMK alleviates pyrotinib-induced diarrhea by reducing cAMP levels, activating the LKB1/AMPK pathway, and inhibiting CFTR activity, as confirmed by enzyme-linked immunosorbent assay (ELISA), qRT-PCR, and western blot analyses. PAMK effectively decreased CFTR-mediated chloride ion secretion in pyrotinib-treated cells, as shown by the MQAE assay. At specific doses, PAMK alleviated pyrotinib-induced diarrhea in rats and significantly restored intestinal barrier integrity. Furthermore, PAMK treatment rebalanced the gut microbiota, reversing the pyrotinib-induced increase in Clostridium and Erysipelotrichi species. Metabolomic profiling further highlighted the involvement of the AMPK signaling pathway. These findings provide a basis for future research aimed at developing cancer treatments with reduced side effects.
Collapse
Affiliation(s)
- Pancen Ran
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Fengxian Jiang
- Department of Radiation Oncology, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan 250031, China
| | - Liying Pan
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Yang Shu
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Fangyan Hu
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yahui Wang
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China
| | - Rui Zhao
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Weihao Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Huaiqian Mu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Juqiong Wang
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Jian Wei
- School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.
| | - Guobin Fu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, China; Department of Medical Oncology, The Third Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250031, China.
| |
Collapse
|
2
|
Suthakaran S, Homami S, Chavez D, Tang S, Moore SKL, Sussman C, Zhang J, Britto CJ, Prince A, May AJ, Kathiriya JJ, Hook JL. CFTR function in alveolar type 1 cells drives lung liquid secretion and host defense. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645303. [PMID: 40196491 PMCID: PMC11974899 DOI: 10.1101/2025.03.25.645303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Loss of the liquid layer that lines the lung's air-facing surface underpins mechanisms of major lung diseases, but the development of therapies that restore liquid secretion is hampered by an incomplete understanding of the cell types that drive it. Here, we show CFTR function in alveolar type 1 (AT1) cells - a cell type that comprises 95% of the lung surface but is presumed to be unimportant in CFTR-related diseases - is critical to lung liquid secretion and the secretion-mediated clearance of particles and S. aureus from lung alveoli. Our findings reveal essential roles for AT1 cells in lung homeostasis and defense, and they call for a reevaluation of the role of AT1 cells in CFTR-related diseases. We suggest AT1 cells be considered key targets of secretion-restoring therapies.
Collapse
|
3
|
Seidler UE. The enigmatic SLC26A6 multifunctional anion transporter: recent advances in structure-function relationship, pathophysiological significance and novel pharmacological inhibitors. Front Pharmacol 2025; 15:1536864. [PMID: 39949394 PMCID: PMC11821980 DOI: 10.3389/fphar.2024.1536864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 12/31/2024] [Indexed: 02/16/2025] Open
Abstract
SLC26A6, a member of the SLC26 family of multifunctional anion transporters, has been particularly enigmatic because of its multiple modes of transport, its expression in organs that are difficult to study physiologically, and the lack of specific antibodies and inhibitors. This has recently changed. SLC26A6 is expressed in the human pancreas, kidney, intestine, heart and some other organs and is involved in fluid absorption, anion secretion, regulation of intracellular pH and elimination of waste products such as oxalate. This review will focus on three topics: Firstly, a molecular structure of human SLC26A6 has recently been obtained by cryo-electron microscopy. Structure-function studies of the reconstituted SLC26A6 in proteoliposomes suggested a 1:1 stoichiometry, resulting in electroneutral Cl-/HCO3 - exchange and electrogenic Cl-/oxalate2- exchange. How do these data help to understand the published functional studies? Secondly, whole exon sequencing of a kidney stone cohort from the United Kingdom database revealed a dominant negative SLC26A6 mutation in a patient with enteric hyperoxaluria, oxalate kidney stones and a low calcium diet. How does this finding fit with previous genetic studies in mice and humans of SLC26A6 gene mutations? Thirdly, progress has been made in identifying specific inhibitors for SLC26A6. Where might this be of clinical relevance?
Collapse
Affiliation(s)
- Ursula E. Seidler
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
4
|
Arreola J, Pérez-Cornejo P, Segura-Covarrubias G, Corral-Fernández N, León-Aparicio D, Guzmán-Hernández ML. Function and Regulation of the Calcium-Activated Chloride Channel Anoctamin 1 (TMEM16A). Handb Exp Pharmacol 2024; 283:101-151. [PMID: 35768554 DOI: 10.1007/164_2022_592] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Various human tissues express the calcium-activated chloride channel Anoctamin 1 (ANO1), also known as TMEM16A. ANO1 allows the passive chloride flux that controls different physiological functions ranging from muscle contraction, fluid and hormone secretion, gastrointestinal motility, and electrical excitability. Overexpression of ANO1 is associated with pathological conditions such as hypertension and cancer. The molecular cloning of ANO1 has led to a surge in structural, functional, and physiological studies of the channel in several tissues. ANO1 is a homodimer channel harboring two pores - one in each monomer - that work independently. Each pore is activated by voltage-dependent binding of two intracellular calcium ions to a high-affinity-binding site. In addition, the binding of phosphatidylinositol 4,5-bisphosphate to sites scattered throughout the cytosolic side of the protein aids the calcium activation process. Furthermore, many pharmacological studies have established ANO1 as a target of promising compounds that could treat several illnesses. This chapter describes our current understanding of the physiological roles of ANO1 and its regulation under physiological conditions as well as new pharmacological compounds with potential therapeutic applications.
Collapse
Affiliation(s)
- Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Guadalupe Segura-Covarrubias
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Nancy Corral-Fernández
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Daniel León-Aparicio
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | |
Collapse
|
5
|
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
|
6
|
Martinovich KM, Kicic A, Stick SM, Johnsen RD, Fletcher S, Wilton SD. Investigating the Implications of CFTR Exon Skipping Using a Cftr Exon 9 Deleted Mouse Model. Front Pharmacol 2022; 13:868863. [PMID: 35392567 PMCID: PMC8981082 DOI: 10.3389/fphar.2022.868863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Severity and disease progression in people with Cystic Fibrosis (CF) is typically dependent on their genotype. One potential therapeutic strategy for people with specific mutations is exon skipping with antisense oligonucleotides (AO). CFTR exon 9 is an in-frame exon and hence the exclusion of this exon would excise only 31 amino acids but not alter the reading frame of the remaining mRNA. Splice mutations 1209 + 1 G > C and 1209 + 2 T > G were documented to cause CFTR exon 9 skipping and these variants were reported to manifest as a milder CF disease, therefore exon 9 skipping could be beneficial for people with class I mutations that affect exon 9 such as p.Trp401X. While the impact of exon 9 skipping on gene expression and cellular pathways can be studied in cells in vitro, trace amount of full-length normal or mutated material could confound the evaluation. To overcome this limitation, the impact of CFTR exon 9 skipping on disease phenotype and severity is more effectively evaluated in a small animal model. It was hypothesised that antisense oligonucleotide-mediated skipping this particular exon could result in a "mild mouse CF phenotype". Methods: Cftr exon 9 deleted mice were generated using homologous recombination. Survival of homozygous (Cftr Δ9/Δ9 ) and heterozygous (Cftr Δ9/+ ) mice was compared to that of other CF mouse models, and lung and intestinal organ histology examined for any pathologies. Primary airway epithelial cells (pAECs) were harvested from Cftr Δ9/Δ9 mice and cultured at the Air Liquid Interface for CFTR functional assessment using Ussing Chamber analysis. Results: A Cftr Δ9/Δ9 mouse model presented with intestinal obstructions, and at time of weaning (21 days). Cftr Δ9/Δ9 mice had a survival rate of 83% that dropped to 38% by day 50. Histological sections of the small intestine from Cftr Δ9/Δ9 mice showed more goblet cells and mucus accumulation than samples from the Cftr Δ9/+ littermates. Airway epithelial cell cultures established from Cftr Δ9/Δ9 mice were not responsive to forskolin stimulation. Summary: The effect of Cftr exon 9 deletion on Cftr function was assessed and it was determined that the encoded Cftr isoform did not result in a milder "mouse CF disease phenotype," suggesting that Cftr exon 9 is not dispensable, although further investigation in human CF pAECs would be required to confirm this observation.
Collapse
Affiliation(s)
- Kelly M Martinovich
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Anthony Kicic
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Childrens Hospital, Nedlands, WA, Australia.,School of Population Health, Curtin University, Bentley, WA, Australia
| | - Stephen M Stick
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Childrens Hospital, Nedlands, WA, Australia
| | - Russell D Johnsen
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia.,PYC Therapeutics, Perth, WA, Australia
| | - Steve D Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
7
|
Takei Y, Ando M, Wong MKS, Tsukada T. Molecular mechanisms underlying guanylin-induced transcellular Cl - secretion into the intestinal lumen of seawater-acclimated eels. Gen Comp Endocrinol 2022; 318:113986. [PMID: 35114197 DOI: 10.1016/j.ygcen.2022.113986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/20/2021] [Accepted: 01/27/2022] [Indexed: 11/19/2022]
Abstract
Guanylin (GN) stimulates Cl- secretion into the intestinal lumen of seawater-acclimated eels, but the molecular mechanisms of transepithelial Cl- transport are still unknown. In Ussing chamber experiments, we confirmed that mucosal application of eel GN reversed intestinal serosa-negative potential difference, indicating Cl- secretion. Serosal application of DNDS or mucosal application of DPC inhibited the GN effect, but serosal application of bumetanide had no effect. Removal of HCO3- from the serosal fluid also inhibited the GN effect. In intestinal sac experiments, mucosal GN stimulated luminal secretion of both Cl- and Na+, which was blocked by serosal DNDS. These results suggest that Cl- is taken up at the serosal side by DNDS-sensitive anion exchanger (AE) coupled with Na+-HCO3- cotransporter (NBC) but not by Na+-K+-2Cl- cotransporter 1 (NKCC1), and Cl- is secreted by unknown DPC-sensitive Cl- channel (ClC) at the mucosal side. The transcriptomic analysis combined with qPCR showed low expression of NKCC1 gene and no upregulation of the gene after seawater transfer, while high expression of ClC2 gene and upregulation after seawater transfer. In addition, SO42- transporters (apical Slc26a3/6 and basolateral Slc26a1) are also candidates for transcellular Cl- secretion in exchange of luminal SO42. Na+ secretion could occur through a paracellular route, as Na+-leaky claudin15 was highly expressed and upregulated after seawater transfer. High local Na+ concentration in the lateral interspace produced by Na+/K+-ATPase (NKA) coupled with K+ channels (Kir5.1b) seems to facilitate the paracellular transport. In situ hybridization confirmed the expression of the candidate genes in the epithelial enterocytes. Together with our previous results, we suggest that GN stimulates basolateral NBCela/AE2 and apical ClC2 to increase transcellular Cl- secretion in seawater eel intestine, which differs from the involvement of apical CFTR and basolateral NKCC1 as suggested in mammals and other teleosts.
