1
|
Hong JS, Tindall JM, Tindall SR, Sorscher EJ. Mutation accumulation in H. sapiens F508del CFTR countermands dN/dS type genomic analysis. PLoS One 2024; 19:e0305832. [PMID: 39024311 PMCID: PMC11257350 DOI: 10.1371/journal.pone.0305832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
Understanding the mechanisms that underlie de novo mutations (DNMs) can be essential for interpreting human evolution, including aspects such as rapidly diverging genes, conservation of non-coding regulatory elements, and somatic DNA adaptation, among others. DNM accumulation in Homo sapiens is often limited to evaluation of human trios or quads across a single generation. Moreover, human SNPs in exons, pseudogenes, or other non-coding elements can be ancient and difficult to date, including polymorphisms attributable to founder effects and identity by descent. In this report, we describe multigenerational evolution of a human coding locus devoid of natural selection, and delineate patterns and principles by which DNMs have accumulated over the past few thousand years. We apply a data set comprising cystic fibrosis transmembrane conductance regulator (CFTR) alleles from 2,393 individuals homozygous for the F508del defect. Additional polymorphism on the F508del background diversified subsequent to a single mutational event during recent human history. Because F508del CFTR is without function, SNPs observed on this haplotype are effectively attributable to factors that govern accumulating de novo mutations. We show profound enhancement of transition, synonymous, and positionally repetitive polymorphisms, indicating appearance of DNMs in a manner evolutionarily designed to protect protein coding DNA against mutational attrition while promoting diversity.
Collapse
Affiliation(s)
- Jeong S. Hong
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Janice M. Tindall
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Samuel R. Tindall
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Eric J. Sorscher
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
2
|
Manzor M, Koutsogiannaki S, DiBlasi M, Schaefers M, Priebe G, Yuki K. Cystic Fibrosis Mice Are Highly Susceptible to Repeated Acute Pseudomonas aeruginosa Pneumonia after Intranasal Inoculation. BIOMED RESEARCH INTERNATIONAL 2024; 2024:4769779. [PMID: 38347907 PMCID: PMC10861279 DOI: 10.1155/2024/4769779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/15/2024]
Abstract
Cystic fibrosis (CF) is a genetic disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) that controls chloride current. A number of different CFTR transgenic mouse lines have been developed and subjected to both acute and chronic infection models. However, prior studies showed no substantial differences in bacterial clearance between CF and non-CF mice after single inoculations. Here, using F508del transgenic CF mice, we examined the role of repeated acute Pseudomonas aeruginosa (PA) infection, with the second inoculation 7 days after the first. We found that CF mice were more susceptible to PA infection than non-CF mice following the second inoculation, with non-CF mice showing better neutrophil recruitment and effector functions. We further investigated the characteristics of lung immune cells using single-cell RNA sequencing, finding that non-CF lung neutrophils had more prominent upregulation of adhesion molecules including intercellular adhesion molecule-1 (ICAM-1) compared to CF lung neutrophils. Although people with CF are often colonized with bacteria and have high numbers of neutrophils in the airways during chronic infection, these data suggest that CF neutrophils have deficient effector functions in the setting of repeated acute infection.
Collapse
Affiliation(s)
- Mariel Manzor
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, USA
| | - Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, USA
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Immunology, Harvard Medical School, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Marco DiBlasi
- Department of Anesthesiology, Critical Care and Pain Medicine, Critical Care Division, Boston Children's Hospital, Boston, USA
| | - Matthew Schaefers
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Critical Care Division, Boston Children's Hospital, Boston, USA
| | - Gregory Priebe
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Critical Care Division, Boston Children's Hospital, Boston, USA
- Department of Pediatrics, Division of Infectious Diseases, Boston Children's Hospital, Boston, USA
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, USA
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Immunology, Harvard Medical School, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| |
Collapse
|
3
|
Salari A, Xiu R, Amiri M, Pallenberg ST, Schreiber R, Dittrich AM, Tümmler B, Kunzelmann K, Seidler U. The Anion Channel TMEM16a/Ano1 Modulates CFTR Activity, but Does Not Function as an Apical Anion Channel in Colonic Epithelium from Cystic Fibrosis Patients and Healthy Individuals. Int J Mol Sci 2023; 24:14214. [PMID: 37762516 PMCID: PMC10531629 DOI: 10.3390/ijms241814214] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Studies in human colonic cell lines and murine intestine suggest the presence of a Ca2+-activated anion channel, presumably TMEM16a. Is there a potential for fluid secretion in patients with severe cystic fibrosis transmembrane conductance regulator (CFTR) mutations by activating this alternative pathway? Two-dimensional nondifferentiated colonoid-myofibroblast cocultures resembling transit amplifying/progenitor (TA/PE) cells, as well as differentiated monolayer (DM) cultures resembling near-surface cells, were established from both healthy controls (HLs) and patients with severe functional defects in the CFTR gene (PwCF). F508del mutant and CFTR knockout (null) mice ileal and colonic mucosa was also studied. HL TA/PE monolayers displayed a robust short-circuit current response (ΔIeq) to UTP (100 µM), forskolin (Fsk, 10 µM) and carbachol (CCH, 100 µM), while ΔIeq was much smaller in differentiated monolayers. The selective TMEM16a inhibitor Ani9 (up to 30 µM) did not alter the response to luminal UTP, significantly decreased Fsk-induced ΔIeq, and significantly increased CCH-induced ΔIeq in HL TA/PE colonoid monolayers. The PwCF TA/PE and the PwCF differentiated monolayers displayed negligible agonist-induced ΔIeq, without a significant effect of Ani9. When TMEM16a was localized in intracellular structures, a staining in the apical membrane was not detected. TMEM16a is highly expressed in human colonoid monolayers resembling transit amplifying cells of the colonic cryptal neck zone, from both HL and PwCF. While it may play a role in modulating agonist-induced CFTR-mediated anion currents, it is not localized in the apical membrane, and it has no function as an apical anion channel in cystic fibrosis (CF) and healthy human colonic epithelium.
Collapse
Affiliation(s)
- Azam Salari
- Department of Gastroenterology, Hannover Medical School, 30625 Hannover, Germany; (A.S.); (R.X.); (M.A.)
| | - Renjie Xiu
- Department of Gastroenterology, Hannover Medical School, 30625 Hannover, Germany; (A.S.); (R.X.); (M.A.)
| | - Mahdi Amiri
- Department of Gastroenterology, Hannover Medical School, 30625 Hannover, Germany; (A.S.); (R.X.); (M.A.)
| | - Sophia Theres Pallenberg
- Department of Pediatric Pneumonology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany (A.-M.D.)
| | - Rainer Schreiber
- Institute of Physiology, University of Regensburg, 93040 Regensburg, Germany; (R.S.); (K.K.)
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumonology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany (A.-M.D.)
| | - Burkhard Tümmler
- Department of Pediatric Pneumonology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany (A.-M.D.)
| | - Karl Kunzelmann
- Institute of Physiology, University of Regensburg, 93040 Regensburg, Germany; (R.S.); (K.K.)
| | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, 30625 Hannover, Germany; (A.S.); (R.X.); (M.A.)
| |
Collapse
|
4
|
Kelly J, Al-Rammahi M, Daly K, Flanagan PK, Urs A, Cohen MC, di Stefano G, Bijvelds MJC, Sheppard DN, de Jonge HR, Seidler UE, Shirazi-Beechey SP. Alterations of mucosa-attached microbiome and epithelial cell numbers in the cystic fibrosis small intestine with implications for intestinal disease. Sci Rep 2022; 12:6593. [PMID: 35449374 PMCID: PMC9023491 DOI: 10.1038/s41598-022-10328-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Defective CFTR leads to accumulation of dehydrated viscous mucus within the small intestine, luminal acidification and altered intestinal motility, resulting in blockage. These changes promote gut microbial dysbiosis, adversely influencing the normal proliferation and differentiation of intestinal epithelial cells. Using Illumina 16S rRNA gene sequencing and immunohistochemistry, we assessed changes in mucosa-attached microbiome and epithelial cell profile in the small intestine of CF mice and a CF patient compared to wild-type mice and non-CF humans. We found increased abundance of pro-inflammatory Escherichia and depletion of beneficial secondary bile-acid producing bacteria in the ileal mucosa-attached microbiome of CFTR-null mice. The ileal mucosa in a CF patient was dominated by a non-aeruginosa Pseudomonas species and lacked numerous beneficial anti-inflammatory and short-chain fatty acid-producing bacteria. In the ileum of both CF mice and a CF patient, the number of absorptive enterocytes, Paneth and glucagon-like peptide 1 and 2 secreting L-type enteroendocrine cells were decreased, whereas stem and goblet cell numbers were increased. These changes in mucosa-attached microbiome and epithelial cell profile suggest that microbiota-host interactions may contribute to intestinal CF disease development with implications for therapy.
Collapse
Affiliation(s)
- Jennifer Kelly
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Miran Al-Rammahi
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.,Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Al-Qadisiyah, Al Diwaniyah, 58002, Iraq
| | - Kristian Daly
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Paul K Flanagan
- Arrowe Park University Teaching Hospital NHS Trust, Wirral, CH49 5PE, UK.,Gastrointestinal and Liver Services, Aintree University Hospital, Lower Lane, Liverpool, Merseyside, L9 7AL, UK
| | - Arun Urs
- Sheffield Children's Hospital NHS Trust, Western Bank, Sheffield, S10 2TH, UK
| | - Marta C Cohen
- Histopathology Department, Sheffield Children's Hospital NHS Trust, Western Bank, Sheffield, S10 2TH, UK
| | - Gabriella di Stefano
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625, Hannover, Germany
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Ursula E Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625, Hannover, Germany
| | - Soraya P Shirazi-Beechey
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| |
Collapse
|
5
|
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
|
6
|
Brinkert K, Hedtfeld S, Burhop A, Gastmeier R, Gad P, Wedekind D, Kloth C, Rothschuh J, Lachmann N, Hetzel M, Jirmo AC, Lopez-Rodriguez E, Brandenberger C, Hansen G, Schambach A, Ackermann M, Tümmler B, Munder A. Rescue from Pseudomonas aeruginosa Airway Infection via Stem Cell Transplantation. Mol Ther 2020; 29:1324-1334. [PMID: 33279724 DOI: 10.1016/j.ymthe.2020.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/21/2020] [Accepted: 11/29/2020] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which lead to impaired ion transport in epithelial cells. Although lung failure due to chronic infection is the major comorbidity in individuals with cystic fibrosis, the role of CFTR in non-epithelial cells has not been definitively resolved. Given the important role of host defense cells, we evaluated the Cftr deficiency in pulmonary immune cells by hematopoietic stem cell transplantation in cystic fibrosis mice. We transplanted healthy bone marrow stem cells and could reveal a stable chimerism of wild-type cells in peripheral blood. The outcome of stem cell transplantation and the impact of healthy immune cells were evaluated in acute Pseudomonas aeruginosa airway infection. In this study, mice transplanted with wild-type cells displayed better survival, lower lung bacterial numbers, and a milder disease course. This improved physiology of infected mice correlated with successful intrapulmonary engraftment of graft-derived alveolar macrophages, as seen by immunofluorescence microscopy and flow cytometry of graft-specific leucocyte surface marker CD45 and macrophage marker CD68. Given the beneficial effect of hematopoietic stem cell transplantation and stable engraftment of monocyte-derived CD68-positive macrophages, we conclude that replacement of mutant Cftr macrophages attenuates airway infection in cystic fibrosis mice.
