1
|
da Silva LVRF, Athanazio RA, Tonon CR, Ferreira JC, Tanni SE. Use of elexacaftor+tezacaftor+ivacaftor in individuals with cystic fibrosis and at least one F508del allele: a systematic review and meta-analysis. J Bras Pneumol 2024; 49:e20230187. [PMID: 38198345 PMCID: PMC10760416 DOI: 10.36416/1806-3756/e20230187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 08/11/2023] [Indexed: 01/12/2024] Open
Abstract
OBJECTIVE To evaluate the effect of treatment with the combination of three cystic fibrosis transmembrane conductance regulator (CFTR) modulators-elexacaftor+tezacaftor+ivacaftor (ETI)-on important clinical endpoints in individuals with cystic fibrosis. METHODS This was a systematic review and meta-analysis of randomized clinical trials that compared the use of ETI in individuals with CF and at least one F508del allele with that of placebo or with an active comparator such as other combinations of CFTR modulators, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) recommendations and the Patients of interest, Intervention to be studied, Comparison of interventions, and Outcome of interest (PICO) methodology. We searched the following databases: MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov from their inception to December 26th, 2022. The risk of bias was assessed using the Cochrane risk-of-bias tool, and the quality of evidence was based on the Grading of Recommendations Assessment, Development and Evaluation (GRADE). RESULTS We retrieved 54 studies in the primary search. Of these, 6 met the inclusion criteria and were analyzed (1,127 patients; 577 and 550 in the intervention and control groups, respectively). The meta-analysis revealed that the use of ETI increased FEV1% [risk difference (RD), +10.47%; 95% CI, 6.88-14.06], reduced the number of acute pulmonary exacerbations (RD, -0.16; 95% CI, -0.28 to -0.04), and improved quality of life (RD, +14.93; 95% CI, 9.98-19.89) and BMI (RD, +1.07 kg/m2; 95% CI, 0.90-1.25). Adverse events did not differ between groups (RD, -0.03; 95% CI, -0.08 to 0.01), and none of the studies reported deaths. CONCLUSIONS Our findings demonstrate that ETI treatment substantially improves clinically significant, patient-centered outcomes.
Collapse
Affiliation(s)
- Luiz Vicente Ribeiro Ferreira da Silva
- . Unidade de Pneumologia Pediátrica, Instituto da Criança e do Adolescente, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Rodrigo Abensur Athanazio
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | | | - Juliana Carvalho Ferreira
- . Divisão de Pneumologia, Instituto do Coração - InCor - Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Suzana Erico Tanni
- . Departamento de Clínica Médica, Faculdade de Medicina de Botucatu, Botucatu, (SP) Brasil
| |
Collapse
|
2
|
Haque MS, Emi Y, Sakaguchi M. A conserved WXXE motif is an apical delivery determinant of ABC transporter C subfamily isoforms. Cell Struct Funct 2023; 48:71-82. [PMID: 36696993 PMCID: PMC10721954 DOI: 10.1247/csf.22049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 01/25/2023] Open
Abstract
ATP-binding cassette transporter isoform C7 (ABCC7), also designated as cystic fibrosis transmembrane conductance regulator (CFTR), is exclusively targeted to the apical plasma membrane of polarized epithelial cells. Although the apical localization of ABCC7 in epithelia is crucial for the Cl- excretion into lumens, the mechanism regulating its apical localization is poorly understood. In the present study, an apical localization determinant was identified in the N-terminal 80-amino acid long cytoplasmic region of ABCC7 (NT80). In HepG2 cells, overexpression of NT80 significantly disturbed the apical expression of ABCC7 in a competitive manner, suggesting the presence of a sorting determinant in this region. Deletion analysis identified a potential sorting information within a 20-amino acid long peptide (aa 41-60) of NT80. Alanine scanning mutagenesis of this region in full-length ABCC7 further narrowed down the apical localization determinant to four amino acids, W57DRE60. This WDRE sequence was conserved among vertebrate ABCC7 orthologs. Site-directed mutagenesis showed that W57 and E60 were critical for the apical expression of ABCC7, confirming a novel apical sorting determinant of ABCC7. Furthermore, a WXXE motif (tryptophan and glutamic acid residues with two-amino acid spacing) was found to be conserved among the N-terminal regions of apically localized ABCC members with 12-TM configuration. The significance of the WXXE motif was demonstrated for proper trafficking of ABCC4 to the apical plasma membrane.Key words: apical plasma membrane, sorting, ATP-binding cassette transporter, CFTR, MRP4.
Collapse
Affiliation(s)
- Md Shajedul Haque
- Graduate School of Science, University of Hyogo, Harima Science Park City, Hyogo 678-1297, Japan
| | - Yoshikazu Emi
- Graduate School of Science, University of Hyogo, Harima Science Park City, Hyogo 678-1297, Japan
| | - Masao Sakaguchi
- Graduate School of Science, University of Hyogo, Harima Science Park City, Hyogo 678-1297, Japan
| |
Collapse
|
3
|
Shukla MK, Dubey A, Pandey S, Singh SK, Gupta G, Prasher P, Chellappan DK, Oliver BG, Kumar D, Dua K. Managing Apoptosis in Lung Diseases using Nano-assisted Drug Delivery System. Curr Pharm Des 2022; 28:3202-3211. [PMID: 35422206 DOI: 10.2174/1381612828666220413103831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/25/2022] [Indexed: 01/28/2023]
Abstract
Several factors exist that limit the efficacy of lung cancer treatment. These may be tumor-specific delivery of therapeutics, airway geometry, humidity, clearance mechanisms, presence of lung diseases, and therapy against tumor cell resistance. Advancements in drug delivery using nanotechnology based multifunctional nanocarriers, have emerged as a viable method for treating lung cancer with more efficacy and fewer adverse effects. This review does a thorough and critical examination of effective nano-enabled approaches for lung cancer treatment, such as nano-assisted drug delivery systems. In addition, to therapeutic effectiveness, researchers have been working to determine several strategies to produce nanotherapeutics by adjusting the size, drug loading, transport, and retention. Personalized lung tumor therapies using sophisticated nano modalities have the potential to provide great therapeutic advantages based on individual unique genetic markers and disease profiles. Overall, this review provides comprehensive information on newer nanotechnological prospects for improving the management of apoptosis in lung cancer.
Collapse
Affiliation(s)
- Monu K Shukla
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan-173229, Himachal Pradesh, India
| | - Amit Dubey
- Computational Chemistry and Drug Discovery Division, Quanta Calculus Pvt. Ltd., Kushinagar-274203, India.,Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Sciences, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Sachin K Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India.,Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Gaurav Gupta
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India.,School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, 302017, India.,Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Brian G Oliver
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, NSW 2037, Australia
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan-173229, Himachal Pradesh, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, NSW 2037, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
4
|
Splicing mutations in the CFTR gene as therapeutic targets. Gene Ther 2022; 29:399-406. [PMID: 35650428 PMCID: PMC9385490 DOI: 10.1038/s41434-022-00347-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 05/07/2022] [Accepted: 05/12/2022] [Indexed: 11/08/2022]
Abstract
The marketing approval, about ten years ago, of the first disease modulator for patients with cystic fibrosis harboring specific CFTR genotypes (~5% of all patients) brought new hope for their treatment. To date, several therapeutic strategies have been approved and the number of CFTR mutations targeted by therapeutic agents is increasing. Although these drugs do not reverse the existing disease, they help to increase the median life expectancy. However, on the basis of their CFTR genotype, ~10% of patients presently do not qualify for any of the currently available CFTR modulator therapies, particularly patients with splicing mutations (~12% of the reported CFTR mutations). Efforts are currently made to develop therapeutic agents that target disease-causing CFTR variants that affect splicing. This highlights the need to fully identify them by scanning non-coding regions and systematically determine their functional consequences. In this review, we present some examples of CFTR alterations that affect splicing events and the different therapeutic options that are currently developed and tested for splice switching.
Collapse
|
5
|
Oren YS, Avizur-Barchad O, Ozeri-Galai E, Elgrabli R, Schirelman MR, Blinder T, Stampfer CD, Ordan M, Laselva O, Cohen-Cymberknoh M, Kerem E, Bear CE, Kerem B. Antisense oligonucleotide splicing modulation as a novel Cystic Fibrosis therapeutic approach for the W1282X nonsense mutation. J Cyst Fibros 2021; 21:630-636. [PMID: 34972649 DOI: 10.1016/j.jcf.2021.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Antisense oligonucleotide- based drugs for splicing modulation were recently approved for various genetic diseases with unmet need. Here we aimed to generate skipping over exon 23 of the CFTR transcript, to eliminate the W1282X nonsense mutation and avoid RNA degradation induced by the nonsense mediated mRNA decay mechanism, allowing production of partially active CFTR proteins lacking exon 23. METHODS ∼80 ASOs were screened in 16HBEge W1282X cells. ASO candidates showing significant exon skipping were assessed for their W1282X allele selectivity and the increase of CFTR protein maturation and function. The effect of a highly potent ASO candidates was further analyzed in well differentiated primary human nasal epithelial cells, derived from a W1282X homozygous patient. RESULTS ASO screening led to identification of several ASOs that significantly decrease the level of CFTR transcripts including exon 23. These ASOs resulted in significant levels of mature CFTR protein and together with modulators restore the channel function following free uptake into these cells. Importantly, a highly potent lead ASOs, efficiently delivered by free uptake, was able to increase the level of transcripts lacking exon 23 and restore the CFTR function in cells from a W1282X homozygote patient. CONCLUSION The highly efficient exon 23 skipping induced by free uptake of the lead ASO and the resulting levels of mature CFTR protein exhibiting channel function in the presence of modulators, demonstrate the ASO therapeutic potential benefit for CF patients carrying the W1282X mutation with the objective to advance the lead candidate SPL23-2 to proof-of-concept clinical study.
Collapse
Affiliation(s)
- Yifat S Oren
- SpliSense Biohouse Labs, Hadassah Ein Kerem, Jerusalem, Israel
| | | | | | - Renana Elgrabli
- SpliSense Biohouse Labs, Hadassah Ein Kerem, Jerusalem, Israel
| | | | - Tehilla Blinder
- SpliSense Biohouse Labs, Hadassah Ein Kerem, Jerusalem, Israel
| | | | - Merav Ordan
- SpliSense Biohouse Labs, Hadassah Ein Kerem, Jerusalem, Israel
| | - Onofrio Laselva
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy; Division of Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Malena Cohen-Cymberknoh
- Pediatric Pulmonology Unit and CF Center, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Eitan Kerem
- CF Center, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Christine E Bear
- Division of Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Batsheva Kerem
- SpliSense Biohouse Labs, Hadassah Ein Kerem, Jerusalem, Israel; Department of Genetics, The Hebrew University, Jerusalem, Israel.
| |
Collapse
|
6
|
Liu M, Zaman R, Sawczak V, Periasamy A, Sun F, Zaman K. S-nitrosothiols signaling in cystic fibrosis airways. J Biosci 2021. [DOI: 10.1007/s12038-021-00223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
7
|
Abstract
Pathogenic variants of the CFTR gene are responsible for a broad phenotypic spectrum characterized by malfunction of some exocrine tissues, with an autosomal recessive mode of inheritance. More than 2,000 variants, distributed throughout the CFTR gene, have been identified, with different effects on the gene and protein expression and function. Genotype-phenotype correlation studies have associated severe variants with a typical multi-organ form of cystic fibrosis, while mild variants are involved in monosymptomatic or adult-onset diseases, called CFTR-related disorders. However, the interpretation of rare variants remains challenging. This review presents an overview of the epidemiology of CFTR variants worldwide and in France and describes the functional classification. Finally, some frequent cystic fibrosis-causing and mild CFTR variants are used as example to depict the molecular pathology of the CFTR locus. Finally, we give the recommendations concerning nomenclature and classification that are useful for appropriate genetic counseling. © 2020 French Society of Pediatrics. Published by Elsevier Masson SAS. All rights reserved.
