51
|
Villamizar O, Waters SA, Scott T, Grepo N, Jaffe A, Morris KV. Mesenchymal Stem Cell exosome delivered Zinc Finger Protein activation of cystic fibrosis transmembrane conductance regulator. J Extracell Vesicles 2021; 10:e12053. [PMID: 33532041 PMCID: PMC7825549 DOI: 10.1002/jev2.12053] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis is a genetic disorder that results in a multi-organ disease with progressive respiratory decline which leads to premature death. Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene disrupts the capacity of the protein to function as a channel, transporting chloride ions and bicarbonate across epithelial cell membranes. Small molecule treatments targeted at potentiating or correcting CFTR have shown clinical benefits, but are only effective for a small percentage of individuals with specific CFTR mutations. To overcome this limitation, we engineered stromal-derived mesenchymal stem cells (MSC) and HEK293 cells to produce exosomes containing a novel CFTR Zinc Finger Protein fusion with transcriptional activation domains VP64, P65 and Rta to target the CFTR promoter (CFZF-VPR) and activate transcription. Treatment with CFZF-VPR results in robust activation of CFTR transcription in patient derived Human Bronchial Epithelial cells (HuBEC). We also find that CFZF-VPR can be packaged into MSC and HEK293 cell exosomes and delivered to HuBEC cells to potently activate CFTR expression. Connexin 43 appeared to be required for functional release of CFZF-VPR from exosomes. The observations presented here demonstrate that MSC derived exosomes can be used to deliver a packaged zinc finger activator to target cells and activate CFTR. The novel approach presented here offers a next-generation genetic therapy that may one day prove effective in treating patients afflicted with Cystic fibrosis.
Collapse
Affiliation(s)
- Olga Villamizar
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA
| | - Shafagh A Waters
- Faculty of Medicine School of Women's & Children's Health University of New South Wales (UNSW) Sydney NSW Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC) Faculty of Medicine University of New South Wales Sydney NSW Australia.,Department of Respiratory Medicine Sydney Children's Hospital Sydney NSW Australia
| | - Tristan Scott
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA
| | - Nicole Grepo
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA
| | - Adam Jaffe
- Faculty of Medicine School of Women's & Children's Health University of New South Wales (UNSW) Sydney NSW Australia.,Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC) Faculty of Medicine University of New South Wales Sydney NSW Australia.,Department of Respiratory Medicine Sydney Children's Hospital Sydney NSW Australia
| | - Kevin V Morris
- Center for Gene Therapy City of Hope-Beckman Research Institute at the City of Hope Duarte California USA.,School of Medical Science Griffith University, Gold Coast Campus 1 Parklands Dr Southport QLD Australia
| |
Collapse
|
52
|
Ng RN, Tai AS, Chang BJ, Stick SM, Kicic A. Overcoming Challenges to Make Bacteriophage Therapy Standard Clinical Treatment Practice for Cystic Fibrosis. Front Microbiol 2021; 11:593988. [PMID: 33505366 PMCID: PMC7829477 DOI: 10.3389/fmicb.2020.593988] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
Individuals with cystic fibrosis (CF) are given antimicrobials as prophylaxis against bacterial lung infection, which contributes to the growing emergence of multidrug resistant (MDR) pathogens isolated. Pathogens such as Pseudomonas aeruginosa that are commonly isolated from individuals with CF are armed with an arsenal of protective and virulence mechanisms, complicating eradication and treatment strategies. While translation of phage therapy into standard care for CF has been explored, challenges such as the lack of an appropriate animal model demonstrating safety in vivo exist. In this review, we have discussed and provided some insights in the use of primary airway epithelial cells to represent the mucoenvironment of the CF lungs to demonstrate safety and efficacy of phage therapy. The combination of phage therapy and antimicrobials is gaining attention and has the potential to delay the onset of MDR infections. It is evident that efforts to translate phage therapy into standard clinical practice have gained traction in the past 5 years. Ultimately, collaboration, transparency in data publications and standardized policies are needed for clinical translation.
Collapse
Affiliation(s)
- Renee N. Ng
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia
| | - Anna S. Tai
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Institute for Respiratory Health, School of Medicine, The University of Western Australia, Perth, WA, Australia
| | - Barbara J. Chang
- The Marshall Center for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Stephen M. Stick
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth, WA, Australia
- Center for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Perth, WA, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Center, Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Perth, WA, Australia
- Center for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Perth, WA, Australia
- Occupation and the Environment, School of Public Health, Curtin University, Perth, WA, Australia
| |
Collapse
|
53
|
A bird eye view on cystic fibrosis: An underestimated multifaceted chronic disorder. Life Sci 2020; 268:118959. [PMID: 33383045 DOI: 10.1016/j.lfs.2020.118959] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 01/19/2023]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease which involves the mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CF involves in the inflammatory processes and is considered as a multisystem disorder that is not confined to lungs, but it also affects other vital organs that leads to numerous co-morbidities. The respiratory disorder in the CF results in mortality and morbidity which is characterized by series of serious events involving mucus hypersecretion, microbial infections, airways obstruction, inflammation, destruction of epithelium, tissue remodeling and terminal lung diseases. Mucins are the high molecular weight glycoproteins important for the viscoelastic properties of the mucus, play a significant role in the disease mechanisms. Determining the functional association between the CFTR and mucins might help to identify the putative target for specific therapeutic approach. In fact, furin enzyme which helps in the entry of novel COVID-19 virus into the cell, is upregulated in CF and this can also serve as a potential target for CF treatment. Moreover, the use of nano-formulations for CF treatment is an area of research being widely studied as they have also demonstrated promising outcomes. The in-depth knowledge of non-coding RNAs like miRNAs and lncRNAs and their functional association with CFTR gene expression and mutation can provide a different range of opportunity to identify the promising therapeutic approaches for CF.
Collapse
|
54
|
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
|
55
|
Checa J, Aran JM. Airway Redox Homeostasis and Inflammation Gone Awry: From Molecular Pathogenesis to Emerging Therapeutics in Respiratory Pathology. Int J Mol Sci 2020; 21:E9317. [PMID: 33297418 PMCID: PMC7731288 DOI: 10.3390/ijms21239317] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/05/2020] [Indexed: 02/06/2023] Open
Abstract
As aerobic organisms, we are continuously and throughout our lifetime subjected to an oxidizing atmosphere and, most often, to environmental threats. The lung is the internal organ most highly exposed to this milieu. Therefore, it has evolved to confront both oxidative stress induced by reactive oxygen species (ROS) and a variety of pollutants, pathogens, and allergens that promote inflammation and can harm the airways to different degrees. Indeed, an excess of ROS, generated intrinsically or from external sources, can imprint direct damage to key structural cell components (nucleic acids, sugars, lipids, and proteins) and indirectly perturb ROS-mediated signaling in lung epithelia, impairing its homeostasis. These early events complemented with efficient recognition of pathogen- or damage-associated recognition patterns by the airway resident cells alert the immune system, which mounts an inflammatory response to remove the hazards, including collateral dead cells and cellular debris, in an attempt to return to homeostatic conditions. Thus, any major or chronic dysregulation of the redox balance, the air-liquid interface, or defects in epithelial proteins impairing mucociliary clearance or other defense systems may lead to airway damage. Here, we review our understanding of the key role of oxidative stress and inflammation in respiratory pathology, and extensively report current and future trends in antioxidant and anti-inflammatory treatments focusing on the following major acute and chronic lung diseases: acute lung injury/respiratory distress syndrome, asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and cystic fibrosis.
Collapse
Affiliation(s)
| | - Josep M. Aran
- Immune-Inflammatory Processes and Gene Therapeutics Group, IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| |
Collapse
|
56
|
Righetti G, Casale M, Tonelli M, Liessi N, Fossa P, Pedemonte N, Millo E, Cichero E. New Insights into the Binding Features of F508del CFTR Potentiators: A Molecular Docking, Pharmacophore Mapping and QSAR Analysis Approach. Pharmaceuticals (Basel) 2020; 13:ph13120445. [PMID: 33291847 PMCID: PMC7762081 DOI: 10.3390/ph13120445] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis (CF) is the autosomal recessive disorder most recurrent in Caucasian populations. To combat this disease, many life-prolonging therapies are required and deeply investigated, including the development of the so-called cystic fibrosis transmembrane conductance regulator (CFTR) modulators, such as correctors and potentiators. Combination therapy with the two series of drugs led to the approval of several multi-drug effective treatments, such as Orkambi, and to the recent promising evaluation of the triple-combination Elexacaftor-Tezacaftor-Ivacaftor. This scenario enlightened the effectiveness of the multi-drug approach to pave the way for the discovery of novel therapeutic agents to contrast CF. The recent X-crystallographic data about the human CFTR in complex with the well-known potentiator Ivacaftor (VX-770) opened the possibility to apply a computational study aimed to explore the key features involved in the potentiator binding. Herein, we discussed molecular docking studies performed onto the chemotypes so far discussed in the literature as CFTR potentiator, reporting the most relevant interactions responsible for their mechanism of action, involving Van der Waals interactions and π–π stacking with F236, Y304, F305 and F312, as well as H-bonding F931, Y304, S308 and R933. This kind of positioning will stabilize the effective potentiator at the CFTR channel. These data have been accompanied by pharmacophore analyses, which promoted the design of novel derivatives endowed with a main (hetero)aromatic core connected to proper substituents, featuring H-bonding moieties. A highly predictive quantitative-structure activity relationship (QSAR) model has been developed, giving a cross-validated r2 (r2cv) = 0.74, a non-cross validated r2 (r2ncv) = 0.90, root mean square error (RMSE) = 0.347, and a test set r2 (r2pred) = 0.86. On the whole, the results are expected to gain useful information to guide the further development and optimization of new CFTR potentiators.
Collapse
Affiliation(s)
- Giada Righetti
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (G.R.); (M.T.)
| | - Monica Casale
- Department of Pharmacy, Section of Chemistry and Food and Pharmaceutical Technologies, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy;
| | - Michele Tonelli
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (G.R.); (M.T.)
| | - Nara Liessi
- Center of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV 9, 16132 Genoa, Italy; (N.L.); (E.M.)
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Paola Fossa
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (G.R.); (M.T.)
- Correspondence: (P.F.); (E.C.); Tel.: +39-010-353-8238 (P.F.); +39-010-353-8370 (E.C.); Fax: +39-010-353-8399 (P.F.); +39-010-353-8399 (E.C.)
| | | | - Enrico Millo
- Center of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV 9, 16132 Genoa, Italy; (N.L.); (E.M.)
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (G.R.); (M.T.)