Collapse
Affiliation(s)
- Kerstin Brinkert
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Silke Hedtfeld
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Annina Burhop
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Rena Gastmeier
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Pauline Gad
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Dirk Wedekind
- Institute of Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Christina Kloth
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Justin Rothschuh
- Institute of Pharmacology, Hannover Medical School, 30625 Hannover, Germany
| | - Nico Lachmann
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational and Regenerative Medicine, 30625 Hannover, Germany
| | - Miriam Hetzel
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational and Regenerative Medicine, 30625 Hannover, Germany
| | - Adan Chari Jirmo
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Elena Lopez-Rodriguez
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany; Institute of Functional Anatomy, Charité Universitätsmedizin Berlin, 10115 Berlin, Germany
| | - Christina Brandenberger
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany; Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Gesine Hansen
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational and Regenerative Medicine, 30625 Hannover, Germany; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mania Ackermann
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Research Center for Translational and Regenerative Medicine, 30625 Hannover, Germany
| | - Burkhard Tümmler
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Antje Munder
- Clinic for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany.
| |
Collapse
|
7
|
Clunes LA, McMillan-Castanares N, Mehta N, Mesadieu A, Rodriguez J, Maj M, Clunes MT. Epithelial vectorial ion transport in cystic fibrosis: Dysfunction, measurement, and pharmacotherapy to target the primary deficit. SAGE Open Med 2020; 8:2050312120933807. [PMID: 32637102 PMCID: PMC7323271 DOI: 10.1177/2050312120933807] [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: 02/11/2019] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
Cystic fibrosis patients display multi-organ system dysfunction (e.g. pancreas, gastrointestinal tract, and lung) with pathogenesis linked to a failure of Cl- secretion from the epithelial surfaces of these organs. If unmanaged, organ dysfunction starts early and patients experience chronic respiratory infection with reduced lung function and a failure to thrive due to gastrointestinal malabsorption. Early mortality is typically caused by respiratory failure. In the past 40 years of newborn screening and improved disease management have driven the median survival up from the mid-teens to 43-53, with most of that improvement coming from earlier and more aggressive management of the symptoms. In the last decade, promising pharmacotherapies have been developed for the correction of the underlying epithelial dysfunction, namely, Cl- secretion. A new generation of systemic drugs target the mutated Cl- channels in cystic fibrosis patients and allow trafficking of the immature mutated protein to the cell membrane (correctors), restore function to the channel once in situ (potentiators), or increase protein levels in the cells (amplifiers). Restoration of channel function prior to symptom development has the potential to significantly change the trajectory of disease progression and their evidence suggests that a modest restoration of Cl- secretion may delay disease progression by decades. In this article, we review epithelial vectorial ion and fluid transport, its quantification and measurement as a marker for cystic fibrosis ion transport dysfunction, and highlight some of the recent therapies targeted at the dysfunctional ion transport of cystic fibrosis.
Collapse
Affiliation(s)
- Lucy A Clunes
- Department of Pharmacology, St. George's University, Grenada, West Indies
| | | | - Neil Mehta
- Medical Student Research Institute, St. George's University, Grenada, West Indies
| | - Afia Mesadieu
- Medical Student Research Institute, St. George's University, Grenada, West Indies
| | - Jorge Rodriguez
- Medical Student Research Institute, St. George's University, Grenada, West Indies
| | - Mary Maj
- Department of Biochemistry, St. George's University, Grenada, West Indies
| | - Mark T Clunes
- Department of Physiology, Neuroscience and Behavioral Sciences, St. George's University, Grenada, West Indies
| |
Collapse
|
8
|
Engevik AC, Coutts AW, Kaji I, Rodriguez P, Ongaratto F, Saqui-Salces M, Medida RL, Meyer AR, Kolobova E, Engevik MA, Williams JA, Shub MD, Carlson DF, Melkamu T, Goldenring JR. Editing Myosin VB Gene to Create Porcine Model of Microvillus Inclusion Disease, With Microvillus-Lined Inclusions and Alterations in Sodium Transporters. Gastroenterology 2020; 158:2236-2249.e9. [PMID: 32112796 PMCID: PMC7282982 DOI: 10.1053/j.gastro.2020.02.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS Microvillus inclusion disease (MVID) is caused by inactivating mutations in the myosin VB gene (MYO5B). MVID is a complex disorder characterized by chronic, watery, life-threatening diarrhea that usually begins in the first hours to days of life. We developed a large animal model of MVID to better understand its pathophysiology. METHODS Pigs were cloned by transfer of chromatin from swine primary fetal fibroblasts, which were edited with TALENs and single-strand oligonucleotide to introduce a P663-L663 substitution in the endogenous swine MYO5B (corresponding to the P660L mutation in human MYO5B, associated with MVID) to fertilized oocytes. We analyzed duodenal tissues from patients with MVID (with the MYO5B P660L mutation) and without (controls), and from pigs using immunohistochemistry. Enteroids were generated from pigs with MYO5B(P663L) and without the substitution (control pigs). RESULTS Duodenal tissues from patients with MVID lacked MYO5B at the base of the apical membrane of intestinal cells; instead MYO5B was intracellular. Intestinal tissues and derived enteroids from MYO5B(P663L) piglets had reduced apical levels and diffuse subapical levels of sodium hydrogen exchanger 3 and SGLT1, which regulate transport of sodium, glucose, and water, compared with tissues from control piglets. However, intestinal tissues and derived enteroids from MYO5B(P663L) piglets maintained CFTR on apical membranes, like tissues from control pigs. Liver tissues from MYO5B(P663L) piglets had alterations in bile salt export pump, a transporter that facilitates bile flow, which is normally expressed in the bile canaliculi in the liver. CONCLUSIONS We developed a large animal model of MVID that has many features of the human disease. Studies of this model could provide information about the functions of MYO5B and MVID pathogenesis, and might lead to new treatments.
Collapse
Affiliation(s)
- Amy C Engevik
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee.
| | | | - Izumi Kaji
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | - Milena Saqui-Salces
- Department of Animal Science, University of Minnesota, Saint Paul, Minnesota
| | - Ramya Lekha Medida
- Department of Animal Science, University of Minnesota, Saint Paul, Minnesota
| | - Anne R Meyer
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elena Kolobova
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Melinda A Engevik
- Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Janice A Williams
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mitchell D Shub
- Phoenix Children's Hospital and University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | | | | | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; The Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
9
|
Towards next generation therapies for cystic fibrosis: Folding, function and pharmacology of CFTR. J Cyst Fibros 2020; 19 Suppl 1:S25-S32. [PMID: 31902693 PMCID: PMC7052731 DOI: 10.1016/j.jcf.2019.12.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022]
Abstract
The treatment of cystic fibrosis (CF) has been transformed by orally-bioavailable small molecule modulators of the cystic fibrosis transmembrane conductance regulator (CFTR), which restore function to CF mutants. However, CFTR modulators are not available to all people with CF and better modulators are required to prevent disease progression. Here, we review selectively recent advances in CFTR folding, function and pharmacology. We highlight ensemble and single-molecule studies of CFTR folding, which provide new insight into CFTR assembly, its perturbation by CF mutations and rescue by CFTR modulators. We discuss species-dependent differences in the action of the F508del-CFTR mutation on CFTR expression, stability and function, which might influence pharmacological studies of CFTR modulators in CF animal models. Finally, we illuminate the identification of combinations of two CFTR potentiators (termed co-potentiators), which restore therapeutically-relevant levels of CFTR activity to rare CF mutations. Thus, mechanistic studies of CFTR folding, function and pharmacology inform the development of highly effective CFTR modulators.
Collapse
|
10
|
Beumer W, Swildens J, Leal T, Noel S, Anthonijsz H, van der Horst G, Kuiperij-Boersma H, Potman M, van Putten C, Biasutto P, Platenburg G, de Jonge H, Henig N, Ritsema T. Evaluation of eluforsen, a novel RNA oligonucleotide for restoration of CFTR function in in vitro and murine models of p.Phe508del cystic fibrosis. PLoS One 2019; 14:e0219182. [PMID: 31251792 PMCID: PMC6599119 DOI: 10.1371/journal.pone.0219182] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/18/2019] [Indexed: 01/14/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the gene encoding the epithelial chloride channel CF transmembrane conductance regulator (CFTR) protein. The most common mutation is a deletion of three nucleotides leading to the loss of phenylalanine at position 508 (p.Phe508del) in the protein. This study evaluates eluforsen, a novel, single-stranded, 33-nucleotide antisense oligonucleotide designed to restore CFTR function, in in vitro and in vivo models of p.Phe508del CF. The aims of the study were to demonstrate cellular uptake of eluforsen, and its efficacy in functional restoration of p.Phe508del-CFTR both in vitro and in vivo. In vitro, the effect of eluforsen was investigated in human CF pancreatic adenocarcinoma cells and human bronchial epithelial cells. Two mouse models were used to evaluate eluforsen in vivo. In vitro, eluforsen improved chloride efflux in CF pancreatic adenocarcinoma cell cultures and increased short-circuit current in primary human bronchial epithelial cells, both indicating restoration of CFTR function. In vivo, eluforsen was taken up by airway epithelium following oro-tracheal administration in mice, resulting in systemic exposure of eluforsen. In female F508del-CFTR mice, eluforsen significantly increased CFTR-mediated saliva secretion (used as a measure of CFTR function, equivalent to the sweat test in humans). Similarly, intranasal administration of eluforsen significantly improved nasal potential difference (NPD), and therefore CFTR conductance, in two CF mouse models. These findings indicate that eluforsen improved CFTR function in cell and animal models of p.Phe508del-CFTR-mediated CF and supported further development of eluforsen in human clinical trials, where eluforsen has also been shown to improve CFTR activity as measured by NPD.
Collapse
Affiliation(s)
- Wouter Beumer
- ProQR Therapeutics, Leiden, The Netherlands
- * E-mail:
| | | | - Teresinha Leal
- Université Catholique de Louvain, Louvain Centre for Toxicology and Applied Pharmacology, Brussels, Belgium
| | - Sabrina Noel
- Université Catholique de Louvain, Louvain Centre for Toxicology and Applied Pharmacology, Brussels, Belgium
| | | | | | | | | | | | | | | | - Hugo de Jonge
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
11
|
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
|
12
|
Edlund A, Barghouth M, Huhn M, Abels M, Esguerra J, Mollet I, Svedin E, Wendt A, Renstrom E, Zhang E, Wierup N, Scholte BJ, Flodström-Tullberg M, Eliasson L. Defective exocytosis and processing of insulin in a cystic fibrosis mouse model. J Endocrinol 2019; 241:JOE-18-0570.R1. [PMID: 30721137 DOI: 10.1530/joe-18-0570] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/05/2019] [Indexed: 01/21/2023]
Abstract
Cystic fibrosis-related diabetes (CFRD) is a common complication for patients with cystic fibrosis (CF), a disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). The cause of CFRD is unclear, but a commonly observed reduction in first-phase insulin secretion suggests defects at the beta cell level. Here we aimed to examine beta- and alpha-cell function in the Cftrtm1EUR/F508del mouse model (C57BL/6J), which carries the most common human mutation in CFTR, the F508del mutation. CFTR expression, beta cell mass, insulin granule distribution, hormone secretion and single cell capacitance changes were evaluated using islets (or beta cells) from F508del mice and age-matched wild-type mice aged 7-10 weeks. Granular pH was measured with DND-189 fluorescence. Serum glucose, insulin and glucagon levels were measured in vivo, and glucose tolerance was assessed using IPGTT. We show increased secretion of proinsulin and concomitant reduced secretion of C-peptide in islets from F508del mice compared to WT mice. Exocytosis and number of docked granules was reduced. We confirmed reduced granular pH by CFTR stimulation. We detected decreased pancreatic beta cell area, but unchanged beta cell number. Moreover, the F508del mutation caused failure to suppress glucagon secretion leading to hyperglucagonemia. In conclusion, F508del mice have beta cell defects resulting in 1) reduced number of docked insulin granules and reduced exocytosis, and 2) potential defective proinsulin cleavage and secretion of immature insulin. These observations provide insight into the functional role of CFTR in pancreatic islets and contribute to increased understanding of the pathogenesis of CFRD.