Collapse
Affiliation(s)
- C Bareil
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Montpellier, France
| | - A Bergougnoux
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Montpellier, France; EA7402 Laboratoire de Génétique de Maladies Rares, Institut Universitaire de Recherche Clinique, Université de Montpellier, Montpellier, France..
| |
Collapse
|
8
|
Zhang YP, Ye LL, Yuan H, Duan DD. CFTR plays an important role in the regulation of vascular resistance and high-fructose/salt-diet induced hypertension in mice. J Cyst Fibros 2020; 20:516-524. [PMID: 33279469 DOI: 10.1016/j.jcf.2020.11.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND The pathophysiological roles of cystic fibrosis transmembrane-conductance regulator (CFTR) Cl- channels in the regulation of blood pressure (BP) remain controversial. Here we studied the function of CFTR Cl- channels in regulation of BP and in the high-fructose-salt-diet (HFSD) induced hypertension in mice. METHODS The systolic, diastolic and mean BP (SBP, DBP and MBP, respectively) were continuously monitored from unrestricted conscious wild-type (cftr+/+) FVB and CFTR-knockout (cftr-/-) mice (8-week old, male). HFSD (64.7% fructose, 2% NaCl water) or control normal starch diet (CNSD, 58.9% corn starch, 0 NaCl water) was given for 8 weeks and vascular Doppler were performed. Real-time PCR and Western blot were used to examine mRNA and protein expression, respectively. RESULTS The aortic stiffness, daytime and nighttime SBP, DBP, and MBP of the cftr-/- mice were significantly higher than those in the age- and gender-matched cftr+/+ mice, which is consistent with the findings of increased vascular resistance in cystic fibrosis patients. The aortic stiffness, daytime and nighttime SBP, DBP, and MBP of cftr+/+ mice fed with HFSD were all significantly higher than those fed with CNSD. Importantly, HFSD caused a significant decrease in mRNA and protein expression of WINK1, WINK4 and CFTR in aorta and mesenteric arteries, but not in the kidney, corroborating that HSFD-induced downregulation of WINKs and loss of CFTR function specifically in the arteries may mediate the increased BP. CONCLUSIONS CFTR regulates peripheral arterial resistance and BP in vivo. HFSD-induced CFTR downregulation specifically in the arteries may be a novel mechanism for hypertension.
Collapse
Affiliation(s)
- Ya-Ping Zhang
- The Heart Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Lingyu Linda Ye
- Center for Phenomics of Traditional Chinese Medicine, The Affiliated Hospital (Traditional Chinese Medicine) to Southwest Medical University, Luzhou, Sichuan 646000, China; Institute of Cardiovascular Research, Education Ministry Key Laboratory of Electrophysiology, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hong Yuan
- Center of Clinical Pharmacology, the Third Xiang-Ya Hospital, Central South University, Changsha 410013, China
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine, The Affiliated Hospital (Traditional Chinese Medicine) to Southwest Medical University, Luzhou, Sichuan 646000, China; Institute of Cardiovascular Research, Education Ministry Key Laboratory of Electrophysiology, Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, NV 89557, USA.
| |
Collapse
|
9
|
Loureiro CA, Pinto FR, Barros P, Matos P, Jordan P. A SYK/SHC1 pathway regulates the amount of CFTR in the plasma membrane. Cell Mol Life Sci 2020; 77:4997-5015. [PMID: 31974654 PMCID: PMC11105000 DOI: 10.1007/s00018-020-03448-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/06/2019] [Accepted: 01/02/2020] [Indexed: 11/24/2022]
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause the recessive genetic disease cystic fibrosis, where the chloride transport across the apical membrane of epithelial cells mediated by the CFTR protein is impaired. CFTR protein trafficking to the plasma membrane (PM) is the result of a complex interplay between the secretory and membrane recycling pathways that control the number of channels present at the membrane. In addition, the ion transport activity of CFTR at the PM is modulated through post-translational protein modifications. Previously we described that spleen tyrosine kinase (SYK) phosphorylates a specific tyrosine residue in the nucleotide-binding domain 1 domain and this modification can regulate the PM abundance of CFTR. Here we identified the underlying biochemical mechanism using peptide pull-down assays followed by mass spectrometry. We identified in bronchial epithelial cells that the adaptor protein SHC1 recognizes tyrosine-phosphorylated CFTR through its phosphotyrosine-binding domain and that the formation of a complex between SHC1 and CFTR is induced at the PM in the presence of activated SYK. The depletion of endogenous SHC1 expression was sufficient to promote an increase in CFTR at the PM of these cells. The results identify a SYK/SHC1 pathway that regulates the PM levels of CFTR channels, contributing to a better understanding of how CFTR-mediated chloride secretion is regulated.
Collapse
Affiliation(s)
- Cláudia Almeida Loureiro
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Francisco R Pinto
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Patrícia Barros
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Paulo Matos
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Peter Jordan
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal.
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
10
|
Zabana Y, Lorén V, Domènech E, Aterido A, Garcia-Jaraquemada A, Julià A, Vicario M, Pedrosa E, Ferreiro M, Troya J, Lozano JJ, Sarrias MR, Cabré E, Mañosa M, Manyé J. Transcriptomic identification of TMIGD1 and its relationship with the ileal epithelial cell differentiation in Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2020; 319:G109-G120. [PMID: 32508154 DOI: 10.1152/ajpgi.00027.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Crohn's disease (CD) is a complex and multifactorial illness. There are still considerable gaps in our knowledge regarding its pathophysiology. A transcriptomic approach could shed some light on little-known biological alterations of the disease. We therefore aimed to explore the ileal transcriptome to gain knowledge about CD. We performed whole transcriptome gene expression analysis on ileocecal resections from CD patients and inflammatory bowel disease-free controls, as well as on a CD-independent cohort to replicate selected results. Normalized data were hierarchically clustered, and gene ontology and the molecular network were studied. Cell cultures and molecular methods were used for further evaluations. Genome-wide expression data analysis identified a robust transmembrane immunoglobulin domain-containing 1 (TMIGD1) gene underexpression in CD tissue, which was even more marked in inflamed ileum, and which was replicated in the validation cohort. Immunofluorescence showed TMIGD1 to be located in the apical microvilli of well-differentiated enterocytes but not in intestinal crypt. This apical TMIGD1 was lower in the noninflamed tissue and almost disappeared in the inflamed mucosa of surgical resections. In vitro studies showed hypoxic-dependent TMIGD1 decreased its expression in enterocyte-like cells. The gene enrichment analysis linked TMIGD1 with cell recovery and tissue remodeling in CD settings, involving guanylate cyclase activities. Transcriptomics may be useful for finding new targets that facilitate studies of the CD pathology. This is how TMIGD1 was identified in CD patients, which was related to multiciliate ileal epithelial cell differentiation.NEW & NOTEWORTHY This is a single-center translational research study that aimed to look for key targets involved in Crohn's disease and define molecular pathways through different functional analysis strategies. With this approach, we have identified and described a novel target, the almost unknown TMIGD1 gene, which may be key in the recovery of injured mucosa involving intestinal epithelial cell differentiation.
Collapse
Affiliation(s)
- Yamile Zabana
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Violeta Lorén
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Eugeni Domènech
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Gastroenterology Department, Germans Trias i Pujol University Hospital, Badalona, Catalonia, Spain
| | - Adrià Aterido
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Catalonia, Spain.,Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Arce Garcia-Jaraquemada
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Antonio Julià
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Catalonia, Spain
| | - Maria Vicario
- Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Laboratory of Translational Mucosal Immunology & Department of Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Research University Hospital, Badalona, Catalonia, Spain
| | - Elisabet Pedrosa
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Miriam Ferreiro
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - José Troya
- Colorectal Surgery Unit, General and Digestive Surgery Department, Germans Trias i Pujol University Hospital, Badalona, Catalonia, Spain
| | - Juan J Lozano
- Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Maria R Sarrias
- Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Innate Immunity Group, IGTP (AGAUR 2017-SGR-490 group), Badalona, Catalonia, Spain
| | - Eduard Cabré
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Gastroenterology Department, Germans Trias i Pujol University Hospital, Badalona, Catalonia, Spain
| | - Miriam Mañosa
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Gastroenterology Department, Germans Trias i Pujol University Hospital, Badalona, Catalonia, Spain
| | - Josep Manyé
- IBD Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| |
Collapse
|
11
|
McCarron A, Cmielewski P, Reyne N, McIntyre C, Finnie J, Craig F, Rout-Pitt N, Delhove J, Schjenken JE, Chan HY, Boog B, Knight E, Gilmore RC, O'Neal WK, Boucher RC, Parsons D, Donnelley M. Phenotypic Characterization and Comparison of Cystic Fibrosis Rat Models Generated Using CRISPR/Cas9 Gene Editing. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:977-993. [PMID: 32084371 DOI: 10.1016/j.ajpath.2020.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/20/2019] [Accepted: 01/21/2020] [Indexed: 01/10/2023]
Abstract
Animal models of cystic fibrosis (CF) are essential for investigating disease mechanisms and trialing potential therapeutics. This study generated two CF rat models using clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated protein 9 gene editing. One rat model carries the common human Phe508del (ΔF508) CF transmembrane conductance regulator (CFTR) mutation, whereas the second is a CFTR knockout model. Phenotype was characterized using a range of functional and histologic assessments, including nasal potential difference to measure electrophysiological function in the upper airways, RNAscope in situ hybridization and quantitative PCR to assess CFTR mRNA expression in the lungs, immunohistochemistry to localize CFTR protein in the airways, and histopathologic assessments in a range of tissues. Both rat models revealed a range of CF manifestations, including reduced survival, intestinal obstruction, bioelectric defects in the nasal epithelium, histopathologic changes in the trachea, large intestine, and pancreas, and abnormalities in the development of the male reproductive tract. The CF rat models presented herein will prove useful for longitudinal assessments of pathophysiology and therapeutics.
Collapse
Affiliation(s)
- Alexandra McCarron
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.
| | - Patricia Cmielewski
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicole Reyne
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Chantelle McIntyre
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - John Finnie
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Division of Anatomical Pathology, SA Pathology, Adelaide, South Australia, Australia
| | - Fiona Craig
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Nathan Rout-Pitt
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Juliette Delhove
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - John E Schjenken
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Hon Y Chan
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Bernadette Boog
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Emma Knight
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Rodney C Gilmore
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Wanda K O'Neal
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Richard C Boucher
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David Parsons
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Martin Donnelley
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
12
|
Anglès F, Hutt DM, Balch WE. HDAC inhibitors rescue multiple disease-causing CFTR variants. Hum Mol Genet 2020; 28:1982-2000. [PMID: 30753450 DOI: 10.1093/hmg/ddz026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/21/2018] [Accepted: 01/16/2019] [Indexed: 12/14/2022] Open
Abstract
Understanding the role of the epigenome in protein-misfolding diseases remains a challenge in light of genetic diversity found in the world-wide population revealed by human genome sequencing efforts and the highly variable response of the disease population to therapeutics. An ever-growing body of evidence has shown that histone deacetylase (HDAC) inhibitors (HDACi) can have significant benefit in correcting protein-misfolding diseases that occur in response to both familial and somatic mutation. Cystic fibrosis (CF) is a familial autosomal recessive disease, caused by genetic diversity in the CF transmembrane conductance regulator (CFTR) gene, a cyclic Adenosine MonoPhosphate (cAMP)-dependent chloride channel expressed at the apical plasma membrane of epithelial cells in multiple tissues. The potential utility of HDACi in correcting the phenylalanine 508 deletion (F508del) CFTR variant as well as the over 2000 CF-associated variants remains controversial. To address this concern, we examined the impact of US Food and Drug Administration-approved HDACi on the trafficking and function of a panel of CFTR variants. Our data reveal that panobinostat (LBH-589) and romidepsin (FK-228) provide functional correction of Class II and III CFTR variants, restoring cell surface chloride channel activity in primary human bronchial epithelial cells. We further demonstrate a synergistic effect of these HDACi with Vx809, which can significantly restore channel activity for multiple CFTR variants. These data suggest that HDACi can serve to level the cellular playing field for correcting CF-causing mutations, a leveling effect that might also extend to other protein-misfolding diseases.
Collapse
Affiliation(s)
- Frédéric Anglès
- Department of Molecular Medicine, Scripps Research, North Torrey Pines Rd, La Jolla, CA, USA
| | - Darren M Hutt
- Department of Molecular Medicine, Scripps Research, North Torrey Pines Rd, La Jolla, CA, USA
| | - William E Balch
- Department of Molecular Medicine, Scripps Research, North Torrey Pines Rd, La Jolla, CA, USA.,Skaggs Institute of Chemical Biology, North Torrey Pines Rd, La Jolla, CA, USA
| |
Collapse
|
13
|
Kim Chiaw P, Hantouche C, Wong MJH, Matthes E, Robert R, Hanrahan JW, Shrier A, Young JC. Hsp70 and DNAJA2 limit CFTR levels through degradation. PLoS One 2019; 14:e0220984. [PMID: 31408507 PMCID: PMC6692068 DOI: 10.1371/journal.pone.0220984] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/26/2019] [Indexed: 11/18/2022] Open
Abstract
Cystic Fibrosis is caused by mutations in the CFTR anion channel, many of which cause its misfolding and degradation. CFTR folding depends on the Hsc70 and Hsp70 chaperones and their co-chaperone DNAJA1, but Hsc70/Hsp70 is also involved in CFTR degradation. Here, we address how these opposing functions are balanced. DNAJA2 and DNAJA1 were both important for CFTR folding, however overexpressing DNAJA2 but not DNAJA1 enhanced CFTR degradation at the endoplasmic reticulum by Hsc70/Hsp70 and the E3 ubiquitin ligase CHIP. Excess Hsp70 also promoted CFTR degradation, but this occurred through the lysosomal pathway and required CHIP but not complex formation with HOP and Hsp90. Notably, the Hsp70 inhibitor MKT077 enhanced levels of mature CFTR and the most common disease variant ΔF508-CFTR, by slowing turnover and allowing delayed maturation, respectively. MKT077 also boosted the channel activity of ΔF508-CFTR when combined with the corrector compound VX809. Thus, the Hsp70 system is the major determinant of CFTR degradation, and its modulation can partially relieve the misfolding phenotype.
