1
|
Zemanick ET, Ramsey B, Sands D, McKone EF, Fajac I, Taylor-Cousar JL, Mall MA, Konstan MW, Nair N, Zhu J, Arteaga-Solis E, Van Goor F, McGarry L, Prieto-Centurion V, Sosnay PR, Bozic C, Waltz D, Mayer-Hamblett N. Sweat chloride reflects CFTR function and correlates with clinical outcomes following CFTR modulator treatment. J Cyst Fibros 2025:S1569-1993(24)01854-X. [PMID: 39755444 DOI: 10.1016/j.jcf.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Highly effective CFTR modulators improve CFTR function and lead to dramatic improvements in health outcomes in many people with cystic fibrosis (pwCF). The relationship between measures of CFTR function, such as sweat chloride concentration, and clinical outcomes in pwCF treated with CFTR modulators is poorly defined. We conducted analyses to better understand the relationships between sweat chloride and CFTR function in vitro, and between sweat chloride and clinical outcomes following CFTR modulator treatment. METHODS Mean sweat chloride values in healthy people, CF carriers, and pwCF treated with CFTR modulators at different doses were compared to chloride transport in corresponding human bronchial epithelial (HBE) cells. A pooled analysis of phase 3 CFTR modulator studies was performed to evaluate the relationship between attained values of sweat chloride and improvements in lung function, body mass index (BMI), patient reported outcomes, pulmonary exacerbations, and lung function change over time. RESULTS Sweat chloride concentrations in vivo correlated strongly with CFTR-dependent chloride current in HBE cells in vitro. Sweat chloride values of <30 mmol/L and ≥30 to <60 mmol/L in pwCF following CFTR modulator treatment were associated with better clinical outcomes than sweat chloride ≥60 to <80 mmol/L and ≥80 mmol/L. CONCLUSIONS In pwCF treated with CFTR modulators, lower sweat chloride levels (reflecting greater CFTR function) are associated with better clinical outcomes. These results support the therapeutic strategy of further restoring CFTR function towards normal, as reflected in lowering sweat chloride to below the diagnostic threshold for CF (<60 mmol/L) and to normal (<30 mmol/L), with CFTR modulators.
Collapse
Affiliation(s)
- Edith T Zemanick
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO, USA.
| | - Bonnie Ramsey
- Cystic Fibrosis Therapeutics Development Network Coordinating Center, Seattle Children's Hospital, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Dorota Sands
- Cystic Fibrosis Department, Institute of Mother and Child, Warsaw, Poland
| | - Edward F McKone
- St. Vincent's University Hospital and University College, Dublin, Ireland
| | | | - Jennifer L Taylor-Cousar
- Departments of Internal Medicine and Pediatrics, Pulmonary Divisions National Jewish Health, Denver, CO, USA
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin and German Center for Lung Research, Associated Partner, Berlin, Germany and German Center for Child and Adolescent Health (DZKJ), partner site Berlin, Berlin, Germany
| | - Michael W Konstan
- Department of Pediatrics, Case Western Reserve University School of Medicine and Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Nitin Nair
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Jiaqiang Zhu
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | | | | | - Lisa McGarry
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | | | | | - Carmen Bozic
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - David Waltz
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Nicole Mayer-Hamblett
- Cystic Fibrosis Therapeutics Development Network Coordinating Center, Seattle Children's Hospital, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Qiu B, Li Y, He Z, Li Z, Terreau S, Wang X, Lyu J, Wang W, Lara-Sáez I. Formulate a concentrated highly branched poly(β-amino ester)/DNA polyplex - one step closer to application in lung cystic fibrosis disease. Drug Dev Ind Pharm 2025:1-15. [PMID: 39743829 DOI: 10.1080/03639045.2024.2448271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/27/2024] [Accepted: 12/14/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE Highly branched poly(β-amino ester) (HPAEs)-based gene therapy holds promise for treating lung cystic fibrosis (CF). However, the translation of HPAEs/DNA nanoparticles into clinical applications poses a significant challenge due to the requirement for high concentrations of the formulation. METHODS In this work, a straightforward and scalable concentration method was developed for concentrating HPAEs/DNA polyplexes. A series of different buffers with various pH values and ionic components were initially tested to develop the optimized HPAEs/DNA polyplex formulation. Subsequently, the optimized HPAEs/DNA polyplex formulation was concentrated through lyophilization and ultrafiltration. RESULTS The ultrafiltration outperformed the lyophilization in concentration capacity, showing a 24-fold increase in the concentrated formulation compared to the original non-concentrated formulation. The concentration does not disturb the transfection efficiency in lung CF epithelial cells, indicating its potential for lung delivery applications. Moreover, the concentrated HPAEs/DNA polyplex successfully restored the production of cystic fibrosis transmembrane conductance regulator (CFTR) protein in primary lung CF epithelial cells, surpassing the performance of the non-concentrated common gene transfection reagents such as Lipofectamine 3000 and Xfect. CONCLUSIONS The concentrated HPAEs/DNA formulation represents a promising step forward for preclinical testing (e.g., in vivo evaluation), with further research needed to confirm its potential for clinical use.