Collapse
Affiliation(s)
- Anna Edlund
- A Edlund, Clinical sciences in Malmo, Lund University, Malmo, 21428, Sweden
| | - Mohammad Barghouth
- M Barghouth, Dept Clinical Sciences in Malmö, Lunds Universitet, Malmö, Sweden
| | - Michael Huhn
- M Huhn, of medicine Huddinge, Karolinska institute, Center for infectious medicine, Stockholm, Sweden
| | - Mia Abels
- M Abels, Department of clinical sciencies in Malmo, Lunds Universitet Institutionen for kliniska vetenskaper i Malmo, Malmo, Sweden
| | - Jonathan Esguerra
- J Esguerra, Clinical Sciences - Malmö, Lund University, Malmö, 21428, Sweden
| | - Ines Mollet
- I Mollet, CEDOC - Chronic Diseases Research Center, NOVA Medical School - Faculdade de Ciências Médicas, Lisboa, 1150-082, Portugal
| | - Emma Svedin
- E Svedin, Department of Medicine Huddinge, Karolinska Institutet Department of Medicine Huddinge, Stockholm, Sweden
| | - Anna Wendt
- A Wendt, Dept Clinical Sciences in Malmö, Lunds Universitet, Malmö, Sweden
| | - Erik Renstrom
- E Renstrom, Clinical Sciences Malmo, Lund University, Malmo, SE-20502, Sweden
| | - Enming Zhang
- E Zhang, Department of Clinical Science, Lund Uinversity, Malmö, 20502, Sweden
| | - Nils Wierup
- N Wierup, Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, 20502, Sweden
| | - Bob J Scholte
- B Scholte, Department of Cellbiology, Pediatric Pulmonology, Erasmus MC, Rotterdam, Netherlands
| | - Malin Flodström-Tullberg
- M Flodström-Tullberg, Dept of Medicine Huddinge, Karolinska institute, Center for Infectious Medicine, Stockholm, Sweden
| | - Lena Eliasson
- L Eliasson, Dept Clinical Sciences in Malmö, Lunds Universitet, Malmö, 214 28, Sweden
| |
Collapse
|
13
|
FKBP8 Enhances Protein Stability of the CLC-1 Chloride Channel at the Plasma Membrane. Int J Mol Sci 2018; 19:ijms19123783. [PMID: 30487393 PMCID: PMC6320802 DOI: 10.3390/ijms19123783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/18/2018] [Accepted: 11/26/2018] [Indexed: 01/23/2023] Open
Abstract
Mutations in the skeletal muscle-specific CLC-1 chloride channel are associated with the human hereditary disease myotonia congenita. The molecular pathophysiology underlying some of the disease-causing mutations can be ascribed to defective human CLC-1 protein biosynthesis. CLC-1 protein folding is assisted by several molecular chaperones and co-chaperones, including FK506-binding protein 8 (FKBP8). FKBP8 is generally considered an endoplasmic reticulum- and mitochondrion-resident membrane protein, but is not thought to contribute to protein quality control at the cell surface. Herein, we aim to test the hypothesis that FKBP8 may regulate CLC-1 protein at the plasma membrane. Surface biotinylation and subcellular fractionation analyses reveal that a portion of FKBP8 is present at the plasma membrane, and that co-expression with CLC-1 enhances surface localization of FKBP8. Immunoblotting analyses of plasma membrane proteins purified from skeletal muscle further confirm surface localization of FKBP8. Importantly, FKBP8 promotes CLC-1 protein stability at the plasma membrane. Together, our data underscore the importance of FKBP8 in the peripheral quality control of CLC-1 channel.
Collapse
|
14
|
Veit G, Xu H, Dreano E, Avramescu RG, Bagdany M, Beitel LK, Roldan A, Hancock MA, Lay C, Li W, Morin K, Gao S, Mak PA, Ainscow E, Orth AP, McNamara P, Edelman A, Frenkiel S, Matouk E, Sermet-Gaudelus I, Barnes WG, Lukacs GL. Structure-guided combination therapy to potently improve the function of mutant CFTRs. Nat Med 2018; 24:1732-1742. [PMID: 30297908 PMCID: PMC6301090 DOI: 10.1038/s41591-018-0200-x] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 08/08/2018] [Indexed: 12/17/2022]
Abstract
Available corrector drugs are unable to effectively rescue the folding defects of CFTR-ΔF508 (or CFTR-F508del), the most common disease-causing mutation of the cystic fibrosis transmembrane conductance regulator, a plasma membrane (PM) anion channel, and thus to substantially ameliorate clinical phenotypes of cystic fibrosis (CF). To overcome the corrector efficacy ceiling, here we show that compounds targeting distinct structural defects of CFTR can synergistically rescue mutant expression and function at the PM. High-throughput cell-based screens and mechanistic analysis identified three small-molecule series that target defects at nucleotide-binding domain (NBD1), NBD2 and their membrane-spanning domain (MSD) interfaces. Although individually these compounds marginally improve ΔF508-CFTR folding efficiency, function and stability, their combinations lead to ~50-100% of wild-type-level correction in immortalized and primary human airway epithelia and in mouse nasal epithelia. Likewise, corrector combinations were effective against rare missense mutations in various CFTR domains, probably acting via structural allostery, suggesting a mechanistic framework for their broad application.
Collapse
Affiliation(s)
- Guido Veit
- Department of Physiology, McGill University, Montréal, Quebec, Canada.
| | - Haijin Xu
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Elise Dreano
- Institut Necker-Enfants Malades (INEM)-INSERM U1151, Paris, France
| | - Radu G Avramescu
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Miklos Bagdany
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Lenore K Beitel
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Ariel Roldan
- Department of Physiology, McGill University, Montréal, Quebec, Canada
| | - Mark A Hancock
- SPR-MS Facility, McGill University, Montréal, Quebec, Canada
| | - Cecilia Lay
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Wei Li
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Katelin Morin
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Sandra Gao
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Puiying A Mak
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Edward Ainscow
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Anthony P Orth
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Peter McNamara
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | | | - Saul Frenkiel
- Department of Otolaryngology - Head and Neck Surgery, McGill University, Montréal, Quebec, Canada
| | - Elias Matouk
- Adult Cystic Fibrosis Clinic, Montreal Chest Institute, McGill University, Montréal, Quebec, Canada
| | | | - William G Barnes
- Genomic Institute of the Novartis Research Foundation, San Diego, CA, USA
| | - Gergely L Lukacs
- Department of Physiology, McGill University, Montréal, Quebec, Canada. .,Department of Biochemistry, McGill University, Montréal, Quebec, Canada. .,Groupe de Recherche Axé sur la Structure des Protéines (GRASP), McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
15
|
Ahmadi S, Xia S, Wu YS, Di Paola M, Kissoon R, Luk C, Lin F, Du K, Rommens J, Bear CE. SLC6A14, an amino acid transporter, modifies the primary CF defect in fluid secretion. eLife 2018; 7:37963. [PMID: 30004386 PMCID: PMC6054531 DOI: 10.7554/elife.37963] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/12/2018] [Indexed: 01/29/2023] Open
Abstract
The severity of intestinal disease associated with Cystic Fibrosis (CF) is variable in the patient population and this variability is partially conferred by the influence of modifier genes. Genome-wide association studies have identified SLC6A14, an electrogenic amino acid transporter, as a genetic modifier of CF-associated meconium ileus. The purpose of the current work was to determine the biological role of Slc6a14, by disrupting its expression in CF mice bearing the major mutation, F508del. We found that disruption of Slc6a14 worsened the intestinal fluid secretion defect, characteristic of these mice. In vitro studies of mouse intestinal organoids revealed that exacerbation of the primary defect was associated with reduced arginine uptake across the apical membrane, with aberrant nitric oxide and cyclic GMP-mediated regulation of the major CF-causing mutant protein. Together, these studies highlight the role of this apical transporter in modifying cellular nitric oxide levels, residual function of the major CF mutant and potentially, its promise as a therapeutic target.
Collapse
Affiliation(s)
- Saumel Ahmadi
- Department of Physiology, University of Toronto, Toronto, Canada.,Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Sunny Xia
- Department of Physiology, University of Toronto, Toronto, Canada.,Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Yu-Sheng Wu
- Department of Physiology, University of Toronto, Toronto, Canada.,Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Michelle Di Paola
- Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Randolph Kissoon
- Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Catherine Luk
- Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Fan Lin
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Kai Du
- Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Johanna Rommens
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Programme in Genetics and Genome Biology, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Christine E Bear
- Department of Physiology, University of Toronto, Toronto, Canada.,Programme in Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
16
|
Tazi MF, Dakhlallah DA, Caution K, Gerber MM, Chang SW, Khalil H, Kopp BT, Ahmed AE, Krause K, Davis I, Marsh C, Lovett-Racke AE, Schlesinger LS, Cormet-Boyaka E, Amer AO. Elevated Mirc1/Mir17-92 cluster expression negatively regulates autophagy and CFTR (cystic fibrosis transmembrane conductance regulator) function in CF macrophages. Autophagy 2017; 12:2026-2037. [PMID: 27541364 DOI: 10.1080/15548627.2016.1217370] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cystic fibrosis (CF) is a fatal, genetic disorder that critically affects the lungs and is directly caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in defective CFTR function. Macroautophagy/autophagy is a highly regulated biological process that provides energy during periods of stress and starvation. Autophagy clears pathogens and dysfunctional protein aggregates within macrophages. However, this process is impaired in CF patients and CF mice, as their macrophages exhibit limited autophagy activity. The study of microRNAs (Mirs), and other noncoding RNAs, continues to offer new therapeutic targets. The objective of this study was to elucidate the role of Mirs in dysregulated autophagy-related genes in CF macrophages, and then target them to restore this host-defense function and improve CFTR channel function. We identified the Mirc1/Mir17-92 cluster as a potential negative regulator of autophagy as CF macrophages exhibit decreased autophagy protein expression and increased cluster expression when compared to wild-type (WT) counterparts. The absence or reduced expression of the cluster increases autophagy protein expression, suggesting the canonical inverse relationship between Mirc1/Mir17-92 and autophagy gene expression. An in silico study for targets of Mirs that comprise the cluster suggested that the majority of the Mirs target autophagy mRNAs. Those targets were validated by luciferase assays. Notably, the ability of macrophages expressing mutant F508del CFTR to transport halide through their membranes is compromised and can be restored by downregulation of these inherently elevated Mirs, via restoration of autophagy. In vivo, downregulation of Mir17 and Mir20a partially restored autophagy expression and hence improved the clearance of Burkholderia cenocepacia. Thus, these data advance our understanding of mechanisms underlying the pathobiology of CF and provide a new therapeutic platform for restoring CFTR function and autophagy in patients with CF.