Collapse
Affiliation(s)
- Patrick Kim Chiaw
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
| | - Christine Hantouche
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Michael J. H. Wong
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
| | - Elizabeth Matthes
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Renaud Robert
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - John W. Hanrahan
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Alvin Shrier
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Jason C. Young
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Groupe de Recherche Axé sur la Structure des Protéines, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
14
|
Loureiro CA, Santos JD, Matos AM, Jordan P, Matos P, Farinha CM, Pinto FR. Network Biology Identifies Novel Regulators of CFTR Trafficking and Membrane Stability. Front Pharmacol 2019; 10:619. [PMID: 31231217 PMCID: PMC6559121 DOI: 10.3389/fphar.2019.00619] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022] Open
Abstract
In cystic fibrosis, the most common disease-causing mutation is F508del, which causes not only intracellular retention and degradation of CFTR, but also defective channel gating and decreased membrane stability of the small amount that reaches the plasma membrane (PM). Thus, pharmacological correction of mutant CFTR requires targeting of multiple cellular defects in order to achieve clinical benefit. Although small-molecule compounds have been identified and commercialized that can correct its folding or gating, an efficient retention of F508del CFTR at the PM has not yet been explored pharmacologically despite being recognized as a crucial factor for improving functional rescue of chloride transport. In ongoing efforts to determine the CFTR interactome at the PM, we used three complementary approaches: targeting proteins binding to tyrosine-phosphorylated CFTR, protein complexes involved in cAMP-mediated CFTR stabilization at the PM, and proteins selectively interacting at the PM with rescued F508del-CFTR but not wt-CFTR. Using co-immunoprecipitation or peptide–pull down strategies, we identified around 400 candidate proteins through sequencing of complex protein mixtures using the nano-LC Triple TOF MS technique. Key candidate proteins were validated for their robust interaction with CFTR-containing protein complexes and for their ability to modulate the amount of CFTR expressed at the cell surface of bronchial epithelial cells. Here, we describe how we explored the abovementioned experimental datasets to build a protein interaction network with the aim of identifying novel pharmacological targets to rescue CFTR function in cystic fibrosis (CF) patients. We identified and validated novel candidate proteins that were essential components of the network but not detected in previous proteomic analyses.
Collapse
Affiliation(s)
- Cláudia Almeida Loureiro
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.,Department of Human Genetics, National Health Institute "Dr. Ricardo Jorge," Lisbon, Portugal
| | - João D Santos
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.,Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Ana Margarida Matos
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.,Department of Human Genetics, National Health Institute "Dr. Ricardo Jorge," Lisbon, Portugal
| | - Peter Jordan
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.,Department of Human Genetics, National Health Institute "Dr. Ricardo Jorge," Lisbon, Portugal
| | - Paulo Matos
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.,Department of Human Genetics, National Health Institute "Dr. Ricardo Jorge," Lisbon, Portugal
| | - Carlos M Farinha
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.,Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Francisco R Pinto
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.,Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
15
|
Kerschner JL, Ghosh S, Paranjapye A, Cosme WR, Audrézet MP, Nakakuki M, Ishiguro H, Férec C, Rommens J, Harris A. Screening for Regulatory Variants in 460 kb Encompassing the CFTR Locus in Cystic Fibrosis Patients. J Mol Diagn 2018; 21:70-80. [PMID: 30296588 DOI: 10.1016/j.jmoldx.2018.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 07/18/2018] [Accepted: 08/10/2018] [Indexed: 12/30/2022] Open
Abstract
It is estimated that up to 5% of cystic fibrosis transmembrane conductance regulator (CFTR) pathogenic alleles are unidentified. Some of these errors may lie in noncoding regions of the locus and affect gene expression. To identify regulatory element variants in the CFTR locus, SureSelect targeted enrichment of 460 kb encompassing the gene was optimized to deep sequence genomic DNA from 80 CF patients with an unequivocal clinical diagnosis but only one or no CFTR-coding region pathogenic variants. Bioinformatics tools were used to identify sequence variants and predict their impact, which were then assayed in transient reporter gene luciferase assays. The effect of five variants in the CFTR promoter and four in an intestinal enhancer of the gene were assayed in relevant cell lines. The initial analysis of sequence data revealed previously known CF-causing variants, validating the robustness of the SureSelect design, and showed that 85 of 160 CF alleles were undefined. Of a total 1737 variants revealed across the extended 460-kb CFTR locus, 51 map to known CFTR cis-regulatory elements, and many of these are predicted to alter transcription factor occupancy. Four promoter variants and all those in the intestinal enhancer significantly repress reporter gene activity. These data suggest that CFTR regulatory elements may harbor novel CF disease-causing variants that warrant further investigation, both for genetic screening protocols and functional assays.
Collapse
Affiliation(s)
- Jenny L Kerschner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Sujana Ghosh
- Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, Illinois; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Wilmel R Cosme
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | | | - Miyuki Nakakuki
- Department of Human Nutrition, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Ishiguro
- Department of Human Nutrition, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Johanna Rommens
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio; Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, Illinois; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
16
|
Hegde RN, Subramanian A, Pothukuchi P, Parashuraman S, Luini A. Rare ER protein misfolding-mistrafficking disorders: Therapeutic developments. Tissue Cell 2017; 49:175-185. [PMID: 28222887 DOI: 10.1016/j.tice.2017.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 02/02/2017] [Accepted: 02/04/2017] [Indexed: 12/16/2022]
Abstract
The presence of a functional protein at the appropriate location in the cell is the result of the processes of transcription, translation, folding and trafficking to the correct destination. There are numerous diseases that are caused by protein misfolding, mainly due to mutations in the respective gene. The consequences of this misfolding may be that proteins effectively lose their function, either by being removed by the cellular quality control machinery or by accumulating at the incorrect intracellular or extracellular location. A number of mutations that lead to protein misfolding and affect trafficking to the final destination, e.g. Cystic fibrosis, Wilson's disease, and Progressive Familial Intrahepatic 1 cholestasis, result in proteins that retain partial function if their folding and trafficking is restored either by molecular or pharmacological means. In this review, we discuss several mutant proteins within this class of misfolding diseases and provide an update on the status of molecular and therapeutic developments and potential therapeutic strategies being developed to counter these diseases.
Collapse
Affiliation(s)
| | - Advait Subramanian
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | | | | | - Alberto Luini
- Institute of Protein Biochemistry, National Research Council, Naples, Italy; Istituto di Ricovero e Cura a Carattere Scientifico SDN, Naples, Italy
| |
Collapse
|
17
|
Farinha CM, Canato S. From the endoplasmic reticulum to the plasma membrane: mechanisms of CFTR folding and trafficking. Cell Mol Life Sci 2017; 74:39-55. [PMID: 27699454 PMCID: PMC11107782 DOI: 10.1007/s00018-016-2387-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 01/10/2023]
Abstract
CFTR biogenesis starts with its co-translational insertion into the membrane of endoplasmic reticulum and folding of the cytosolic domains, towards the acquisition of a fully folded compact native structure. Efficiency of this process is assessed by the ER quality control system that allows the exit of folded proteins but targets unfolded/misfolded CFTR to degradation. If allowed to leave the ER, CFTR is modified at the Golgi and reaches the post-Golgi compartments to be delivered to the plasma membrane where it functions as a cAMP- and phosphorylation-regulated chloride/bicarbonate channel. CFTR residence at the membrane is a balance of membrane delivery, endocytosis, and recycling. Several adaptors, motor, and scaffold proteins contribute to the regulation of CFTR stability and are involved in continuously assessing its structure through peripheral quality control systems. Regulation of CFTR biogenesis and traffic (and its dysregulation by mutations, such as the most common F508del) determine its overall activity and thus contribute to the fine modulation of chloride secretion and hydration of epithelial surfaces. This review covers old and recent knowledge on CFTR folding and trafficking from its synthesis to the regulation of its stability at the plasma membrane and highlights how several of these steps can be modulated to promote the rescue of mutant CFTR.
Collapse
Affiliation(s)
- Carlos M Farinha
- BioISI-Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal.
| | - Sara Canato
- BioISI-Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal
| |
Collapse
|
18
|
Farinha CM, Swiatecka-Urban A, Brautigan DL, Jordan P. Regulatory Crosstalk by Protein Kinases on CFTR Trafficking and Activity. Front Chem 2016; 4:1. [PMID: 26835446 PMCID: PMC4718993 DOI: 10.3389/fchem.2016.00001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/04/2016] [Indexed: 12/12/2022] Open
Abstract
Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) is a member of the ATP binding cassette (ABC) transporter superfamily that functions as a cAMP-activated chloride ion channel in fluid-transporting epithelia. There is abundant evidence that CFTR activity (i.e., channel opening and closing) is regulated by protein kinases and phosphatases via phosphorylation and dephosphorylation. Here, we review recent evidence for the role of protein kinases in regulation of CFTR delivery to and retention in the plasma membrane. We review this information in a broader context of regulation of other transporters by protein kinases because the overall functional output of transporters involves the integrated control of both their number at the plasma membrane and their specific activity. While many details of the regulation of intracellular distribution of CFTR and other transporters remain to be elucidated, we hope that this review will motivate research providing new insights into how protein kinases control membrane transport to impact health and disease.