Collapse
Affiliation(s)
- Yoshio Takei
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan.
| | - Masaaki Ando
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan
| | - Marty K S Wong
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba 277-8564, Japan
| | - Takehiro Tsukada
- Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
8
|
Methodological tools to study species of the genus Burkholderia. Appl Microbiol Biotechnol 2021; 105:9019-9034. [PMID: 34755214 PMCID: PMC8578011 DOI: 10.1007/s00253-021-11667-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/26/2022]
Abstract
Bacteria belonging to the Burkholderia genus are extremely versatile and diverse. They can be environmental isolates, opportunistic pathogens in cystic fibrosis, immunocompromised or chronic granulomatous disease patients, or cause disease in healthy people (e.g., Burkholderia pseudomallei) or animals (as in the case of Burkholderia mallei). Since the genus was separated from the Pseudomonas one in the 1990s, the methodological tools to study and characterize these bacteria are evolving fast. Here we reviewed the techniques used in the last few years to update the taxonomy of the genus, to study gene functions and regulations, to deepen the knowledge on the drug resistance which characterizes these bacteria, and to elucidate their mechanisms to establish infections. The availability of these tools significantly impacts the quality of research on Burkholderia and the choice of the most appropriated is fundamental for a precise characterization of the species of interest. Key points • Updated techniques to study the genus Burkholderia were reviewed. • Taxonomy, genomics, assays, and animal models were described. • A comprehensive overview on recent advances in Burkholderia studies was made.
Collapse
|
9
|
Choudhary I, Vo T, Paudel K, Yadav R, Mao Y, Patial S, Saini Y. Postnatal Ozone Exposure Disrupts Alveolar Development, Exaggerates Mucoinflammatory Responses, and Suppresses Bacterial Clearance in Developing Scnn1b-Tg + Mice Lungs. THE JOURNAL OF IMMUNOLOGY 2021; 207:1165-1179. [PMID: 34330754 DOI: 10.4049/jimmunol.2001286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/03/2021] [Indexed: 11/19/2022]
Abstract
Increased levels of ambient ozone, one of the six criteria air pollutants, result in respiratory tract injury and worsening of ongoing lung diseases. However, the effect of ozone exposure on the respiratory tract undergoing active lung development and simultaneously experiencing mucoinflammatory lung diseases, such as cystic fibrosis, remains unclear. To address these questions, we exposed Scnn1b transgenic (Scnn1b-Tg+) mice, a mouse model of cystic fibrosis-like lung disease, and littermate wild-type (WT) mice to ozone from postnatal days (PND) 3-20 and examined the lung phenotypes at PND21. As compared with filtered air (FA)-exposed WT mice, the ozone-exposed WT mice exhibited marked alveolar space enlargement, in addition to significant eosinophilic infiltration, type 2 inflammation, and mucous cell metaplasia. Ozone-exposed Scnn1b-Tg+ mice also exhibited significantly increased alveolar space enlargement, which was also accompanied by exaggerated granulocytic infiltration, type 2 inflammation, and a greater degree of mucus obstruction. The alveolar space enlargement in ozone-exposed WT, FA-exposed Scnn1b-Tg+, and ozone-exposed Scnn1b-Tg+ mice was accompanied by elevated levels of MMP12 protein in macrophages and Mmp12 mRNA in the lung homogenates. Finally, although bacterial burden was largely resolved by PND21 in FA-exposed Scnn1b-Tg+ mice, ozone-exposed Scnn1b-Tg+ mice exhibited compromised bacterial clearance, which was also associated with increased levels of IL-10, an immunosuppressive cytokine, and marked mucus obstruction. Taken together, our data show that ozone exposure results in alveolar space remodeling during active phases of lung development and markedly exaggerates the mucoinflammatory outcomes of pediatric-onset lung disease, including bacterial infections, granulocytic inflammation, mucus obstruction, and alveolar space enlargement.
Collapse
Affiliation(s)
- Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Thao Vo
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Kshitiz Paudel
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Radha Yadav
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Yun Mao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| |
Collapse
|
10
|
Pai AC, Parekh KR, Engelhardt JF, Lynch TJ. Ferret respiratory disease models for the study of lung stem cells. LUNG STEM CELLS IN DEVELOPMENT, HEALTH AND DISEASE 2021. [DOI: 10.1183/2312508x.10010320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
11
|
Tan Q, di Stefano G, Tan X, Renjie X, Römermann D, Talbot SR, Seidler UE. Inhibition of Na + /H + exchanger isoform 3 improves gut fluidity and alkalinity in cystic fibrosis transmembrane conductance regulator-deficient and F508del mutant mice. Br J Pharmacol 2021; 178:1018-1036. [PMID: 33179259 DOI: 10.1111/bph.15323] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Constipation and intestinal obstructive episodes are major health problems in cystic fibrosis (CF) patients. Three FDA-approved drugs against constipation-prone irritable bowel syndrome were tested for their ability to increase luminal fluidity and alkalinity in cystic fibrosis transmembrane conductance regulator (CFTR) null (cftr-/- ) and F508del mutant (F508delmut/mut ) murine intestine. EXPERIMENTAL APPROACH Guanylate cyclase C agonist linaclotide, PGE1 analogue lubiprostone and intestine-specific NHE3 inhibitor tenapanor were perfused through a ~3 cm jejunal, proximal or mid-distal colonic segment in anaesthetized cftr-/- , F508delmut/mut and WT mice. Net fluid balance was determined gravimetrically and alkaline output by pH-stat back titration. KEY RESULTS Basal jejunal fluid absorptive rates were significantly higher and basal HCO3 - output was significantly lower in cftr-/- and F508delmut/mut compared to WT mice. In cftr-/- and F508delmut/mut mice, all three drugs significantly inhibited the fluid absorptive rate and increased alkaline output in the jejunum and tenapanor and lubiprostone, but not linaclotide, in the colon. After tenapanor pre-incubation, linaclotide elicited a robust fluid secretory response in WT jejunum, while no further change in absorptive rates was observed in cftr-/- and F508delmut/mut jejunum, suggesting that the increase in gut fluidity and alkalinity by linaclotide in CF gut is mediated via NHE3 inhibition. Lubiprostone also inhibited fluid absorption in cftr-/- and F508delmut/mut jejunum via NHE3 inhibition but had a residual NHE3-independent effect. CONCLUSION AND IMPLICATIONS Linaclotide, lubiprostone and tenapanor reduced fluid absorption and increased alkaline output in the CF gut. Their application may ameliorate constipation and reduce obstructive episodes in CF patients.
Collapse
Affiliation(s)
- Qinghai Tan
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | | | - Xinjie Tan
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | - Xiu Renjie
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | - Dorothee Römermann
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | - Steven R Talbot
- Institute of Veterinary Research, Hannover Medical School, Hanover, Germany
| | - Ursula E Seidler
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
12
|
Mondejar-Parreño G, Perez-Vizcaino F, Cogolludo A. Kv7 Channels in Lung Diseases. Front Physiol 2020; 11:634. [PMID: 32676036 PMCID: PMC7333540 DOI: 10.3389/fphys.2020.00634] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/18/2020] [Indexed: 12/23/2022] Open
Abstract
Lung diseases constitute a global health concern causing disability. According to WHO in 2016, respiratory diseases accounted for 24% of world population mortality, the second cause of death after cardiovascular diseases. The Kv7 channels family is a group of voltage-dependent K+ channels (Kv) encoded by KCNQ genes that are involved in various physiological functions in numerous cell types, especially, cardiac myocytes, smooth muscle cells, neurons, and epithelial cells. Kv7 channel α-subunits are regulated by KCNE1–5 ancillary β-subunits, which modulate several characteristics of Kv7 channels such as biophysical properties, cell-location, channel trafficking, and pharmacological sensitivity. Kv7 channels are mainly expressed in two large groups of lung tissues: pulmonary arteries (PAs) and bronchial tubes. In PA, Kv7 channels are expressed in pulmonary artery smooth muscle cells (PASMCs); while in the airway (trachea, bronchus, and bronchioles), Kv7 channels are expressed in airway smooth muscle cells (ASMCs), airway epithelial cells (AEPs), and vagal airway C-fibers (VACFs). The functional role of Kv7 channels may vary depending on the cell type. Several studies have demonstrated that the impairment of Kv7 channel has a strong impact on pulmonary physiology contributing to the pathophysiology of different respiratory diseases such as cystic fibrosis, asthma, chronic obstructive pulmonary disease, chronic coughing, lung cancer, and pulmonary hypertension. Kv7 channels are now recognized as playing relevant physiological roles in many tissues, which have encouraged the search for Kv7 channel modulators with potential therapeutic use in many diseases including those affecting the lung. Modulation of Kv7 channels has been proposed to provide beneficial effects in a number of lung conditions. Therefore, Kv7 channel openers/enhancers or drugs acting partly through these channels have been proposed as bronchodilators, expectorants, antitussives, chemotherapeutics and pulmonary vasodilators.
Collapse
Affiliation(s)
- Gema Mondejar-Parreño
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| | - Angel Cogolludo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Ciber Enfermedades Respiratorias (Ciberes), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
| |
Collapse
|
13
|
He M, Wu B, Ye W, Le DD, Sinclair AW, Padovano V, Chen Y, Li KX, Sit R, Tan M, Caplan MJ, Neff N, Jan YN, Darmanis S, Jan LY. Chloride channels regulate differentiation and barrier functions of the mammalian airway. eLife 2020; 9:e53085. [PMID: 32286221 PMCID: PMC7182432 DOI: 10.7554/elife.53085] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 04/13/2020] [Indexed: 12/16/2022] Open
Abstract
The conducting airway forms a protective mucosal barrier and is the primary target of airway disorders. The molecular events required for the formation and function of the airway mucosal barrier, as well as the mechanisms by which barrier dysfunction leads to early onset airway diseases, remain unclear. In this study, we systematically characterized the developmental landscape of the mouse airway using single-cell RNA sequencing and identified remarkably conserved cellular programs operating during human fetal development. We demonstrated that in mouse, genetic inactivation of chloride channel Ano1/Tmem16a compromises airway barrier function, results in early signs of inflammation, and alters the airway cellular landscape by depleting epithelial progenitors. Mouse Ano1-/-mutants exhibited mucus obstruction and abnormal mucociliary clearance that resemble the airway defects associated with cystic fibrosis. The data reveal critical and non-redundant roles for Ano1 in organogenesis, and show that chloride channels are essential for mammalian airway formation and function.