Collapse
Affiliation(s)
- Mia F Tazi
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Duaa A Dakhlallah
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Kyle Caution
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Madelyn M Gerber
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Sheng-Wei Chang
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA.,c Department of Veterinary Biosciences , The Ohio State University , Columbus , OH , USA
| | - Hany Khalil
- d Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute , University of Sadat City , Egypt
| | | | - Amr E Ahmed
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Kathrin Krause
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Ian Davis
- c Department of Veterinary Biosciences , The Ohio State University , Columbus , OH , USA
| | - Clay Marsh
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Amy E Lovett-Racke
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA
| | - Larry S Schlesinger
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| | - Estelle Cormet-Boyaka
- b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA.,c Department of Veterinary Biosciences , The Ohio State University , Columbus , OH , USA
| | - Amal O Amer
- a Department of Microbial Infection and Immunity, Center for Microbial Interface Biology , The Ohio State University , Columbus , OH , USA.,b Davis Heart and Lung Research Institute , The Ohio State University , Columbus , OH , USA
| |
Collapse
|
17
|
Rapp C, Bai X, Reithmeier RAF. Molecular analysis of human solute carrier SLC26 anion transporter disease-causing mutations using 3-dimensional homology modeling. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2420-2434. [PMID: 28941661 DOI: 10.1016/j.bbamem.2017.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 06/08/2017] [Accepted: 09/14/2017] [Indexed: 12/22/2022]
Abstract
The availability of the first crystal structure of a bacterial member (SLC26Dg) of the solute carrier SLC26 family of anion transporters has allowed us to create 3-dimensional models of all 10 human members (SLC26A1-A11, A10 being a pseudogene) of these membrane proteins using the Phyre2 bioinformatic tool. The homology modeling predicted that the SLC26 human proteins, like the SLC26Dg template, all consist of 14 transmembrane segments (TM) arranged in a 7+7 inverted topology with the amino-termini of two half-helices (TM3 and 10) facing each other in the centre of the protein to create the anion-binding site, linked to a C-terminal cytosolic sulfate transporter anti-sigma factor antagonist (STAS) domain. A plethora of human diseases are associated with mutations in the genes encoding human SLC26 transporters, including chondrodysplasias with varying severity in SLC26A2 (~50 mutations, 27 point mutations), congenital chloride-losing diarrhea in SLC26A3 (~70 mutations, 31 point mutations) and Pendred Syndrome or deafness autosomal recessive type 4 in SLC26A4 (~500 mutations, 203 point mutations). We have localized all of these point mutations in the 3-dimensional structures of the respective SLC26A2, A3 and A4 proteins and systematically analyzed their effect on protein structure. While most disease-causing mutations may cause folding defects resulting in impaired trafficking of these membrane glycoproteins from the endoplasmic reticulum to the cell surface - as demonstrated in a number of functional expression studies - the modeling also revealed that a number of pathogenic mutations are localized to the anion-binding site, which may directly affect transport function.
Collapse
Affiliation(s)
- Chloe Rapp
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Xiaoyun Bai
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | |
Collapse
|
18
|
Defective CFTR leads to aberrant β-catenin activation and kidney fibrosis. Sci Rep 2017; 7:5233. [PMID: 28701694 PMCID: PMC5507915 DOI: 10.1038/s41598-017-05435-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/30/2017] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR), known as a cAMP-activated Cl− channel, is widely expressed at the apical membrane of epithelial cells in a wide variety of tissues. Of note, despite the abundant expression of CFTR in mammalian kidney, the role of CFTR in kidney disease development is unclear. Here, we report that CFTR expression is downregulated in the UUO (unilateral ureteral obstruction)-induced kidney fibrosis mouse model and human fibrotic kidneys. Dysfunction or downregulation of CFTR in renal epithelial cells leads to alteration of genes involved in Epithelial-Mesenchymal Transition (EMT) and kidney fibrosis. In addition, dysregulation of CFTR activates canonical Wnt/β-catenin signaling pathways, whereas the β-catenin inhibitor reverses the effects of CFTR downregulation on EMT marker. More interestingly, CFTR interacts with Dishevelled 2 (Dvl2), a key component of Wnt signaling, thereby suppressing the activation of β-catenin. Compared to wild type, deltaF508 mice with UUO treatment exhibit significantly higher β-catenin activity with aggregated kidney fibrogenesis, which is reduced by forced overexpression of CFTR. Taken together, our study reveals a novel mechanism by which CFTR regulates Wnt/β-catenin signaling pertinent to progression of kidney fibrosis and indicates a potential treatment target.
Collapse
|
19
|
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
|
20
|
Veltman M, Stolarczyk M, Radzioch D, Wojewodka G, De Sanctis JB, Dik WA, Dzyubachyk O, Oravecz T, de Kleer I, Scholte BJ. Correction of lung inflammation in a F508del CFTR murine cystic fibrosis model by the sphingosine-1-phosphate lyase inhibitor LX2931. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1000-L1014. [PMID: 27663991 DOI: 10.1152/ajplung.00298.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/19/2016] [Indexed: 01/08/2023] Open
Abstract
Progressive lung disease with early onset is the main cause of mortality and morbidity in cystic fibrosis patients. Here we report a reduction of sphingosine-1-phosphate (S1P) in the lung of unchallenged Cftrtm1EUR F508del CFTR mutant mice. This correlates with enhanced infiltration by inducible nitric oxide synthase (iNOS)-expressing granulocytes, B cells, and T cells. Furthermore, the ratio of macrophage-derived dendritic cells (MoDC) to conventional dendritic cells (cDC) is higher in mutant mouse lung, consistent with unprovoked inflammation. Oral application of a S1P lyase inhibitor (LX2931) increases S1P levels in mutant mouse tissues. This normalizes the lung MoDC/cDC ratio and reduces B and T cell counts. Lung granulocytes are enhanced, but iNOS expression is reduced in this population. Although lung LyC6+ monocytes are enhanced by LX2931, they apparently do not differentiate to MoDC and macrophages. After challenge with bacterial toxins (LPS-fMLP) we observe enhanced levels of proinflammatory cytokines TNF-α, KC, IFNγ, and IL-12 and the inducible mucin MUC5AC in mutant mouse lung, evidence of deficient resolution of inflammation. LX2931 does not prevent transient inflammation or goblet cell hyperplasia after challenge, but it reduces MUC5AC and proinflammatory cytokine levels toward normal values. We conclude that lung pathology in homozygous mice expressing murine F508del CFTR, which represents the most frequent mutation in CF patients, is characterized by abnormal behavior of infiltrating myeloid cells and delayed resolution of induced inflammation. This phenotype can be partially corrected by a S1P lyase inhibitor, providing a rationale for therapeutic targeting of the S1P signaling pathway in CF patients.
Collapse
Affiliation(s)
- Mieke Veltman
- Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Danuta Radzioch
- Departments of Medicine and Human Genetics, McGill University, Montreal, Canada
| | - Gabriella Wojewodka
- Departments of Medicine and Human Genetics, McGill University, Montreal, Canada
| | - Juan B De Sanctis
- Faculty of Medicine. Universidad Central de Venezuela, Institute of Immunology, Caracas, Venezuela
| | - Willem A Dik
- Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Oleh Dzyubachyk
- Department of Radiology, Division of Image Processing, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ismé de Kleer
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus MC, Rotterdam, The Netherlands; and.,Laboratory of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Bob J Scholte
- Cell Biology, Erasmus MC, Rotterdam, The Netherlands;
| |
Collapse
|
21
|
Zhang Y, O'Brien WG, Zhao Z, Lee CC. 5'-adenosine monophosphate mediated cooling treatment enhances ΔF508-Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) stability in vivo. J Biomed Sci 2015; 22:72. [PMID: 26335336 PMCID: PMC4559075 DOI: 10.1186/s12929-015-0178-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/20/2015] [Indexed: 12/26/2022] Open
Abstract
Background Gene mutations that produce misprocessed proteins are linked to many human disorders. Interestingly, some misprocessed proteins retained their biological function when stabilized by low temperature treatment of cultured cells in vitro. Here we investigate whether low temperature treatment in vivo can rescue misfolded proteins by applying 5’-AMP mediated whole body cooling to a Cystic Fibrosis (CF) mouse model carrying a mutant cystic fibrosis transmembrane conductance regulator (CFTR) with a deletion of the phenylalanine residue in position 508 (ΔF508-CFTR). Low temperature treatment of cultured cells was previously shown to be able to alleviate the processing defect of ΔF508-CFTR, enhancing its plasma membrane localization and its function in mediating chloride ion transport. Results Here, we report that whole body cooling enhanced the retention of ΔF508-CFTR in intestinal epithelial cells. Functional analysis based on β-adrenergic dependent salivary secretion and post-natal mortality rate revealed a moderate but significant improvement in treated compared with untreated CF mice. Conclusions Our findings demonstrate that temperature sensitive processing of mutant proteins can be responsive to low temperature treatment in vivo.
Collapse
Affiliation(s)
- Yueqiang Zhang
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - William G O'Brien
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhaoyang Zhao
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Cheng Chi Lee
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
22
|
Ermund A, Meiss LN, Scholte BJ, Hansson GC. Hypertonic saline releases the attached small intestinal cystic fibrosis mucus. Clin Exp Pharmacol Physiol 2015; 42:69-75. [PMID: 25311799 PMCID: PMC4303949 DOI: 10.1111/1440-1681.12322] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 01/01/2023]
Abstract
Hypertonic saline inhalation has become a cornerstone in the treatment of cystic fibrosis (CF), but its effect on CF mucus is still not understood. In CF, mucus stagnates in the airways, causing mucus plugging, and forming a substrate for bacterial invasion. Using horizontal Ussing-type chambers to allow easy access to the tissue, we have recently shown that the small intestinal mucus of CF mice is attached to the epithelium and not freely movable as opposed to normal mucus, thus pointing to a similarity between the CF mucus in the ileum and airways. In the same type of system, we investigated how hypertonic saline affects mucus thickness, attachment and penetrability to fluorescent beads the size of bacteria in ileal explants from the cystic fibrosis transmembrane conductance regulator mutant (ΔF508) mouse, in order to characterize how this common therapy affects mucus properties. Hypertonic saline (1.75–5%) detached the mucus from the epithelium, but the mucus remained impenetrable to beads the size of bacteria. This approach might be used to test other mucolytic interventions in CF.
Collapse
Affiliation(s)
- Anna Ermund
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | | | | |
Collapse
|
23
|
Tomati V, Sondo E, Armirotti A, Caci E, Pesce E, Marini M, Gianotti A, Ju Jeon Y, Cilli M, Pistorio A, Mastracci L, Ravazzolo R, Scholte B, Ronai Z, Galietta LJV, Pedemonte N. Genetic Inhibition Of The Ubiquitin Ligase Rnf5 Attenuates Phenotypes Associated To F508del Cystic Fibrosis Mutation. Sci Rep 2015; 5:12138. [PMID: 26183966 PMCID: PMC4505316 DOI: 10.1038/srep12138] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/17/2015] [Indexed: 12/28/2022] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the CFTR chloride channel. Deletion of phenylalanine 508 (F508del), the most frequent CF mutation, impairs CFTR trafficking and gating. F508del-CFTR mistrafficking may be corrected by acting directly on mutant CFTR itself or by modulating expression/activity of CFTR-interacting proteins, that may thus represent potential drug targets. To evaluate possible candidates for F508del-CFTR rescue, we screened a siRNA library targeting known CFTR interactors. Our analysis identified RNF5 as a protein whose inhibition promoted significant F508del-CFTR rescue and displayed an additive effect with the investigational drug VX-809. Significantly, RNF5 loss in F508del-CFTR transgenic animals ameliorated intestinal malabsorption and concomitantly led to an increase in CFTR activity in intestinal epithelial cells. In addition, we found that RNF5 is differentially expressed in human bronchial epithelia from CF vs. control patients. Our results identify RNF5 as a target for therapeutic modalities to antagonize mutant CFTR proteins.