Collapse
Affiliation(s)
- Carlos M Farinha
- Faculty of Sciences, Biosystems and Integrative Sciences Institute, University of Lisboa Lisbon, Portugal
| | - Agnieszka Swiatecka-Urban
- Department of Cell Biology, University of Pittsburgh School of MedicinePittsburgh, PA, USA; Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of MedicinePittsburgh, PA, USA
| | - David L Brautigan
- Center for Cell Signaling and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine Charlottesville, VA, USA
| | - Peter Jordan
- Faculty of Sciences, Biosystems and Integrative Sciences Institute, University of LisboaLisbon, Portugal; Department of Human Genetics, National Health Institute Dr Ricardo JorgeLisbon, Portugal
| |
Collapse
|
19
|
Taylor-Cousar J, Niknian M, Gilmartin G, Pilewski JM. Effect of ivacaftor in patients with advanced cystic fibrosis and a G551D-CFTR mutation: Safety and efficacy in an expanded access program in the United States. J Cyst Fibros 2016; 15:116-22. [DOI: 10.1016/j.jcf.2015.01.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 10/24/2022]
|
20
|
Hegde RN, Parashuraman S, Iorio F, Ciciriello F, Capuani F, Carissimo A, Carrella D, Belcastro V, Subramanian A, Bounti L, Persico M, Carlile G, Galietta L, Thomas DY, Di Bernardo D, Luini A. Unravelling druggable signalling networks that control F508del-CFTR proteostasis. eLife 2015; 4. [PMID: 26701908 PMCID: PMC4749566 DOI: 10.7554/elife.10365] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 11/26/2015] [Indexed: 01/17/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in CF transmembrane conductance regulator (CFTR). The most frequent mutation (F508del-CFTR) results in altered proteostasis, that is, in the misfolding and intracellular degradation of the protein. The F508del-CFTR proteostasis machinery and its homeostatic regulation are well studied, while the question whether ‘classical’ signalling pathways and phosphorylation cascades might control proteostasis remains barely explored. Here, we have unravelled signalling cascades acting selectively on the F508del-CFTR folding-trafficking defects by analysing the mechanisms of action of F508del-CFTR proteostasis regulator drugs through an approach based on transcriptional profiling followed by deconvolution of their gene signatures. Targeting multiple components of these signalling pathways resulted in potent and specific correction of F508del-CFTR proteostasis and in synergy with pharmacochaperones. These results provide new insights into the physiology of cellular proteostasis and a rational basis for developing effective pharmacological correctors of the F508del-CFTR defect. DOI:http://dx.doi.org/10.7554/eLife.10365.001 Cystic fibrosis is a genetic disease that commonly affects people of European descent. The condition is caused by mutations in the gene encoding a protein called “cystic fibrosis transmembrane conductance regulator” (or CFTR for short). CFTR forms a channel in the membrane of cells in the lungs that help transport salt across the membrane. Mutated versions of the protein are not as efficient at transporting salts, and eventually this damages the lung tissue. As the damage progresses, individuals become very vulnerable to bacterial infections that further damage the lungs and may eventually lead to death. One of the reasons CFTR mutations are harmful is that they cause the protein to fold up incorrectly and remain trapped inside the cell. Cells have quality control systems that recognize and destroy poorly folded proteins, and so only a few of the mutated CFTR proteins ever make it to the membrane to move salts. New therapies have been developed that improve folding of the protein and/or help the CFTR proteins that make it to the membrane work better. But more and better treatment options are needed. Hegde, Parashuraman et al. have now tested drugs that control how proteins fold and move to the membrane to see how they affect gene expression in cells with the most common cystic fibrosis-causing mutation. These drugs are known to improve the activity of the CFTR mutant, but do so too weakly to be of clinical interest. The experiments revealed that the expression of a few hundred genes was changed in response the drugs. Many of these genes were involved in major signalling pathways that control how CFTR is folded and trafficked within cells. Next, Hegde, Parashuraman et al. tested drugs that inhibit these signalling pathways to see if they improve salt handling in the mutated cells. The experiments demonstrated that these inhibitor drugs efficiently block the breakdown of misfolded CFTR, or boost the likelihood of CFTR making it to the membrane, helping improve salt trafficking in the cells. The inhibitors produced even better results when used in combination with a known CFTR-protecting drug. The results suggest that identifying and targeting signalling pathways involved in the folding, trafficking, and breakdown of CFTR may prove a promising way to treat cystic fibrosis. DOI:http://dx.doi.org/10.7554/eLife.10365.002
Collapse
Affiliation(s)
- Ramanath Narayana Hegde
- Institute of Protein Biochemistry, National Research Council, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Seetharaman Parashuraman
- Institute of Protein Biochemistry, National Research Council, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Francesco Iorio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Fabiana Ciciriello
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Biology and Biotechnology Department "Charles Darwin", Sapienza University, Rome, Italy.,Department of Biochemistry, McIntyre Medical Sciences Building, McGill University, Montréal, Canada
| | | | | | - Diego Carrella
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | - Advait Subramanian
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Laura Bounti
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Maria Persico
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Graeme Carlile
- Department of Biochemistry, McIntyre Medical Sciences Building, McGill University, Montréal, Canada
| | - Luis Galietta
- U.O.C. Genetica Medica, Institute of Giannina Gaslini, Genova, Italy
| | - David Y Thomas
- Department of Biochemistry, McIntyre Medical Sciences Building, McGill University, Montréal, Canada
| | - Diego Di Bernardo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | - Alberto Luini
- Institute of Protein Biochemistry, National Research Council, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico SDN, Naples, Italy
| |
Collapse
|
21
|
Comparative ex vivo, in vitro and in silico analyses of a CFTR splicing mutation: Importance of functional studies to establish disease liability of mutations. J Cyst Fibros 2015; 15:21-33. [PMID: 25735457 DOI: 10.1016/j.jcf.2015.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/09/2015] [Accepted: 02/04/2015] [Indexed: 11/23/2022]
Abstract
The Cystic Fibrosis p.Ile1234Val missense mutation actually creates a new dual splicing site possibly used either as a new acceptor or donor. Here, we aimed to test the accuracy of in silico predictions by comparing them with in vitro and ex vivo functional analyses of this mutation for an accurate CF diagnosis/prognosis. To this end, we applied a new in vitro strategy using a CFTR mini-gene which includes the complete CFTR coding sequence plus intron 22 (short version) which allows the assessment of alternatively spliced mRNA levels as well as the properties of the resulting abnormal CFTR protein regarding processing, intracellular localization and function. Our data demonstrate that p.Ile1234Val leads to usage of the alternative splicing donor (but not acceptor) resulting in alternative CFTR transcripts lacking 18 nts of exon 22 which produce a truncated CFTR protein with residual Cl- channel function. These results recapitulate data from native tissues of a CF patient. In conclusion, the existing in silico prediction models have limited application and ex vivo functional assessment of mutation effects should be made. Alternatively the in vitro strategy adopted here can be applied to assess the disease liability of mutations for an accurate CF diagnosis/prognosis.
Collapse
|
22
|
Amaral MD. Novel personalized therapies for cystic fibrosis: treating the basic defect in all patients. J Intern Med 2015; 277:155-166. [PMID: 25266997 DOI: 10.1111/joim.12314] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cystic fibrosis (CF) is the most common genetic life-shortening condition in Caucasians. Despite being a multi-organ disease, CF is classically diagnosed by symptoms of acute/chronic respiratory disease, with persistent pulmonary infections and mucus plugging of the airways and failure to thrive. These multiple symptoms originate from dysfunction of the CF transmembrane conductance regulator (CFTR) protein, a channel that mediates anion transport across epithelia. Indeed, establishment of a definite CF diagnosis requires proof of CFTR dysfunction, commonly through the so-called sweat Cl(-) test. Many drug therapies, including mucolytics and antibiotics, aim to alleviate the symptoms of CF lung disease. However, new therapies to modulate defective CFTR, the basic defect underlying CF, have started to reach the clinic, and several others are in development or in clinical trials. The novelty of these therapies is that, besides targeting the basic defect underlying CF, they are mutation specific. Indeed, even this monogenic disease is influenced by a large number of different genes and biological pathways as well as by environmental factors that are difficult to assess. Accordingly, every person with CF is unique and so functional assessment of patients' tissues ex vivo is key for diagnosing and predicting the severity of this disease. Of note, such assessment will also be crucial to assess drug responses, in order to effectively treat all CF patients. It is not because it is a monogenic disorder that personalized treatment for CF is much easier than for complex disorders.
Collapse
Affiliation(s)
- M D Amaral
- BioFIG-Center for Biodiversity, Functional and Integrative Genomics, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| |
Collapse
|
23
|
Abstract
The availability of the human genome sequence and tools for interrogating individual genomes provide an unprecedented opportunity to apply genetics to medicine. Mendelian conditions, which are caused by dysfunction of a single gene, offer powerful examples that illustrate how genetics can provide insights into disease. Cystic fibrosis, one of the more common lethal autosomal recessive Mendelian disorders, is presented here as an example. Recent progress in elucidating disease mechanism and causes of phenotypic variation, as well as in the development of treatments, demonstrates that genetics continues to play an important part in cystic fibrosis research 25 years after the discovery of the disease-causing gene.
Collapse
|
24
|
Boyle MP, Bell SC, Konstan MW, McColley SA, Rowe SM, Rietschel E, Huang X, Waltz D, Patel NR, Rodman D. A CFTR corrector (lumacaftor) and a CFTR potentiator (ivacaftor) for treatment of patients with cystic fibrosis who have a phe508del CFTR mutation: a phase 2 randomised controlled trial. THE LANCET. RESPIRATORY MEDICINE 2014; 2:527-38. [PMID: 24973281 DOI: 10.1016/s2213-2600(14)70132-8] [Citation(s) in RCA: 340] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The phe508del CFTR mutation causes cystic fibrosis by limiting the amount of CFTR protein that reaches the epithelial cell surface. We tested combination treatment with lumacaftor, an investigational CFTR corrector that increases trafficking of phe508del CFTR to the cell surface, and ivacaftor, a CFTR potentiator that enhances chloride transport of CFTR on the cell surface. METHODS In this phase 2 clinical trial, we assessed three successive cohorts, with the results of each cohort informing dose selection for the subsequent cohort. We recruited patients from 24 cystic fibrosis centres in Australia, Belgium, Germany, New Zealand, and the USA. Eligibility criteria were: confirmed diagnosis of cystic fibrosis, age at least 18 years, and a forced expiratory volume in 1 s (FEV1) of 40% or more than predicted. Cohort 1 included phe508del CFTR homozygous patients randomly assigned to either lumacaftor 200 mg once per day for 14 days followed by addition of ivacaftor 150 mg or 250 mg every 12 h for 7 days, or 21 days of placebo. Together, cohorts 2 and 3 included phe508del CFTR homozygous and heterozygous patients, randomly assigned to either 56 days of lumacaftor (cohort 2: 200 mg, 400 mg, or 600 mg once per day, cohort 3: 400 mg every 12 h) with ivacaftor 250 mg every 12 h added after 28 days, or 56 days of placebo. The primary outcomes for all cohorts were change in sweat chloride concentration during the combination treatment period in the intention-to-treat population and safety (laboratory measurements and adverse events). The study is registered with ClinicalTrials.gov, number NCT01225211, and EudraCT, number 2010-020413-90. FINDINGS Cohort 1 included 64 participants. Cohort 2 and 3 combined contained 96 phe508del CFTR homozygous patients and 28 compound heterozygotes. Treatment with lumacaftor 200 mg once daily and ivacaftor 250 mg every 12 h decreased mean sweat chloride concentration by 9.1 mmol/L (p<0.001) during the combination treatment period in cohort 1. In cohorts 2 and 3, mean sweat chloride concentration did not decrease significantly during combination treatment in any group. Frequency and nature of adverse events were much the same in the treatment and placebo groups during the combination treatment period; the most commonly reported events were respiratory. 12 of 97 participants had chest tightness or dyspnoea during treatment with lumacaftor alone. In pre-planned secondary analyses, a significant decrease in sweat chloride concentration occurred in the treatment groups between day 1 and day 56 (lumacaftor 400 mg once per day group -9.1 mmol/L, p<0.001; lumacaftor 600 mg once per day group -8.9 mmol/L, p<0.001; lumacaftor 400 mg every 12 h group -10.3 mmol/L, p=0.002). These changes were significantly greater than the change in the placebo group. In cohort 2, the lumacaftor 600 mg once per day significantly improved FEV1 from day 1 to 56 (difference compared with placebo group: +5.6 percentage points, p=0.013), primarily during the combination period. In cohort 3, FEV1 did not change significantly across the entire study period compared with placebo (difference +4.2 percentage points, p=0.132), but did during the combination period (difference +7.7 percentage points, p=0·003). Phe508del CFTR heterozygous patients did not have a significant improvement in FEV1. INTERPRETATION We provide evidence that combination lumacaftor and ivacaftor improves FEV1 for patients with cystic fibrosis who are homozygous for phe508del CFTR, with a modest effect on sweat chloride concentration. These results support the further exploration of combination lumacaftor and ivacaftor as a treatment in this setting. FUNDING Vertex Pharmaceuticals, Cystic Fibrosis Foundation Therapeutics Development Network.
Collapse
Affiliation(s)
| | - Scott C Bell
- The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Michael W Konstan
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Steven M Rowe
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ernst Rietschel
- Children's Hospital, University of Cologne, Cologne, Germany
| | | | | | | | | |
Collapse
|
25
|
Vali P, Shahcheraghi F, Seyfipour M, Zamani MA, Allahyartorkaman M, Feizabadi MM. Phenotypic and Genetic Characterization of Carbapenemase and ESBLs Producing Gram-negative Bacteria (GNB) Isolated from Patients with Cystic Fibrosis (CF) in Tehran Hospitals. J Clin Diagn Res 2014; 8:26-30. [PMID: 24596716 PMCID: PMC3939568 DOI: 10.7860/jcdr/2014/6877.3916] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/24/2013] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cystic Fibrosis (CF) is an autosomal recessive genetic disorder in white populations caused by mutation in a gene that encodes Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) protein. Since frequent respiratory tract infections are the major problem in patients with CF, obligation to identify the causative bacteria and determining their antibiotic resistance pattern is crucial. The purpose of this project was to detect Gram-negative bacteria (GNB) isolated from sputa of CF patients and to determine their antibiotic resistance pattern. MATERIALS AND METHODS The sputum of 52 CF patients, treated as inpatients at hospitals in Tehran, was obtained between November 2011 and June 2012. Samples cultured in selective and non-selective media and GNB recognized by biochemical tests. Antimicrobial susceptibility testing to cephalosporins, aminoglycosides and carbapenems was performed by disk diffusion method and MICs of them were measured. For phenotypic detection of carbapenemase and ESBLs production, the Modified Hodge test, double disk synergy test and the combined disk methods were performed. Subsequently, the genes encoding the extended spectrum beta-lactamases (blaPER, blaCTX-M) and carbapenemases (blaIMP-1, blaGES, blaKPC, blaNDM, blaVIM-1, blaVIM-2, blaSPM, blaSIM) in Gram negative bacteria were targeted among the resistant isolates by using PCR. PFGE was used to determine any genetic relationship among the Pseudomonas aeruginosa isolated from these patients. RESULTS Fifty five GNB were isolated from 52 sputum samples including Pseudomonas aeruginosa, Klebsiella ozaenae, Alcaligenes xylosoxidans, Achromobacter denitrificans, Klebsiella pneumonia and Stenotrophomonas maltophilia. The rates of resistance to different antibiotic were as follows: cefixime (%80), ceftriaxone (%43), ceftazidime (%45) and meropenem (%7). The prevalence of genes encoding the ESBLs and Carbapenemases among the the phenotypically positive strains were as follows: blaCTX-M (19), blaIMP-1 (2), blaVIM-1 (2) and blaVIM-2 (3) genes respectively. No other genes were detected. PFGE analysis revealed 8 genotypes. Six isolates had mutually 3 similar patterns. CONCLUSION This study showed the existence of important ESBLs and carbapenemases genes among the GNB isolated from patients with CF. Continuous surveillance of ESBLs and Carbapenemases, also identification of their types, in bacteria isolated from these patients have an important clinical impact, since, it can often provide valuable information for effective infection control measures and for the choice of appropriate antimicrobial therapy.