- Correspondence: (P.F.); (E.C.); Tel.: +39-010-353-8238 (P.F.); +39-010-353-8370 (E.C.); Fax: +39-010-353-8399 (P.F.); +39-010-353-8399 (E.C.)
| |
Collapse
|
57
|
Jackson L, Waters V. Factors influencing the acquisition and eradication of early Pseudomonas aeruginosa infection in cystic fibrosis. J Cyst Fibros 2020; 20:8-16. [PMID: 33172756 DOI: 10.1016/j.jcf.2020.10.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/02/2020] [Accepted: 10/27/2020] [Indexed: 12/21/2022]
Abstract
In recent years considerable improvements have been made in increasing the life expectancy of patients with cystic fibrosis. New highly effective modulator therapies targeting the underlying defect in the cystic fibrosis transmembrane conductance regulator protein are expected to enhance lifespan even further. However, chronic Pseudomonas aeruginosa pulmonary infections continue to threaten CF patient lung health and mortality rates. Early and aggressive antibiotic eradication therapies targeting P. aeruginosa are standard practice, but these eradication therapies fail in 10-40% of patients. The reasons for P. aeruginosa eradication failure remain unclear. Thus, this review summarizes the evidence to date for pseudomonal acquisition and eradication failure in the cystic fibrosis lung. A complex combination of host and bacterial factors are responsible for initial establishment of P. aeruginosa pulmonary infections. Moreover, host and pseudomonal factors, polymicrobial interactions, and antimicrobial limitations in relation to P. aeruginosa eradication therapy failure are summarized.
Collapse
Affiliation(s)
- Lindsay Jackson
- Translational Medicine, Hospital for Sick Children, Toronto, Canada.
| | - Valerie Waters
- Translational Medicine, Hospital for Sick Children, Toronto, Canada; Infectious Diseases, Department of Pediatrics, Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
58
|
Molecular Docking and QSAR Studies as Computational Tools Exploring the Rescue Ability of F508del CFTR Correctors. Int J Mol Sci 2020; 21:ijms21218084. [PMID: 33138251 PMCID: PMC7663332 DOI: 10.3390/ijms21218084] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is the autosomal recessive disorder most recurrent in Caucasian populations. Different mutations involving the cystic fibrosis transmembrane regulator protein (CFTR) gene, which encodes the CFTR channel, are involved in CF. A number of life-prolonging therapies have been conceived and deeply investigated to combat this disease. Among them, the administration of the so-called CFTR modulators, such as correctors and potentiators, have led to quite beneficial effects. Recently, based on QSAR (quantitative structure activity relationship) studies, we reported the rational design and synthesis of compound 2, an aminoarylthiazole-VX-809 hybrid derivative exhibiting promising F508del-CFTR corrector ability. Herein, we explored the docking mode of the prototype VX-809 as well as of the aforementioned correctors in order to derive useful guidelines for the rational design of further analogues. In addition, we refined our previous QSAR analysis taking into account our first series of in-house hybrids. This allowed us to optimize the QSAR model based on the chemical structure and the potency profile of hybrids as F508del-CFTR correctors, identifying novel molecular descriptors explaining the SAR of the dataset. This study is expected to speed up the discovery process of novel potent CFTR modulators.
Collapse
|
59
|
Characterization of the mechanism of action of RDR01752, a novel corrector of F508del-CFTR. Biochem Pharmacol 2020; 180:114133. [DOI: 10.1016/j.bcp.2020.114133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/16/2022]
|
60
|
Çobanoğlu N, Özçelik U, Çakır E, Şişmanlar Eyüboğlu T, Pekcan S, Cinel G, Yalçın E, Kiper N, Emiralioğlu N, Şen V, Şen HS, Ercan Ö, Çokuğraş H, Kılınç AA, Al Shadfan LM, Yazan H, Altıntaş DU, Karagöz D, Demir E, Kartal Öztürk G, Bingöl A, Başaran AE, Sapan N, Çekiç Ş, Çelebioğlu E, Aslan AT, Gürsoy TR, Tuğcu G, Özdemir A, Harmancı K, Yıldırım GK, Köse M, Hangül M, Tamay Z, Süleyman A, Yüksel H, Yılmaz Ö, Özcan G, Topal E, Can D, Korkmaz Ekren P, Çaltepe G, Kılıç M, Özdoğan Ş, Doğru D. Patients eligible for modulator drugs: Data from cystic fibrosis registry of Turkey. Pediatr Pulmonol 2020; 55:2302-2306. [PMID: 32453906 DOI: 10.1002/ppul.24854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/22/2020] [Accepted: 05/09/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND A better understanding of cystic fibrosis transmembrane conductance regulator biology has led to the development of modulator drugs such as ivacaftor, lumacaftor-ivacaftor, tezacaftor-ivacaftor, and elexacaftor-tezacaftor-ivacaftor. This cross-sectional study evaluated cystic fibrosis (CF) patients eligible for modulator drugs. METHODS Data for age and genetic mutations from the Cystic Fibrosis Registry of Turkey collected in 2018 were used to find out the number of patients who are eligible for modulator therapy. RESULTS Of registered 1488 CF patients, genetic analysis was done for 1351. The numbers and percentages of patients and names of the drugs, that the patients are eligible for, are as follows: 122 (9.03%) for ivacaftor, 156 (11.54%) for lumacaftor-ivacaftor, 163 (11.23%) for tezacaftor-ivacaftor, and 57 (4.21%) for elexacaftor-tezacaftor-ivacaftor. Among 1351 genotyped patients total of 313 (23.16%) patients are eligible for currently licensed modulator therapies (55 patients were shared by ivacaftor and tezacaftor-ivacaftor, 108 patients were shared by lumacaftor-ivacaftor and tezacaftor-ivacaftor, and 22 patients were shared by tezacaftor-ivacaftor and elexacaftor-tezacaftor-ivacaftor groups). CONCLUSIONS The present study shows that approximately one-fourth of the registered CF patients in Turkey are eligible for modulator drugs. As, frequent mutations that CF patients have in Turkey are different from North American and European CF patients, developing modulator drugs effective for those mutations is necessary. Furthermore, as modulator drugs are very expensive currently, financial support of the government in developing countries like Turkey is noteworthy.
Collapse
Affiliation(s)
- Nazan Çobanoğlu
- Division of Pediatric Pulmonology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Uğur Özçelik
- Division of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Erkan Çakır
- Division of Pediatric Pulmonology, Faculty of Medicine, Bezmialem University, İstanbul, Turkey
| | | | - Sevgi Pekcan
- Division of Pediatric Pulmonology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Güzin Cinel
- Division of Pediatric Pulmonology, Ankara Children's Hematology Oncology Training and Research Hospital, Ankara, Turkey
| | - Ebru Yalçın
- Division of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Nural Kiper
- Division of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Nagehan Emiralioğlu
- Division of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Velat Şen
- Division of Pediatric Pulmonology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Hadice Selimoğlu Şen
- Department of Pulmonology, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Ömür Ercan
- Division of Pediatric Pulmonology, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Haluk Çokuğraş
- Division of Pediatric Pulmonology, Cerrahpaşa Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | - Ayşe Ayzıt Kılınç
- Division of Pediatric Pulmonology, Cerrahpaşa Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | | | - Hakan Yazan
- Division of Pediatric Pulmonology, Faculty of Medicine, Bezmialem University, İstanbul, Turkey
| | - Derya Ufuk Altıntaş
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Dilek Karagöz
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Esen Demir
- Division of Pediatric Pulmonology, Faculty of Medicine, Ege University, İzmir, Turkey
| | - Gökçen Kartal Öztürk
- Division of Pediatric Pulmonology, Faculty of Medicine, Ege University, İzmir, Turkey
| | - Ayşen Bingöl
- Division of Pediatric Pulmonology, Allergy and Immunology, Faculty of Medicine Akdeniz University, Antalya, Turkey
| | - Abdurrahman Erdem Başaran
- Division of Pediatric Pulmonology, Allergy and Immunology, Faculty of Medicine Akdeniz University, Antalya, Turkey
| | - Nihat Sapan
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Uludağ University, Bursa, Turkey
| | - Şükrü Çekiç
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Uludağ University, Bursa, Turkey
| | - Ebru Çelebioğlu
- Department of Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ayşe Tana Aslan
- Division of Pediatric Pulmonology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Tuğba Ramaslı Gürsoy
- Division of Pediatric Pulmonology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Gökçen Tuğcu
- Division of Pediatric Pulmonology, Ankara Children's Hematology Oncology Training and Research Hospital, Ankara, Turkey
| | - Ali Özdemir
- Division of Pediatric Pulmonology, Mersin City Training and Research Hospital, Mersin, Turkey
| | - Koray Harmancı
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Gonca Kılıç Yıldırım
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Eskişehir Osmangazi University, Eskişehir, Turkey
| | - Mehmet Köse
- Division of Pediatric Pulmonology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Melih Hangül
- Division of Pediatric Pulmonology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Zeynep Tamay
- Division of Pediatric Allergy and Pulmonology, İstanbul Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | - Ayşe Süleyman
- Division of Pediatric Allergy and Pulmonology, İstanbul Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | - Hasan Yüksel
- Division of Pediatric Pulmonology, Allergy and Immunology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Özge Yılmaz
- Division of Pediatric Pulmonology, Allergy and Immunology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Gizem Özcan
- Division of Pediatric Pulmonology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Erdem Topal
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Demet Can
- Division of Pediatric Pulmonology, Faculty of Medicine, Balıkesir University, Balıkesir, Turkey
| | | | - Gönül Çaltepe
- Division of Pediatric Gastroenterology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Mehmet Kılıç
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Fırat University, Elazığ, Turkey
| | - Şebnem Özdoğan
- Division of Pediatric Pulmonology, Sarıyer Hamidiye Etfal Training and Research Hospital, İstanbul, Turkey
| | - Deniz Doğru
- Division of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
61
|
Olivença DV, Voit EO, Pinto FR. ENaC regulation by phospholipids and DGK explained through mathematical modeling. Sci Rep 2020; 10:13952. [PMID: 32811866 PMCID: PMC7435262 DOI: 10.1038/s41598-020-70630-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/21/2020] [Indexed: 01/16/2023] Open
Abstract
Cystic fibrosis is a condition caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). It is also thought to increase the activity of epithelial sodium channels (ENaC). The altered function of these ion channels is one of the causes of the thick dehydrated mucus that characterizes the disease and is partially responsible for recurrent pulmonary infections and inflammation events that ultimately destroy the lungs of affected subjects. Phosphoinositides are signaling lipids that regulate numerous cellular processes and membrane proteins, including ENaC. Inhibition of diacylglycerol kinase (DGK), an enzyme of the phosphoinositide pathway, reduces ENaC function. We propose a computational analysis that is based on the combination of two existing mathematical models: one representing the dynamics of phosphoinositides and the other explaining how phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) influences ENaC activity and, consequently, airway surface liquid. This integrated model permits, for the first time, a detailed assessment of the intricate interactions between DGK and ENaC and is consistent with available literature data. In particular, the computational approach allows comparisons of two competing hypotheses regarding the regulation of ENaC. The results strongly suggest that the regulation of ENaC is primarily exerted through the control of PI(4,5)P2 production by type-I phosphatidylinositol-4-phosphate 5-kinase (PIP5KI), which in turn is controlled by phosphatidic acid (PA), the product of the DGK reaction.