Collapse
Affiliation(s)
| | | | - Andrea Armirotti
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genova, Italy
| | | | | | | | | | - Young Ju Jeon
- The Burnham Institute for Medical Research, La Jolla, California, USA
| | | | | | - Luca Mastracci
- IRCCS AOU San Martino–IST, Genova, Italy
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics, University of Genova, Italy
| | - Roberto Ravazzolo
- Istituto Giannina Gaslini, Genova, Italy
- DINOGMI Department, University of Genova, Italy
| | - Bob Scholte
- Cell Biology Department, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ze’ev Ronai
- The Burnham Institute for Medical Research, La Jolla, California, USA
| | | | | |
Collapse
|
24
|
Sphingosine-1-Phosphate Is a Novel Regulator of Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Activity. PLoS One 2015; 10:e0130313. [PMID: 26079370 PMCID: PMC4469317 DOI: 10.1371/journal.pone.0130313] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 05/18/2015] [Indexed: 01/12/2023] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) attenuates sphingosine-1-phosphate (S1P) signaling in resistance arteries and has emerged as a prominent regulator of myogenic vasoconstriction. This investigation demonstrates that S1P inhibits CFTR activity via adenosine monophosphate-activated kinase (AMPK), establishing a potential feedback link. In Baby Hamster Kidney (BHK) cells expressing wild-type human CFTR, S1P (1μmol/L) attenuates forskolin-stimulated, CFTR-dependent iodide efflux. S1P's inhibitory effect is rapid (within 30 seconds), transient and correlates with CFTR serine residue 737 (S737) phosphorylation. Both S1P receptor antagonism (4μmol/L VPC 23019) and AMPK inhibition (80μmol/L Compound C or AMPK siRNA) attenuate S1P-stimluated (i) AMPK phosphorylation, (ii) CFTR S737 phosphorylation and (iii) CFTR activity inhibition. In BHK cells expressing the ΔF508 CFTR mutant (CFTRΔF508), the most common mutation causing cystic fibrosis, both S1P receptor antagonism and AMPK inhibition enhance CFTR activity, without instigating discernable correction. In summary, we demonstrate that S1P/AMPK signaling transiently attenuates CFTR activity. Since our previous work positions CFTR as a negative S1P signaling regulator, this signaling link may positively reinforce S1P signals. This discovery has clinical ramifications for the treatment of disease states associated with enhanced S1P signaling and/or deficient CFTR activity (e.g. cystic fibrosis, heart failure). S1P receptor/AMPK inhibition could synergistically enhance the efficacy of therapeutic strategies aiming to correct aberrant CFTR trafficking.
Collapse
|
25
|
Bazett M, Honeyman L, Stefanov AN, Pope CE, Hoffman LR, Haston CK. Cystic fibrosis mouse model-dependent intestinal structure and gut microbiome. Mamm Genome 2015; 26:222-34. [PMID: 25721416 DOI: 10.1007/s00335-015-9560-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/13/2015] [Indexed: 01/15/2023]
Abstract
Mice with a null mutation in the cystic fibrosis transmembrane conductance regulator (Cftr) gene show intestinal structure alterations and bacterial overgrowth. To determine whether these changes are model-dependent and whether the intestinal microbiome is altered in cystic fibrosis (CF) mouse models, we characterized the ileal tissue and intestinal microbiome of mice with the clinically common ΔF508 Cftr mutation (FVB/N Cftr(tm1Eur)) and with Cftr null mutations (BALB/c Cftr(tm1UNC) and C57BL/6 Cftr(tm1UNC)). Intestinal disease in 12-week-old CF mice, relative to wild-type strain controls, was measured histologically. The microbiome was characterized by pyrosequencing of the V4-V6 region of the 16S rRNA gene and intestinal load was measured by RT-PCR of the 16S rRNA gene. The CF-associated increases in ileal crypt to villus axis distention, goblet cell hyperplasia, and muscularis externa thickness were more severe in the BALB/c and C57BL/6 Cftr(tm1UNC) mice than in the FVB/N Cftr(tm1Eur) mice. Intestinal bacterial load was significantly increased in all CF models, compared to levels in controls, and positively correlated with circular muscle thickness in CF, but not wild-type, mice. Microbiome profiling identified Bifidobacterium and groups of Lactobacillus to be of altered abundance in the CF mice but overall bacterial frequencies were not common to the three CF strains and were not correlative of major histological changes. In conclusion, intestinal structure alterations, bacterial overgrowth, and dysbiosis were each more severe in BALB/c and C57BL/6 Cftr(tm1UNC) mice than in the FVB/N Cftr(tm1Eur) mice. The intestinal microbiome differed among the three CF mouse models.
Collapse
Affiliation(s)
- Mark Bazett
- Meakins-Christie Laboratories, Departments of Medicine and Human Genetics, McGill University, 3626 St. Urbain, Montreal, QC, H2X 2P2, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Palomo J, Marchiol T, Piotet J, Fauconnier L, Robinet M, Reverchon F, Le Bert M, Togbe D, Buijs-Offerman R, Stolarczyk M, Quesniaux VFJ, Scholte BJ, Ryffel B. Role of IL-1β in experimental cystic fibrosis upon P. aeruginosa infection. PLoS One 2014; 9:e114884. [PMID: 25500839 PMCID: PMC4264861 DOI: 10.1371/journal.pone.0114884] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 11/14/2014] [Indexed: 11/18/2022] Open
Abstract
Cystic fibrosis is associated with increased inflammatory responses to pathogen challenge. Here we revisited the role of IL-1β in lung pathology using the experimental F508del-CFTR murine model on C57BL/6 genetic background (Cftr(tm1eur) or d/d), on double deficient for d/d and type 1 interleukin-1 receptor (d/d X IL-1R1-/-), and antibody neutralization. At steady state, young adult d/d mice did not show any signs of spontaneous lung inflammation. However, IL-1R1 deficiency conferred partial protection to repeated P. aeruginosa endotoxins/LPS lung instillation in d/d mice, as 50% of d/d mice succumbed to inflammation, whereas all d/d x IL-1R1-/- double mutants survived with lower initial weight loss and less pulmonary collagen and mucus production, suggesting that the absence of IL-1R1 signaling is protective in d/d mice in LPS-induced lung damage. Using P. aeruginosa acute lung infection we found heightened neutrophil recruitment in d/d mice with higher epithelial damage, increased bacterial load in BALF, and augmented IL-1β and TNF-α in parenchyma as compared to WT mice. Thus, F508del-CFTR mice show enhanced IL-1β signaling in response to P. aeruginosa. IL-1β antibody neutralization had no effect on lung homeostasis in either d/d or WT mice, however P. aeruginosa induced lung inflammation and bacterial load were diminished by IL-1β antibody neutralization. In conclusion, enhanced susceptibility to P. aeruginosa in d/d mice correlates with an excessive inflammation and with increased IL-1β production and reduced bacterial clearance. Further, we show that neutralization of IL-1β in d/d mice through the double mutation d/d x IL-1R1-/- and in WT via antibody neutralization attenuates inflammation. This supports the notion that intervention in the IL-1R1/IL-1β pathway may be detrimental in CF patients.
Collapse
Affiliation(s)
- Jennifer Palomo
- CNRS, UMR7355, Orleans, France
- Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | | | - Julie Piotet
- CNRS, UMR7355, Orleans, France
- Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | | | | | - Flora Reverchon
- CNRS, UMR7355, Orleans, France
- Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | | | | | | | | | - Valérie F. J. Quesniaux
- CNRS, UMR7355, Orleans, France
- Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Bob J. Scholte
- Erasmus MC, Cell Biology department, Rotterdam, The Netherlands
- * E-mail: (BS); (BR)
| | - Bernhard Ryffel
- CNRS, UMR7355, Orleans, France
- Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
- Institute of Infectious Disease and Molecular Medicine, IDM, Cape Town, South Africa
- * E-mail: (BS); (BR)
| |
Collapse
|
27
|
Pfister S, Weber T, Härtig W, Schwerdel C, Elsaesser R, Knuesel I, Fritschy JM. Novel role of cystic fibrosis transmembrane conductance regulator in maintaining adult mouse olfactory neuronal homeostasis. J Comp Neurol 2014; 523:406-30. [PMID: 25271146 DOI: 10.1002/cne.23686] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 02/03/2023]
Abstract
The olfactory epithelium (OE) of mice deficient in cystic fibrosis transmembrane conductance regulator (CFTR) exhibits ion transport deficiencies reported in human CF airways, as well as progressive neuronal loss, suggesting defects in olfactory neuron homeostasis. Microvillar cells, a specialized OE cell-subtype, have been implicated in maintaining tissue homeostasis. These cells are endowed with a PLCβ2/IP3 R3/TRPC6 signal transduction pathway modulating release of neuropeptide Y (NPY), which stimulates OE stem cell activity. It is unknown, however, whether microvillar cells also mediate the deficits observed in CFTR-null mice. Here we show that Cftr mRNA in mouse OE is exclusively localized in microvillar cells and CFTR immunofluorescence is coassociated with the scaffolding protein NHERF-1 and PLCβ2 in microvilli. In CFTR-null mice, PLCβ2 was undetectable, NHERF-1 mislocalized, and IP3 R3 more intensely stained, along with increased levels of NPY, suggesting profound alteration of the PLCβ2/IP3 R3 signaling pathway. In addition, basal olfactory neuron homeostasis was altered, shown by increased progenitor cell proliferation, differentiation, and apoptosis and by reduced regenerative capacity following methimazole-induced neurodegeneration. The importance of CFTR in microvillar cells was further underscored by decreased thickness of the OE mucus layer and increased numbers of immune cells within this tissue in CFTR-KO mice. Finally, we observed enhanced immune responses to an acute viral-like infection, as well as hyper-responsiveness to chemical and physical stimuli applied intranasally. Taken together, these data strengthen the notion that microvillar cells in the OE play a key role in maintaining tissue homeostasis and identify several mechanisms underlying this regulation through the multiple functions of CFTR.
Collapse
Affiliation(s)
- Sandra Pfister
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
28
|
Gustafsson JK, Lindén SK, Alwan AH, Scholte BJ, Hansson GC, Sjövall H. Carbachol-induced colonic mucus formation requires transport via NKCC1, K⁺ channels and CFTR. Pflugers Arch 2014; 467:1403-1415. [PMID: 25139191 DOI: 10.1007/s00424-014-1595-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 08/01/2014] [Accepted: 08/06/2014] [Indexed: 12/26/2022]
Abstract
The colonic mucosa protects itself from the luminal content by secreting mucus that keeps the bacteria at a distance from the epithelium. For this barrier to be effective, the mucus has to be constantly replenished which involves exocytosis and expansion of the secreted mucins. Mechanisms involved in regulation of mucus exocytosis and expansion are poorly understood, and the aim of this study was to investigate whether epithelial anion secretion regulates mucus formation in the colon. The muscarinic agonist carbachol was used to induce parallel secretion of anions and mucus, and by using established inhibitors of ion transport, we studied how inhibition of epithelial transport affected mucus formation in mouse colon. Anion secretion and mucin exocytosis were measured by changes in membrane current and epithelial capacitance, respectively. Mucus thickness measurements were used to determine the carbachol effect on mucus growth. The results showed that the carbachol-induced increase in membrane current was dependent on NKCC1 co-transport, basolateral K(+) channels and Cftr activity. In contrast, the carbachol-induced increase in capacitance was partially dependent on NKCC1 and K(+) channel activity, but did not require Cftr activity. Carbachol also induced an increase in mucus thickness that was inhibited by the NKCC1 blocker bumetanide. However, mice that lacked a functional Cftr channel did not respond to carbachol with an increase in mucus thickness, suggesting that carbachol-induced mucin expansion requires Cftr channel activity. In conclusion, these findings suggest that colonic epithelial transport regulates mucus formation by affecting both exocytosis and expansion of the mucin molecules.