Collapse
Affiliation(s)
- Parisa Vali
- Student, Department of Microbiology and Microbiology Research Centre, Pasteur Institute, Tehran, Iran
| | - Fereshteh Shahcheraghi
- Associate Professor, Department of Microbiology and Microbiology Research Centre, Pasteur Institute, Tehran, Iran
| | - Maryam Seyfipour
- M.Sc. Student, Department of Microbiology and Microbiology Research Centre, Pasteur Institute, Tehran, Iran
| | - Maryam Alsadat Zamani
- M.Sc. Student, Department of Microbiology and Microbiology Research Centre, Pasteur Institute, Tehran, Iran
| | | | - Mohammad Mehdi Feizabadi
- Professor, Department of Microbiology, Faculty of Medicine, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
26
|
Blanchard E, Zlock L, Lao A, Mika D, Namkung W, Xie M, Scheitrum C, Gruenert DC, Verkman AS, Finkbeiner WE, Conti M, Richter W. Anchored PDE4 regulates chloride conductance in wild-type and ΔF508-CFTR human airway epithelia. FASEB J 2013; 28:791-801. [PMID: 24200884 DOI: 10.1096/fj.13-240861] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) that impair its expression and/or chloride channel function. Here, we provide evidence that type 4 cyclic nucleotide phosphodiesterases (PDE4s) are critical regulators of the cAMP/PKA-dependent activation of CFTR in primary human bronchial epithelial cells. In non-CF cells, PDE4 inhibition increased CFTR activity under basal conditions (ΔISC 7.1 μA/cm(2)) and after isoproterenol stimulation (increased ΔISC from 13.9 to 21.0 μA/cm(2)) and slowed the return of stimulated CFTR activity to basal levels by >3-fold. In cells homozygous for ΔF508-CFTR, the most common mutation found in CF, PDE4 inhibition alone produced minimal channel activation. However, PDE4 inhibition strongly amplified the effects of CFTR correctors, drugs that increase expression and membrane localization of CFTR, and/or CFTR potentiators, drugs that increase channel gating, to reach ∼ 25% of the chloride conductance observed in non-CF cells. Biochemical studies indicate that PDE4s are anchored to CFTR and mediate a local regulation of channel function. Taken together, our results implicate PDE4 as an important determinant of CFTR activity in airway epithelia, and support the use of PDE4 inhibitors to potentiate the therapeutic benefits of CFTR correctors and potentiators.
Collapse
Affiliation(s)
- Elise Blanchard
- 1Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, 513 Parnassus Ave., Box 0556, San Francisco, CA 94143-0556, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Assessing the residual CFTR gene expression in human nasal epithelium cells bearing CFTR splicing mutations causing cystic fibrosis. Eur J Hum Genet 2013; 22:784-91. [PMID: 24129438 DOI: 10.1038/ejhg.2013.238] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 09/06/2013] [Accepted: 09/10/2013] [Indexed: 01/18/2023] Open
Abstract
The major purpose of the present study was to quantify correctly spliced CFTR transcripts in human nasal epithelial cells from cystic fibrosis (CF) patients carrying the splicing mutations c.580-1G>T (712-1G>T) and c.2657+5G>A (2789+5G>A) and to assess the applicability of this model in CFTR therapeutic approaches. We performed the relative quantification of CFTR mRNA by reverse transcription quantitative PCR (RT-qPCR) of these splicing mutations in four groups (wild type, CF-F508del controls, CF patients and CF carriers) of individuals. In addition, in vitro assays using minigene constructs were performed to evaluate the effect of a new CF complex allele c.[2657+5G>A; 2562T>G]. Ex vivo qPCR data show that the primary consequence of both mutations at the RNA level is the skipping of their neighboring exon (6 and 16, respectively). The CFTR minigenes results mimicked the ex vivo data, as exon 16 skipping is the main aberrant transcript, and the correctly spliced transcript level was observed in a similar proportion when the c.2657+5G>A mutation is present. In summary, we provide evidence that ex vivo quantitative transcripts analysis using RT/qPCR is a robust technology that could be useful for measuring the efficacy of therapeutic approaches that attempt to achieve an increase in CFTR gene expression.
Collapse
|
28
|
Farinha CM, Matos P, Amaral MD. Control of cystic fibrosis transmembrane conductance regulator membrane trafficking: not just from the endoplasmic reticulum to the Golgi. FEBS J 2013; 280:4396-406. [PMID: 23773658 DOI: 10.1111/febs.12392] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/25/2013] [Accepted: 06/11/2013] [Indexed: 12/18/2022]
Abstract
Biogenesis of cystic fibrosis transmembrane conductance regulator (CFTR) starts with its cotranslational insertion into the membrane of the endoplasmic reticulum (ER) and core glycosylation. These initial events are followed by a complex succession of steps with the main goal of checking the overall quality of CFTR conformation in order to promote its exit from the ER through the secretory pathway. Failure to pass the various checkpoints of the ER quality control targets the most frequent disease-causing mutant protein (F508del-CFTR) for premature degradation. For wild-type CFTR that exits the ER, trafficking through the Golgi is the major site for glycan processing, although nonconventional trafficking pathways have also been described for CFTR. Once CFTR is at the cell surface, its stability is also controlled by multiple protein interactors, including Rab proteins, Rho small GTPases, and PDZ proteins. These regulate not only anterograde trafficking to the cell surface, but also endocytosis and recycling, thus achieving fine and tight modulation of CFTR plasma membrane levels. Exciting recent data have related autophagy and epithelial differentiation to the regulation of CFTR trafficking. Herein, we review the various checkpoints of the complex quality control along the secretory trafficking pathway and the associated pathways that are starting to be explored for the benefit of cystic fibrosis patients.
Collapse
Affiliation(s)
- Carlos M Farinha
- Faculty of Sciences, BioFIG - Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Portugal
| | | | | |
Collapse
|
29
|
Moniz S, Sousa M, Moraes BJ, Mendes AI, Palma M, Barreto C, Fragata JI, Amaral MD, Matos P. HGF stimulation of Rac1 signaling enhances pharmacological correction of the most prevalent cystic fibrosis mutant F508del-CFTR. ACS Chem Biol 2013; 8:432-42. [PMID: 23148778 DOI: 10.1021/cb300484r] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cystic fibrosis (CF), a major life-limiting genetic disease leading to severe respiratory symptoms, is caused by mutations in CF transmembrane conductance regulator (CFTR), a chloride (Cl(-)) channel expressed at the apical membrane of epithelial cells. Absence of functional CFTR from the surface of respiratory cells reduces mucociliary clearance, promoting airways obstruction, chronic infection, and ultimately lung failure. The most frequent mutation, F508del, causes the channel to misfold, triggering its premature degradation and preventing it from reaching the cell surface. Recently, novel small-molecule correctors rescuing plasma membrane localization of F508del-CFTR underwent clinical trials but with limited success. Plausibly, this may be due to the mutant intrinsic plasma membrane (PM) instability. Herein, we show that restoration of F508del-CFTR PM localization by correctors can be dramatically improved through a novel pathway involving stimulation of signaling by the endogenous small GTPase Rac1 via hepatocyte growth factor (HGF). We first show that CFTR anchors to apical actin cytoskeleton (via Ezrin) upon activation of Rac1 signaling through PIP5K and Arp2/3. We then found that such anchoring retains pharmacologically rescued F508del-CFTR at the cell surface, boosting functional restoration by correctors up to 30% of wild-type channel levels in human airway epithelial cells. Our findings reveal that surface anchoring and retention is a major target pathway for CF pharmacotherapy, namely, to achieve maximal restoration of F508del-CFTR in patients in combination with correctors. Moreover, this approach may also translate to other disorders caused by trafficking-deficient surface proteins.
Collapse
Affiliation(s)
- Sónia Moniz
- Department of Genetics, National Health Institute ‘Dr. Ricardo Jorge’, Av. Padre Cruz, 1649-016 Lisboa;
Portugal
- Faculty of Sciences,
BioFIG
- Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Campo Grande-C8, 1749-016 Lisboa,
Portugal
| | - Marisa Sousa
- Department of Genetics, National Health Institute ‘Dr. Ricardo Jorge’, Av. Padre Cruz, 1649-016 Lisboa;
Portugal
- Faculty of Sciences,
BioFIG
- Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Campo Grande-C8, 1749-016 Lisboa,
Portugal
| | - Bruno José Moraes
- Department of Genetics, National Health Institute ‘Dr. Ricardo Jorge’, Av. Padre Cruz, 1649-016 Lisboa;
Portugal
- Faculty of Sciences,
BioFIG
- Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Campo Grande-C8, 1749-016 Lisboa,
Portugal
| | - Ana Isabel Mendes
- Department of Genetics, National Health Institute ‘Dr. Ricardo Jorge’, Av. Padre Cruz, 1649-016 Lisboa;
Portugal
- Faculty of Sciences,
BioFIG
- Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Campo Grande-C8, 1749-016 Lisboa,
Portugal
| | - Marta Palma
- Faculty of Sciences,
BioFIG
- Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Campo Grande-C8, 1749-016 Lisboa,
Portugal
| | - Celeste Barreto
- Department of Pediatrics, Hospital de Santa Maria, Avenida Professor Egas Moniz,
1649-035 Lisboa, Portugal
| | - José I. Fragata
- Department of Cardiothoracic
Surgery, Hospital de Santa Marta, R. de
Santa Marta 50, 1169-024 Lisboa, Portugal
| | - Margarida D. Amaral
- Department of Genetics, National Health Institute ‘Dr. Ricardo Jorge’, Av. Padre Cruz, 1649-016 Lisboa;
Portugal
- Faculty of Sciences,
BioFIG
- Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Campo Grande-C8, 1749-016 Lisboa,
Portugal
| | - Paulo Matos
- Department of Genetics, National Health Institute ‘Dr. Ricardo Jorge’, Av. Padre Cruz, 1649-016 Lisboa;
Portugal
- Faculty of Sciences,
BioFIG
- Centre for Biodiversity, Functional and Integrative Genomics, University of Lisboa, Campo Grande-C8, 1749-016 Lisboa,
Portugal
| |
Collapse
|
30
|
Leonard A, Leal T, Lebecque P. [Mucoviscidosis: CFTR mutation-specific therapy: a ray of sunshine in a cloudy sky]. Arch Pediatr 2012. [PMID: 23199563 DOI: 10.1016/j.arcped.2012.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is a need to find a cure for pulmonary disease in cystic fibrosis (CF), though full benefit of this approach will be restricted to those patients with well-preserved lungs. The most promising route is currently that of a pharmacological mutation-specific approach aiming at correcting the mechanism by which mutations lead to impairment of chloride conductance across respiratory epithelial cells. In the past 14years, 7 candidate drugs (CPX, 4PBA, gentamicin, PTC124, VX-770 or Ivacaftor, VX-809 or Lumacaftor, and Miglustat) have been investigated in CF patients. A postulate of 14 out of the 15 published studies has been that an effective agent had to improve total chloride secretion as assessed in vivo by nasal potential difference measurements. The present review casts a critical look at these studies. Apparent inconsistencies are discussed as well as possible limitations of nasal potential difference measurements as outcome parameters in these trials. Primarily targeting a mutation carried by less than 2% of French CF patients, the 2 Ivacaftor studies could well be a milestone on the long road toward a cure for CF. However, further data on safety and long-term efficacy are obviously needed and the current price of this medication in the US would make it unaffordable for European patients.