Collapse
Affiliation(s)
- Daniel V. Olivença
- Faculty of Sciences, BioISI – Biosystems and Integrative Sciences Institute, University of Lisboa, 1749-016 Lisbon, Portugal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Atlanta, GA 30332-2000 USA
| | - Eberhard O. Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Atlanta, GA 30332-2000 USA
| | - Francisco R. Pinto
- Faculty of Sciences, BioISI – Biosystems and Integrative Sciences Institute, University of Lisboa, 1749-016 Lisbon, Portugal
| |
Collapse
|
62
|
Coelho CC, Aquino EDS, Diniz ALR, Santos MDS, Oliveira LCD, Poeiras PTC, Pereira DAG. Tissue oxygenation in peripheral muscles and functional capacity in cystic fibrosis: a cross-sectional study. Exp Physiol 2020; 105:1571-1578. [PMID: 32770583 DOI: 10.1113/ep088764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/06/2020] [Indexed: 12/21/2022]
Abstract
NEW FINDINGS What is the central question of this study? How do peripheral muscle tissue oxygenation and physical conditioning levels of children and adolescents with cystic fibrosis compare to demographically matched controls? What is the main finding and its importance? Children and adolescents with cystic fibrosis consumed more oxygen, more quickly and exhibited slower recovery, demonstrating that there may have been deficiencies in oxygen supply related to both oxygen uptake and oxygen transport. ABSTRACT Cystic fibrosis affects skeletal muscle performance and functional capacity. However, it is currently unclear how peripheral muscle behaviour is affected, especially in children and adolescents. To examine this, we compared tissue oxygenation of children and adolescents with cystic fibrosis against healthy volunteers. We also evaluated the functional capacity of participants via the modified shuttle test (MST) and assessed for associations between performance and near-infrared spectroscopy. A total of 124 participants enrolled. Participants were divided into either the cystic fibrosis group (CFG) or the healthy group (HG). Statistical comparisons between groups were evaluated with the Mann-Whitney U test and associations with functional capacity were evaluated using Spearman's correlation coefficient. CFG volunteers scored lower on the MST compared to the HG. They walked shorter distances (P = 0.001) with less efficiency because they performed the tests with a less efficient walking economy (P = 0.001) and a greater deoxyhaemoglobin concentration (P = 0.001). Further, they experienced reduced tissue oxygen saturation (P = 0.037) faster than the HG. As a result, they presented lower respiratory (P = 0.001) and lower heart (P = 0.001) rate values at the end of the MST, with a longer post-test heart rate recovery time (P = 0.005). There was a significant association between deoxygenation time and functional capacity. The CFG consumed more oxygen, more quickly, with a slower recovery, reflecting impairments in the dynamics of muscle oxygen extraction. The results suggest differences in functional capacity and haemodynamic recovery in children and adolescents with cystic fibrosis.
Collapse
Affiliation(s)
- Cristiane Cenachi Coelho
- Hospital Infantil João Paulo II - FHEMIG, Brazil.,Program in Rehabilitation Sciences, Universidade Federal de Minas Gerais, Brazil
| | | | | | | | | | | | | |
Collapse
|
63
|
Almughem FA, Aldossary AM, Tawfik EA, Alomary MN, Alharbi WS, Alshahrani MY, Alshehri AA. Cystic Fibrosis: Overview of the Current Development Trends and Innovative Therapeutic Strategies. Pharmaceutics 2020; 12:E616. [PMID: 32630625 PMCID: PMC7407299 DOI: 10.3390/pharmaceutics12070616] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Cystic Fibrosis (CF), an autosomal recessive genetic disease, is caused by a mutation in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR). This mutation reduces the release of chloride ions (Cl-) in epithelial tissues, and hyperactivates the epithelial sodium channels (ENaC) which aid in the absorption of sodium ions (Na+). Consequently, the mucus becomes dehydrated and thickened, making it a suitable medium for microbial growth. CF causes several chronic lung complications like thickened mucus, bacterial infection and inflammation, progressive loss of lung function, and ultimately, death. Until recently, the standard of clinical care in CF treatment had focused on preventing and treating the disease complications. In this review, we have summarized the current knowledge on CF pathogenesis and provided an outlook on the current therapeutic approaches relevant to CF (i.e., CFTR modulators and ENaC inhibitors). The enormous potential in targeting bacterial biofilms using antibiofilm peptides, and the innovative therapeutic strategies in using the CRISPR/Cas approach as a gene-editing tool to repair the CFTR mutation have been reviewed. Finally, we have discussed the wide range of drug delivery systems available, particularly non-viral vectors, and the optimal properties of nanocarriers which are essential for successful drug delivery to the lungs.
Collapse
Affiliation(s)
- Fahad A. Almughem
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (F.A.A.); (E.A.T.)
| | - Ahmad M. Aldossary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (A.M.A.); (M.N.A.)
| | - Essam A. Tawfik
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (F.A.A.); (E.A.T.)
| | - Mohammad N. Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (A.M.A.); (M.N.A.)
| | - Waleed S. Alharbi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
| | - Mohammad Y. Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, P.O. Box 61413, Abha 9088, Saudi Arabia;
| | - Abdullah A. Alshehri
- National Centre for Pharmaceutical Technology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia; (F.A.A.); (E.A.T.)
| |
Collapse
|
64
|
Hu JY, Yang P, Wegner DJ, Heins HB, Luke CJ, Li F, White FV, Silverman GA, Cole FS, Wambach JA. Functional characterization of four ATP-binding cassette transporter A3 gene (ABCA3) variants. Hum Mutat 2020; 41:1298-1307. [PMID: 32196812 PMCID: PMC7292786 DOI: 10.1002/humu.24014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 12/30/2022]
Abstract
ABCA3 transports phospholipids across lamellar body membranes in pulmonary alveolar type II cells and is required for surfactant assembly. Rare, biallelic, pathogenic ABCA3 variants result in lethal neonatal respiratory distress syndrome and childhood interstitial lung disease. Qualitative functional characterization of ABCA3 missense variants suggests two pathogenic classes: disrupted intracellular trafficking (type I mutant) or impaired ATPase-mediated phospholipid transport into the lamellar bodies (type II mutant). We qualitatively compared wild-type (WT-ABCA3) with four uncharacterized ABCA3 variants (c.418A>C;p.Asn140His, c.3609_3611delCTT;p.Phe1203del, c.3784A>G;p.Ser1262Gly, and c.4195G>A;p.Val1399Met) in A549 cells using protein processing, colocalization with intracellular organelles, lamellar body ultrastructure, and ATPase activity. We quantitatively measured lamellar body-like vesicle diameter and intracellular ABCA3 trafficking using fluorescence-based colocalization. Three ABCA3 variants (p.Asn140His, p.Ser1262Gly, and p.Val1399Met) were processed and trafficked normally and demonstrated well-organized lamellar body-like vesicles, but had reduced ATPase activity consistent with type II mutants. P.Phe1203del was processed normally, had reduced ATPase activity, and well-organized lamellar body-like vesicles, but quantitatively colocalized with both endoplasmic reticulum and lysosomal markers, an intermediate phenotype suggesting disruption of both intracellular trafficking and phospholipid transport. All ABCA3 mutants demonstrated mean vesicle diameters smaller than WT-ABCA3. Qualitative and quantitative functional characterization of ABCA3 variants informs mechanisms of pathogenicity.
Collapse
Affiliation(s)
- June Y. Hu
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Ping Yang
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel J. Wegner
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Hillary B. Heins
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Cliff J. Luke
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Fuhai Li
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri
| | - Frances V. White
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Gary A. Silverman
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - F. Sessions Cole
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer A. Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
65
|
Guerra L, Favia M, Di Gioia S, Laselva O, Bisogno A, Casavola V, Colombo C, Conese M. The preclinical discovery and development of the combination of ivacaftor + tezacaftor used to treat cystic fibrosis. Expert Opin Drug Discov 2020; 15:873-891. [PMID: 32290721 DOI: 10.1080/17460441.2020.1750592] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Cystic Fibrosis (CF) is caused by mutations in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene. The most common mutation, F508del, induces protein misprocessing and loss of CFTR function. The discovery through in vitro studies of the CFTR correctors (i.e. lumacaftor, tezacaftor) that partially rescue the misprocessing of F508del-CFTR with the potentiator ivacaftor is promising in giving an unprecedented clinical benefit in affected patients. AREAS COVERED Online databases were searched using key phrases for CF and CFTR modulators. Tezacaftor-ivacaftor treatment has proved to be safer than lumacaftor-ivacaftor, although clinical efficacy is similar. Further clinical efficacy has ensued with the introduction of triple therapy, i.e. applying second-generation correctors, such as VX-569 and VX-445 (elexacaftor) to tezacaftor-ivacaftor. The triple combinations will herald the availability of etiologic therapies for patients for whom no CFTR modulators are currently applied (i.e. F508del/minimal function mutations) and enhance CFTR modulator therapy for patients homozygous for F508del. EXPERT OPINION CF patient-derived tissue models are being explored to determine donor-specific response to current approved and future novel CFTR modulators for F508del and other rare mutations. The discovery and validation of biomarkers of CFTR modulation will complement these studies in the long term and in real-life world.
Collapse
Affiliation(s)
- Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia , Foggia, Italy
| | - Onofrio Laselva
- Programme in Molecular Medicine, Research Institute, Hospital for Sick Children , Toronto, Ontario, Canada.,Department of Physiology, University of Toronto , Toronto, Ontario, Canada
| | - Arianna Bisogno
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Regionale di Riferimento per la Fibrosi Cistica, Università degli Studi di Milano , Milan, Italy
| | - Valeria Casavola
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari , Bari, Italy
| | - Carla Colombo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Regionale di Riferimento per la Fibrosi Cistica, Università degli Studi di Milano , Milan, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia , Foggia, Italy
| |
Collapse
|
66
|
Small Molecule Anion Carriers Correct Abnormal Airway Surface Liquid Properties in Cystic Fibrosis Airway Epithelia. Int J Mol Sci 2020; 21:ijms21041488. [PMID: 32098269 PMCID: PMC7073096 DOI: 10.3390/ijms21041488] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 01/03/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disease characterized by the lack of cystic fibrosis transmembrane conductance regulator (CFTR) protein expressed in epithelial cells. The resulting defective chloride and bicarbonate secretion and imbalance of the transepithelial homeostasis lead to abnormal airway surface liquid (ASL) composition and properties. The reduced ASL volume impairs ciliary beating with the consequent accumulation of sticky mucus. This situation prevents the normal mucociliary clearance, favouring the survival and proliferation of bacteria and contributing to the genesis of CF lung disease. Here, we have explored the potential of small molecules capable of facilitating the transmembrane transport of chloride and bicarbonate in order to replace the defective transport activity elicited by CFTR in CF airway epithelia. Primary human bronchial epithelial cells obtained from CF and non-CF patients were differentiated into a mucociliated epithelia in order to assess the effects of our compounds on some key properties of ASL. The treatment of these functional models with non-toxic doses of the synthetic anionophores improved the periciliary fluid composition, reducing the fluid re-absorption, correcting the ASL pH and reducing the viscosity of the mucus, thus representing promising drug candidates for CF therapy.