Collapse
Affiliation(s)
- Jenny K Gustafsson
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden.
| | - Sara K Lindén
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden
| | - Ala H Alwan
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden
| | - Bob J Scholte
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Gunnar C Hansson
- Department of Medical Biochemistry, University of Gothenburg, Medicinaregatan 9A, Box 440, Gothenburg, 405 30, Sweden
| | - Henrik Sjövall
- Department of Internal Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
29
|
Liu X, Li T, Riederer B, Lenzen H, Ludolph L, Yeruva S, Tuo B, Soleimani M, Seidler U. Loss of Slc26a9 anion transporter alters intestinal electrolyte and HCO3(-) transport and reduces survival in CFTR-deficient mice. Pflugers Arch 2014; 467:1261-75. [PMID: 24965066 PMCID: PMC4434866 DOI: 10.1007/s00424-014-1543-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/19/2014] [Accepted: 05/20/2014] [Indexed: 12/16/2022]
Abstract
Slc26a9 is an anion transporter that is strongly expressed in the stomach and lung. Slc26a9 variants were recently found associated with a higher incidence of meconium ileus in cystic fibrosis (CF) infants, raising the question whether Slc26a9 is expressed in the intestine and what its functional role is. Slc26a9 messenger RNA (mRNA) was found highly expressed in the mucosae of the murine and human upper gastrointestinal tract, with an abrupt decrease in expression levels beyond the duodenum. Absence of SLC26a9 expression strongly increased the intestinally related mortality in cystic fibrosis transmembrane conductance regulator (CFTR)-deficient mice. Proximal duodenal JHCO3(-) and fluid secretion were reduced in the absence of Slc26a9 expression. In the proximal duodenum of young Slc26a9 KO mice, the glands and villi/crypts were elongated and proliferation was enhanced. This difference was lost with ageing, as were the alterations in fluid movement, whereas the reduction in JHCO3(-) remained. Laser dissection followed by qPCR suggested Slc26a9 expression to be crypt-predominant in the duodenum. In summary, deletion of Slc26a9 caused bicarbonate secretory and fluid absorptive changes in the proximal duodenal mucosa and increased the postweaning death rates in CFTR-deficient mice. Functional alterations in the duodenum were most prominent at young ages. We assume that the association of meconium ileus and Slc26a9 variants may be related to maldigestion and impaired downstream signaling caused by loss of upper GI tract digestive functions, aggravating the situation of lack of secretion and sticky mucus at the site of obstruction in CF intestine.
Collapse
Affiliation(s)
- Xuemei Liu
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Multicenter intestinal current measurements in rectal biopsies from CF and non-CF subjects to monitor CFTR function. PLoS One 2013; 8:e73905. [PMID: 24040112 PMCID: PMC3769519 DOI: 10.1371/journal.pone.0073905] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/23/2013] [Indexed: 11/30/2022] Open
Abstract
Intestinal current measurements (ICM) from rectal biopsies are a sensitive means to detect cystic fibrosis transmembrane conductance regulator (CFTR) function, but have not been optimized for multicenter use. We piloted multicenter standard operating procedures (SOPs) to detect CFTR activity by ICM and examined key questions for use in clinical trials. SOPs for ICM using human rectal biopsies were developed across three centers and used to characterize ion transport from non-CF and CF subjects (two severe CFTR mutations). All data were centrally evaluated by a blinded interpreter. SOPs were then used across four centers to examine the effect of cold storage on CFTR currents and compare CFTR currents in biopsies from one subject studied simultaneously either at two sites (24 hours post-biopsy) or when biopsies were obtained by either forceps or suction. Rectal biopsies from 44 non-CF and 17 CF subjects were analyzed. Mean differences (µA/cm2; 95% confidence intervals) between CF and non-CF were forskolin/IBMX=102.6(128.0 to 81.1), carbachol=96.3(118.7 to 73.9), forskolin/IBMX+carbachol=200.9(243.1 to 158.6), and bumetanide=-44.6 (-33.7 to -55.6) (P<0.005, CF vs non-CF for all parameters). Receiver Operating Characteristic curves indicated that each parameter discriminated CF from non-CF subjects (area under the curve of 0.94-0.98). CFTR dependent currents following 18-24 hours of cold storage for forskolin/IBMX, carbachol, and forskolin/IBMX+carbachol stimulation (n=17 non-CF subjects) were 44%, 47.5%, and 47.3%, respectively of those in fresh biopsies. CFTR-dependent currents from biopsies studied after cold storage at two sites simultaneously demonstrated moderate correlation (n=14 non-CF subjects, Pearson correlation coefficients 0.389, 0.484, and 0.533). Similar CFTR dependent currents were detected from fresh biopsies obtained by either forceps or suction (within-subject comparisons, n=22 biopsies from three non-CF subjects). Multicenter ICM is a feasible CFTR outcome measure that discriminates CF from non-CF ion transport, offers unique advantages over other CFTR bioassays, and warrants further development as a potential CFTR biomarker.
Collapse
|
31
|
Ermund A, Schütte A, Johansson MEV, Gustafsson JK, Hansson GC. Studies of mucus in mouse stomach, small intestine, and colon. I. Gastrointestinal mucus layers have different properties depending on location as well as over the Peyer's patches. Am J Physiol Gastrointest Liver Physiol 2013; 305:G341-7. [PMID: 23832518 PMCID: PMC3761247 DOI: 10.1152/ajpgi.00046.2013] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Colon has been shown to have a two-layered mucus system where the inner layer is devoid of bacteria. However, a complete overview of the mouse gastrointestinal mucus system is lacking. We now characterize mucus release, thickness, growth over time, adhesive properties, and penetrability to fluorescent beads from stomach to distal colon. Colon displayed spontaneous mucus release and all regions released mucus in response to carbachol and PGE2, except the distal colon and domes of Peyer's patches. Stomach and colon had an inner mucus layer that was adherent to the epithelium. In contrast, the small intestine and Peyer's patches had a single mucus layer that was easily aspirated. The inner mucus layer of the distal colon was not penetrable to beads the size of bacteria and the inner layer of the proximal colon was only partly penetrable. In contrast, the inner mucus layer of stomach was fully penetrable, as was the small intestinal mucus. This suggests a functional organization of the intestinal mucus system, where the small intestine has loose and penetrable mucus that may allow easy penetration of nutrients, in contrast to the stomach, where the mucus provides physical protection, and the colon, where the mucus separates bacteria from the epithelium. This knowledge of the mucus system and its organization improves our understanding of the gastrointestinal tract physiology.
Collapse
Affiliation(s)
- Anna Ermund
- Dept. Medical Biochemistry, Univ. of Gothenburg, Box 440, 40530 Gothenburg, Sweden.
| | | | | | | | | |
Collapse
|
32
|
Dekkers JF, Wiegerinck CL, de Jonge HR, Bronsveld I, Janssens HM, de Winter-de Groot KM, Brandsma AM, de Jong NWM, Bijvelds MJC, Scholte BJ, Nieuwenhuis EES, van den Brink S, Clevers H, van der Ent CK, Middendorp S, Beekman JM. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat Med 2013; 19:939-45. [PMID: 23727931 DOI: 10.1038/nm.3201] [Citation(s) in RCA: 740] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 12/20/2012] [Indexed: 01/02/2023]
Abstract
We recently established conditions allowing for long-term expansion of epithelial organoids from intestine, recapitulating essential features of the in vivo tissue architecture. Here we apply this technology to study primary intestinal organoids of people suffering from cystic fibrosis, a disease caused by mutations in CFTR, encoding cystic fibrosis transmembrane conductance regulator. Forskolin induces rapid swelling of organoids derived from healthy controls or wild-type mice, but this effect is strongly reduced in organoids of subjects with cystic fibrosis or in mice carrying the Cftr F508del mutation and is absent in Cftr-deficient organoids. This pattern is phenocopied by CFTR-specific inhibitors. Forskolin-induced swelling of in vitro-expanded human control and cystic fibrosis organoids corresponds quantitatively with forskolin-induced anion currents in freshly excised ex vivo rectal biopsies. Function of the CFTR F508del mutant protein is restored by incubation at low temperature, as well as by CFTR-restoring compounds. This relatively simple and robust assay will facilitate diagnosis, functional studies, drug development and personalized medicine approaches in cystic fibrosis.
Collapse
Affiliation(s)
- Johanna F Dekkers
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Mucins--large, highly glycosylated proteins--are important for the luminal protection of the gastrointestinal tract. Enterocytes have their apical surface covered by transmembrane mucins and goblet cells produce the secreted gel-forming mucins that form mucus. The small intestine has a single unattached mucus layer, which in cystic fibrosis becomes attached, accounting for the intestinal manifestations of this disease. The stomach and colon have two layers of mucus; the inner layer is attached and the outer layer is less dense and unattached. In the colon, the outer mucus layer is the habitat for commensal bacteria. The inner mucus layer is impervious to bacteria and is renewed every hour by surface goblet cells. The crypt goblet cells have the ability to restitute the mucus layer by secretion, for example after an ischaemic challenge. Proteases of certain parasites and some bacteria can cleave mucins and dissolve the mucus as part of their pathogenicity. The inner mucus layer can, however, also become penetrable to bacteria by several other mechanisms, including aberrations in the immune system. When bacteria reach the epithelial surface, the immune system is activated and inflammation is triggered. This mechanism might occur in some types of ulcerative colitis.
Collapse
|
34
|
Figueiras-Fierro D, Acevedo JJ, Martínez-López P, Escoffier J, Sepúlveda FV, Balderas E, Orta G, Visconti PE, Darszon A. Electrophysiological evidence for the presence of cystic fibrosis transmembrane conductance regulator (CFTR) in mouse sperm. J Cell Physiol 2013; 228:590-601. [PMID: 22833409 DOI: 10.1002/jcp.24166] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 07/17/2012] [Indexed: 01/25/2023]
Abstract
Mammalian sperm must undergo a maturational process, named capacitation, in the female reproductive tract to fertilize the egg. Sperm capacitation is regulated by a cAMP/protein kinase A (PKA) pathway and involves increases in intracellular Ca(2+), pH, Cl(-), protein tyrosine phosphorylation, and in mouse and some other mammals a membrane potential hyperpolarization. The cystic fibrosis transmembrane conductance regulator (CFTR), a Cl(-) channel modulated by cAMP/PKA and ATP, was detected in mammalian sperm and proposed to modulate capacitation. Our whole-cell patch-clamp recordings from testicular mouse sperm now reveal a Cl(-) selective component to membrane current that is ATP-dependent, stimulated by cAMP, cGMP, and genistein (a CFTR agonist, at low concentrations), and inhibited by DPC and CFTR(inh) -172, two well-known CFTR antagonists. Furthermore, the Cl(-) current component activated by cAMP and inhibited by CFTR(inh) -172 is absent in recordings on testicular sperm from mice possessing the CFTR ΔF508 loss-of-function mutation, indicating that CFTR is responsible for this component. A Cl(-) selective like current component displaying CFTR characteristics was also found in wild type epididymal sperm bearing the cytoplasmatic droplet. Capacitated sperm treated with CFTR(inh) -172 undergo a shape change, suggesting that CFTR is involved in cell volume regulation. These findings indicate that functional CFTR channels are present in mouse sperm and their biophysical properties are consistent with their proposed participation in capacitation.
Collapse
Affiliation(s)
- Dulce Figueiras-Fierro
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Morelos, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Lee TT, Zhang XD, Chuang CC, Chen JJ, Chen YA, Chen SC, Chen TY, Tang CY. Myotonia congenita mutation enhances the degradation of human CLC-1 chloride channels. PLoS One 2013; 8:e55930. [PMID: 23424641 PMCID: PMC3570542 DOI: 10.1371/journal.pone.0055930] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 01/03/2013] [Indexed: 11/18/2022] Open
Abstract
Myotonia congenita is a hereditary muscle disorder caused by mutations in the human voltage-gated chloride (Cl−) channel CLC-1. Myotonia congenita can be inherited in an autosomal recessive (Becker type) or dominant (Thomsen type) fashion. One hypothesis for myotonia congenita is that the inheritance pattern of the disease is determined by the functional consequence of the mutation on the gating of CLC-1 channels. Several disease-related mutations, however, have been shown to yield functional CLC-1 channels with no detectable gating defects. In this study, we have functionally and biochemically characterized a myotonia mutant: A531V. Despite a gating property similar to that of wild-type (WT) channels, the mutant CLC-1 channel displayed a diminished whole-cell current density and a reduction in the total protein expression level. Our biochemical analyses further demonstrated that the reduced expression of A531V can be largely attributed to an enhanced proteasomal degradation as well as a defect in protein trafficking to surface membranes. Moreover, the A531V mutant protein also appeared to be associated with excessive endosomal-lysosomal degradation. Neither the reduced protein expression nor the diminished current density was rescued by incubating A531V-expressing cells at 27°C. These results demonstrate that the molecular pathophysiology of A531V does not involve anomalous channel gating, but rather a disruption of the balance between the synthesis and degradation of the CLC-1 channel protein.