Collapse
Affiliation(s)
- Mu He
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Bing Wu
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Wenlei Ye
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Daniel D Le
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Adriane W Sinclair
- Department of Urology, University of California, San FranciscoSan FranciscoUnited States
- Division of Pediatric Urology, University of California, San Francisco, Benioff Children's HospitalSan FranciscoUnited States
| | - Valeria Padovano
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HeavenUnited States
| | - Yuzhang Chen
- Department of Anesthesia and Perioperative Care, University of California, San FranciscoSan FranciscoUnited States
| | - Ke-Xin Li
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Rene Sit
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Michelle Tan
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HeavenUnited States
| | - Norma Neff
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Yuh Nung Jan
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical Institute, University of California, San FranciscoSan FranciscoUnited States
| | | | - Lily Yeh Jan
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical Institute, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
14
|
Cho DY, Skinner D, Zhang S, Lazrak A, Lim DJ, Weeks CG, Banks CG, Han CK, Kim SK, Tearney GJ, Matalon S, Rowe SM, Woodworth BA. Korean Red Ginseng aqueous extract improves markers of mucociliary clearance by stimulating chloride secretion. J Ginseng Res 2019; 45:66-74. [PMID: 33437158 PMCID: PMC7790903 DOI: 10.1016/j.jgr.2019.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 08/30/2019] [Accepted: 09/06/2019] [Indexed: 11/26/2022] Open
Abstract
Background Abnormal chloride (Cl-) transport has a detrimental impact on mucociliary clearance in both cystic fibrosis (CF) and non-CF chronic rhinosinusitis. Ginseng is a medicinal plant noted to have anti-inflammatory and antimicrobial properties. The present study aims to assess the capability of red ginseng aqueous extract (RGAE) to promote transepithelial Cl- secretion in nasal epithelium. Methods Primary murine nasal septal epithelial (MNSE) [wild-type (WT) and transgenic CFTR-/-], fisher-rat-thyroid (FRT) cells expressing human WT CFTR, and TMEM16A-expressing human embryonic kidney cultures were utilized for the present experiments. Ciliary beat frequency (CBF) and airway surface liquid (ASL) depth measurements were performed using micro-optical coherence tomography (μOCT). Mechanisms underlying transepithelial Cl- transport were determined using pharmacologic manipulation in Ussing chambers and whole-cell patch clamp analysis. Results RGAE (at 30μg/mL of ginsenosides) significantly increased Cl- transport [measured as change in short-circuit current (ΔISC = μA/cm2)] when compared with control in WT and CFTR-/- MNSE (WT vs control = 49.8±2.6 vs 0.1+/-0.2, CFTR-/- = 33.5±1.5 vs 0.2±0.3, p < 0.0001). In FRT cells, the CFTR-mediated ΔISC attributed to RGAE was small (6.8 ± 2.5 vs control, 0.03 ± 0.01, p < 0.05). In patch clamp, TMEM16A-mediated currents were markedly improved with co-administration of RGAE and uridine 5-triphosphate (8406.3 +/- 807.7 pA) over uridine 5-triphosphate (3524.1 +/- 292.4 pA) or RGAE alone (465.2 +/- 90.7 pA) (p < 0.0001). ASL and CBF were significantly greater with RGAE (6.2+/-0.3 μm vs control, 3.9+/-0.09 μm; 10.4+/-0.3 Hz vs control, 7.3 ± 0.2 Hz; p < 0.0001) in MNSE. Conclusion RGAE augments ASL depth and CBF by stimulating Cl- secretion through CaCC, which suggests therapeutic potential in both CF and non-CF chronic rhinosinusitis.
Collapse
Affiliation(s)
- Do-Yeon Cho
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Daniel Skinner
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Shaoyan Zhang
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ahmed Lazrak
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States.,Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Dong Jin Lim
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Christopher G Weeks
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Catherine G Banks
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Chang Kyun Han
- Korea Ginseng Research Institute, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Si-Kwan Kim
- Department of Biomedical Chemistry, Konkuk University, Chungju, Republic of Korea
| | - Guillermo J Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Sadis Matalon
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States.,Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Steven M Rowe
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States.,Departments of Medicine, Pediatrics, Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Bradford A Woodworth
- Department of Otolaryngology Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
15
|
Lee B, Hong GS, Lee SH, Kim H, Kim A, Hwang EM, Kim J, Lee MG, Yang JY, Kweon MN, Tse CM, Mark D, Oh U. Anoctamin 1/TMEM16A controls intestinal Cl - secretion induced by carbachol and cholera toxin. Exp Mol Med 2019; 51:1-14. [PMID: 31383845 PMCID: PMC6802608 DOI: 10.1038/s12276-019-0287-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/07/2019] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
Calcium-activated chloride channels (CaCCs) mediate numerous physiological functions and are best known for the transport of electrolytes and water in epithelia. In the intestine, CaCC currents are considered necessary for the secretion of fluid to protect the intestinal epithelium. Although genetic ablation of ANO1/TMEM16A, a gene encoding a CaCC, reduces the carbachol-induced secretion of intestinal fluid, its mechanism of action is still unknown. Here, we confirm that ANO1 is essential for the secretion of intestinal fluid. Carbachol-induced transepithelial currents were reduced in the proximal colon of Ano1-deficient mice. Surprisingly, cholera toxin-induced and cAMP-induced fluid secretion, believed to be mediated by CFTR, were also significantly reduced in the intestine of Ano1-deficient mice. ANO1 is largely expressed in the apical membranes of intestines, as predicted for CaCCs. The Ano1-deficient colons became edematous under basal conditions and had a greater susceptibility to dextran sodium sulfate-induced colitis. However, Ano1 depletion failed to affect tumor development in a model of colorectal cancer. We thus conclude that ANO1 is necessary for cAMP- and carbachol-induced Cl− secretion in the intestine, which is essential for the protection of the intestinal epithelium from colitis. An ion channel, a membrane protein allowing ion transport, that controls the flow of chloride is needed for proper secretion of protective fluids in the intestine. Uhtaek Oh from the Korea Institute of Science & Technology in Seoul, South Korea, and colleagues showed that cells lining the intestinal surface express a calcium-activated chloride channel called anoctamin-1 (ANO1) that regulates fluid secretion in the gut. Compared to control animals, ANO1-deficient mice released less fluid into their intestines following exposure to a diarrhea-inducing toxin or to a chloride transport–stimulating signaling molecule. This fluid secretion was previously thought to be mediated via a different ion channel. The ANO1-deficient mice accumulated fluid within colonic tissues, which increased their susceptibility to colitis. The findings point to ANO1 activation as a potential therapeutic strategy for treating colitis.
Collapse
Affiliation(s)
- Byeongjun Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Gyu-Sang Hong
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Korea
| | - Hyungsup Kim
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Ajung Kim
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Jiyoon Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jin-Young Yang
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, 05505, Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, 05505, Korea
| | - Chung-Ming Tse
- Departments of Physiology and Medicine, Division of Gastroenterois maintained by the opening of plasmalogy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donowitz Mark
- Departments of Physiology and Medicine, Division of Gastroenterois maintained by the opening of plasmalogy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Uhtaek Oh
- College of Pharmacy, Seoul National University, Seoul, 08826, Korea. .,Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea.
| |
Collapse
|
16
|
Vega G, Guequén A, Johansson MEV, Arike L, Martínez-Abad B, Nyström EEL, Scudieri P, Pedemonte N, Millar-Büchner P, Philp AR, Galietta LJ, Hansson GC, Flores CA. Normal Calcium-Activated Anion Secretion in a Mouse Selectively Lacking TMEM16A in Intestinal Epithelium. Front Physiol 2019; 10:694. [PMID: 31263421 PMCID: PMC6585864 DOI: 10.3389/fphys.2019.00694] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/16/2019] [Indexed: 01/24/2023] Open
Abstract
Calcium-activated anion secretion is expected to ameliorate cystic fibrosis, a genetic disease that carries an anion secretory defect in exocrine tissues. Human patients and animal models of the disease that present a mild intestinal phenotype have been postulated to bear a compensatory calcium-activated anion secretion in the intestine. TMEM16A is calcium-activated anion channel whose presence in the intestinal epithelium is contradictory. We aim to test the functional expression of TMEM16A using animal models with Cftr and/or Tmem16a intestinal silencing. Expression of TMEM16A was studied in a wild type and intestinal Tmem16a knockout mice by mRNA-seq, mass-spectrometry, q-PCR, Western blotting and immunolocalization. Calcium-activated anion secretion was recorded in the ileum and proximal colon of these animals including intestinal Cftr knockout and double mutants with dual Tmem16a and Cftr intestinal ablation. Mucus homeostasis was studied by immune-analysis of Mucin-2 (Muc2) and survival curves were recorded. Tmem16a transcript was found in intestine. Nevertheless, protein was barely detected in colon samples. Electrophysiological measurements demonstrated that the intestinal deletion of Tmem16a did not change calcium-activated anion secretion induced by carbachol or ATP in ileum and proximal colon. Muc2 architecture was not altered by Tmem16a silencing as was observed when Cftr was deleted from mouse intestine. Tmem16a silencing neither affected animal survival nor modified the lethality observed in the intestinal Cftr-null mouse. Our results demonstrate that TMEM16A function in the murine intestine is not related to electrogenic calcium-activated anion transport and does not affect mucus homeostasis and survival of animals.
Collapse
Affiliation(s)
- Génesis Vega
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Anita Guequén
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | | | - Liisa Arike
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Paolo Scudieri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | | | - Amber R. Philp
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Luis J. Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy
| | - Gunnar C. Hansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
17
|
Rao MC. Physiology of Electrolyte Transport in the Gut: Implications for Disease. Compr Physiol 2019; 9:947-1023. [PMID: 31187895 DOI: 10.1002/cphy.c180011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We now have an increased understanding of the genetics, cell biology, and physiology of electrolyte transport processes in the mammalian intestine, due to the availability of sophisticated methodologies ranging from genome wide association studies to CRISPR-CAS technology, stem cell-derived organoids, 3D microscopy, electron cryomicroscopy, single cell RNA sequencing, transgenic methodologies, and tools to manipulate cellular processes at a molecular level. This knowledge has simultaneously underscored the complexity of biological systems and the interdependence of multiple regulatory systems. In addition to the plethora of mammalian neurohumoral factors and their cross talk, advances in pyrosequencing and metagenomic analyses have highlighted the relevance of the microbiome to intestinal regulation. This article provides an overview of our current understanding of electrolyte transport processes in the small and large intestine, their regulation in health and how dysregulation at multiple levels can result in disease. Intestinal electrolyte transport is a balance of ion secretory and ion absorptive processes, all exquisitely dependent on the basolateral Na+ /K+ ATPase; when this balance goes awry, it can result in diarrhea or in constipation. The key transporters involved in secretion are the apical membrane Cl- channels and the basolateral Na+ -K+ -2Cl- cotransporter, NKCC1 and K+ channels. Absorption chiefly involves apical membrane Na+ /H+ exchangers and Cl- /HCO3 - exchangers in the small intestine and proximal colon and Na+ channels in the distal colon. Key examples of our current understanding of infectious, inflammatory, and genetic diarrheal diseases and of constipation are provided. © 2019 American Physiological Society. Compr Physiol 9:947-1023, 2019.