Collapse
|
67
|
Lopes-Pacheco M. CFTR Modulators: The Changing Face of Cystic Fibrosis in the Era of Precision Medicine. Front Pharmacol 2020; 10:1662. [PMID: 32153386 PMCID: PMC7046560 DOI: 10.3389/fphar.2019.01662] [Citation(s) in RCA: 298] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022] Open
Abstract
Cystic fibrosis (CF) is a lethal inherited disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, which result in impairment of CFTR mRNA and protein expression, function, stability or a combination of these. Although CF leads to multifaceted clinical manifestations, the respiratory disorder represents the major cause of morbidity and mortality of these patients. The life expectancy of CF patients has substantially lengthened due to early diagnosis and improvements in symptomatic therapeutic regimens. Quality of life remains nevertheless limited, as these individuals are subjected to considerable clinical, psychosocial and economic burdens. Since the discovery of the CFTR gene in 1989, tremendous efforts have been made to develop therapies acting more upstream on the pathogenesis cascade, thereby overcoming the underlying dysfunctions caused by CFTR mutations. In this line, the advances in cell-based high-throughput screenings have been facilitating the fast-tracking of CFTR modulators. These modulator drugs have the ability to enhance or even restore the functional expression of specific CF-causing mutations, and they have been classified into five main groups depending on their effects on CFTR mutations: potentiators, correctors, stabilizers, read-through agents, and amplifiers. To date, four CFTR modulators have reached the market, and these pharmaceutical therapies are transforming patients' lives with short- and long-term improvements in clinical outcomes. Such breakthroughs have paved the way for the development of novel CFTR modulators, which are currently under experimental and clinical investigations. Furthermore, recent insights into the CFTR structure will be useful for the rational design of next-generation modulator drugs. This review aims to provide a summary of recent developments in CFTR-directed therapeutics. Barriers and future directions are also discussed in order to optimize treatment adherence, identify feasible and sustainable solutions for equitable access to these therapies, and continue to expand the pipeline of novel modulators that may result in effective precision medicine for all individuals with CF.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
68
|
Ivey G, Youker RT. Disease-relevant mutations alter amino acid co-evolution networks in the second nucleotide binding domain of CFTR. PLoS One 2020; 15:e0227668. [PMID: 31978131 PMCID: PMC6980524 DOI: 10.1371/journal.pone.0227668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 12/25/2019] [Indexed: 01/23/2023] Open
Abstract
Cystic Fibrosis (CF) is an inherited disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel. Mutations in CFTR cause impaired chloride ion transport in the epithelial tissues of patients leading to cardiopulmonary decline and pancreatic insufficiency in the most severely affected patients. CFTR is composed of twelve membrane-spanning domains, two nucleotide-binding domains (NBDs), and a regulatory domain. The most common mutation in CFTR is a deletion of phenylalanine at position 508 (ΔF508) in NBD1. Previous research has primarily concentrated on the structure and dynamics of the NBD1 domain; However numerous pathological mutations have also been found in the lesser-studied NBD2 domain. We have investigated the amino acid co-evolved network of interactions in NBD2, and the changes that occur in that network upon the introduction of CF and CF-related mutations (S1251N(T), S1235R, D1270N, N1303K(T)). Extensive coupling between the α- and β-subdomains were identified with residues in, or near Walker A, Walker B, H-loop and C-loop motifs. Alterations in the predicted residue network varied from moderate for the S1251T perturbation to more severe for N1303T. The S1235R and D1270N networks varied greatly compared to the wildtype, but these CF mutations only affect ion transport preference and do not severely disrupt CFTR function, suggesting dynamic flexibility in the network of interactions in NBD2. Our results also suggest that inappropriate interactions between the β-subdomain and Q-loop could be detrimental. We also identified mutations predicted to stabilize the NBD2 residue network upon introduction of the CF and CF-related mutations, and these predicted mutations are scored as benign by the MUTPRED2 algorithm. Our results suggest the level of disruption of the co-evolution predictions of the amino acid networks in NBD2 does not have a straightforward correlation with the severity of the CF phenotypes observed.
Collapse
Affiliation(s)
- Gabrianne Ivey
- Kyder Christian Academy, Franklin, North Carolina, United States of America
- Southwestern Community College, Sylva, North Carolina, United States of America
| | - Robert T. Youker
- Department of Biology, Western Carolina University, Cullowhee, North Carolina, United States of America
| |
Collapse
|
69
|
Bellisola G, Caldrer S, Cestelli-Guidi M, Cinque G. Infrared biomarkers of impaired cystic fibrosis transmembrane regulator protein biogenesis. JOURNAL OF BIOPHOTONICS 2020; 13:e201900174. [PMID: 31654605 DOI: 10.1002/jbio.201900174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/26/2019] [Accepted: 10/03/2019] [Indexed: 06/10/2023]
Abstract
The mid-infrared (IR) spectra of human cystic fibrosis (CF) cells acquired by Fourier transform infrared microspectroscopy were compared with those of non-CF cells. Within the 1700 to 1480 cm-1 spectral domain of amides, unsupervised explorative principal component analysis identified a few variables reflecting quantitative and qualitative vibrations arising from protein secondary structures and amino acid side chains. Their pattern reflected α-helix to β-sheet transitions in bronchial epithelial cells and in immortalized peripheral blood mononuclear cells from patients with R1162X missense or in-frame F508del mutations in the cystic fibrosis transmembrane regulator gene (Cftr). Similar transitions have been described in IR spectra of cells, tissues and body fluids of patients affected with some neurodegenerative diseases characterized by the accumulation of misfolded protein aggregates. The variables pattern was able to distinguish CF cells from non-CF cells and was modified by molecular compounds used to rescue the unbalanced folding process of mutated cystic fibrosis transmembrane regulator (CFTR) anion channel. To our knowledge, this is the first experimental evidence of spectroscopic biomarkers of the impaired biogenesis of CFTR by IR microanalysis in the spectra of human CF bronchial epithelial and lymphoblastoid cells.
Collapse
Affiliation(s)
- Giuseppe Bellisola
- Istituto Nazionale di Fisica Nucleare - Laboratori Nazionali di Frascati, Frascati, Rome, Italy
| | - Sara Caldrer
- IRCSS Sacro Cuore - Don Calabria, Centro Malattie Tropicali, Negrar, Verona, Italy
| | | | - Gianfelice Cinque
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, Oxfordshire, UK
| |
Collapse
|
70
|
Tosco A, Villella VR, Raia V, Kroemer G, Maiuri L. Cystic Fibrosis: New Insights into Therapeutic Approaches. CURRENT RESPIRATORY MEDICINE REVIEWS 2020. [DOI: 10.2174/1573398x15666190702151613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Since the identification of Cystic Fibrosis (CF) as a disease in 1938 until 2012, only
therapies to treat symptoms rather than etiological therapies have been used to treat the disease. Over
the last few years, new technologies have been developed, and gene editing strategies are now
moving toward a one-time cure. This review will summarize recent advances in etiological therapies
that target the basic defect in the CF Transmembrane Receptor (CFTR), the protein that is mutated in
CF. We will discuss how newly identified compounds can directly target mutated CFTR to improve
its function. Moreover, we will discuss how proteostasis regulators can modify the environment in
which the mutant CFTR protein is synthesized and decayed, thus restoring CFTR function. The
future of CF therapies lies in combinatory therapies that may be personalized for each CF patient.
Collapse
Affiliation(s)
- Antonella Tosco
- Department of Translational Medical Sciences, Pediatric Unit, Regional Cystic Fibrosis Center, Federico II University, Naples 80131, Italy
| | - Valeria R. Villella
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Valeria Raia
- Department of Translational Medical Sciences, Pediatric Unit, Regional Cystic Fibrosis Center, Federico II University, Naples 80131, Italy
| | - Guido Kroemer
- Equipe11 labellisee Ligue Nationale Contrele Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - Luigi Maiuri
- Division of Genetics and Cell Biology, European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan 20132, Italy
| |
Collapse
|
71
|
Velino C, Carella F, Adamiano A, Sanguinetti M, Vitali A, Catalucci D, Bugli F, Iafisco M. Nanomedicine Approaches for the Pulmonary Treatment of Cystic Fibrosis. Front Bioeng Biotechnol 2019; 7:406. [PMID: 31921811 PMCID: PMC6927921 DOI: 10.3389/fbioe.2019.00406] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease affecting today nearly 70,000 patients worldwide and characterized by a hypersecretion of thick mucus difficult to clear arising from the defective CFTR protein. The over-production of the mucus secreted in the lungs, along with its altered composition and consistency, results in airway obstruction that makes the lungs susceptible to recurrent and persistent bacterial infections and endobronchial chronic inflammation, which are considered the primary cause of bronchiectasis, respiratory failure, and consequent death of patients. Despite the difficulty of treating the continuous infections caused by pathogens in CF patients, various strategies focused on the symptomatic therapy have been developed during the last few decades, showing significant positive impact on prognosis. Moreover, nowadays, the discovery of CFTR modulators as well as the development of gene therapy have provided new opportunity to treat CF. However, the lack of effective methods for delivery and especially targeted delivery of therapeutics specifically to lung tissues and cells limits the efficiency of the treatments. Nanomedicine represents an extraordinary opportunity for the improvement of current therapies and for the development of innovative treatment options for CF previously considered hard or impossible to treat. Due to the peculiar environment in which the therapies have to operate characterized by several biological barriers (pulmonary tract, mucus, epithelia, bacterial biofilm) the use of nanotechnologies to improve and enhance drug delivery or gene therapies is an extremely promising way to be pursued. The aim of this review is to revise the currently used treatments and to outline the most recent progresses about the use of nanotechnology for the management of CF.