Collapse
Affiliation(s)
- A Leonard
- Unité de pneumologie pédiatrique et mucoviscidose, cliniques Saint-Luc, université de Louvain, Bruxelles, Belgique.
| | | | | |
Collapse
|
31
|
Sousa M, Servidoni MF, Vinagre AM, Ramalho AS, Bonadia LC, Felício V, Ribeiro MA, Uliyakina I, Marson FA, Kmit A, Cardoso SR, Ribeiro JD, Bertuzzo CS, Sousa L, Kunzelmann K, Ribeiro AF, Amaral MD. Measurements of CFTR-mediated Cl- secretion in human rectal biopsies constitute a robust biomarker for Cystic Fibrosis diagnosis and prognosis. PLoS One 2012; 7:e47708. [PMID: 23082198 PMCID: PMC3474728 DOI: 10.1371/journal.pone.0047708] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 09/14/2012] [Indexed: 01/14/2023] Open
Abstract
Background Cystic Fibrosis (CF) is caused by ∼1,900 mutations in the CF transmembrane conductance regulator (CFTR) gene encoding for a cAMP-regulated chloride (Cl−) channel expressed in several epithelia. Clinical features are dominated by respiratory symptoms, but there is variable organ involvement thus causing diagnostic dilemmas, especially for non-classic cases. Methodology/Principal Findings To further establish measurement of CFTR function as a sensitive and robust biomarker for diagnosis and prognosis of CF, we herein assessed cholinergic and cAMP-CFTR-mediated Cl− secretion in 524 freshly excised rectal biopsies from 118 individuals, including patients with confirmed CF clinical diagnosis (n = 51), individuals with clinical CF suspicion (n = 49) and age-matched non-CF controls (n = 18). Conclusive measurements were obtained for 96% of cases. Patients with “Classic CF”, presenting earlier onset of symptoms, pancreatic insufficiency, severe lung disease and low Shwachman-Kulczycki scores were found to lack CFTR-mediated Cl− secretion (<5%). Individuals with milder CF disease presented residual CFTR-mediated Cl− secretion (10–57%) and non-CF controls show CFTR-mediated Cl− secretion ≥30–35% and data evidenced good correlations with various clinical parameters. Finally, comparison of these values with those in “CF suspicion” individuals allowed to confirm CF in 16/49 individuals (33%) and exclude it in 28/49 (57%). Statistical discriminant analyses showed that colonic measurements of CFTR-mediated Cl− secretion are the best discriminator among Classic/Non-Classic CF and non-CF groups. Conclusions/Significance Determination of CFTR-mediated Cl− secretion in rectal biopsies is demonstrated here to be a sensitive, reproducible and robust predictive biomarker for the diagnosis and prognosis of CF. The method also has very high potential for (pre-)clinical trials of CFTR-modulator therapies.
Collapse
Affiliation(s)
- Marisa Sousa
- BioFIG - Centre for Biodiversity, Functional and Integrative Genomics; Faculty of Sciences, University of Lisboa, Lisboa, Portugal
- Department of Genetics - National Institute of Health, Lisboa, Portugal
| | - Maria F. Servidoni
- Gastrocentro - Endoscopy Unit - State University of Campinas, Campinas, Brazil
- Pediatrics Department - State University of Campinas, Campinas, Brazil
| | - Adriana M. Vinagre
- Faculty of Medical Sciences - State University of Campinas, Campinas, Brazil
| | - Anabela S. Ramalho
- BioFIG - Centre for Biodiversity, Functional and Integrative Genomics; Faculty of Sciences, University of Lisboa, Lisboa, Portugal
- Department of Genetics - National Institute of Health, Lisboa, Portugal
| | - Luciana C. Bonadia
- Faculty of Medical Sciences - State University of Campinas, Campinas, Brazil
| | - Verónica Felício
- BioFIG - Centre for Biodiversity, Functional and Integrative Genomics; Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Maria A. Ribeiro
- CIPED - Research Center in Pediatrics - State University of Campinas, Campinas, Brazil
| | - Inna Uliyakina
- BioFIG - Centre for Biodiversity, Functional and Integrative Genomics; Faculty of Sciences, University of Lisboa, Lisboa, Portugal
- Department of Genetics - National Institute of Health, Lisboa, Portugal
| | - Fernando A. Marson
- Faculty of Medical Sciences - State University of Campinas, Campinas, Brazil
| | - Arthur Kmit
- Faculty of Medical Sciences - State University of Campinas, Campinas, Brazil
| | - Silvia R. Cardoso
- Pediatrics Department - State University of Campinas, Campinas, Brazil
- Endoscopy Unit – University Hospital of Campinas, Campinas, Brazil
| | - José D. Ribeiro
- Pediatrics Department - State University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences - State University of Campinas, Campinas, Brazil
- CIPED - Research Center in Pediatrics - State University of Campinas, Campinas, Brazil
| | - Carmen S. Bertuzzo
- Faculty of Medical Sciences - State University of Campinas, Campinas, Brazil
| | - Lisete Sousa
- CEAUL - Center of Statistics and Applications of the University of Lisboa; Department of Statistics and Operation Research, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Karl Kunzelmann
- Institut für Physiologie – University of Regensburg, Regensburg, Germany
| | - Antônio F. Ribeiro
- Pediatrics Department - State University of Campinas, Campinas, Brazil
- Faculty of Medical Sciences - State University of Campinas, Campinas, Brazil
- CIPED - Research Center in Pediatrics - State University of Campinas, Campinas, Brazil
| | - Margarida D. Amaral
- BioFIG - Centre for Biodiversity, Functional and Integrative Genomics; Faculty of Sciences, University of Lisboa, Lisboa, Portugal
- Department of Genetics - National Institute of Health, Lisboa, Portugal
- * E-mail:
| |
Collapse
|
32
|
Keeling KM, Wang D, Conard SE, Bedwell DM. Suppression of premature termination codons as a therapeutic approach. Crit Rev Biochem Mol Biol 2012; 47:444-63. [PMID: 22672057 PMCID: PMC3432268 DOI: 10.3109/10409238.2012.694846] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this review, we describe our current understanding of translation termination and pharmacological agents that influence the accuracy of this process. A number of drugs have been identified that induce suppression of translation termination at in-frame premature termination codons (PTCs; also known as nonsense mutations) in mammalian cells. We discuss efforts to utilize these drugs to suppress disease-causing PTCs that result in the loss of protein expression and function. In-frame PTCs represent a genotypic subset of mutations that make up ~11% of all known mutations that cause genetic diseases, and millions of patients have diseases attributable to PTCs. Current approaches aimed at reducing the efficiency of translation termination at PTCs (referred to as PTC suppression therapy) have the goal of alleviating the phenotypic consequences of a wide range of genetic diseases. Suppression therapy is currently in clinical trials for treatment of several genetic diseases caused by PTCs, and preliminary results suggest that some patients have shown clinical improvements. While current progress is promising, we discuss various approaches that may further enhance the efficiency of this novel therapeutic approach.
Collapse
Affiliation(s)
- Kim M. Keeling
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dan Wang
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sara E. Conard
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David M. Bedwell
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
33
|
Luciani A, Villella VR, Esposito S, Gavina M, Russo I, Silano M, Guido S, Pettoello-Mantovani M, Carnuccio R, Scholte B, De Matteis A, Maiuri MC, Raia V, Luini A, Kroemer G, Maiuri L. Targeting autophagy as a novel strategy for facilitating the therapeutic action of potentiators on ΔF508 cystic fibrosis transmembrane conductance regulator. Autophagy 2012; 8:1657-72. [PMID: 22874563 DOI: 10.4161/auto.21483] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Channel activators (potentiators) of cystic fibrosis (CF) transmembrane conductance regulator (CFTR), can be used for the treatment of the small subset of CF patients that carry plasma membrane-resident CFTR mutants. However, approximately 90% of CF patients carry the misfolded ΔF508-CFTR and are poorly responsive to potentiators, because ΔF508-CFTR is intrinsically unstable at the plasma membrane (PM) even if rescued by pharmacological correctors. We have demonstrated that human and mouse CF airways are autophagy deficient due to functional sequestration of BECN1 and that the tissue transglutaminase-2 inhibitor, cystamine, or antioxidants restore BECN1-dependent autophagy and reduce SQSTM1/p62 levels, thus favoring ΔF508-CFTR trafficking to the epithelial surface. Here, we investigated whether these treatments could facilitate the beneficial action of potentiators on ΔF508-CFTR homozygous airways. Cystamine or the superoxide dismutase (SOD)/catalase-mimetic EUK-134 stabilized ΔF508-CFTR at the plasma membrane of airway epithelial cells and sustained the expression of CFTR at the epithelial surface well beyond drug withdrawal, overexpressing BECN1 and depleting SQSTM1. This facilitates the beneficial action of potentiators in controlling inflammation in ex vivo ΔF508-CFTR homozygous human nasal biopsies and in vivo in mouse ΔF508-CFTR lungs. Direct depletion of Sqstm1 by shRNAs in vivo in ΔF508-CFTR mice synergized with potentiators in sustaining surface CFTR expression and suppressing inflammation. Cystamine pre-treatment restored ΔF508-CFTR response to the CFTR potentiators genistein, Vrx-532 or Vrx-770 in freshly isolated brushed nasal epithelial cells from ΔF508-CFTR homozygous patients. These findings delineate a novel therapeutic strategy for the treatment of CF patients with the ΔF508-CFTR mutation in which patients are first treated with cystamine and subsequently pulsed with CFTR potentiators.
Collapse
Affiliation(s)
- Alessandro Luciani
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Li H, Yang W, Mendes F, Amaral MD, Sheppard DN. Impact of the cystic fibrosis mutation F508del-CFTR on renal cyst formation and growth. Am J Physiol Renal Physiol 2012; 303:F1176-86. [PMID: 22874761 DOI: 10.1152/ajprenal.00130.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In autosomal dominant polycystic kidney disease (ADPKD), cystic fibrosis transmembrane conductance regulator (CFTR), the protein product of the gene defective in cystic fibrosis (CF), plays a crucial role in fluid accumulation, which promotes cyst swelling. Several studies have identified individuals afflicted by both ADPKD and CF. Two studies suggested that CF mutations might attenuate the severity of ADPKD, whereas a third found no evidence of a protective effect. In this study, we investigated the impact of the commonest CF mutation F508del-CFTR on the formation and growth of renal cysts. As a model system, we used Madin-Darby canine kidney (MDCK) epithelial cells engineered to express wild-type and F508del human CFTR. We grew MDCK cysts in collagen gels in the presence of the cAMP agonist forskolin and measured transepithelial resistance and Cl(-) secretion with the Ussing chamber technique and assayed cell proliferation using nonpolarized MDCK cells. When compared with untransfected MDCK cells, cells expressing wild-type CFTR generated substantial numbers of large cysts, which grew markedly over time. By contrast, MDCK cells expressing F508del-CFTR formed very few tiny cysts that failed to enlarge. Interestingly, treatment of F508del-CFTR cysts with the CFTR corrector VRT-325 and the CFTR corrector-potentiator VRT-532 increased the number, but not size, of F508del-CFTR cysts, possibly because VRT-325 inhibited strongly cell proliferation. Based on its effects on transepithelial resistance, Cl(-) secretion, and cell proliferation, we conclude that the F508del-CFTR mutation disrupts cyst formation and growth by perturbing strongly fluid accumulation within the cyst lumen without compromising epithelial integrity.
Collapse
Affiliation(s)
- Hongyu Li
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, UK
| | | | | | | | | |
Collapse
|
35
|
Cho HJ, Gee HY, Baek KH, Ko SK, Park JM, Lee H, Kim ND, Lee MG, Shin I. A small molecule that binds to an ATPase domain of Hsc70 promotes membrane trafficking of mutant cystic fibrosis transmembrane conductance regulator. J Am Chem Soc 2011; 133:20267-76. [PMID: 22074182 DOI: 10.1021/ja206762p] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is a cell-surface anion channel that permeates chloride and bicarbonate ions. The most frequent mutation of CFTR that causes cystic fibrosis is the deletion of phenylalanine at position 508 (ΔF508), which leads to defects in protein folding and cellular trafficking to the plasma membrane. The lack of the cell-surface CFTR results in a reduction in the lifespan due to chronic lung infection with progressive deterioration of lung function. Hsc70 plays a crucial role in degradation of mutant CFTR by the ubiquitin-proteasome system. To date, various Hsc70 inhibitors and transcription regulators have been tested to determine whether they correct the defective activity of mutant CFTR. However, they exhibited limited or questionable effects on restoring the chloride channel activity in cystic fibrosis cells. Herein, we show that a small molecule apoptozole (Az) has high cellular potency to promote membrane trafficking of mutant CFTR and its chloride channel activity in cystic fibrosis cells. Results from affinity chromatography and ATPase activity assay indicate that Az inhibits the ATPase activity of Hsc70 by binding to its ATPase domain. In addition, a ligand-directed protein labeling and molecular modeling studies also suggest the binding of Az to an ATPase domain, in particular, an ATP-binding pocket. It is proposed that Az suppresses ubiquitination of ΔF508-CFTR maybe by blocking interaction of the mutant with Hsc70 and CHIP, and, as a consequence, it enhances membrane trafficking of the mutant.