Collapse
Affiliation(s)
- Ting-Ting Lee
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Xiao-Dong Zhang
- Neuroscience Center, University of California Davis, Davis, California, United States of America
| | - Chao-Chin Chuang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jing-Jer Chen
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-An Chen
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Ching Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsung-Yu Chen
- Neuroscience Center, University of California Davis, Davis, California, United States of America
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
36
|
Wilke M, Bot A, Jorna H, Scholte BJ, de Jonge HR. Rescue of murine F508del CFTR activity in native intestine by low temperature and proteasome inhibitors. PLoS One 2012; 7:e52070. [PMID: 23284872 PMCID: PMC3528711 DOI: 10.1371/journal.pone.0052070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 11/15/2012] [Indexed: 11/19/2022] Open
Abstract
Most patients with Cystic Fibrosis (CF) carry at least one allele with the F508del mutation, resulting in a CFTR chloride channel protein with a processing, gating and stability defect, but with substantial residual activity when correctly sorted to the apical membranes of epithelial cells. New therapies are therefore aimed at improving the folding and trafficking of F508del CFTR, (CFTR correctors) or at enhancing the open probability of the CFTR chloride channel (CFTR potentiators). Preventing premature breakdown of F508del CFTR is an alternative or additional strategy, which is investigated in this study. We established an ex vivo assay for murine F508del CFTR rescue in native intestinal epithelium that can be used as a pre-clinical test for candidate therapeutics. Overnight incubation of muscle stripped ileum in modified William's E medium at low temperature (26°C), and 4 h or 6 h incubation at 37°C with different proteasome inhibitors (PI: ALLN, MG-132, epoxomicin, PS341/bortezomib) resulted in fifty to hundred percent respectively of the wild type CFTR mediated chloride secretion (forskolin induced short-circuit current). The functional rescue was accompanied by enhanced expression of the murine F508del CFTR protein at the apical surface of intestinal crypts and a gain in the amount of complex-glycosylated CFTR (band C) up to 20% of WT levels. Sustained rescue in the presence of brefeldin A shows the involvement of a post-Golgi compartment in murine F508del CFTR degradation, as was shown earlier for its human counterpart. Our data show that proteasome inhibitors are promising candidate compounds for improving rescue of human F508del CFTR function, in combination with available correctors and potentiators.
Collapse
Affiliation(s)
- Martina Wilke
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
37
|
Zaydman MA, Silva JR, Cui J. Ion channel associated diseases: overview of molecular mechanisms. Chem Rev 2012; 112:6319-33. [PMID: 23151230 DOI: 10.1021/cr300360k] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mark A Zaydman
- Department of Biomedical Engineering, Washington University, Saint Louis, Missouri 63130, United States
| | | | | |
Collapse
|
38
|
Zhang D, Ciciriello F, Anjos SM, Carissimo A, Liao J, Carlile GW, Balghi H, Robert R, Luini A, Hanrahan JW, Thomas DY. Ouabain Mimics Low Temperature Rescue of F508del-CFTR in Cystic Fibrosis Epithelial Cells. Front Pharmacol 2012; 3:176. [PMID: 23060796 PMCID: PMC3463858 DOI: 10.3389/fphar.2012.00176] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 09/14/2012] [Indexed: 11/23/2022] Open
Abstract
Most cases of cystic fibrosis (CF) are caused by the deletion of a single phenylalanine residue at position 508 of the cystic fibrosis transmembrane conductance regulator (CFTR). The mutant F508del-CFTR is retained in the endoplasmic reticulum and degraded, but can be induced by low temperature incubation (29°C) to traffic to the plasma membrane where it functions as a chloride channel. Here we show that, cardiac glycosides, at nanomolar concentrations, can partially correct the trafficking of F508del-CFTR in human CF bronchial epithelial cells (CFBE41o-) and in an F508del-CFTR mouse model. Comparison of the transcriptional profiles obtained with polarized CFBE41o-cells after treatment with ouabain and by low temperature has revealed a striking similarity between the two corrector treatments that is not shared with other correctors. In summary, our study shows a novel function of ouabain and its analogs in the regulation of F508del-CFTR trafficking and suggests that compounds that mimic this low temperature correction of trafficking will provide new avenues for the development of therapeutics for CF.
Collapse
Affiliation(s)
- Donglei Zhang
- Department of Biochemistry, McGill University Montréal, QC, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Carlile G, Keyzers R, Teske K, Robert R, Williams D, Linington R, Gray C, Centko R, Yan L, Anjos S, Sampson H, Zhang D, Liao J, Hanrahan J, Andersen R, Thomas D. Correction of F508del-CFTR Trafficking by the Sponge Alkaloid Latonduine Is Modulated by Interaction with PARP. ACTA ACUST UNITED AC 2012; 19:1288-99. [DOI: 10.1016/j.chembiol.2012.08.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 08/04/2012] [Accepted: 08/16/2012] [Indexed: 12/31/2022]
|
40
|
Xiao F, Li J, Singh AK, Riederer B, Wang J, Sultan A, Park H, Lee MG, Lamprecht G, Scholte BJ, De Jonge HR, Seidler U. Rescue of epithelial HCO3- secretion in murine intestine by apical membrane expression of the cystic fibrosis transmembrane conductance regulator mutant F508del. J Physiol 2012; 590:5317-34. [PMID: 22802588 DOI: 10.1113/jphysiol.2012.232124] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study investigated whether expression of the common cystic fibrosis transmembrane conductance regulator (CFTR) mutant F508del in the apical membrane of enterocytes confers increased bicarbonate secretory capacity on the intestinal epithelium of F508del mutant mice compared to that of CFTR knockout (KO) mice. CFTR KO mice, F508del mutant mice (F508del) and wild-type (WT) littermates were bred on the FVB/N background. F508del isolated brush border membrane (BBM) contained approximately 5-10% fully glycosylated band C protein compared to WT BBM. Similarly, the forskolin (FSK)-induced, CFTR-dependent short-circuit current (I(sc)) of F508del mucosa was approximately 5-10% of WT, whereas the HCO(3)(-) secretory response ( ) was almost half that of WT in both duodenum and mid-colon studied in vitro and in vivo. While WT intestine retained full FSK-induced in the absence of luminal Cl(-), the markedly higher than I(sc) in F508del intestine was dependent on the presence of luminal Cl(-), and was blocked by CFTR inhibitors. The Ste20-related proline-alanine-rich kinases (SPAK/OSR1), which are downstream of the with-no-lysine (K) protein kinases (WNK), were rapidly phosphorylated by FSK in WT and F508del, but significantly more slowly in CFTR KO intestine. In conclusion, the data demonstrate that low levels of F508del membrane expression in the intestine of F508del mice significantly increased FSK-induced HCO(3)(-) secretion mediated by Cl(-)/HCO(3)(-) exchange. However, in WT mucosa FSK elicited strong SPAK/OSR1 phosphorylation and Cl(-)-independent HCO(3)(-) efflux. This suggests that therapeutic strategies which deliver F508del to the apical membrane have the potential to significantly enhance epithelial HCO(3)(-) secretion.
Collapse
Affiliation(s)
- Fang Xiao
- Department of Gastoenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Meissner A, Yang J, Kroetsch JT, Sauvé M, Dax H, Momen A, Noyan-Ashraf MH, Heximer S, Husain M, Lidington D, Bolz SS. Tumor necrosis factor-α-mediated downregulation of the cystic fibrosis transmembrane conductance regulator drives pathological sphingosine-1-phosphate signaling in a mouse model of heart failure. Circulation 2012; 125:2739-50. [PMID: 22534621 DOI: 10.1161/circulationaha.111.047316] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P) signaling is a central regulator of resistance artery tone. Therefore, S1P levels need to be tightly controlled through the delicate interplay of its generating enzyme sphingosine kinase 1 and its functional antagonist S1P phosphohydrolase-1. The intracellular localization of S1P phosphohydrolase-1 necessitates the import of extracellular S1P into the intracellular compartment before its degradation. The present investigation proposes that the cystic fibrosis transmembrane conductance regulator transports extracellular S1P and hence modulates microvascular S1P signaling in health and disease. METHODS AND RESULTS In cultured murine vascular smooth muscle cells in vitro and isolated murine mesenteric and posterior cerebral resistance arteries ex vivo, the cystic fibrosis transmembrane conductance regulator (1) is critical for S1P uptake; (2) modulates S1P-dependent responses; and (3) is downregulated in vitro and in vivo by tumor necrosis factor-α, with significant functional consequences for S1P signaling and vascular tone. In heart failure, tumor necrosis factor-α downregulates the cystic fibrosis transmembrane conductance regulator across several organs, including the heart, lung, and brain, suggesting that it is a fundamental mechanism with implications for systemic S1P effects. CONCLUSIONS We identify the cystic fibrosis transmembrane conductance regulator as a critical regulatory site for S1P signaling; its tumor necrosis factor-α-dependent downregulation in heart failure underlies an enhancement in microvascular tone. This molecular mechanism potentially represents a novel and highly strategic therapeutic target for cardiovascular conditions involving inflammation.
Collapse
Affiliation(s)
- Anja Meissner
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mouse models of cystic fibrosis: Phenotypic analysis and research applications. J Cyst Fibros 2011; 10 Suppl 2:S152-71. [DOI: 10.1016/s1569-1993(11)60020-9] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
43
|
Abstract
The mutation F508del is the commonest cause of the genetic disease cystic fibrosis (CF). CF disrupts the function of many organs in the body, most notably the lungs, by perturbing salt and water transport across epithelial surfaces. F508del causes harm in two principal ways. First, the mutation prevents delivery of the cystic fibrosis transmembrane conductance regulator (CFTR) to its correct cellular location, the apical (lumen-facing) membrane of epithelial cells. Second, F508del perturbs the Cl(-) channel function of CFTR by disrupting channel gating. Here, we discuss the development of rational new therapies for CF that target F508del-CFTR. We highlight how structural studies provide new insight into the role of F508 in the regulation of channel gating by cycles of ATP binding and hydrolysis. We emphasize the use of high-throughput screening to identify lead compounds for therapy development. These compounds include CFTR correctors that restore the expression of F508del-CFTR at the apical membrane of epithelial cells and CFTR potentiators that rescue the F508del-CFTR gating defect. Initial results from clinical trials of CFTR correctors and potentiators augur well for the development of small molecule therapies that target the root cause of CF: mutations in CFTR.