Collapse
Affiliation(s)
- Cecilia Velino
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Francesca Carella
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Alessio Adamiano
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Maurizio Sanguinetti
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alberto Vitali
- Institute for the Chemistry of Molecular Recognition (ICRM), National Research Council (CNR), c/o Institute of Biochemistry and Clinical Biochemistry, Catholic University, Rome, Italy
| | - Daniele Catalucci
- Humanitas Clinical and Research Center, Rozzano, Italy
- Institute of Genetic and Biomedical Research (IRGB) - UOS Milan, National Research Council (CNR), Milan, Italy
| | - Francesca Bugli
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| |
Collapse
|
72
|
Lopes-Pacheco M, Pedemonte N, Kicic A. Editorial: Emerging Therapeutic Approaches for Cystic Fibrosis. Front Pharmacol 2019; 10:1440. [PMID: 31849681 PMCID: PMC6895245 DOI: 10.3389/fphar.2019.01440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 01/31/2023] Open
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute (BioISI), Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | | | - Anthony Kicic
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| |
Collapse
|
73
|
Meehan-Atrash J, Korzun T, Ziegler A. Cannabis Inhalation and Voice Disorders. JAMA Otolaryngol Head Neck Surg 2019; 145:956-964. [DOI: 10.1001/jamaoto.2019.1986] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Tetiana Korzun
- Department of Otolaryngology, Oregon Health & Science University, Portland
| | - Aaron Ziegler
- School of Medicine, Department of Otolaryngology–Head & Neck Surgery, Oregon Health & Science University, Portland
| |
Collapse
|
74
|
Daley T, Hughan K, Rayas M, Kelly A, Tangpricha V. Vitamin D deficiency and its treatment in cystic fibrosis. J Cyst Fibros 2019; 18 Suppl 2:S66-S73. [DOI: 10.1016/j.jcf.2019.08.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022]
|
75
|
Spanò V, Montalbano A, Carbone A, Scudieri P, Galietta LJV, Barraja P. An overview on chemical structures as ΔF508-CFTR correctors. Eur J Med Chem 2019; 180:430-448. [PMID: 31326599 DOI: 10.1016/j.ejmech.2019.07.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 10/26/2022]
Abstract
Deletion of phenylalanine at position 508 (F508del) in the CFTR protein, is the most common mutation causing cystic fibrosis (CF). F508del causes misfolding and rapid degradation of CFTR protein a defect that can be targeted with pharmacological agents termed "correctors". Correctors belong to various chemical classes but are generally small molecules based on nitrogen sulfur or oxygen heterocycles. The mechanism of action of correctors is generally unknown but there is experimental evidence that some of them can directly act on mutant CFTR improving folding and stability. Here we overview the characteristics of the various F508del correctors described so far to obtain indications on key chemical structures and modifications that are required for mutant protein rescue.
Collapse
Affiliation(s)
- Virginia Spanò
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Alessandra Montalbano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Anna Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Paolo Scudieri
- Telethon Institute of Genetics and Medicine (TIGEM), Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Campi Flegrei 34, 80078, Pozzuoli, NA, Italy; Department of Translational Medical Sciences (DISMET), University of Naples, "Federico II", 80131, Naples, Italy
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123, Palermo, Italy.
| |
Collapse
|
76
|
Hanrahan JW, Sato Y, Carlile GW, Jansen G, Young JC, Thomas DY. Cystic Fibrosis: Proteostatic correctors of CFTR trafficking and alternative therapeutic targets. Expert Opin Ther Targets 2019; 23:711-724. [PMID: 31169041 DOI: 10.1080/14728222.2019.1628948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Cystic fibrosis (CF) is the most frequent lethal orphan disease and is caused by mutations in the CFTR gene. The most frequent mutation F508del-CFTR affects multiple organs; infections and subsequent infections and complications in the lung lead to death. Areas covered: This review focuses on new targets and mechanisms that are attracting interest for the development of CF therapies. The F508del-CFTR protein is retained in the endoplasmic reticulum (ER) but has some function if it can traffic to the plasma membrane. Cell-based assays have been used to screen chemical libraries for small molecule correctors that restore its trafficking. Pharmacological chaperones are correctors that bind directly to the F508del-CFTR mutant and promote its folding and trafficking. Other correctors fall into a heterogeneous class of proteostasis modulators that act indirectly by altering cellular homeostasis. Expert opinion: Pharmacological chaperones have so far been the most successful correctors of F508del-CFTR trafficking, but their level of correction means that more than one corrector is required. Proteostasis modulators have low levels of correction but hold promise because some can correct several different CFTR mutations. Identification of their cellular targets and the potential for development may lead to new therapies for CF.
Collapse
Affiliation(s)
- John W Hanrahan
- a Department of Physiology , McGill University , Montréal , QC , Canada.,c Research Institute of the McGill University Health Centre , McGill University , Montréal , QC , Canada
| | - Yukiko Sato
- a Department of Physiology , McGill University , Montréal , QC , Canada.,b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada
| | - Graeme W Carlile
- b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada.,d Department of Biochemistry , McGill University , Montréal , QC , Canada
| | - Gregor Jansen
- d Department of Biochemistry , McGill University , Montréal , QC , Canada
| | - Jason C Young
- b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada.,d Department of Biochemistry , McGill University , Montréal , QC , Canada
| | - David Y Thomas
- b Cystic Fibrosis Translational Research centre , McGill University , Montréal , QC , Canada.,d Department of Biochemistry , McGill University , Montréal , QC , Canada.,e Department of Human Genetics , McGill University , Montréal , QC , Canada
| |
Collapse
|
77
|
Cystic Fibrosis Foundation Pulmonary Guidelines. Use of Cystic Fibrosis Transmembrane Conductance Regulator Modulator Therapy in Patients with Cystic Fibrosis. Ann Am Thorac Soc 2019; 15:271-280. [PMID: 29342367 DOI: 10.1513/annalsats.201707-539ot] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
RATIONALE Cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulators are a new class of medications targeting the underlying defect in CF. Ivacaftor (IVA) and IVA combined with lumacaftor (LUM; IVA/LUM) have been approved by the U.S. Food and Drug Administration (FDA) for use in patients with CF. However, the FDA label for these medications encompasses patient groups that were not studied as part of the drug approval process. CF clinicians, patients, and their families have recognized a need for recommendations to guide the use of these medications. OBJECTIVE Develop evidence-based guidelines for CFTR modulator therapy in patients with CF. METHODS A multidisciplinary committee of CF caregivers and patient representatives was assembled. A methodologist, an epidemiologist, a medical librarian, and a biostatistician were recruited to assist with the literature search, evidence grading, and generation of recommendations. The committee developed clinical questions using the Patient-Intervention-Comparison-Outcome format. A systematic review was conducted to find relevant publications. The evidence was then evaluated using the GRADE (Grading of Recommendations, Assessment, Development, and Evaluation) approach, and recommendations were made based on this analysis. RESULTS For adults and children aged 6 years and older with CF due to gating mutations other than G551D or R117H, the guideline panel made a conditional recommendation for treatment with IVA. For those with the R117H mutation, the guideline panel made a conditional recommendation for treatment with IVA for 1) adults aged 18 years or older, and 2) children aged 6-17 years with a forced expiratory volume in 1 second (FEV1) less than 90% predicted. For those with the R117H mutation, the guideline panel made a conditional recommendation against treatment with IVA for 1) children aged 12-17 years with an FEV1 greater than 90% predicted, and 2) children less than 6 years of age. Among those with two copies of F508del, the guideline panel made a strong recommendation for treatment with IVA/LUM for adults and children aged 12 years and older with an FEV1 less than 90% predicted; and made a conditional recommendation for treatment with IVA/LUM for 1) adults and children aged 12 years or older with an FEV1 greater than 90% predicted, and 2) children aged 6-11 years. CONCLUSIONS Using the GRADE approach, we have made recommendations for the use of CFTR modulators in patients with CF. These recommendations will be of help to CF clinicians, patients, and their families in guiding decisions regarding use of these medications.
Collapse
|
78
|
Chioccioli M, Feriani L, Kotar J, Bratcher PE, Cicuta P. Phenotyping ciliary dynamics and coordination in response to CFTR-modulators in Cystic Fibrosis respiratory epithelial cells. Nat Commun 2019; 10:1763. [PMID: 30992452 PMCID: PMC6467870 DOI: 10.1038/s41467-019-09798-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/29/2019] [Indexed: 12/22/2022] Open
Abstract
Personalized approaches for systematically assessing ciliary beat dynamics and for drug testing would improve the challenging task of diagnosing and treating respiratory disorders. In this pilot study, we show how multiscale differential dynamic microscopy (multi-DDM) can be used to characterize collective ciliary beating in a non-biased automated manner. We use multi-DDM to assess the efficacy of different CFTR-modulating drugs in human airway epithelial cells derived from subjects with cystic fibrosis (ΔF508/ΔF508 and ∆F508/-) based on ciliary beat frequency and coordination. Similar to clinical observations, drug efficacy is variable across donors, even within the same genotype. We show how our assay can quantitatively identify the most efficient drugs for restoring ciliary beating for each individual donor. Multi-DDM provides insight into ciliary beating responses following treatment with drugs, and has application in the broader context of respiratory disease and for drug screening.
Collapse
Affiliation(s)
- M Chioccioli
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06510, USA
| | - L Feriani
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
- Institute of Clinical Sciences, Imperial College London, London, SW7 2AZ, UK
- MRC London Institute of Medical Sciences, London, W12 0NN, UK
| | - J Kotar
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
| | - P E Bratcher
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, 80206, USA.
| | - P Cicuta
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK.
| |
Collapse
|
79
|
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease characterized by pancreatic insufficiency and chronic endobronchial airway infection. This latter feature results in progressive bronchiectasis and ultimately respiratory failure, which is the leading cause of death in patients with CF. Other complications include sinusitis, diabetes mellitus, bowel obstruction, hepatobiliary disease, hyponatremic dehydration, and infertility. Diagnosis of CF is confirmed by demonstration of elevated sweat chloride. Most cases of CF are identified through newborn screening (NBS). There are also infants with positive NBS but inconclusive diagnostic testing; a small proportion of these infants may go on to develop CF. CF is a lifelong, life-limiting disease, but an organized care center network with multidisciplinary approach, quality improvement initiatives, and research has led to markedly increased survival and development of adult CF care programs. In the past few years, medications that directly target the underlying CF defect have been developed, which should result in even greater survival benefits. [Pediatr Ann. 2019;48(4):e154-e161.].