Collapse
Affiliation(s)
- Mrinalini C Rao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
18
|
Bose SJ, Bijvelds MJC, Wang Y, Liu J, Cai Z, Bot AGM, de Jonge HR, Sheppard DN. Differential thermostability and response to cystic fibrosis transmembrane conductance regulator potentiators of human and mouse F508del-CFTR. Am J Physiol Lung Cell Mol Physiol 2019; 317:L71-L86. [PMID: 30969810 PMCID: PMC6689747 DOI: 10.1152/ajplung.00034.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cross-species comparative studies have highlighted differences between human and mouse cystic fibrosis transmembrane conductance regulator (CFTR), the epithelial Cl- channel defective in cystic fibrosis (CF). Here, we compare the impact of the most common CF mutation F508del on the function of human and mouse CFTR heterologously expressed in mammalian cells and their response to CFTR modulators using the iodide efflux and patch-clamp techniques. Once delivered to the plasma membrane, human F508del-CFTR exhibited a severe gating defect characterized by infrequent channel openings and was thermally unstable, deactivating within minutes at 37°C. By contrast, the F508del mutation was without effect on the gating pattern of mouse CFTR, and channel activity demonstrated thermostability at 37°C. Strikingly, at all concentrations tested, the clinically approved CFTR potentiator ivacaftor was without effect on the mouse F508del-CFTR Cl- channel. Moreover, eight CFTR potentiators, including ivacaftor, failed to generate CFTR-mediated iodide efflux from CHO cells expressing mouse F508del-CFTR. However, they all produced CFTR-mediated iodide efflux with human F508del-CFTR-expressing CHO cells, while fifteen CFTR correctors rescued the plasma membrane expression of both human and mouse F508del-CFTR. Interestingly, the CFTR potentiator genistein enhanced CFTR-mediated iodide efflux from CHO cells expressing either human or mouse F508del-CFTR, whereas it only potentiated human F508del-CFTR Cl- channels in cell-free membrane patches, suggesting that its action on mouse F508del-CFTR is indirect. Thus, the F508del mutation has distinct effects on human and mouse CFTR Cl- channels.
Collapse
Affiliation(s)
- Samuel J Bose
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - Yiting Wang
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Jia Liu
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Zhiwei Cai
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Alice G M Bot
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| |
Collapse
|
19
|
Semaniakou A, Croll RP, Chappe V. Animal Models in the Pathophysiology of Cystic Fibrosis. Front Pharmacol 2019; 9:1475. [PMID: 30662403 PMCID: PMC6328443 DOI: 10.3389/fphar.2018.01475] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/03/2018] [Indexed: 01/28/2023] Open
Abstract
Our understanding of the multiorgan pathology of cystic fibrosis (CF) has improved impressively during the last decades, but we still lack a full comprehension of the disease progression. Animal models have greatly contributed to the elucidation of specific mechanisms involved in CF pathophysiology and the development of new therapies. Soon after the cloning of the CF transmembrane conductance regulator (CFTR) gene in 1989, the first mouse model was generated and this model has dominated in vivo CF research ever since. Nonetheless, the failure of murine models to mirror human disease severity in the pancreas and lung has led to the generation of larger animal models such as pigs and ferrets. The following review presents and discusses data from the current animal models used in CF research.
Collapse
Affiliation(s)
- Anna Semaniakou
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Valerie Chappe
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
20
|
Ryan K, Byrd TF. Mycobacterium abscessus: Shapeshifter of the Mycobacterial World. Front Microbiol 2018; 9:2642. [PMID: 30443245 PMCID: PMC6221961 DOI: 10.3389/fmicb.2018.02642] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/16/2018] [Indexed: 01/23/2023] Open
Abstract
In this review we will focus on unique aspects of Mycobacterium abscessus (MABS) which we feel earn it the designation of "shapeshifter of the mycobacterial world." We will review its emergence as a distinct species, the recognition and description of MABS subspecies which are only now being clearly defined in terms of pathogenicity, its ability to exist in different forms favoring a saprophytic lifestyle or one more suitable to invasion of mammalian hosts, as well as current challenges in terms of antimicrobial therapy and future directions for research. One can see in the various phases of MABS, a species transitioning from a free living saprophyte to a host-adapted pathogen.
Collapse
Affiliation(s)
- Keenan Ryan
- Department of Pharmacy, University of New Mexico Hospital, Albuquerque, NM, United States
| | - Thomas F. Byrd
- Department of Medicine, The University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
21
|
Imberti R, Garavaglia ML, Verduci I, Cannavale G, Balduzzi G, Papetti S, Mazzanti M. Antiestrogen- and tamoxifen-induced effects on calcium-activated chloride currents in epithelial cells carrying the ∆F508-CFTR point mutation. Respir Res 2018; 19:198. [PMID: 30290809 PMCID: PMC6173901 DOI: 10.1186/s12931-018-0901-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/25/2018] [Indexed: 11/15/2022] Open
Abstract
Background Although pharmacological treatment has increased the average life expectancy of patients with cystic fibrosis, the median survival of females is shorter than that of males. In vitro and in vivo studies have shown that estrogens play a relevant role in the disease progression. The aim of this study was to investigate the effects of 17β-estradiol and tamoxifen citrate (TMX) on calcium-activated chloride channel (CaCC) currents in human bronchial epithelial cells carrying the ΔPhe508-CFTR mutation both in homozygosis and in heterozygosis. Methods Perforated patch clamp experiments were performed on single cells of the immortalized cell lines CFBE and IB3–1. Gramicidin (10 or 20 μM) was added to the electrode solution to reach the whole cell configuration. The electrical stimulation protocol consisted of square voltages ranging from − 80 to + 80 mV, in steps of 20 mV and with a duration of 800 msec. Results The presence of 17β-estradiol significantly reduced the CaCC currents, both in basal conditions and in the presence of ATP (100 μM). The addition of TMX (10 μM) completely restored the currents abolished by 17β-estradiol, in basal conditions and after stimulation with ATP in both CFBE and IB3–1 cells. TMX had a strong, direct action on membrane current density, which significantly increased more than 4-fold in both cases. The membrane current stimulation produced by TMX was further enhanced by the addition of ATP. CFBE cells incubated for 24 h with 3 μM VX-809 (a CFTR corrector) and then acutely stimulated with VX-770 (a CFTR potentiator) in the presence of forskolin, showed an increase of chloride currents which were abolished by Inh-172. The chloride current density induced by TMX + ATP was, on average, greater than that obtained with VX-809 + VX-770 + forskolin. The currents elicited by TMX + ATP were abolished by the addition of NPPB, a CaCC inhibitor. The combined administration of TMX/ATP and VXs/FSK had an additional effect on chloride currents. Conclusions Our results show that TMX restores CaCC currents inhibited by 17ß-estradiol and directly activates the transmembrane chloride currents potentiated by ATP, an effect which is mutation independent. The combined effect of TMX with current used treatments for cystic fibrosis could be of benefit to patients.
Collapse
Affiliation(s)
- Roberto Imberti
- Phase I Clinical Trials Unit and Experimental Therapy, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Maria Lisa Garavaglia
- Department of Biosciences, Laboratory of Cellular and Molecular Physiology, University of Milano, via Celoria 26, I-20133, Milan, Italy
| | - Ivan Verduci
- Department of Biosciences, Laboratory of Cellular and Molecular Physiology, University of Milano, via Celoria 26, I-20133, Milan, Italy
| | - Gaetano Cannavale
- Department of Biosciences, Laboratory of Cellular and Molecular Physiology, University of Milano, via Celoria 26, I-20133, Milan, Italy
| | | | - Sara Papetti
- GB Pharma S.r.l., via Ferreri 11, 27100, Pavia, Italy
| | - Michele Mazzanti
- Department of Biosciences, Laboratory of Cellular and Molecular Physiology, University of Milano, via Celoria 26, I-20133, Milan, Italy.
| |
Collapse
|
22
|
Hahn A, Salomon JJ, Leitz D, Feigenbutz D, Korsch L, Lisewski I, Schrimpf K, Millar-Büchner P, Mall MA, Frings S, Möhrlen F. Expression and function of Anoctamin 1/TMEM16A calcium-activated chloride channels in airways of in vivo mouse models for cystic fibrosis research. Pflugers Arch 2018; 470:1335-1348. [DOI: 10.1007/s00424-018-2160-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/30/2018] [Accepted: 05/23/2018] [Indexed: 01/17/2023]
|
23
|
Rottgen TS, Nickerson AJ, Rajendran VM. Calcium-Activated Cl - Channel: Insights on the Molecular Identity in Epithelial Tissues. Int J Mol Sci 2018; 19:E1432. [PMID: 29748496 PMCID: PMC5983713 DOI: 10.3390/ijms19051432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022] Open
Abstract
Calcium-activated chloride secretion in epithelial tissues has been described for many years. However, the molecular identity of the channel responsible for the Ca2+-activated Cl− secretion in epithelial tissues has remained a mystery. More recently, TMEM16A has been identified as a new putative Ca2+-activated Cl− channel (CaCC). The primary goal of this article will be to review the characterization of TMEM16A, as it relates to the physical structure of the channel, as well as important residues that confer voltage and Ca2+-sensitivity of the channel. This review will also discuss the role of TMEM16A in epithelial physiology and potential associated-pathophysiology. This will include discussion of developed knockout models that have provided much needed insight on the functional localization of TMEM16A in several epithelial tissues. Finally, this review will examine the implications of the identification of TMEM16A as it pertains to potential novel therapies in several pathologies.
Collapse
Affiliation(s)
- Trey S Rottgen
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| | - Andrew J Nickerson
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| | - Vazhaikkurichi M Rajendran
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| |
Collapse
|
24
|
McCarron A, Donnelley M, Parsons D. Airway disease phenotypes in animal models of cystic fibrosis. Respir Res 2018; 19:54. [PMID: 29609604 PMCID: PMC5879563 DOI: 10.1186/s12931-018-0750-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/13/2018] [Indexed: 12/20/2022] Open
Abstract
In humans, cystic fibrosis (CF) lung disease is characterised by chronic infection, inflammation, airway remodelling, and mucus obstruction. A lack of pulmonary manifestations in CF mouse models has hindered investigations of airway disease pathogenesis, as well as the development and testing of potential therapeutics. However, recently generated CF animal models including rat, ferret and pig models demonstrate a range of well characterised lung disease phenotypes with varying degrees of severity. This review discusses the airway phenotypes of currently available CF animal models and presents potential applications of each model in airway-related CF research.