Collapse
Affiliation(s)
- Hyungseoph J Cho
- Center for Biofunctional Molecules, Department of Chemistry, Yonsei University, Seoul 120-749, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Xiong Y, Chen B, Shi L, Fredrickson JK, Bigelow DJ, Squier TC. Targeted Protein Degradation of Outer Membrane Decaheme Cytochrome MtrC Metal Reductase in Shewanella oneidensis MR-1 Measured Using Biarsenical Probe CrAsH-EDT2. Biochemistry 2011; 50:9738-51. [DOI: 10.1021/bi200602f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yijia Xiong
- Biological Sciences Division, Fundamental
Sciences
Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Baowei Chen
- Biological Sciences Division, Fundamental
Sciences
Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Liang Shi
- Biological Sciences Division, Fundamental
Sciences
Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - James K. Fredrickson
- Biological Sciences Division, Fundamental
Sciences
Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Diana J. Bigelow
- Biological Sciences Division, Fundamental
Sciences
Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Thomas C. Squier
- Biological Sciences Division, Fundamental
Sciences
Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| |
Collapse
|
37
|
Abstract
With knowledge of the molecular behaviour of the cystic fibrosis transmembrane conductance regulator (CFTR), its physiological role and dysfunction in cystic fibrosis (CF), therapeutic strategies are now being developed that target the root cause of CF rather than disease symptoms. Here, we review progress towards the development of rational new therapies for CF. We highlight the discovery of small molecules that rescue the cell surface expression and defective channel gating of CF mutants, termed CFTR correctors and CFTR potentiators, respectively. We draw attention to alternative approaches to restore epithelial ion transport to CF epithelia, including inhibitors of the epithelial Na(+) channel (ENaC) and activators of the Ca(2+)-activated Cl(-) channel TMEM16A. The expertise required to translate small molecules identified in the laboratory to drugs for CF patients depends on our ability to coordinate drug development at an international level and our ability to provide pertinent biological information using suitable disease models.
Collapse
|
38
|
Antagonistic regulation of cystic fibrosis transmembrane conductance regulator cell surface expression by protein kinases WNK4 and spleen tyrosine kinase. Mol Cell Biol 2011; 31:4076-86. [PMID: 21807898 DOI: 10.1128/mcb.05152-11] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Members of the WNK (with-no-lysine [K]) subfamily of protein kinases regulate various ion channels involved in sodium, potassium, and chloride homeostasis by either inducing their phosphorylation or regulating the number of channel proteins expressed at the cell surface. Here, we describe findings demonstrating that the cell surface expression of the cystic fibrosis transmembrane conductance regulator (CFTR) is also regulated by WNK4 in mammalian cells. This effect of WNK4 is independent of the presence of kinase and involves interaction with and inhibition of spleen tyrosine kinase (Syk), which phosphorylates Tyr512 in the first nucleotide-binding domain 1 (NBD1) of CFTR. Transfection of catalytically active Syk into CFTR-expressing baby hamster kidney cells reduces the cell surface expression of CFTR, whereas that of WNK4 promotes it. This is shown by biotinylation of cell surface proteins, immunofluorescence microscopy, and functional efflux assays. Mutation of Tyr512 to either glutamic acid or phenylalanine is sufficient to alter CFTR surface levels. In human airway epithelial cells, downregulation of endogenous Syk and WNK4 confirms their roles as physiologic regulators of CFTR surface expression. Together, our results show that Tyr512 phosphorylation is a novel signal regulating the prevalence of CFTR at the cell surface and that WNK4 and Syk perform an antagonistic role in this process.
Collapse
|
39
|
Henderson MJ, Singh OV, Zeitlin PL. Applications of proteomic technologies for understanding the premature proteolysis of CFTR. Expert Rev Proteomics 2010; 7:473-86. [PMID: 20653504 DOI: 10.1586/epr.10.42] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, which encodes an ATP-dependent anion channel. Disease-causing mutations can affect channel biogenesis, trafficking or function, and result in reduced ion transport at the apical surface of many tissues. The most common CFTR mutation is a deletion of phenylalanine at position 508 (DeltaF508), which results in a misfolded protein that is prematurely targeted for degradation. This article focuses on how proteomic approaches have been utilized to explore the mechanisms of premature proteolysis in CF. Additionally, we emphasize the potential for proteomic-based technologies in expanding our understanding of CF pathophysiology and therapeutic approaches.
Collapse
Affiliation(s)
- Mark J Henderson
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
40
|
Kim Chiaw P, Wellhauser L, Huan LJ, Ramjeesingh M, Bear CE. A chemical corrector modifies the channel function of F508del-CFTR. Mol Pharmacol 2010; 78:411-8. [PMID: 20501743 DOI: 10.1124/mol.110.065862] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
The deletion of Phe-508 (F508del) constitutes the most prevalent cystic fibrosis-causing mutation. This mutation leads to cystic fibrosis transmembrane conductance regulator (CFTR) misfolding and retention in the endoplasmic reticulum and altered channel activity in mammalian cells. This folding defect can however be partially overcome by growing cells expressing this mutant protein at low (27 degrees C) temperature. Chemical "correctors" have been identified that are also effective in rescuing the biosynthetic defect in F508del-CFTR, thereby permitting its functional expression at the cell surface. The mechanism of action of chemical correctors remains unclear, but it has been suggested that certain correctors [including 4-cyclohexyloxy-2-(1-[4-(4-methoxy-benzenesulfonyl)-piperazin-1-yl]-ethyl)-quinazoline (VRT-325)] may act to promote trafficking by interacting directly with the mutant protein. To test this hypothesis, we assessed the effect of VRT-325 addition on the channel activity of F508del-CFTR after its surface expression had been "rescued" by low temperature. It is noteworthy that short-term pretreatment with VRT-325 [but not with an inactive analog, 4-hydroxy-2-(1-[4-(4-methoxy-benzenesulfonyl)-piperazin-1-yl]-ethyl)-quinazoline (VRT-186)], caused a modest but significant inhibition of cAMP-mediated halide flux. Furthermore, VRT-325 decreased the apparent ATP affinity of purified and reconstituted F508del-CFTR in our ATPase activity assay, an effect that may account for the decrease in channel activity by temperature-rescued F508del-CFTR. These findings suggest that biosynthetic rescue mediated by VRT-325 may be conferred (at least in part) by direct modification of the structure of the mutant protein, leading to a decrease in its ATP-dependent conformational dynamics. Therefore, the challenge for therapy discovery will be the design of small molecules that bind to promote biosynthetic maturation of the major mutant without compromising its activity in vivo.
Collapse
Affiliation(s)
- Patrick Kim Chiaw
- Programme in Molecular Structure and Function, Research Institute, Hospital for Sick Children and Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
41
|
Mishra S, Wang X, Smiley N, Xia P, Hong CM, Senadheera D, Bui KC, Lutzko C. Genetic modification of airway progenitors after lentiviral gene delivery to the amniotic fluid of murine fetuses. Am J Respir Cell Mol Biol 2010; 44:562-70. [PMID: 20581098 DOI: 10.1165/rcmb.2009-0235oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lentiviral vectors with the firefly luciferase or enhanced green fluorescent protein (EGFP) transgenes were delivered to the amniotic fluid of murine fetuses at Embryonic Day (E) 14.5 or E16.5. Whole-body imaging of luciferase recipients after birth demonstrated transgene expression in the peritoneal and thoracic regions. Organ imaging showed luciferase expression in lung, skin, stomach, and/or intestine. Histological immunofluorescence analysis of EGFP recipients demonstrated that small clusters (≤ three cells) of EGFP-positive epithelial cells were present in the large and small airways of recipients at up to 7 months (n = 11). There was no difference in the frequency of transgene expression in mice injected at E14.5 or E16.5 in respiratory or nonrespiratory organs. Analysis of the bronchoalveolar duct junctions on tissue sections of recipient mice identified multiple EGFP-positive epithelial cells. Cells coexpressing EGFP, Clara cell 10-kd protein, and surfactant protein C (SPC) were also found in lungs, consistent with the transduction of bronchoalveolar stem cells. Next, naphthalene lung injury in both luciferase and EGFP recipients was performed to determine whether transduced cells could contribute to tissue repair. In luciferase recipients, the whole-body luciferase signal increased 2- to 20-fold at 2 weeks after naphthalene treatment. Remarkably, immunohistological analysis of the lungs of EGFP recipients after lung injury repair demonstrated repopulation of airways with long stretches of EGFP-positive epithelial cells (n = 4). Collectively, these data demonstrate that lentiviral gene delivery to the amniotic fluid of murine fetuses genetically modifies long-lived epithelial progenitors capable of contributing to lung injury repair.
Collapse
Affiliation(s)
- Suparna Mishra
- Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Respiratory syncytial virus engineered to express the cystic fibrosis transmembrane conductance regulator corrects the bioelectric phenotype of human cystic fibrosis airway epithelium in vitro. J Virol 2010; 84:7770-81. [PMID: 20504917 DOI: 10.1128/jvi.00346-10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Cystic fibrosis (CF) is the most common lethal recessive genetic disease in the Caucasian population. It is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene that is normally expressed in ciliated airway epithelial cells and the submucosal glands of the lung. Since the CFTR gene was first characterized in 1989, a major goal has been to develop an effective gene therapy for CF lung disease, which has the potential to ameliorate morbidity and mortality. Respiratory syncytial virus (RSV) naturally infects the ciliated cells in the human airway epithelium. In addition, the immune response mounted against an RSV infection does not prevent subsequent infections, suggesting that an RSV-based vector might be effectively readministered. To test whether the large 4.5-kb CFTR gene could be expressed by a recombinant RSV and whether infectious virus could be used to deliver CFTR to ciliated airway epithelium derived from CF patients, we inserted the CFTR gene into four sites in a recombinant green fluorescent protein-expressing RSV (rgRSV) genome to generate virus expressing four different levels of CFTR protein. Two of these four rgRSV-CFTR vectors were capable of expressing CFTR with little effect on viral replication. rgRSV-CFTR infection of primary human airway epithelial cultures derived from CF patients resulted in expression of CFTR protein that was properly localized at the luminal surface and corrected the chloride ion channel defect in these cells.
Collapse
|
43
|
Hutt DM, Herman D, Rodrigues APC, Noel S, Pilewski JM, Matteson J, Hoch B, Kellner W, Kelly JW, Schmidt A, Thomas PJ, Matsumura Y, Skach WR, Gentzsch M, Riordan JR, Sorscher EJ, Okiyoneda T, Lukacs GL, Frizzell RA, Manning G, Gottesfeld JM, Balch WE. Reduced histone deacetylase 7 activity restores function to misfolded CFTR in cystic fibrosis. Nat Chem Biol 2010; 6:25-33. [PMID: 19966789 PMCID: PMC2901172 DOI: 10.1038/nchembio.275] [Citation(s) in RCA: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 09/22/2009] [Indexed: 12/20/2022]
Abstract
Chemical modulation of histone deacetylase (HDAC) activity by HDAC inhibitors (HDACi) is an increasingly important approach for modifying the etiology of human disease. Loss-of-function diseases arise as a consequence of protein misfolding and degradation, which lead to system failures. The DeltaF508 mutation in cystic fibrosis transmembrane conductance regulator (CFTR) results in the absence of the cell surface chloride channel and a loss of airway hydration, leading to the premature lung failure and reduced lifespan responsible for cystic fibrosis. We now show that the HDACi suberoylanilide hydroxamic acid (SAHA) restores surface channel activity in human primary airway epithelia to levels that are 28% of those of wild-type CFTR. Biological silencing of all known class I and II HDACs reveals that HDAC7 plays a central role in restoration of DeltaF508 function. We suggest that the tunable capacity of HDACs can be manipulated by chemical biology to counter the onset of cystic fibrosis and other human misfolding disorders.