Collapse
|
80
|
Villella VR, Tosco A, Esposito S, Bona G, Raia V, Maiuri L. Mutation-specific therapies and drug repositioning in cystic fibrosis. Minerva Pediatr 2019; 71:287-296. [DOI: 10.23736/s0026-4946.19.05506-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
81
|
Villella VR, Esposito S, Ferrari E, Monzani R, Tosco A, Rossin F, Castaldo A, Silano M, Marseglia GL, Romani L, Barlev NA, Piacentini M, Raia V, Kroemer G, Maiuri L. Autophagy suppresses the pathogenic immune response to dietary antigens in cystic fibrosis. Cell Death Dis 2019; 10:258. [PMID: 30874543 PMCID: PMC6420598 DOI: 10.1038/s41419-019-1500-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/22/2019] [Accepted: 02/28/2019] [Indexed: 12/30/2022]
Abstract
Under physiological conditions, a finely tuned system of cellular adaptation allows the intestinal mucosa to maintain the gut barrier function while avoiding excessive immune responses to non-self-antigens from dietary origin or from commensal microbes. This homeostatic function is compromised in cystic fibrosis (CF) due to loss-of-function mutations in the CF transmembrane conductance regulator (CFTR). Recently, we reported that mice bearing defective CFTR are abnormally susceptible to a celiac disease-like enteropathy, in thus far that oral challenge with the gluten derivative gliadin elicits an inflammatory response. However, the mechanisms through which CFTR malfunction drives such an exaggerated response to dietary protein remains elusive. Here we demonstrate that the proteostasis regulator/transglutaminase 2 (TGM2) inhibitor cysteamine restores reduced Beclin 1 (BECN1) protein levels in mice bearing cysteamine-rescuable F508del-CFTR mutant, either in homozygosis or in compound heterozygosis with a null allele, but not in knock-out CFTR mice. When cysteamine restored BECN1 expression, autophagy was increased and gliadin-induced inflammation was reduced. The beneficial effects of cysteamine on F508del-CFTR mice were lost when these mice were backcrossed into a Becn1 haploinsufficient/autophagy-deficient background. Conversely, the transfection-enforced expression of BECN1 in human intestinal epithelial Caco-2 cells mitigated the pro-inflammatory cellular stress response elicited by the gliadin-derived P31–43 peptide. In conclusion, our data provide the proof-of-concept that autophagy stimulation may mitigate the intestinal malfunction of CF patients.
Collapse
Affiliation(s)
- Valeria R Villella
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Speranza Esposito
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Ferrari
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy.,Department of Health Sciences, University of Eastern Piedmont, Novara, 28100, Italy
| | - Romina Monzani
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy.,Department of Health Sciences, University of Eastern Piedmont, Novara, 28100, Italy
| | - Antonella Tosco
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University Naples, Naples, 80131, Italy
| | - Federica Rossin
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Alice Castaldo
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University Naples, Naples, 80131, Italy
| | - Marco Silano
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Roma, Italy
| | - Gian Luigi Marseglia
- Dipartimento di Pediatria, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Nikolai A Barlev
- Gene Expression Laboratory, Institute of Citology, Saint-Petersburg, Russia
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University Naples, Naples, 80131, Italy
| | - Guido Kroemer
- Equipe11 labellisée Ligue Nationale contrele Cancer, Centre de Recherche des Cordeliers, Paris, France. .,INSERM U1138, Centre de Recherche des Cordeliers, Paris, France. .,Université Paris Descartes, Paris, France. .,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France. .,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France. .,Suzhou Institute for Systems Biology, Chinese Academy of Sciences, Suzhou, China. .,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, 17176, Sweden.
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy.,Department of Health Sciences, University of Eastern Piedmont, Novara, 28100, Italy
| |
Collapse
|
82
|
Patel W, Moore PJ, Sassano MF, Lopes-Pacheco M, Aleksandrov AA, Amaral MD, Tarran R, Gray MA. Increases in cytosolic Ca 2+ induce dynamin- and calcineurin-dependent internalisation of CFTR. Cell Mol Life Sci 2019; 76:977-994. [PMID: 30547226 PMCID: PMC6394554 DOI: 10.1007/s00018-018-2989-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/28/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-regulated, apical anion channel that regulates ion and fluid transport in many epithelia including the airways. We have previously shown that cigarette smoke (CS) exposure to airway epithelia causes a reduction in plasma membrane CFTR expression which correlated with a decrease in airway surface hydration. The effect of CS on CFTR was dependent on an increase in cytosolic Ca2+. However, the underlying mechanism for this Ca2+-dependent, internalisation of CFTR is unknown. To gain a better understanding of the effect of Ca2+ on CFTR, we performed whole cell current recordings to study the temporal effect of raising cytosolic Ca2+ on CFTR function. We show that an increase in cytosolic Ca2+ induced a time-dependent reduction in whole cell CFTR conductance, which was paralleled by a loss of cell surface CFTR expression, as measured by confocal and widefield fluorescence microscopy. The decrease in CFTR conductance and cell surface expression were both dynamin-dependent. Single channel reconstitution studies showed that raising cytosolic Ca2+ per se had no direct effect on CFTR. In fact, the loss of CFTR plasma membrane activity correlated with activation of calcineurin, a Ca2+-dependent phosphatase, suggesting that dephosphorylation of CFTR was linked to the loss of surface expression. In support of this, the calcineurin inhibitor, cyclosporin A, prevented the Ca2+-induced decrease in cell surface CFTR. These results provide a hitherto unrecognised role for cytosolic Ca2+ in modulating the residency of CFTR at the plasma membrane through a dynamin- and calcineurin-dependent mechanism.
Collapse
Affiliation(s)
- Waseema Patel
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick J Moore
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Flori Sassano
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miquéias Lopes-Pacheco
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| | - Andrei A Aleksandrov
- Department of Biochemistry and Biophysics, Cystic Fibrosis Research and Treatment Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Margarida D Amaral
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisbon, Portugal
| | - Robert Tarran
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, Cystic Fibrosis Research and Treatment Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael A Gray
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
83
|
Bodas M, Vij N. Adapting Proteostasis and Autophagy for Controlling the Pathogenesis of Cystic Fibrosis Lung Disease. Front Pharmacol 2019; 10:20. [PMID: 30774592 PMCID: PMC6367269 DOI: 10.3389/fphar.2019.00020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Cystic fibrosis (CF), a fatal genetic disorder predominant in the Caucasian population, is caused by mutations in the cystic fibrosis transmembrane conductance regulator (Cftr) gene. The most common mutation is the deletion of phenylalanine from the position-508 (F508del-CFTR), resulting in a misfolded-CFTR protein, which is unable to fold, traffic and retain its plasma membrane (PM) localization. The resulting CFTR dysfunction, dysregulates variety of key cellular mechanisms such as chloride ion transport, airway surface liquid (ASL) homeostasis, mucociliary-clearance, inflammatory-oxidative signaling, and proteostasis that includes ubiquitin-proteasome system (UPS) and autophagy. A collective dysregulation of these key homoeostatic mechanisms contributes to the development of chronic obstructive cystic fibrosis lung disease, instead of the classical belief focused exclusively on ion-transport defect. Hence, therapeutic intervention(s) aimed at rescuing chronic CF lung disease needs to correct underlying defect that mediates homeostatic dysfunctions and not just chloride ion transport. Since targeting all the myriad defects individually could be quite challenging, it will be prudent to identify a process which controls almost all disease-promoting processes in the CF airways including underlying CFTR dysfunction. There is emerging experimental and clinical evidence that supports the notion that impaired cellular proteostasis and autophagy plays a central role in regulating pathogenesis of chronic CF lung disease. Thus, correcting the underlying proteostasis and autophagy defect in controlling CF pulmonary disease, primarily via correcting the protein processing defect of F508del-CFTR protein has emerged as a novel intervention strategy. Hence, we discuss here both the rationale and significant therapeutic utility of emerging proteostasis and autophagy modulating drugs/compounds in controlling chronic CF lung disease, where targeted delivery is a critical factor-influencing efficacy.
Collapse
Affiliation(s)
- Manish Bodas
- Department of Medicine, University of Oklahoma, Oklahoma City, OK, United States
| | - Neeraj Vij
- Department of Pediatric Pulmonary Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- 4Dx Limited, Los Angeles, CA, United States
- VIJ Biotech LLC, Baltimore, MD, United States
| |
Collapse
|
84
|
[Patients with cystic fibrosis become adults : Treatment hopes and disappointments]. Internist (Berl) 2019; 60:98-108. [PMID: 30627755 DOI: 10.1007/s00108-018-0536-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Mucoviscidosis or cystic fibrosis (CF) is one of the most frequent monogenetic diseases in middle Europe. It is inherited in an autosomal recessive manner. A defect in the cystic fibrosis transmembrane conductance regulator (CFTR) channel reduces chloride ion transport to the cell membrane, which leads to malfunctions in all exocrine glands. This results in a progressive multiorgan disease, which leads to chronic inflammation and infections of the lungs. The progressive destruction of lung tissue with respiratory insufficiency is the most common cause of death in CF. Progress in symptomatic treatment over the past decades has led to a dramatic improvement in life expectation and quality of life for those affected, so that nowadays in nearly all industrial countries the majority of patients are adults. In 2012 the era of causal therapy of the CFTR protein defects was opened with the approval of ivacaftor. Long-term data now confirm the benefits. There is reason to hope that the success story of CF treatment will be continued, particularly by further CFTR modulators with innovative modes of action and improved efficacy; however, so far these are not available for all mutation classes, so that not all patients can reap the benefits. Therefore, the further development of symptomatic treatment becomes of great importance due to the complications that have already occurred before the implementation of the CFTR modulators. The implementation of modulators in early childhood can attenuate or prevent early irreversible complications. Therefore, in this article special emphasis is placed on new developments in symptomatic treatment and on new treatment options.