Collapse
Affiliation(s)
- Alexandra McCarron
- Adelaide Medical School, Discipline of Paediatrics, University of Adelaide, Adelaide, SA Australia
- Department of Respiratory and Sleep Medicine, Women’s and Children’s Hospital, Adelaide, SA Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA Australia
| | - Martin Donnelley
- Adelaide Medical School, Discipline of Paediatrics, University of Adelaide, Adelaide, SA Australia
- Department of Respiratory and Sleep Medicine, Women’s and Children’s Hospital, Adelaide, SA Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA Australia
| | - David Parsons
- Adelaide Medical School, Discipline of Paediatrics, University of Adelaide, Adelaide, SA Australia
- Department of Respiratory and Sleep Medicine, Women’s and Children’s Hospital, Adelaide, SA Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA Australia
| |
Collapse
|
25
|
Rotti PG, Xie W, Poudel A, Yi Y, Sun X, Tyler SR, Uc A, Norris AW, Hara M, Engelhardt JF, Gibson-Corley KN. Pancreatic and Islet Remodeling in Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Knockout Ferrets. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:876-890. [PMID: 29366680 DOI: 10.1016/j.ajpath.2017.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/05/2017] [Accepted: 12/28/2017] [Indexed: 12/25/2022]
Abstract
In cystic fibrosis (CF), there is early destruction of the exocrine pancreas, and this results in a unique form of diabetes that affects approximately half of adult CF individuals. An animal model of cystic fibrosis-related diabetes has been developed in the ferret, which progresses through phases of glycemic abnormalities because of islet remodeling during and after exocrine destruction. Herein, we quantified the pancreatic histopathological changes that occur during these phases. There was an increase in percentage ductal, fat, and islet area in CF ferrets over time compared with age-matched wild-type controls. We also quantified islet size, shape, islet cell composition, cell proliferation (Ki-67), and expression of remodeling markers (matrix metalloprotease-7, desmin, and α-smooth muscle actin). Pancreatic ducts were dilated with scattered proliferating cells and were surrounded by activated stellate cells, indicative of tissue remodeling. The timing of islet and duct proliferation, stellate cell activation, and matrix remodeling coincided with the previously published stages of glycemic crisis and inflammation. This mapping of remodeling events in the CF ferret pancreas provides insights into early changes that control glycemic intolerance and subsequent recovery during the evolution of CF pancreatic disease.
Collapse
Affiliation(s)
- Pavana G Rotti
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa; Department of Biomedical Engineering, The University of Iowa, Iowa City, Iowa
| | - Weiliang Xie
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Ananta Poudel
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Yaling Yi
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Xingshen Sun
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Scott R Tyler
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | - Aliye Uc
- Stead Family Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Andrew W Norris
- Stead Family Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Manami Hara
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, Iowa
| | | |
Collapse
|
26
|
Sandoval-Skeet N, Kaufman JA, Castro MJ, Al-Nakkash L. Genistein diet does not modify crypt morphology in the ob/ob mouse jejunum: a comparison of cryostat and clearing techniques. Diabetes Metab Syndr Obes 2018; 11:863-873. [PMID: 30568474 PMCID: PMC6276911 DOI: 10.2147/dmso.s182501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Diabetes is commonly associated with gastrointestinal dysfunction. We have previously shown that transepithelial short circuit current, Isc (chloride secretion), is significantly reduced in the jejunum from ob/ob mice vs lean controls, and consumption of 600 mg genistein/kg of diet (600 G) for 4 weeks significantly rescues Isc. We aimed to evaluate whether morphological changes in the jejunal crypts contribute to the rescue of Isc. METHODS Male mice (ob/ob and lean controls) were fed either a genistein-free diet or genistein-containing diet (600 G). Comparisons of crypt morphology were made for crypt depth, length, and numbers of proliferative cells. Assessments of crypt measures using DAPI and 5-ethynyl-2'-deoxyuridine (EdU) were performed using traditional cryostat sectioning and an innovative 3D optical clearing method. RESULTS We found that crypt length in the ob/ob genistein-fed group was significantly greater when measured with cleared tissue (85.19±4.73 µm, P<0.05, n=8) compared to lengths measured with cryostat (65.42±3.48 µm, n=8). In addition, proliferative EdU+ counts were approximately fivefold greater with clearing, compared to counts obtained via single plane images from cryostat sections for all groups measured. The average length to EdU+ ratio was unchanged between groups. CONCLUSION Thus, we conclude that genistein diet does not affect overall cellular proliferation or crypt morphology, other than for the modest increased crypt length measured via clearing in the ob/ob genistein group. The increase in crypt length is likely indicative of the greater accuracy of the 3D measures compared to single plane. Genistein diet-induced increases in the intestinal Isc are therefore likely not attributed to changes in intestinal crypt morphology.
Collapse
Affiliation(s)
| | | | | | - Layla Al-Nakkash
- Department of Physiology, Midwestern University, Glendale, AZ 85308, USA,
| |
Collapse
|
27
|
LaRanger R, Peters-Hall JR, Coquelin M, Alabi BR, Chen CT, Wright WE, Shay JW. Reconstituting Mouse Lungs with Conditionally Reprogrammed Human Bronchial Epithelial Cells. Tissue Eng Part A 2017; 24:559-568. [PMID: 28726588 DOI: 10.1089/ten.tea.2017.0114] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We developed methods for conditionally reprogramming (CR) primary human bronchial epithelial cells (HBECs) to extend their functional lifespan and permit their differentiation into both upper and lower airway lung epithelium. We also developed a bioreactor to support vascular perfusion and rhythmic breathing of decellularized mouse lungs reconstituted with CR HBECs isolated from patients with and without cystic fibrosis (CF). While conditionally reprogrammed cells only differentiate into an upper airway epithelium after 35 days at the air-liquid interface, in reconstituted lungs these cells differentiate into upper airway bronchial epithelium and lower airway alveolar structures after 12 days. Rapid scale-up and the ability to obtain clonal derivatives of primary patient-derived HBECs without the need for genetic manipulation may permit rapid reconstitution of the lung epithelium; facilitating the study of lung disease in tissue-engineered models.
Collapse
Affiliation(s)
- Ryan LaRanger
- 1 Department of Cell Biology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Jennifer R Peters-Hall
- 1 Department of Cell Biology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Melissa Coquelin
- 1 Department of Cell Biology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Busola R Alabi
- 1 Department of Cell Biology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Christopher T Chen
- 2 Biomedical Engineering Program, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Woodring E Wright
- 1 Department of Cell Biology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Jerry W Shay
- 1 Department of Cell Biology, University of Texas Southwestern Medical Center , Dallas, Texas
| |
Collapse
|
28
|
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
|
29
|
Hahn A, Faulhaber J, Srisawang L, Stortz A, Salomon JJ, Mall MA, Frings S, Möhrlen F. Cellular distribution and function of ion channels involved in transport processes in rat tracheal epithelium. Physiol Rep 2017; 5:e13290. [PMID: 28642338 PMCID: PMC5492199 DOI: 10.14814/phy2.13290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 01/17/2023] Open
Abstract
Transport of water and electrolytes in airway epithelia involves chloride-selective ion channels, which are controlled either by cytosolic Ca2+ or by cAMP The contributions of the two pathways to chloride transport differ among vertebrate species. Because rats are becoming more important as animal model for cystic fibrosis, we have examined how Ca2+- dependent and cAMP- dependent Cl- secretion is organized in the rat tracheal epithelium. We examined the expression of the Ca2+-gated Cl- channel anoctamin 1 (ANO1), the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel, the epithelial Na+ channel ENaC, and the water channel aquaporin 5 (AQP5) in rat tracheal epithelium. The contribution of ANO1 channels to nucleotide-stimulated Cl- secretion was determined using the channel blocker Ani9 in short-circuit current recordings obtained from primary cultures of rat tracheal epithelial cells in Ussing chambers. We found that ANO1, CFTR and AQP5 proteins were expressed in nonciliated cells of the tracheal epithelium, whereas ENaC was expressed in ciliated cells. Among nonciliated cells, ANO1 occurred together with CFTR and Muc5b and, in addition, in a different cell type without CFTR and Muc5b. Bioelectrical studies with the ANO1-blocker Ani9 indicated that ANO1 mediated the secretory response to the nucleotide uridine-5'-triphosphate. Our data demonstrate that, in rat tracheal epithelium, Cl- secretion and Na+ absorption are routed through different cell types, and that ANO1 channels form the molecular basis of Ca2+-dependent Cl- secretion in this tissue. These characteristic features of Cl--dependent secretion reveal similarities and distinct differences to secretory processes in human airways.
Collapse
Affiliation(s)
- Anne Hahn
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Johannes Faulhaber
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Lalita Srisawang
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Andreas Stortz
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Johanna J Salomon
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC) German Center for Lung Research (DZL) University of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC) German Center for Lung Research (DZL) University of Heidelberg, Heidelberg, Germany
| | - Stephan Frings
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Frank Möhrlen
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
30
|
Romani L, Oikonomou V, Moretti S, Iannitti RG, D'Adamo MC, Villella VR, Pariano M, Sforna L, Borghi M, Bellet MM, Fallarino F, Pallotta MT, Servillo G, Ferrari E, Puccetti P, Kroemer G, Pessia M, Maiuri L, Goldstein AL, Garaci E. Thymosin α1 represents a potential potent single-molecule-based therapy for cystic fibrosis. Nat Med 2017; 23:590-600. [PMID: 28394330 PMCID: PMC5420451 DOI: 10.1038/nm.4305] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 02/17/2017] [Indexed: 12/17/2022]
Abstract
Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) that compromise its chloride-channel activity. The most common mutation, p.Phe508del, results in the production of a misfolded CFTR protein, which has residual channel activity but is prematurely degraded. Because of the inherent complexity of the pathogenetic mechanisms involved in CF —which include impaired chloride permeability and persistent lung inflammation—a multidrug approach is required for efficacious CF therapy. To date, no individual, drug with pleiotropic beneficial effects for CF is available. Here we report on the ability of thymosin alpha 1 (Tα1)—a naturally occurring polypeptide with an excellent safety profile in the clinic when used as an adjuvant or an immunotherapeutic agent—to rectify the multiple tissue defects in CF mice as well as in cells from subjects with the p.Phe508del mutation. Tα1 displayed two combined properties that favorably opposed CF symptomatology; namely, it reduced inflammation and increased CFTR maturation, stability and activity. By virtue of this two-pronged action, Tα1 offers a strong potential to be an efficacious single molecule-based therapeutic agent in CF.