Collapse
|
44
|
Johnson JS, Gentzsch M, Zhang L, Ribeiro CMP, Kantor B, Kafri T, Pickles RJ, Samulski RJ. AAV exploits subcellular stress associated with inflammation, endoplasmic reticulum expansion, and misfolded proteins in models of cystic fibrosis. PLoS Pathog 2011; 7:e1002053. [PMID: 21625534 PMCID: PMC3098238 DOI: 10.1371/journal.ppat.1002053] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 03/16/2011] [Indexed: 12/11/2022] Open
Abstract
Barriers to infection act at multiple levels to prevent viruses, bacteria, and parasites from commandeering host cells for their own purposes. An intriguing hypothesis is that if a cell experiences stress, such as that elicited by inflammation, endoplasmic reticulum (ER) expansion, or misfolded proteins, then subcellular barriers will be less effective at preventing viral infection. Here we have used models of cystic fibrosis (CF) to test whether subcellular stress increases susceptibility to adeno-associated virus (AAV) infection. In human airway epithelium cultured at an air/liquid interface, physiological conditions of subcellular stress and ER expansion were mimicked using supernatant from mucopurulent material derived from CF lungs. Using this inflammatory stimulus to recapitulate stress found in diseased airways, we demonstrated that AAV infection was significantly enhanced. Since over 90% of CF cases are associated with a misfolded variant of Cystic Fibrosis Transmembrane Conductance Regulator (ΔF508-CFTR), we then explored whether the presence of misfolded proteins could independently increase susceptibility to AAV infection. In these models, AAV was an order of magnitude more efficient at transducing cells expressing ΔF508-CFTR than in cells expressing wild-type CFTR. Rescue of misfolded ΔF508-CFTR under low temperature conditions restored viral transduction efficiency to that demonstrated in controls, suggesting effects related to protein misfolding were responsible for increasing susceptibility to infection. By testing other CFTR mutants, G551D, D572N, and 1410X, we have shown this phenomenon is common to other misfolded proteins and not related to loss of CFTR activity. The presence of misfolded proteins did not affect cell surface attachment of virus or influence expression levels from promoter transgene cassettes in plasmid transfection studies, indicating exploitation occurs at the level of virion trafficking or processing. Thus, we surmised that factors enlisted to process misfolded proteins such as ΔF508-CFTR in the secretory pathway also act to restrict viral infection. In line with this hypothesis, we found that AAV trafficked to the microtubule organizing center and localized near Golgi/ER transport proteins. Moreover, AAV infection efficiency could be modulated with siRNA-mediated knockdown of proteins involved in processing ΔF508-CFTR or sorting retrograde cargo from the Golgi and ER (calnexin, KDEL-R, β-COP, and PSMB3). In summary, our data support a model where AAV exploits a compromised secretory system and, importantly, underscore the gravity with which a stressed subcellular environment, under internal or external insults, can impact infection efficiency. Misfolded proteins have been associated with a variety of disorders such as cystic fibrosis, diabetes insipidus, alpha-antitrypsin deficiency, Parkinson's disease, and cancer. In this study, by using cellular models of events in cystic fibrosis lung disease we have revealed an effect of misfolded proteins on increasing susceptibility to infection with a parvovirus. Infection efficiency was an order of magnitude higher in cells expressing misfolded Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) mutant proteins than in cells expressing the correctly folded protein. During infection, virus capsids accumulated near cellular factors that normally process misfolded proteins and are involved in retrograde trafficking from the Golgi to endoplasmic reticulum. Furthermore, we have demonstrated that infection efficiency can be attenuated by restoring correct protein folding or augmented by siRNA-mediated knockdown of secretory pathway components. Taken together our results indicate that converging cellular systems operate to clear misfolded proteins and virus capsids from an infected cell. We raise the possibility that parvoviruses and perhaps other viruses exploit congested cellular secretory pathways during entry, and that viral infection could be a contributing factor in the progression of diseases associated with misfolded proteins.
Collapse
Affiliation(s)
- Jarrod S Johnson
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Suaud L, Miller K, Alvey L, Yan W, Robay A, Kebler C, Kreindler JL, Guttentag S, Hubbard MJ, Rubenstein RC. ERp29 regulates DeltaF508 and wild-type cystic fibrosis transmembrane conductance regulator (CFTR) trafficking to the plasma membrane in cystic fibrosis (CF) and non-CF epithelial cells. J Biol Chem 2011; 286:21239-53. [PMID: 21525008 DOI: 10.1074/jbc.m111.240267] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sodium 4-phenylbutyrate (4PBA) improves the intracellular trafficking of ΔF508-CFTR in cystic fibrosis (CF) epithelial cells. The underlying mechanism is uncertain, but 4PBA modulates the expression of some cytosolic molecular chaperones. To identify other 4PBA-regulated proteins that might regulate ΔF508-CFTR trafficking, we performed a differential display RT-PCR screen on IB3-1 CF bronchiolar epithelial cells exposed to 4PBA. One transcript up-regulated by 4PBA encoded ERp29, a luminal resident of the endoplasmic reticulum (ER) thought to be a novel molecular chaperone. We tested the hypothesis that ERp29 is a 4PBA-regulated ER chaperone that influences ΔF508-CFTR trafficking. ERp29 mRNA and protein expression was significantly increased (∼1.5-fold) in 4PBA-treated IB3-1 cells. In Xenopus oocytes, ERp29 overexpression increased the functional expression of both wild-type and ΔF508-CFTR over 3-fold and increased wild-type cystic fibrosis transmembrane conductance regulator (CFTR) plasma membrane expression. In CFBE41o- WT-CFTR cells, expression of and short circuit currents mediated by CFTR decreased upon depletion of ERp29 as did maturation of newly synthesized CFTR. In IB3-1 cells, ΔF508-CFTR co-immunoprecipitated with endogenous ERp29, and overexpression of ERp29 led to increased ΔF508-CFTR expression at the plasma membrane. These data suggest that ERp29 is a 4PBA-regulated ER chaperone that regulates WT-CFTR biogenesis and can promote ΔF508-CFTR trafficking in CF epithelial cells.
Collapse
Affiliation(s)
- Laurence Suaud
- Division of Pulmonary Medicine and Cystic Fibrosis Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hashimoto Y, Shuto T, Mizunoe S, Tomita A, Koga T, Sato T, Takeya M, Suico MA, Niibori A, Sugahara T, Shimasaki S, Sugiyama T, Scholte B, Kai H. CFTR-deficiency renders mice highly susceptible to cutaneous symptoms during mite infestation. J Transl Med 2011; 91:509-18. [PMID: 21135815 DOI: 10.1038/labinvest.2010.193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Pruritus, also known as itch, is a sensation that causes a desire to scratch. Prolonged scratching exacerbates skin lesions in several skin diseases such as atopic dermatitis. Here, we identify the cystic fibrosis transmembrane conductance regulator (CFTR/Cftr), an integral membrane protein that mediates transepithelial chloride transport, as a determinant factor in mice for the susceptibility to several cutaneous symptoms during mite infestation. Mice that endogenously express dysfunctional Cftr (Cftr(ΔF508/ΔF508)) show significant increase of scratching behavior and skin fibrosis after mite exposure. These phenotypes were due to the increased expression of nerve growth factor (NGF) that augments the sensitization of peripheral nerve fibers. Moreover, protein gene product 9.5 (PGP9.5)-positive neurites were abundant in the epidermis of mite-infested Cftr(ΔF508/ΔF508) mice. Furthermore, mite-infested Cftr(+/+) mice orally administered with a chloride channel inhibitor glibenclamide had higher scratching count and increased level of NGF than vehicle-treated mice. Consistently, mite extract-exposed primary and transformed human keratinocytes, treated with CFTR inhibitor, had significantly higher level of NGF mRNA compared with vehicle-treated, mite extract-exposed cells. These results reveal that CFTR in keratinocytes plays a critical role for the regulation of peripheral nerve function and pruritus sensation, and suggest that Cftr(ΔF508/ΔF508) mice may serve as a novel mouse model that represents NGF-dependent generation of pruritus.
Collapse
Affiliation(s)
- Yasuaki Hashimoto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Global COE Cell Fate Regulation Research and Education Unit, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sampson H, Robert R, Liao J, Matthes E, Carlile G, Hanrahan J, Thomas D. Identification of a NBD1-Binding Pharmacological Chaperone that Corrects the Trafficking Defect of F508del-CFTR. ACTA ACUST UNITED AC 2011; 18:231-42. [DOI: 10.1016/j.chembiol.2010.11.016] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 11/09/2010] [Accepted: 11/29/2010] [Indexed: 11/28/2022]
|
48
|
Abstract
Animal models of human diseases are critical for dissecting mechanisms of pathophysiology and developing therapies. In the context of cystic fibrosis (CF), mouse models have been the dominant species by which to study CF disease processes in vivo for the past two decades. Although much has been learned through these CF mouse models, limitations in the ability of this species to recapitulate spontaneous lung disease and several other organ abnormalities seen in CF humans have created a need for additional species on which to study CF. To this end, pig and ferret CF models have been generated by somatic cell nuclear transfer and are currently being characterized. These new larger animal models have phenotypes that appear to closely resemble human CF disease seen in newborns, and efforts to characterize their adult phenotypes are ongoing. This chapter will review current knowledge about comparative lung cell biology and cystic fibrosis transmembrane conductance regulator (CFTR) biology among mice, pigs, and ferrets that has implications for CF disease modeling in these species. We will focus on methods used to compare the biology and function of CFTR between these species and their relevance to phenotypes seen in the animal models. These cross-species comparisons and the development of both the pig and the ferret CF models may help elucidate pathophysiologic mechanisms of CF lung disease and lead to new therapeutic approaches.
Collapse
Affiliation(s)
- John T Fisher
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| | | | | |
Collapse
|
49
|
Paradis J, Wilke M, Haston CK. Osteopenia in Cftr-deltaF508 mice. J Cyst Fibros 2010; 9:239-45. [DOI: 10.1016/j.jcf.2010.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 04/23/2010] [Accepted: 05/05/2010] [Indexed: 11/28/2022]
|
50
|
Freudenberg F, Leonard MR, Liu SA, Glickman JN, Carey MC. Pathophysiological preconditions promoting mixed "black" pigment plus cholesterol gallstones in a DeltaF508 mouse model of cystic fibrosis. Am J Physiol Gastrointest Liver Physiol 2010; 299:G205-14. [PMID: 20430874 PMCID: PMC2904121 DOI: 10.1152/ajpgi.00341.2009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gallstones are frequent in patients with cystic fibrosis (CF). These stones are generally "black" pigment (i.e., Ca bilirubinate) with an appreciable cholesterol admixture. The pathophysiology and molecular mechanisms for this "mixed" gallstone in CF are unknown. Here we investigate in a CF mouse model with no overt liver or gallbladder disease whether pathophysiological changes in the physical chemistry of gallbladder bile might predict the occurrence of "mixed" cholelithiasis. Employing a DeltaF508 mouse model with documented increased fecal bile acid loss and induced enterohepatic cycling of bilirubin (Am J Physiol Gastrointest Liver Physiol 294: G1411-G1420, 2008), we assessed gallbladder bile chemistry, morphology, and microscopy in CF and wild-type mice, with focus on the concentrations and compositions of the common biliary lipids, bilirubins, Ca(2+), and pH. Our results demonstrate that gallbladder bile of CF mice contains significantly higher levels of all bilirubin conjugates and unconjugated bilirubin with lower gallbladder bile pH values. Significant elevations in Ca bilirubinate ion products in bile of CF mice increase the likelihood of supersaturating bile and forming black pigment gallstones. The risk of potential pigment cholelithogenesis is coupled with higher cholesterol saturations and bile salt hydrophobicity indexes, consistent with a proclivity to cholesterol phase separation during pigment gallstone formation. This is an initial step toward unraveling the molecular basis of CF gallstone disease and constitutes a framework for investigating animal models of CF with more severe biliary disease, as well as the human disease.
Collapse
Affiliation(s)
- Folke Freudenberg
- 1Department of Medicine, Harvard Medical School and Harvard Digestive Diseases Center; ,2Department of Medicine, Gastroenterology Division, Brigham and Women's Hospital, and
| | - Monika R. Leonard
- 2Department of Medicine, Gastroenterology Division, Brigham and Women's Hospital, and
| | - Shou-An Liu
- 2Department of Medicine, Gastroenterology Division, Brigham and Women's Hospital, and
| | - Jonathan N. Glickman
- 3Pathology Department, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Martin C. Carey
- 1Department of Medicine, Harvard Medical School and Harvard Digestive Diseases Center; ,2Department of Medicine, Gastroenterology Division, Brigham and Women's Hospital, and
| |
Collapse
|