Collapse
|
85
|
Rate and predictors of prescription of lumacaftor – Ivacaftor in the 18 months following approval in the United States. J Cyst Fibros 2018; 17:742-746. [DOI: 10.1016/j.jcf.2018.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 11/20/2022]
|
86
|
Prevalence of abnormal glucose metabolism in pediatric acute, acute recurrent and chronic pancreatitis. PLoS One 2018; 13:e0204979. [PMID: 30379828 PMCID: PMC6209152 DOI: 10.1371/journal.pone.0204979] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 09/09/2018] [Indexed: 12/16/2022] Open
Abstract
Type 3C Diabetes, or diseases of the exocrine pancreas has been reported to occur in approximately 30% of adult patient with pancreatitis. The incidence of glucose abnormalities or risk factors that may predict the development of abnormal glucose in the pediatric pancreatitis population is not known. We performed a retrospective chart review from 1998–2016 for patients who carry the diagnosis of acute pancreatitis (AP), acute recurrent pancreatitis (ARP), and chronic pancreatitis (CP). We extracted glucose values, HbA1c%, and data from oral glucose tolerance and mixed meal testing with timing in relation to pancreatic exacerbations. Patient characteristic data such as age, gender, body proportions, family history of pancreatitis, exocrine function and genetic mutations were also assessed. Abnormal glucose was based on definitions put forth by the American Diabetes Society for pre-diabetes and diabetes. Fifty-two patients had AP and met criteria. Of those, 15 (29%) had glucose testing on or after the first attack, 21 (40%) were tested on or after the second attack (in ARP patients) and 16 (31%) were tested after a diagnosis of CP. Of the patients tested for glucose abnormalities, 25% (13/52) had abnormal glucose testing (testing indicating pre-DM or DM as defined by ADA guidelines. A significantly higher proportion of the abnormal glucose testing was seen in patients (85%, 11/13) with a BMI at or greater than the 85th percentile compared to the normal glucose patients (28%, 11/39) (p = 0.0007). A significantly higher proportion of the abnormal glucose patients (77%, 10/13) had SAP during the prior AP episode to testing compared to the 10% (4/39) of the normal glucose patients (p<0.0001). Older age at DM testing was associated with a higher prevalence of abnormal glucose testing (p = 0.04). In our patient population, a higher proportion of glucose abnormalities were after the second episode of pancreatitis, however 62% (8/13) with abnormalities was their first time tested. We identified obesity and having severe acute pancreatitis (SAP) during the prior AP episode to testing could be associated with abnormal glucose. We propose that systematic screening for abnormal glucose after the first episode of acute pancreatitis in order to better establish the timing of diabetes progression.
Collapse
|
87
|
Fukuda R, Okiyoneda T. Peripheral Protein Quality Control as a Novel Drug Target for CFTR Stabilizer. Front Pharmacol 2018; 9:1100. [PMID: 30319426 PMCID: PMC6170605 DOI: 10.3389/fphar.2018.01100] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Conformationally defective cystic fibrosis transmembrane conductance regulator (CFTR) including rescued ΔF508-CFTR is rapidly eliminated from the plasma membrane (PM) even in the presence of a CFTR corrector and potentiator, limiting the therapeutic effort of the combination therapy. CFTR elimination from the PM is determined by the conformation-dependent ubiquitination as a part of the peripheral quality control (PQC) mechanism. Recently, the molecular machineries responsible for the CFTR PQC mechanism which includes molecular chaperones and ubiquitination enzymes have been revealed. This review summarizes the molecular mechanism of the CFTR PQC and discusses the possibility that the peripheral ubiquitination mechanism becomes a novel drug target to develop the CFTR stabilizer as a novel class of CFTR modulator.
Collapse
Affiliation(s)
- Ryosuke Fukuda
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Nishinomiya, Japan
| | - Tsukasa Okiyoneda
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Nishinomiya, Japan
| |
Collapse
|
88
|
Calhoun JD, Carvill GL. Unravelling the genetic architecture of autosomal recessive epilepsy in the genomic era. J Neurogenet 2018; 32:295-312. [PMID: 30247086 DOI: 10.1080/01677063.2018.1513509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The technological advancement of next-generation sequencing has greatly accelerated the pace of variant discovery in epilepsy. Despite an initial focus on autosomal dominant epilepsy due to the tractable nature of variant discovery with trios under a de novo model, more and more variants are being reported in families with epilepsies consistent with autosomal recessive (AR) inheritance. In this review, we touch on the classical AR epilepsy variants such as the inborn errors of metabolism and malformations of cortical development. However, we also highlight recently reported genes that are being identified by next-generation sequencing approaches and online 'matchmaking' platforms. Syndromes mainly characterized by seizures and complex neurodevelopmental disorders comorbid with epilepsy are discussed as an example of the wide phenotypic spectrum associated with the AR epilepsies. We conclude with a foray into the future, from the application of whole-genome sequencing to identify elusive epilepsy variants, to the promise of precision medicine initiatives to provide novel targeted therapeutics specific to the individual based on their clinical genetic testing.
Collapse
Affiliation(s)
- Jeffrey D Calhoun
- a Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Gemma L Carvill
- a Department of Neurology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| |
Collapse
|
89
|
Lopes-Pacheco M, Kitoko JZ, Morales MM, Petrs-Silva H, Rocco PRM. Self-complementary and tyrosine-mutant rAAV vectors enhance transduction in cystic fibrosis bronchial epithelial cells. Exp Cell Res 2018; 372:99-107. [PMID: 30244179 DOI: 10.1016/j.yexcr.2018.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/13/2018] [Accepted: 09/20/2018] [Indexed: 10/28/2022]
Abstract
Recombinant adeno-associated virus (rAAV) vector platforms have shown considerable therapeutic success in gene therapy for inherited disorders. In cystic fibrosis (CF), administration of first-generation rAAV2 was safe, but clinical benefits were not clearly demonstrated. Therefore, next-generation vectors that overcome rate-limiting steps in rAAV transduction are needed to obtain successful gene therapy for this devastating disease. In this study, we evaluated the effects of single-strand or self-complementary (sc) rAAV vectors containing single or multiple tyrosine-to-phenylalanine (Y-F) mutations in capsid surface-exposed residues on serotypes 2, 8 or 9. For this purpose, CF bronchial epithelial (CFBE) cells were transduced with rAAV vectors, and the transgene expression of enhanced green fluorescence protein (eGFP) was analyzed at different time points. The effects of vectors on the cell viability, host cell cycle and in association with co-adjuvant drugs that modulate intracellular vector trafficking were also investigated. Six rAAV vectors demonstrated greater percentage of eGFP+ cells compared to their counterparts at days 4, 7 and 10 post-transduction: rAAV2 Y(272,444,500,730)F, with 1.95-, 3.5- and 3.06-fold increases; rAAV2 Y(252,272,444,500,704,730)F, with 1.65-, 2.12-, and 2-fold increases; scrAAV2 WT, with 1.69-, 2.68-, and 2.32-fold increases; scrAAV8 Y773F, with 57-, 6.06-, and 7-fold increases; scrAAV9 WT, with 7.47-, 4.64-, and 3.66-fold increases; and scrAAV9 Y446F, with 8.39-, 4.62-, and 4.4-fold increases. At days 15, 20, and 30 post-transduction, these vectors still demonstrated higher transgene expression than transfected cells. Although the percentage of eGFP+ cells reduced during the time-course analysis, the delta mean fluorescence intensity increased. These vectors also led to increased percentage of cells in G1-phase without eliciting any cytotoxicity. Prior administration of bortezomib or genistein did not increase eGFP expression in cells transduced with either rAAV2 Y(272,444,500,730)F or rAAV2 Y(252,272,444,500,704,730)F. In conclusion, self-complementary and tyrosine capsid mutations on rAAV serotypes 2, 8, and 9 led to more efficient transduction than their counterparts in CFBE cells by overcoming the intracellular trafficking and second-strand DNA synthesis limitations.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Jamil Z Kitoko
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hilda Petrs-Silva
- Laboratory of Neurogenesis, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
90
|
Mota LR, de Melo Filho VM, de Castro LL, Garcia DF, Terse-Ramos R, Toralles MBP, de Lima RLLF, Souza EL. Description of rare mutations and a novel variant in Brazilian patients with Cystic Fibrosis: a case series from a referral center in the Bahia State. Mol Biol Rep 2018; 45:2045-2051. [PMID: 30232781 DOI: 10.1007/s11033-018-4361-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/05/2018] [Indexed: 12/17/2022]
Abstract
Knowledge of the genetic profile of Cystic Fibrosis (CF) contributes to a better understanding of the genotype/phenotype relationship, particularly in mixed populations such as in Brazil. To describe clinical data of CF patients with rare or not yet observed CFTR gene mutations in Brazil. It was a case series of CF patients followed-up at a referral center. Clinical and laboratory data were obtained through medical records. Molecular analysis of the mutations was performed by conventional methods and/or by next-generation sequencing. Ten patients were studied, seven had five pathogenic mutations without previous description in Brazil (Q1100P, Y109C, A107P, E1409K and K162E), one of which has not yet been reported in patients with CF (A107P). Among the seven patients, three (two siblings) had the second mutant allele of rare occurrence among Brazilians patients (G1069R and 2307insA). Three other patients also had at least one rare variant (V201M, S466X and G1069R). The age of the CF diagnosis ranged from 1 to 190 months in the ten cases and the main clinical manifestations were respiratory symptoms and difficulty in gaining weight. All but one patient presented clinical and/or laboratory data compatible with pancreatic insufficiency. The identification of rare or not yet described CFTR mutations in patients with CF in Brazil highlights the high genetic heterogeneity in this population. Knowledge of the genotypic profile of Brazilian CF patients can contribute to the development of specific mutation panels for the genetic investigation targeting each region of the country, as well as helping to understand the complex genotype/phenotype relationship, especially in mixed populations.
Collapse
Affiliation(s)
- Laís Ribeiro Mota
- Post-Graduation Program in Interactive Processes of Systems and Organs, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil
| | | | | | | | - Regina Terse-Ramos
- Department of Pediatrics, School of Medicine of Bahia, Federal University of Bahia, Avenida: Santa Luzia, 379/902. Horto Florestal, Salvador, Bahia, CEP: 40295-050, Brazil
| | - Maria Betânia Pereira Toralles
- Department of Pediatrics, School of Medicine of Bahia, Federal University of Bahia, Avenida: Santa Luzia, 379/902. Horto Florestal, Salvador, Bahia, CEP: 40295-050, Brazil
| | | | - Edna Lúcia Souza
- Department of Pediatrics, School of Medicine of Bahia, Federal University of Bahia, Avenida: Santa Luzia, 379/902. Horto Florestal, Salvador, Bahia, CEP: 40295-050, Brazil.
| |
Collapse
|
91
|
Flume PA, Chalmers JD, Olivier KN. Advances in bronchiectasis: endotyping, genetics, microbiome, and disease heterogeneity. Lancet 2018; 392:880-890. [PMID: 30215383 PMCID: PMC6173801 DOI: 10.1016/s0140-6736(18)31767-7] [Citation(s) in RCA: 260] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/16/2018] [Accepted: 07/25/2018] [Indexed: 12/29/2022]
Abstract
Bronchiectasis is characterised by pathological dilation of the airways. More specifically, the radiographic demonstration of airway enlargement is the common feature of a heterogeneous set of conditions and clinical presentations. No approved therapies exist for the condition other than for bronchiectasis caused by cystic fibrosis. The heterogeneity of bronchiectasis is a major challenge in clinical practice and the main reason for difficulty in achieving endpoints in clinical trials. Recent observations of the past 2 years have improved the understanding of physicians regarding bronchiectasis, and have indicated that it might be more effective to classify patients in a different way. Patients could be categorised according to a heterogeneous group of endotypes (defined by a distinct functional or pathobiological mechanism) or by clinical phenotypes (defined by relevant and common features of the disease). In doing so, more specific therapies needed to effectively treat patients might finally be developed. Here, we describe some of the recent advances in endotyping, genetics, and disease heterogeneity of bronchiectasis including observations related to the microbiome.