Collapse
Affiliation(s)
- Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Rossana G Iannitti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria Cristina D'Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Valeria R Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | | | - Giuseppe Servillo
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Eleonora Ferrari
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Descartes, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Mauro Pessia
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.,Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| |
Collapse
|
31
|
da Cunha MF, Simonin J, Sassi A, Freund R, Hatton A, Cottart CH, Elganfoud N, Zoubairi R, Dragu C, Jais JP, Hinzpeter A, Edelman A, Sermet-Gaudelus I. Analysis of nasal potential in murine cystic fibrosis models. Int J Biochem Cell Biol 2016; 80:87-97. [PMID: 27717840 DOI: 10.1016/j.biocel.2016.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 12/27/2022]
Abstract
The nasal epithelium of the mouse closely mimics the bioelectrical phenotype of the human airways. Ion transport across the nasal epithelium induces a nasal transepithelial potential difference. Its measurement by a relatively non-invasive method adapted from humans allows in vivo longitudinal measurements of CFTR-dependent ionic transport in the murine nasal mucosa. This test offers a useful tool to assess CFTR function in preclinical studies for novel therapeutics modulating CFTR activity. Here we extensively review work done to assess transepithelial transport in the murine respiratory epithelium in the basal state and after administration of CFTR modulators. Factors of variability and discriminative threshold between the CF and the WT mice for different readouts are discussed.
Collapse
Affiliation(s)
- Mélanie Faria da Cunha
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Juliette Simonin
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Ali Sassi
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Romain Freund
- Unité de Biostatistiques, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Aurélie Hatton
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Charles-Henry Cottart
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Nadia Elganfoud
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Rachid Zoubairi
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Corina Dragu
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Jean Philippe Jais
- Unité de Biostatistiques, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Alexandre Hinzpeter
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | - Aleksander Edelman
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Sorbonne, Paris, France
| | | |
Collapse
|
32
|
Prentice B, Hameed S, Verge CF, Ooi CY, Jaffe A, Widger J. Diagnosing cystic fibrosis-related diabetes: current methods and challenges. Expert Rev Respir Med 2016; 10:799-811. [DOI: 10.1080/17476348.2016.1190646] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Bernadette Prentice
- Department of Respiratory Medicine, Sydney Children’s Hospital, Randwick, Australia
- School of Women’s and Children’s Health, The University of New South Wales, Randwick, Australia
| | - Shihab Hameed
- School of Women’s and Children’s Health, The University of New South Wales, Randwick, Australia
- Department of Endocrinology, Sydney Children’s Hospital, Randwick, Australia
| | - Charles F. Verge
- School of Women’s and Children’s Health, The University of New South Wales, Randwick, Australia
- Department of Endocrinology, Sydney Children’s Hospital, Randwick, Australia
| | - Chee Y. Ooi
- School of Women’s and Children’s Health, The University of New South Wales, Randwick, Australia
- Department of Gastroenterology, Sydney Children’s Hospital, Randwick, Australia
| | - Adam Jaffe
- Department of Respiratory Medicine, Sydney Children’s Hospital, Randwick, Australia
- School of Women’s and Children’s Health, The University of New South Wales, Randwick, Australia
| | - John Widger
- Department of Respiratory Medicine, Sydney Children’s Hospital, Randwick, Australia
- School of Women’s and Children’s Health, The University of New South Wales, Randwick, Australia
| |
Collapse
|
33
|
Lavelle GM, White MM, Browne N, McElvaney NG, Reeves EP. Animal Models of Cystic Fibrosis Pathology: Phenotypic Parallels and Divergences. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5258727. [PMID: 27340661 PMCID: PMC4908263 DOI: 10.1155/2016/5258727] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/08/2016] [Indexed: 12/14/2022]
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. The resultant characteristic ion transport defect results in decreased mucociliary clearance, bacterial colonisation, and chronic neutrophil-dominated inflammation. Much knowledge surrounding the pathophysiology of the disease has been gained through the generation of animal models, despite inherent limitations in each. The failure of certain mouse models to recapitulate the phenotypic manifestations of human disease has initiated the generation of larger animals in which to study CF, including the pig and the ferret. This review will summarise the basic phenotypes of three animal models and describe the contributions of such animal studies to our current understanding of CF.
Collapse
Affiliation(s)
- Gillian M. Lavelle
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Michelle M. White
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Niall Browne
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Noel G. McElvaney
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Emer P. Reeves
- Respiratory Research Division, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
34
|
Shah VS, Meyerholz DK, Tang XX, Reznikov L, Abou Alaiwa M, Ernst SE, Karp PH, Wohlford-Lenane CL, Heilmann KP, Leidinger MR, Allen PD, Zabner J, McCray PB, Ostedgaard LS, Stoltz DA, Randak CO, Welsh MJ. Airway acidification initiates host defense abnormalities in cystic fibrosis mice. Science 2016; 351:503-7. [PMID: 26823428 DOI: 10.1126/science.aad5589] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cystic fibrosis (CF) is caused by mutations in the gene that encodes the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel. In humans and pigs, the loss of CFTR impairs respiratory host defenses, causing airway infection. But CF mice are spared. We found that in all three species, CFTR secreted bicarbonate into airway surface liquid. In humans and pigs lacking CFTR, unchecked H(+) secretion by the nongastric H(+)/K(+) adenosine triphosphatase (ATP12A) acidified airway surface liquid, which impaired airway host defenses. In contrast, mouse airways expressed little ATP12A and secreted minimal H(+); consequently, airway surface liquid in CF and non-CF mice had similar pH. Inhibiting ATP12A reversed host defense abnormalities in human and pig airways. Conversely, expressing ATP12A in CF mouse airways acidified airway surface liquid, impaired defenses, and increased airway bacteria. These findings help explain why CF mice are protected from infection and nominate ATP12A as a potential therapeutic target for CF.
Collapse
Affiliation(s)
- Viral S Shah
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA. Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Xiao Xiao Tang
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA. Howard Hughes Medical Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Leah Reznikov
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | - Sarah E Ernst
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA. Howard Hughes Medical Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Philip H Karp
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA. Howard Hughes Medical Institute, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | - Patrick D Allen
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Joseph Zabner
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Paul B McCray
- Department of Pediatrics University of Iowa, Iowa City, IA 52242, USA. Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | | - David A Stoltz
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA. Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA. Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
| | | | - Michael J Welsh
- Department of Medicine, University of Iowa, Iowa City, IA 52242, USA. Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA. Howard Hughes Medical Institute, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
35
|
Mou H, Brazauskas K, Rajagopal J. Personalized medicine for cystic fibrosis: establishing human model systems. Pediatr Pulmonol 2015; 50 Suppl 40:S14-23. [PMID: 26335952 DOI: 10.1002/ppul.23233] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 05/26/2015] [Indexed: 12/16/2022]
Abstract
With over 1,500 identifiable mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that result in distinct functional and phenotypical abnormalities, it is virtually impossible to perform randomized clinical trials to identify the best therapeutics for all patients. Therefore, a personalized medicine approach is essential. The only way to realistically accomplish this is through the development of improved in vitro human model systems. The lack of a readily available and infinite supply of human CFTR-expressing airway epithelial cells is a key bottleneck. We propose that a concerted two-pronged approach is necessary for patient-specific cystic fibrosis research to continue to prosper and realize its potential: (1) more effective culture and differentiation conditions for growing primary human airway and nasal epithelial cells and (2) the development of collective protocols for efficiently differentiating disease- and patient-specific induced pluripotent stem cells (iPSC) into pure populations of adult epithelial cells. Ultimately, we need a personalized human model system for cystic fibrosis with the capacity for uncomplicated bankability, widespread availability, and universal applicability for patient-specific disease modeling, novel pharmacotherapy investigation and screening, and readily executable genetic modification.
Collapse
Affiliation(s)
- Hongmei Mou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Massachusetts
| | - Karissa Brazauskas
- Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Massachusetts
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Pulmonary Critical Care Unit, Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
36
|
Ghosh A, Boucher RC, Tarran R. Airway hydration and COPD. Cell Mol Life Sci 2015; 72:3637-52. [PMID: 26068443 PMCID: PMC4567929 DOI: 10.1007/s00018-015-1946-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the prevalent causes of worldwide mortality and encompasses two major clinical phenotypes, i.e., chronic bronchitis (CB) and emphysema. The most common cause of COPD is chronic tobacco inhalation. Research focused on the chronic bronchitic phenotype of COPD has identified several pathological processes that drive disease initiation and progression. For example, the lung's mucociliary clearance (MCC) system performs the critical task of clearing inhaled pathogens and toxic materials from the lung. MCC efficiency is dependent on: (1) the ability of apical plasma membrane ion channels such as the cystic fibrosis transmembrane conductance regulator (CFTR) and the epithelial Na(+) channel (ENaC) to maintain airway hydration; (2) ciliary beating; and (3) appropriate rates of mucin secretion. Each of these components is impaired in CB and likely contributes to the mucus stasis/accumulation seen in CB patients. This review highlights the cellular components responsible for maintaining MCC and how this process is disrupted following tobacco exposure and with CB. We shall also discuss existing therapeutic strategies for the treatment of chronic bronchitis and how components of the MCC can be used as biomarkers for the evaluation of tobacco or tobacco-like-product exposure.