Collapse
Affiliation(s)
- Bei Qiu
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Yinghao Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Zhonglei He
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
- School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Zishan Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Sébastien Terreau
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Xianqing Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| | | | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
- Institute of Precision Medicine (AUST-IPM), Anhui University of Science and Technology, Huainan 232001, China
- School of Public Health, Anhui University of Science and Technology, Huainan 232001, China
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
3
|
Escobar NS, Ratjen F. An update on multiple breath washout in children with cystic fibrosis. Expert Rev Respir Med 2024:1-11. [PMID: 39709582 DOI: 10.1080/17476348.2024.2445683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
INTRODUCTION Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CF transmembrane regulator (CFTR) gene, leading to progressive lung disease and systemic complications. Lung disease remains the primary cause of morbidity and mortality, making early detection of lung function decline crucial. The Lung Clearance Index (LCI), derived from the multiple breath washout (MBW) test, has emerged as a sensitive measure for identifying early airway disease. AREAS COVERED This review examines the technical aspects and clinical relevance of LCI, its advantages over traditional lung function tests, and its application in CF clinical trials. A focused literature review highlights LCI's utility in evaluating treatment efficacy and its potential integration into routine CF care. EXPERT OPINION LCI is more sensitive than spirometry for detecting early lung function decline and is predominantly used in pediatric settings. Its use is expanding in adult CF populations as advances in treatment allow adults to maintain stable lung function. In clinical trials, LCI is widely recognized as an outcome measure. While implemented into clinical care in many centers in Europe, this is not yet the case in North America. Faster testing protocols and point-of-care interpretation tools will support LCI's integration into routine CF monitoring.
Collapse
Affiliation(s)
- Natalia S Escobar
- Division of Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Felix Ratjen
- Division of Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Michael S, Liotta N, Fei T, Bendall ML, Nixon DF, Dopkins N. Endogenous retroelement expression in modeled airway epithelial repair. Microbes Infect 2024:105465. [PMID: 39681187 DOI: 10.1016/j.micinf.2024.105465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 11/14/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder characterized by impairment of the CF transmembrane conductance regulator (CFTR) via gene mutation. CFTR is expressed at the cellular membrane of epithelial cells and functions as an anion pump which maintains water and salt ion homeostasis. In pulmonary airways of CF patients, pathogens such as P. aeruginosa and subsequent uncontrolled inflammation damage the human airway epithelial cells (HAECs) and can be life-threatening. We previously identified that inhibiting endogenous retroelement (ERE) reverse transcriptase can hamper the inflammatory response to bacterial flagella in THP-1 cells. Here, we investigate how ERE expression is sensitive to HAEC repair and toll-like receptor 5 (TLR5) activation, a primary mechanism by which inflammation impacts disease outcome. Our results demonstrate that several human endogenous retroviruses (HERVs) and long interspersed nuclear elements (LINEs) fluctuate throughout the various stages of repair and that TLR5 activation further influences ERE expression. By considering the impact of the most common CF mutation F508del/F508del on ERE expression in unwounded HAECs, we also found that two specific EREs, L1FLnI_2p23.1c and HERVH_10p12.33, were downregulated in CF-derived HAECs. Collectively, we show that ERE expression in HAECs is sensitive to certain modalities reflective of CF pathogenesis, and specific EREs may be indicative of CF disease state and pathogenesis.