Collapse
Affiliation(s)
- Darren M. Hutt
- Departments of Cell Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| | - David Herman
- Department of Molecular Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| | - Ana P. C. Rodrigues
- Resave Newman Center for Bioinformatics, Salk Institute for Biological Studies, La Jolla, CA, 92037 USA
| | - Sabrina Noel
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Joseph M. Pilewski
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Jeanne Matteson
- Departments of Cell Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| | - Ben Hoch
- Department of Molecular Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| | - Wendy Kellner
- Departments of Cell Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| | - Jeffery W. Kelly
- Department of Chemistry at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
- Skaggs Institute of Chemical Biology at The Scripps Research Institute at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| | - Andre Schmidt
- Molecular Biophysics, University of Texas Southwestern Medical Center, 6001 Forest Park Lane, Dallas, TX 75390
| | - Philip J. Thomas
- Molecular Biophysics, University of Texas Southwestern Medical Center, 6001 Forest Park Lane, Dallas, TX 75390
| | - Yoshihiro Matsumura
- Department of Biochemistry and Molecular Biology, Oregon Health and Sciences University, Portland, OR 97239
| | - William R. Skach
- Department of Biochemistry and Molecular Biology, Oregon Health and Sciences University, Portland, OR 97239
| | - Martina Gentzsch
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, NC 27599
| | - John R. Riordan
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27510
| | - Eric J. Sorscher
- Department of Cell Biology and Physiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Tsukasa Okiyoneda
- Department of Physiology, McGill University, Montreal, QC, H3G1Y6 Canada
| | - Gergely L. Lukacs
- Department of Physiology, McGill University, Montreal, QC, H3G1Y6 Canada
| | - Raymond A. Frizzell
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Gerard Manning
- Resave Newman Center for Bioinformatics, Salk Institute for Biological Studies, La Jolla, CA, 92037 USA
| | - Joel M. Gottesfeld
- Department of Molecular Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| | - William E. Balch
- Departments of Cell Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
- Department of Molecular Biology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
- Department of Chemical Physiology at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
- The Institute for Childhood and Neglected Diseases at The Scripps Research Institute, 10550 North Torrey Pines Rd, La Jolla, CA, 92037 USA
| |
Collapse
|
44
|
Edelman A, Fritsch J, Ollero M. Twenty years after cystic fibrosis gene identification: Where are we and what are we up to? ACTA ACUST UNITED AC 2009; 59:131-3. [PMID: 19896304 DOI: 10.1016/j.patbio.2009.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 05/15/2009] [Indexed: 11/25/2022]
|
45
|
Ramalho AS, Lewandowska MA, Farinha CM, Mendes F, Gonçalves J, Barreto C, Harris A, Amaral MD. Deletion of CFTR translation start site reveals functional isoforms of the protein in CF patients. Cell Physiol Biochem 2009; 24:335-46. [PMID: 19910674 PMCID: PMC2793277 DOI: 10.1159/000257426] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2009] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND/AIMS Mutations in the CFTR gene cause Cystic Fibrosis (CF) the most common life-threatening autosomal recessive disease affecting Caucasians. We identified a CFTR mutation (c.120del23) abolishing the normal translation initiation codon, which occurs in two Portuguese CF patients. This study aims at functionally characterizing the effect of this novel mutation. METHODS RNA and protein techniques were applied to both native tissues from CF patients and recombinant cells expressing CFTR constructs to determine whether c.120del23 allows CFTR protein production through usage of alternative internal codons, and to characterize the putative truncated CFTR form(s). RESULTS Our data show that two shorter forms of CFTR protein are produced when the initiation translation codon is deleted indicating usage of internal initiation codons. The N-truncated CFTR generated by this mutation has decreased stability, very low processing efficiency, and drastically reduced function. Analysis of mutants of four methionine codons downstream to M1 (M82, M150, M152, M156) revealed that each of the codons M150/M152/M156 (exon 4) can mediate CFTR alternative translation. CONCLUSIONS The CFTR N-terminus has an important role in avoiding CFTR turnover and in rendering effective its plasma membrane traffic. These data correlate well with the severe clinical phenotype of CF patients bearing the c.120del23 mutation.
Collapse
Affiliation(s)
- Anabela S. Ramalho
- University of Lisboa, Faculty of Sciences, Centre for Biodiversity, Functional and Integrative Genomics (BioFIG), Lisboa, Portugal
- Department of Genetics, National Institute of Health, Lisboa, Portugal
| | - Marzena A. Lewandowska
- Human Molecular Genetics Program, Children's Memorial Research Center, Northwestern University, Chicago IL, USA
| | - Carlos M. Farinha
- University of Lisboa, Faculty of Sciences, Centre for Biodiversity, Functional and Integrative Genomics (BioFIG), Lisboa, Portugal
- Department of Genetics, National Institute of Health, Lisboa, Portugal
| | - Filipa Mendes
- University of Lisboa, Faculty of Sciences, Centre for Biodiversity, Functional and Integrative Genomics (BioFIG), Lisboa, Portugal
- Department of Genetics, National Institute of Health, Lisboa, Portugal
| | - Juan Gonçalves
- Hospital do Divino Espírito Santo, Ponta Delgada, Portugal
| | | | - Ann Harris
- Human Molecular Genetics Program, Children's Memorial Research Center, Northwestern University, Chicago IL, USA
| | - Margarida D. Amaral
- University of Lisboa, Faculty of Sciences, Centre for Biodiversity, Functional and Integrative Genomics (BioFIG), Lisboa, Portugal
- Department of Genetics, National Institute of Health, Lisboa, Portugal
| |
Collapse
|
46
|
Park HJ, Mylvaganum M, McPherson A, Fewell SW, Brodsky JL, Lingwood CA. A soluble sulfogalactosyl ceramide mimic promotes Delta F508 CFTR escape from endoplasmic reticulum associated degradation. ACTA ACUST UNITED AC 2009; 16:461-70. [PMID: 19389632 DOI: 10.1016/j.chembiol.2009.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 02/19/2009] [Accepted: 02/25/2009] [Indexed: 10/20/2022]
Abstract
AdaSGC binds Hsc70s to inhibit ATPase activity. Using single-turnover assays, adaSGC, a soluble SGC mimic, preferentially inhibited Hsp40-activated Hsc70 ATP hydrolysis (Ki approximately 10 microM) to reduce C-terminal Hsc70-peptide binding and, potentially, chaperone function. ERAD of misfolded Delta F508 CFTR requires Hsc70-Hsp40 chaperones. In transfected baby hamster kidney (BHK) cells, adaSGC increased Delta F508CFTR ERAD escape, and after low-temperature glycerol rescue, maturation, and iodide efflux. Inhibition of SGC biosynthesis reduced Delta F508CFTR but not wtCFTR expression, whereas depletion of other glycosphingolipids had no affect. WtCFTR transfected BHK cells showed increased SGC synthesis compared with Delta F508CFTR/mock-transfected cells. Partial rescue of Delta F508CFTR by low-temperature glycerol increased SGC synthesis. AdaSGC also increased cellular endogenous SGC levels. SGC in the lung, liver, and kidney was severely depleted in Delta F508CFTR compared with wtCFTR mice, suggesting a role for CFTR in SGC biosynthesis.
Collapse
Affiliation(s)
- Hyun-Joo Park
- Research Institute, Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | | | | | | | | | |
Collapse
|
47
|
Functional Rescue of DeltaF508-CFTR by Peptides Designed to Mimic Sorting Motifs. ACTA ACUST UNITED AC 2009; 16:520-30. [DOI: 10.1016/j.chembiol.2009.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 04/05/2009] [Accepted: 04/13/2009] [Indexed: 11/17/2022]
|
48
|
White E, McKenna J, Cavanaugh A, Breitwieser GE. Pharmacochaperone-mediated rescue of calcium-sensing receptor loss-of-function mutants. Mol Endocrinol 2009; 23:1115-23. [PMID: 19389809 DOI: 10.1210/me.2009-0041] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The calcium sensing receptor (CaSR) is a Family C/3 G protein-coupled receptor that translates changes in extracellular Ca(2+) into diverse intracellular signals. Loss-of-function mutations in human CaSR cause familial hypocalciuric hypercalcemia and neonatal severe hyperparathyroidism. CaSR must navigate a number of endoplasmic reticulum quality control checkpoints during biosynthesis, including a conformational/functional checkpoint. Here we examine the biosynthesis of 25 CaSR mutations causing familial hypocalciuric hypercalcemia /neonatal severe hyperparathyroidism using immunoprecipitation, biotinylation, and functional assays. We define classes of CaSR mutants based on their biosynthetic profile. Class I CaSR mutants are not rescued to the plasma membrane. To dissect the organellar compartments that class I mutants can access, we engineered a cleavage site for the proprotein convertase furin into the extracellular domain of wild-type CaSR and class I mutants. Based on absence or presence of cleavage fragments, we find most mutants are degraded from the endoplasmic reticulum (no furin-mediated cleavage), whereas others access the Golgi (furin-mediated cleavage) before degradation. Class II CaSR mutants show increased expression and/or enhanced plasma membrane localization upon treatment with MG132 or the pharmacochaperone NPS R-568, permitting assay of functional activity. Of the 10 CaSR mutants that exhibit plasma membrane localization, only two did not show enhanced functional activity after rescue with NPS R-568. The established approaches can be used with current and newly identified CaSR mutations to identify the location of biosynthetic block and to determine the likelihood of rescue by allosteric agonists.
Collapse
Affiliation(s)
- Elissa White
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania 17822-2604, USA
| | | | | | | |
Collapse
|
49
|
Fu L, Sztul E. ER-associated complexes (ERACs) containing aggregated cystic fibrosis transmembrane conductance regulator (CFTR) are degraded by autophagy. Eur J Cell Biol 2009; 88:215-26. [PMID: 19131141 DOI: 10.1016/j.ejcb.2008.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 11/20/2008] [Accepted: 11/21/2008] [Indexed: 11/18/2022] Open
Abstract
The ubiquitin-proteasome pathway and autophagy are the two major mechanisms responsible for the clearance of cellular proteins. We have used the yeast Saccharomyces cerevisiae as a model system and the cystic fibrosis transmembrane conductance regulator (CFTR) as a model substrate to study the interactive function of these two pathways in the degradation of misfolded proteins. EGFP-tagged human CFTR was introduced into yeast and expressed under a copper-inducible promoter. The localization and degradation of EGFP-CFTR in live cells were monitored by time-lapse imaging following its de novo synthesis. EGFP-CFTR first appears within the perinuclear and sub-cortical ER and is mobile within the plane of the membrane as assessed by fluorescence recovery after photobleaching (FRAP). This pool of EGFP-CFTR is subsequently degraded through a proteasome-dependent pathway that is inhibited in the pre1-1 yeast strain defective in proteasomal degradation. Prolonged expression of EGFP-CFTR leads to the sequestration of EGFP-CFTR molecules into ER structures called ER-associated complexes (ERACs). The sequestration of EGFP-CFTR into ERACs appears to be driven by aggregation since EGFP-CFTR molecules present within ERACs are immobile as measured by FRAP. Individual ERACs are cleared from cells through the autophagic pathway that is blocked in the atg6Delta and atg1Delta yeast strains defective in autophagy. Our results suggest that the proteasomal and the autophagic pathways function together to clear misfolded proteins from the ER.
Collapse
Affiliation(s)
- Lianwu Fu
- Department of Cell Biology, University of Alabama at Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA.
| | | |
Collapse
|
50
|
Wang X, Koulov AV, Kellner WA, Riordan JR, Balch WE. Chemical and biological folding contribute to temperature-sensitive DeltaF508 CFTR trafficking. Traffic 2008; 9:1878-93. [PMID: 18764821 DOI: 10.1111/j.1600-0854.2008.00806.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Proteostasis (Balch WE, Morimoto RI, Dillin A, Kelly JW. Adapting proteostasis for disease intervention. Science 2008;319:916-919) refers to the biology that maintains the proteome in health and disease. Proteostasis is challenged by the most common mutant in cystic fibrosis, DeltaF508, a chloride channel [the cystic fibrosis transmembrane conductance regulator (CFTR)] that exhibits a temperature-sensitive phenotype for coupling to the coatomer complex II (COPII) transport machine for exit from the endoplasmic reticulum. Whether rescue of export of DeltaF508 CFTR at reduced temperature simply reflects energetic stabilization of the chemical fold defined by its primary sequence or requires a unique proteostasis environment is unknown. We now show that reduced temperature (30 degrees C) export of DeltaF508 does not occur in some cell types, despite efficient export of wild-type CFTR. We find that DeltaF508 export requires a local biological folding environment that is sensitive to heat/stress-inducible factors found in some cell types, suggesting that the energetic stabilization by reduced temperature is necessary, but not sufficient, for export of DeltaF508. Thus, the cell may require a proteostasis environment that is in part distinct from the wild-type pathway to restore DeltaF508 coupling to COPII. These results are discussed in the context of the energetics of the protein fold and the potential application of small molecules to achieve a proteostasis environment favoring export of a functional form of DeltaF508.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|