Collapse
Affiliation(s)
- Arunava Ghosh
- Cystic Fibrosis Center/Marsico Lung Institute and the Department of Cell Biology and Physiology, The University of North Carolina, 7102 Marsico Hall, Chapel Hill, NC, 27599-7248, USA
| | - R C Boucher
- Cystic Fibrosis Center/Marsico Lung Institute and the Department of Cell Biology and Physiology, The University of North Carolina, 7102 Marsico Hall, Chapel Hill, NC, 27599-7248, USA
| | - Robert Tarran
- Cystic Fibrosis Center/Marsico Lung Institute and the Department of Cell Biology and Physiology, The University of North Carolina, 7102 Marsico Hall, Chapel Hill, NC, 27599-7248, USA.
| |
Collapse
|
37
|
Seys LJM, Verhamme FM, Dupont LL, Desauter E, Duerr J, Seyhan Agircan A, Conickx G, Joos GF, Brusselle GG, Mall MA, Bracke KR. Airway Surface Dehydration Aggravates Cigarette Smoke-Induced Hallmarks of COPD in Mice. PLoS One 2015; 10:e0129897. [PMID: 26066648 PMCID: PMC4466573 DOI: 10.1371/journal.pone.0129897] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 05/14/2015] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Airway surface dehydration, caused by an imbalance between secretion and absorption of ions and fluid across the epithelium and/or increased epithelial mucin secretion, impairs mucociliary clearance. Recent evidence suggests that this mechanism may be implicated in chronic obstructive pulmonary disease (COPD). However, the role of airway surface dehydration in the pathogenesis of cigarette smoke (CS)-induced COPD remains unknown. OBJECTIVE We aimed to investigate in vivo the effect of airway surface dehydration on several CS-induced hallmarks of COPD in mice with airway-specific overexpression of the β-subunit of the epithelial Na⁺ channel (βENaC). METHODS βENaC-Tg mice and wild-type (WT) littermates were exposed to air or CS for 4 or 8 weeks. Pathological hallmarks of COPD, including goblet cell metaplasia, mucin expression, pulmonary inflammation, lymphoid follicles, emphysema and airway wall remodelling were determined and lung function was measured. RESULTS Airway surface dehydration in βENaC-Tg mice aggravated CS-induced airway inflammation, mucin expression and destruction of alveolar walls and accelerated the formation of pulmonary lymphoid follicles. Moreover, lung function measurements demonstrated an increased compliance and total lung capacity and a lower resistance and hysteresis in βENaC-Tg mice, compared to WT mice. CS exposure further altered lung function measurements. CONCLUSIONS We conclude that airway surface dehydration is a risk factor that aggravates CS-induced hallmarks of COPD.
Collapse
Affiliation(s)
- Leen J. M. Seys
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Fien M. Verhamme
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Lisa L. Dupont
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Elke Desauter
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Julia Duerr
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Ayca Seyhan Agircan
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Griet Conickx
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Guy F. Joos
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Guy G. Brusselle
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Marcus A. Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Ken R. Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
38
|
Olivier AK, Gibson-Corley KN, Meyerholz DK. Animal models of gastrointestinal and liver diseases. Animal models of cystic fibrosis: gastrointestinal, pancreatic, and hepatobiliary disease and pathophysiology. Am J Physiol Gastrointest Liver Physiol 2015; 308:G459-71. [PMID: 25591863 PMCID: PMC4360044 DOI: 10.1152/ajpgi.00146.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Multiple organ systems, including the gastrointestinal tract, pancreas, and hepatobiliary systems, are affected by cystic fibrosis (CF). Many of these changes begin early in life and are difficult to study in young CF patients. Recent development of novel CF animal models has expanded opportunities in the field to better understand CF pathogenesis and evaluate traditional and innovative therapeutics. In this review, we discuss manifestations of CF disease in gastrointestinal, pancreatic, and hepatobiliary systems of humans and animal models. We also compare the similarities and limitations of animal models and discuss future directions for modeling CF.
Collapse
Affiliation(s)
- Alicia K. Olivier
- Department of Pathology and Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Katherine N. Gibson-Corley
- Department of Pathology and Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - David K. Meyerholz
- Department of Pathology and Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
39
|
Yan Z, Stewart ZA, Sinn PL, Olsen JC, Hu J, McCray PB, Engelhardt JF. Ferret and pig models of cystic fibrosis: prospects and promise for gene therapy. HUM GENE THER CL DEV 2015; 26:38-49. [PMID: 25675143 PMCID: PMC4367511 DOI: 10.1089/humc.2014.154] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 12/26/2014] [Indexed: 12/13/2022] Open
Abstract
Large animal models of genetic diseases are rapidly becoming integral to biomedical research as technologies to manipulate the mammalian genome improve. The creation of cystic fibrosis (CF) ferrets and pigs is an example of such progress in animal modeling, with the disease phenotypes in the ferret and pig models more reflective of human CF disease than mouse models. The ferret and pig CF models also provide unique opportunities to develop and assess the effectiveness of gene and cell therapies to treat affected organs. In this review, we examine the organ disease phenotypes in these new CF models and the opportunities to test gene therapies at various stages of disease progression in affected organs. We then discuss the progress in developing recombinant replication-defective adenoviral, adeno-associated viral, and lentiviral vectors to target genes to the lung and pancreas in ferrets and pigs, the two most affected organs in CF. Through this review, we hope to convey the potential of these new animal models for developing CF gene and cell therapies.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa School of Medicine, Iowa City, IA 52242
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
| | - Zoe A. Stewart
- Department of Surgery, University of Iowa School of Medicine, Iowa City, IA 52242
| | - Patrick L. Sinn
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Pediatrics, University of Iowa School of Medicine, Iowa City, IA 52242
| | - John C. Olsen
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jim Hu
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Paul B. McCray
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Pediatrics, University of Iowa School of Medicine, Iowa City, IA 52242
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa School of Medicine, Iowa City, IA 52242
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Internal Medicine, University of Iowa School of Medicine, Iowa City, IA 52242
| |
Collapse
|
40
|
Rayyan E, Polito S, Leung L, Bhakta A, Kang J, Willey J, Mansour W, Drumm ML, Al-Nakkash L. Effect of genistein on basal jejunal chloride secretion in R117H CF mice is sex and route specific. Clin Exp Gastroenterol 2015; 8:77-87. [PMID: 25674010 PMCID: PMC4321419 DOI: 10.2147/ceg.s72111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cystic fibrosis (CF) results from the loss or reduction in function of the CFTR (cystic fibrosis transmembrane conductance regulatory protein) chloride channel. The third most common CFTR mutation seen clinically is R117H. Genistein, a naturally occurring phytoestrogen, is known to stimulate CFTR function in vitro. We aimed to determine whether route of administration of genistein could mediate differential effects in R117H male and female CF mice. Mice were fed (4 weeks) or injected subcutaneously (1 week) with the following: genistein 600 mg/kg diet (600Gd); genistein-free diet (0Gd); genistein injection 600 mg/kg body weight (600Gi); dimethyl sulfoxide control (0Gi). In male R117H mice fed 600Gd, basal short circuit current (Isc) was unchanged. In 600Gd-fed female mice, there was a subgroup that demonstrated a significant increase in basal Isc (53.14±7.92 μA/cm(2), n=6, P<0.05) and a subgroup of nonresponders (12.05±6.59 μA/cm(2), n=4), compared to 0Gd controls (29.3±6.5 μA/cm(2), n=7). In R117H mice injected with 600Gi, basal Isc was unchanged in both male and female mice compared to 0Gi controls. Isc was measured in response to the following: the adenylate cyclase activator forskolin (10 μM, bilateral), bumetanide (100 μM, basolateral) to indicate the Cl(-) secretory component, and acetazolamide (100 μM, bilateral) to indicate the HCO3 (-) secretory component; however, there was no effect of genistein (diet or injection) on any of these parameters. Jejunal morphology (ie, villi length, number of goblet cells per villus, crypt depth, and number of goblet cells per crypt) in R117H mice suggested no genistein-mediated difference among the groups. Serum levels of genistein were significantly elevated, compared to respective controls, by either 600Gd (equally elevated in males and females) or 600Gi (elevated more in females versus males). These data suggest a sex-dependent increase in basal Isc of R117H mice and that the increase is also specific for route of administration.
Collapse
Affiliation(s)
- Esa Rayyan
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Sarah Polito
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Lana Leung
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Ashesh Bhakta
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Jonathan Kang
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Justin Willey
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Wasim Mansour
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Mitchell L Drumm
- Pediatric Pulmonology Division, Case Western Reserve University, Cleveland, OH, USA
| | - Layla Al-Nakkash
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| |
Collapse
|
41
|
Yan Z, Stewart ZA, Sinn PL, Olsen JC, Hu J, McCray, Jr. PB, Engelhardt JF. Ferret and Pig Models of Cystic Fibrosis: Prospects and Promise for Gene Therapy. HUM GENE THER CL DEV 2014. [DOI: 10.1089/hum.2014.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
42
|
Abstract
The availability of the human genome sequence and tools for interrogating individual genomes provide an unprecedented opportunity to apply genetics to medicine. Mendelian conditions, which are caused by dysfunction of a single gene, offer powerful examples that illustrate how genetics can provide insights into disease. Cystic fibrosis, one of the more common lethal autosomal recessive Mendelian disorders, is presented here as an example. Recent progress in elucidating disease mechanism and causes of phenotypic variation, as well as in the development of treatments, demonstrates that genetics continues to play an important part in cystic fibrosis research 25 years after the discovery of the disease-causing gene.
Collapse
|
43
|
d'Angelo I, Conte C, La Rotonda MI, Miro A, Quaglia F, Ungaro F. Improving the efficacy of inhaled drugs in cystic fibrosis: challenges and emerging drug delivery strategies. Adv Drug Deliv Rev 2014; 75:92-111. [PMID: 24842473 DOI: 10.1016/j.addr.2014.05.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/23/2014] [Accepted: 05/09/2014] [Indexed: 02/06/2023]
Abstract
Cystic fibrosis (CF) is the most common autosomal recessive disease in Caucasians associated with early death. Although the faulty gene is expressed in epithelia throughout the body, lung disease is still responsible for most of the morbidity and mortality of CF patients. As a local delivery route, pulmonary administration represents an ideal way to treat respiratory infections, excessive inflammation and other manifestations typical of CF lung disease. Nonetheless, important determinants of the clinical outcomes of inhaled drugs are the concentration/permanence at the lungs as well as the ability of the drug to overcome local extracellular and cellular barriers. This review focuses on emerging delivery strategies used for local treatment of CF pulmonary disease. After a brief description of the disease and formulation rules dictated by CF lung barriers, it describes current and future trends in inhaled drugs for CF. The most promising advanced formulations are discussed, highlighting the advantages along with the major challenges for researchers working in this field.
Collapse
Affiliation(s)
- Ivana d'Angelo
- Di.S.T.A.B.i.F., Second University of Napoli, Via Vivaldi 43, 81100 Caserta, Italy
| | - Claudia Conte
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Maria Immacolata La Rotonda
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Agnese Miro
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Fabiana Quaglia
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Francesca Ungaro
- Laboratory of Drug Delivery, Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| |
Collapse
|
44
|
Abstract
Cystic fibrosis (CF) remains the most common fatal hereditary lung disease. The discovery of the cystic fibrosis transmembrane conductance regulator (CFTR) gene 25 years ago set the stage for: 1) unravelling the molecular and cellular basis of CF lung disease; 2) the generation of animal models to study in vivo pathogenesis; and 3) the development of mutation-specific therapies that are now becoming available for a subgroup of patients with CF. This article highlights major advances in our understanding of how CFTR dysfunction causes chronic mucus obstruction, neutrophilic inflammation and bacterial infection in CF airways. Furthermore, we focus on recent breakthroughs and remaining challenges of novel therapies targeting the basic CF defect, and discuss the next steps to be taken to make disease-modifying therapies available to a larger group of patients with CF, including those carrying the most common mutation ΔF508-CFTR. Finally, we will summarise emerging evidence indicating that acquired CFTR dysfunction may be implicated in the pathogenesis of chronic obstructive pulmonary disease, suggesting that lessons learned from CF may be applicable to common airway diseases associated with mucus plugging.