Collapse
Affiliation(s)
- Stephanie Michael
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nicholas Liotta
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Tongyi Fei
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew L Bendall
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Douglas F Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Nicholas Dopkins
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Liu G, Dong BB, Devanarayana S, Chen RC, Liu Q. Emerging roles of mechanosensitive ion channels in ventilator induced lung injury: a systematic review. Front Immunol 2024; 15:1479230. [PMID: 39664395 PMCID: PMC11631737 DOI: 10.3389/fimmu.2024.1479230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Background The pathogenetic mechanisms of ventilator-induced lung injury (VILI) still need to be elucidated. The mechanical forces during mechanical ventilation are continually sensed and transmitted by mechanosensitive ion channels (MSICs) in pulmonary endothelial, epithelial, and immune cells. In recent years, MSICs have been shown to be involved in VILI. Methods A systematic search across PubMed, the Cochrane Library, Web of Science, and ScienceDirect was performed from inception to March 2024, and the review was conducted in accordance with PRISMA guidelines. The potential eligible studies were evaluated by two authors independently. Study characteristics, quality assessment, and potential mechanisms were analyzed. Results We included 23 eligible studies, most of which were performed with murine animals in vivo. At the in vitro level, 52% and 48% of the experiments were conducted with human or animal cells, respectively. No clinical studies were found. The most reported MSICs include Piezo channels, transient receptor potential channels, potassium channels, and stretch-activated sodium channels. Piezo1 has been the most concerned channel in the recent five years. This study found that signal pathways, such as RhoA/ROCK1, could be enhanced by cyclic stretch-activated MSICs, which contribute to VILI through dysregulated inflammation and immune responses mediated by ion transport. The review indicates the emerging role of MSICs in the pathogenesis of VILI, especially as a signal-transmitting link between mechanical stretch and pathogenesis such as inflammation, disruption of cell junctions, and edema formation. Conclusions Mechanical stretch stimulates MSICs to increase transcellular ion exchange and subsequently generates VILI through inflammation and other pathogeneses mediated by MSICs signal-transmitting pathways. These findings make it possible to identify potential therapeutic targets for the prevention of lung injury through further exploration and more studies. Systematic review registration https://inplasy.com/inplasy-2024-10-0115/, identifier INPLASY2024100115.
Collapse
Affiliation(s)
- Gang Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin-bin Dong
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shalika Devanarayana
- School of International Education, Zhengzhou University, Zhengzhou, Henan, China
| | - Rong-Chang Chen
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Qi Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Mianowski L, Doléans-Jordheim A, Barraud L, Rabilloud M, Richard M, Josserand RN, Durieu I, Reynaud Q. One year of ETI reduces lung bacterial colonisation in adults with cystic fibrosis. Sci Rep 2024; 14:29298. [PMID: 39592637 PMCID: PMC11599715 DOI: 10.1038/s41598-024-77246-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
The triple combination elexacaftor-tezacaftor-ivacaftor (ETI) has provided unprecedented clinical benefits for people with cystic fibrosis (pwCF) and drastically transformed the outcome of this disease. We aimed to describe the evolution of lung bacterial colonization in 198 French adult pwCF taking into account the use of concomitantly respiratory treatment. We collected sputum cultures produced during the entire follow-up period starting 3 years before and ending 1 year after ETI initiation. All sputum cultures were centralized and analyzed at our bacteriological laboratory. Clinical data included pulmonary function, respiratory treatments, physiotherapy, number of IV antibiotics treatment, as well as inpatient stays. We observed a significant decrease in colonization prevalence by any CF pathogen after one year of treatment with ETI (p < 0.001). This decrease was confirmed for Pseudomonas aeruginosa, MRSA and MSSA, Stenotrophomonas maltophilia, Achromobacter spp. and nontuberculous mycobacteria (NTM). The maximal density of bacteria documented in sputum cultures decreased from 2.107 CFU/ml to 1.106 CFU/ml after one year of ETI. We also found a decrease in prevalence of Pseudomonas aeruginosa chronic colonization and in the density of Pseudomonas aeruginosa after one year of ETI. These results confirm the decrease in prevalence and bacterial density of lung colonisation for most of the CF pathogens, including Achromobacter spp, Stenotrophomonas maltophilia concomitantly to the clinical improvement. Further studies are needed to better understand the underlying mechanisms of these microbiological changes.
Collapse
Affiliation(s)
- Lucile Mianowski
- Centre de référence Adulte de la Mucoviscidose, Service de médecine interne, Hospices civils de Lyon, Pierre Bénite, France
| | - Anne Doléans-Jordheim
- Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
- Centre de recherche International en infectiologie, INSERM U1111 Université Claude Bernard Lyon 1, CNRS UMR 5308 Ecole Normale Supérieure de Lyon, Lyon, France
| | - Laurent Barraud
- Service de Biostatistique et Bioinformatique, Hospices Civils de Lyon, Lyon, F-69003, France
| | - Muriel Rabilloud
- Service de Biostatistique et Bioinformatique, Hospices Civils de Lyon, Lyon, F-69003, France
- Laboratoire de Biométrie et Biologie Evolutive, CNRS, UMR 5558, Equipe Biostatistique-Santé, Villeurbanne, F-69100, France
| | - Mael Richard
- Centre de référence Adulte de la Mucoviscidose, Service de médecine interne, Hospices civils de Lyon, Pierre Bénite, France
| | - Raphaele Nove Josserand
- Centre de référence Adulte de la Mucoviscidose, Service de médecine interne, Hospices civils de Lyon, Pierre Bénite, France
| | - Isabelle Durieu
- Centre de référence Adulte de la Mucoviscidose, Service de médecine interne, Hospices civils de Lyon, Pierre Bénite, France
- RESearch on HealthcAre PErformance (RESHAPE), INSERM U1290, Université Claude Bernard Lyon 1, Lyon , France
- ERN-Lung CF network, Frankfurt, Germany
| | - Quitterie Reynaud
- Centre de référence Adulte de la Mucoviscidose, Service de médecine interne, Hospices civils de Lyon, Pierre Bénite, France.