Collapse
Affiliation(s)
- Patrick A. Flume
- Departments of Medicine and Pediatrics, Medical University
of South Carolina, Charleston, SC, USA.
| | | | | |
Collapse
|
92
|
Tosco A, Villella VR, Castaldo A, Kroemer G, Maiuri L, Raia V. Repurposing therapies for the personalised treatment of cystic fibrosis. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1483231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Antonella Tosco
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Valeria R. Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Alice Castaldo
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Guido Kroemer
- Equipe11 labellisée Ligue Nationale Contrele Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes, Paris, Sorbonne Paris Cité, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, HôpitalEuropéen Georges Pompidou, AP-HP, Paris, France
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
93
|
Moreno Ortega M, Quintana Gallego ME, Carrasco Hernández L, Pérez Borrego E, Delgado Pecellín I. Mycobacterium Lentiflavum in Cystic Fibrosis Subjects. A Colonizer or a True Pathogen? Arch Bronconeumol 2018; 54:639-640. [PMID: 29861072 DOI: 10.1016/j.arbres.2018.02.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 02/27/2018] [Indexed: 01/07/2023]
Affiliation(s)
- María Moreno Ortega
- Unidad de Pediatría y áreas específicas, Hospital Universitario Virgen del Rocío, Sevilla, España; Unidad de Fibrosis Quística, Hospital Universitario Virgen del Rocío, Sevilla, España.
| | - María Esther Quintana Gallego
- Unidad de Fibrosis Quística, Hospital Universitario Virgen del Rocío, Sevilla, España; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), España
| | - Laura Carrasco Hernández
- Unidad de Fibrosis Quística, Hospital Universitario Virgen del Rocío, Sevilla, España; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), España
| | - Esther Pérez Borrego
- Unidad de Pediatría y áreas específicas, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Isabel Delgado Pecellín
- Unidad de Fibrosis Quística, Hospital Universitario Virgen del Rocío, Sevilla, España; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), España
| |
Collapse
|
94
|
Wiencek JR, Lo SF. Advances in the Diagnosis and Management of Cystic Fibrosis in the Genomic Era. Clin Chem 2018; 64:898-908. [DOI: 10.1373/clinchem.2017.274670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/17/2018] [Indexed: 01/08/2023]
Abstract
Abstract
BACKGROUND
Cystic fibrosis (CF) is a complex autosomal recessive disease that continues to present unique diagnostic challenges. Because CF was first described in 1938, there has been a substantial growth of genetic and phenotypic information about the disorder. During the past few years, as more evidence has become available, a consortium of international experts determined that the 2008 guidelines from the CF Foundation needed to be reviewed and updated.
CONTENT
The goal of this review is to highlight the latest advances in CF multidisciplinary care, together with the recent updates to the 2017 CF Foundation diagnostic guidelines.
SUMMARY
Data from newborn screening programs, patient registries, clinical databases, and functional research have led to a better understanding of the CF transmembrane conductance regulator (CFTR) gene. Recent consensus guidelines have provided recommendations for clinicians and laboratorians to better assist with interpretation of disease status and related CF mutations. The highly recommended Clinical and Functional Translation of CFTR project should be the first resource in the evaluation of disease severity for CF mutations. Screen-positive newborns and patients with high clinical suspicion for CF are always recommended to undergo confirmatory sweat chloride testing with interpretations based on updated reference intervals. Every patient diagnosed with CF should receive genotyping, as novel molecular therapies are becoming standard of practice. The future of CF management must consider healthcare system disparities as CF transitions from a historically childhood disease to a predominantly adult epidemic.
Collapse
Affiliation(s)
- Joesph R Wiencek
- Division of Laboratory Medicine, Department of Pathology, University of Virginia School of Medicine and Health System, Charlottesville, VA
| | - Stanley F Lo
- Department of Pathology, Medical College of Wisconsin and Department of Pathology and Laboratory Medicine, Children's Hospital of Wisconsin, Milwaukee, WI
| |
Collapse
|
95
|
Van der Plas SE, Kelgtermans H, De Munck T, Martina SLX, Dropsit S, Quinton E, De Blieck A, Joannesse C, Tomaskovic L, Jans M, Christophe T, van der Aar E, Borgonovi M, Nelles L, Gees M, Stouten P, Van Der Schueren J, Mammoliti O, Conrath K, Andrews M. Discovery of N-(3-Carbamoyl-5,5,7,7-tetramethyl-5,7-dihydro-4H-thieno[2,3-c]pyran-2-yl)-lH-pyrazole-5-carboxamide (GLPG1837), a Novel Potentiator Which Can Open Class III Mutant Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Channels to a High Extent. J Med Chem 2018; 61:1425-1435. [DOI: 10.1021/acs.jmedchem.7b01288] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | - Hans Kelgtermans
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Tom De Munck
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | | | - Evelyne Quinton
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Ann De Blieck
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Linda Tomaskovic
- Fidelta Ltd., Prilaz Baruna Filipovića 29, Zagreb HR-10000, Croatia
| | - Mia Jans
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | | | - Monica Borgonovi
- Galapagos SASU, 102
Avenue Gaston Roussel, 93230 Romainville, France
| | - Luc Nelles
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Maarten Gees
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Pieter Stouten
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | | | - Oscar Mammoliti
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Katja Conrath
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| | - Martin Andrews
- Galapagos NV, Generaal De Wittelaan L11 A3, 2800 Mechelen, Belgium
| |
Collapse
|
96
|
Liessi N, Cichero E, Pesce E, Arkel M, Salis A, Tomati V, Paccagnella M, Damonte G, Tasso B, Galietta LJ, Pedemonte N, Fossa P, Millo E. Synthesis and biological evaluation of novel thiazole- VX-809 hybrid derivatives as F508del correctors by QSAR-based filtering tools. Eur J Med Chem 2018; 144:179-200. [DOI: 10.1016/j.ejmech.2017.12.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 12/29/2022]
|
97
|
Giugliani R, Vairo F, Kubaski F, Poswar F, Riegel M, Baldo G, Saute JA. Neurological manifestations of lysosomal disorders and emerging therapies targeting the CNS. THE LANCET CHILD & ADOLESCENT HEALTH 2017; 2:56-68. [PMID: 30169196 DOI: 10.1016/s2352-4642(17)30087-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022]
Abstract
Lysosomal disorders have been an area of interest since intravenous enzyme replacement therapy was successfully introduced for the treatment of Gaucher's disease in the early 1990s. This treatment approach has also been developed for several other lysosomal disorders, including Fabry's disease, Pompe's disease, lysosomal acid lipase deficiency, and five types of mucopolysaccharidosis. Despite the benefits of enzyme replacement therapy, it has limitations-most importantly, its ineffectiveness in treating the neurological components of lysosomal disorders, as only a small proportion of recombinant enzymes can cross the blood-brain barrier. Development of strategies to improve drug delivery to the CNS is now the primary focus in lysosomal disorder research. This Review discusses the neurological manifestations and emerging therapies for the CNS component of these diseases. The therapies in development (which are now in phase 1 or phase 2 clinical trials) might be for specific lysosomal disorders (enzyme replacement therapy via intrathecal or intracerebroventricular routes or with fusion proteins, or gene therapy) or applicable to more than one lysosomal disorder (haemopoietic stem cell transplantation, pharmacological chaperones, substrate reduction therapy, or stop codon readthrough). The combination of early diagnosis with effective therapies should change the outlook for patients with lysosomal disorders with neurological involvement in the next 5-10 years.
Collapse
Affiliation(s)
- Roberto Giugliani
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | | | | | - Fabiano Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mariluce Riegel
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Baldo
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Postgraduate Program in Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jonas Alex Saute
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Postgraduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
98
|
Strategies for the etiological therapy of cystic fibrosis. Cell Death Differ 2017; 24:1825-1844. [PMID: 28937684 PMCID: PMC5635223 DOI: 10.1038/cdd.2017.126] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022] Open
Abstract
Etiological therapies aim at repairing the underlying cause of cystic fibrosis (CF), which is the functional defect of the cystic fibrosis transmembrane conductance regulator (CFTR) protein owing to mutations in the CFTR gene. Among these, the F508del CFTR mutation accounts for more than two thirds of CF cases worldwide. Two somehow antinomic schools of thought conceive CFTR repair in a different manner. According to one vision, drugs should directly target the mutated CFTR protein to increase its plasma membrane expression (correctors) or improve its ion transport function (potentiators). An alternative strategy consists in modulating the cellular environment and proteostasis networks in which the mutated CFTR protein is synthesized, traffics to its final destination, the plasma membrane, and is turned over. We will analyze distinctive advantages and drawbacks of these strategies in terms of their scientific and clinical dimensions, and we will propose a global strategy for CF research and development based on a reconciliatory approach. Moreover, we will discuss the utility of preclinical biomarkers that may guide the personalized, patient-specific implementation of CF therapies.
Collapse
|
99
|
Zemanick ET, Daines CL, Dellon EP, Esther CR, Kinghorn B, Ong T, Muhlebach MS. Highlights from the 2016 North American Cystic Fibrosis Conference. Pediatr Pulmonol 2017; 52:1103-1110. [PMID: 28696526 PMCID: PMC5963883 DOI: 10.1002/ppul.23707] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/23/2017] [Indexed: 01/25/2023]
Abstract
The 30th annual North American Cystic Fibrosis Conference (NACFC) was held in Orlando, FL, on October 27-29, 2016. Abstracts were published in a supplement to Pediatric Pulmonology. This review summarizes several major topic areas addressed at the conference: the pathophysiology of cystic fibrosis (CF) lung disease, clinical trials, clinical management issues, and quality improvement. We sought to provide an overview of emerging concepts in several areas of CF research and care, rather than a comprehensive review of the conference. Citations from the conference are by first author and abstract number or symposium number, as designated in the supplement.
Collapse
Affiliation(s)
- Edith T Zemanick
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Cori L Daines
- Department of Pediatrics, University of Arizona College of Medicine, Tucson, Arizona
| | - Elisabeth P Dellon
- Division of Pulmonology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles R Esther
- Division of Pulmonology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - BreAnna Kinghorn
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - Thida Ong
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - Marianne S Muhlebach
- Division of Pulmonology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
100
|
Vauthier V, Housset C, Falguières T. Targeted pharmacotherapies for defective ABC transporters. Biochem Pharmacol 2017; 136:1-11. [DOI: 10.1016/j.bcp.2017.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 02/23/2017] [Indexed: 02/07/2023]
|