Collapse
Affiliation(s)
- Marcus A Mall
- Dept of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), University of Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany Division of Paediatric Pulmonology and Allergy and Cystic Fibrosis Center, Dept of Paediatrics, University of Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Dominik Hartl
- Paediatric Infectiology and Immunology, Dept of Pediatrics, University of Tübingen, Tübingen, Germany
| |
Collapse
|
45
|
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
|
46
|
Leung L, Kang J, Rayyan E, Bhakta A, Barrett B, Larsen D, Jelinek R, Willey J, Cochran S, Broderick TL, Al-Nakkash L. Decreased basal chloride secretion and altered cystic fibrosis transmembrane conductance regulatory protein, Villin, GLUT5 protein expression in jejunum from leptin-deficient mice. Diabetes Metab Syndr Obes 2014; 7:321-30. [PMID: 25092993 PMCID: PMC4112754 DOI: 10.2147/dmso.s63714] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Patients with diabetes and obesity are at increased risk of developing disturbances in intestinal function. In this study, we characterized jejunal function in the clinically relevant leptin-deficient ob/ob mouse, a model of diabetes and obesity. We measured transepithelial short circuit current (Isc), across freshly isolated segments of jejunum from 12-week-old ob/ob and lean C57BL/6J (female and male) mice. The basal Isc was significantly decreased (~30%) in the ob/ob mice (66.5±5.7 μA/cm(2) [n=20]) (P< 0.05) compared with their lean counterparts (95.1±9.1 μA/cm(2) [n=19]). Inhibition with clotrimazole (100 μM, applied bilaterally) was significantly reduced in the ob/ob mice (-7.92%±3.67% [n=15]) (P<0.05) compared with the lean mice (10.44%±7.92% [n=15]), indicating a decreased contribution of Ca(2+)-activated K(+) (KCa) channels in the ob/ob mice. Inhibition with ouabain (100 μM, applied serosally) was significantly reduced in the ob/ob mice (1.40%±3.61%, n=13) (P< 0.05) versus the lean mice (18.93%±3.76% [n=18]), suggesting a potential defect in the Na(+)/K(+)-adenosine triphosphate (ATP)ase pump with leptin-deficiency. Expression of cystic fibrosis transmembrane conductance regulatory protein (CFTR) (normalized to glyceraldehyde-3-phosphate dehydrogenase [GAPDH]) was significantly decreased ~twofold (P<0.05) in the ob/ob mice compared with the leans, whilst crypt depth was unchanged. Villi length was significantly increased by ~25% (P<0.05) in the ob/ob mice compared with the leans and was associated with an increase in Villin and GLUT5 expression. GLUT2 and SGLT-1 expression were both unchanged. Our data suggests that reduced basal jejunal Isc in ob/ob mice is likely a consequence of reduced CFTR expression and decreased activity of the basolateral KCa channel and Na(+)/K(+)-ATPase. Understanding intestinal dysfunctions in ob/ob jejunum may allow for the development of novel drug targets to treat obesity and diabetes.
Collapse
Affiliation(s)
- Lana Leung
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Jonathan Kang
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Esa Rayyan
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Ashesh Bhakta
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Brennan Barrett
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - David Larsen
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Ryan Jelinek
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Justin Willey
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Scott Cochran
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Tom L Broderick
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Layla Al-Nakkash
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
- Correspondence: Layla Al-Nakkash, Department of Physiology, Midwestern University, 19555 N 59th Avenue, Glendale, AZ, 85308, USA, Tel +1 623 572 3719, Fax +1 623 572 3673, Email
| |
Collapse
|
47
|
Fisher JT, Tyler SR, Zhang Y, Lee BJ, Liu X, Sun X, Sui H, Liang B, Luo M, Xie W, Yi Y, Zhou W, Song Y, Keiser N, Wang K, de Jonge HR, Engelhardt JF. Bioelectric characterization of epithelia from neonatal CFTR knockout ferrets. Am J Respir Cell Mol Biol 2013; 49:837-44. [PMID: 23782101 DOI: 10.1165/rcmb.2012-0433oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cystic fibrosis (CF) is a life-shortening, recessive, multiorgan genetic disorder caused by the loss of CF transmembrane conductance regulator (CFTR) chloride channel function found in many types of epithelia. Animal models that recapitulate the human disease phenotype are critical to understanding pathophysiology in CF and developing therapies. CFTR knockout ferrets manifest many of the phenotypes observed in the human disease, including lung infections, pancreatic disease and diabetes, liver disease, malnutrition, and meconium ileus. In the present study, we have characterized abnormalities in the bioelectric properties of the trachea, stomach, intestine, and gallbladder of newborn CF ferrets. Short-circuit current (ISC) analysis of CF and wild-type (WT) tracheas revealed the following similarities and differences: (1) amiloride-sensitive sodium currents were similar between genotypes; (2) responses to 4,4'-diisothiocyano-2,2'-stilbene disulphonic acid were 3.3-fold greater in CF animals, suggesting elevated baseline chloride transport through non-CFTR channels in a subset of CF animals; and (3) a lack of 3-isobutyl-1-methylxanthine (IBMX)/forskolin-stimulated and N-(2-Naphthalenyl)-((3,5-dibromo-2,4-dihydroxyphenyl)methylene)glycine hydrazide (GlyH-101)-inhibited currents in CF animals due to the lack of CFTR. CFTR mRNA was present throughout all levels of the WT ferret and IBMX/forskolin-inducible ISC was only observed in WT animals. However, despite the lack of CFTR function in the knockout ferret, the luminal pH of the CF ferret gallbladder, stomach, and intestines was not significantly changed relative to WT. The WT stomach and gallbladder exhibited significantly enhanced IBMX/forskolin ISC responses and inhibition by GlyH-101 relative to CF samples. These findings demonstrate that multiple organs affected by disease in the CF ferret have bioelectric abnormalities consistent with the lack of cAMP-mediated chloride transport.
Collapse
|
48
|
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: 31] [Impact Index Per Article: 2.6] [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
|
49
|
Stalvey MS, Clines KL, Havasi V, McKibbin CR, Dunn LK, Chung WJ, Clines GA. Osteoblast CFTR inactivation reduces differentiation and osteoprotegerin expression in a mouse model of cystic fibrosis-related bone disease. PLoS One 2013; 8:e80098. [PMID: 24236172 PMCID: PMC3827431 DOI: 10.1371/journal.pone.0080098] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 09/30/2013] [Indexed: 11/18/2022] Open
Abstract
Low bone mass and increased fracture risk are recognized complications of cystic fibrosis (CF). CF-related bone disease (CFBD) is characterized by uncoupled bone turnover—impaired osteoblastic bone formation and enhanced osteoclastic bone resorption. Intestinal malabsorption, vitamin D deficiency and inflammatory cytokines contribute to CFBD. However, epidemiological investigations and animal models also support a direct causal link between inactivation of skeletal cystic fibrosis transmembrane regulator (CFTR), the gene that when mutated causes CF, and CFBD. The objective of this study was to examine the direct actions of CFTR on bone. Expression analyses revealed that CFTR mRNA and protein were expressed in murine osteoblasts, but not in osteoclasts. Functional studies were then performed to investigate the direct actions of CFTR on osteoblasts using a CFTR knockout (Cftr−/−) mouse model. In the murine calvarial organ culture assay, Cftr−/− calvariae displayed significantly less bone formation and osteoblast numbers than calvariae harvested from wildtype (Cftr+/+) littermates. CFTR inactivation also reduced alkaline phosphatase expression in cultured murine calvarial osteoblasts. Although CFTR was not expressed in murine osteoclasts, significantly more osteoclasts formed in Cftr−/− compared to Cftr+/+ bone marrow cultures. Indirect regulation of osteoclastogenesis by the osteoblast through RANK/RANKL/OPG signaling was next examined. Although no difference in receptor activator of NF-κB ligand (Rankl) mRNA was detected, significantly less osteoprotegerin (Opg) was expressed in Cftr−/− compared to Cftr+/+ osteoblasts. Together, the Rankl:Opg ratio was significantly higher in Cftr−/− murine calvarial osteoblasts contributing to a higher osteoclastogenesis potential. The combined findings of reduced osteoblast differentiation and lower Opg expression suggested a possible defect in canonical Wnt signaling. In fact, Wnt3a and PTH-stimulated canonical Wnt signaling was defective in Cftr−/− murine calvarial osteoblasts. These results support that genetic inactivation of CFTR in osteoblasts contributes to low bone mass and that targeting osteoblasts may represent an effective strategy to treat CFBD.
Collapse
Affiliation(s)
- Michael S. Stalvey
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Katrina L. Clines
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Viktoria Havasi
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Christopher R. McKibbin
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lauren K. Dunn
- Department of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - W. Joon Chung
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Gregory A. Clines
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Veterans Administration Medical Center, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
50
|
Rab A, Rowe SM, Raju SV, Bebok Z, Matalon S, Collawn JF. Cigarette smoke and CFTR: implications in the pathogenesis of COPD. Am J Physiol Lung Cell Mol Physiol 2013; 305:L530-41. [PMID: 23934925 DOI: 10.1152/ajplung.00039.2013] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive respiratory disorder consisting of chronic bronchitis and/or emphysema. COPD patients suffer from chronic infections and display exaggerated inflammatory responses and a progressive decline in respiratory function. The respiratory symptoms of COPD are similar to those seen in cystic fibrosis (CF), although the molecular basis of the two disorders differs. CF is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene encoding a chloride and bicarbonate channel (CFTR), leading to CFTR dysfunction. The majority of COPD cases result from chronic oxidative insults such as cigarette smoke. Interestingly, environmental stresses including cigarette smoke, hypoxia, and chronic inflammation have also been implicated in reduced CFTR function, and this suggests a common mechanism that may contribute to both the CF and COPD. Therefore, improving CFTR function may offer an excellent opportunity for the development of a common treatment for CF and COPD. In this article, we review what is known about the CF respiratory phenotype and discuss how diminished CFTR expression-associated ion transport defects may contribute to some of the pathological changes seen in COPD.
Collapse
Affiliation(s)
- Andras Rab
- Dept. of Cell, Developmental and Integrative Biology, Univ. of Alabama at Birmingham, 1918 Univ. Blvd., MCLM 395, Birmingham, AL 35294.
| | | | | | | | | | | |
Collapse
|