- RESearch on HealthcAre PErformance (RESHAPE), INSERM U1290, Université Claude Bernard Lyon 1, Lyon , France.
- ERN-Lung CF network, Frankfurt, Germany.
| |
Collapse
|
7
|
Steinberg R, Mostacci N, Kieninger E, Frauchiger B, Casaulta C, Usemann J, Moeller A, Trachsel D, Rochat I, Blanchon S, Mueller-Suter D, Kern B, Zanolari M, Frey U, Ramsey KA, Hilty M, Latzin P, Korten I. Early nasal microbiota and subsequent respiratory tract infections in infants with cystic fibrosis. COMMUNICATIONS MEDICINE 2024; 4:246. [PMID: 39580540 PMCID: PMC11585651 DOI: 10.1038/s43856-024-00616-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/18/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Respiratory tract infections (RTIs) drive lung function decline in children with cystic fibrosis (CF). While the respiratory microbiota is clearly associated with RTI pathogenesis in infants without CF, data on infants with CF is scarce. We compared nasal microbiota development between infants with CF and controls and assessed associations between early-life nasal microbiota, RTIs, and antibiotic treatment in infants with CF. METHODS We included 50 infants with CF and 30 controls from two prospective birth cohorts followed throughout the first year of life. We collected 1511 biweekly nasal swabs and analyzed the microbiota after amplifying the V3-V4 region of the 16S rRNA gene. We conducted structured weekly interviews to assess respiratory symptoms and antibiotic treatment. We calculated generalized additive mixed models and permutational analysis of variance. RESULTS Here, we show that the nasal microbiota is already altered before the first RTI or antibiotic treatment in infants with CF. Microbiota diversity differs between infants with CF and controls following RTIs and/or antibiotic treatment. CF infants with lower α-diversity have a higher number of subsequent RTIs. CONCLUSIONS Early nasal microbiota alterations may reflect predisposition or predispose to RTIs in infants with CF, and further change after RTIs and antibiotic treatment. This highlights the potential of targeting the nasal microbiota in CF-related RTI management, while also questioning current practices in the era of novel modulator therapies.
Collapse
Affiliation(s)
- Ruth Steinberg
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Nadja Mostacci
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Elisabeth Kieninger
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bettina Frauchiger
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Carmen Casaulta
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jakob Usemann
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
- Department of Respiratory Medicine and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Alexander Moeller
- Department of Respiratory Medicine and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Daniel Trachsel
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Isabelle Rochat
- Pediatric Pulmonology and Cystic Fibrosis Unit, Division of Pediatrics, Department of Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sylvain Blanchon
- Pediatric Pulmonology and Cystic Fibrosis Unit, Division of Pediatrics, Department of Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Barbara Kern
- Division of Pediatric Pneumology, Kantonsspital Aarau, Aarau, Switzerland
| | - Maura Zanolari
- Division of Pediatrics, Hospital Bellinzona, Bellinzona, Switzerland
| | - Urs Frey
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Kathryn A Ramsey
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Markus Hilty
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Insa Korten
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Akinnurun OM, Riedel T, Müller S, Bunk B, Schröttner P. Current knowledge on Inquilinus limosus, a scarcely researched human pathogen. BMC Microbiol 2024; 24:474. [PMID: 39538164 PMCID: PMC11558892 DOI: 10.1186/s12866-024-03617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Inquilinus limosus belongs to the class of the Alphaproteobacteria and was first described in 2002. So far, the species has mainly been isolated from respiratory specimens of patients with cystic fibrosis. A main characteristic of Inquilinus limosus is the prolonged time until bacterial colony growth is detectable. As the defined incubation times in many laboratories are too short to detect the growth of Inquilinus limosus, it is likely that the species is less frequently detected in the clinical setting than it actually occurs. This also explains why there are currently only very few data on the incidence available. Furthermore, as an uncommon pathogen, Inquilinus limosus may be familiar to only a few specialised clinicians. Due to these reasons, only little research (e.g. case reports and research papers) have been published on this species to date. However, given that a clear human pathogenic significance can be deduced from the existing literature, we have decided to present the current state of knowledge in this review and to address further aspects for the future elucidation of the pathogenesis of Inquilinus limosus.
Collapse
Affiliation(s)
- Oluwafemi M Akinnurun
- Institute for Medical Microbiology and Virology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thomas Riedel
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Brunswick, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Brunswick, Germany
| | - Stephanie Müller
- Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Boyke Bunk
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Brunswick, Germany
| | - Percy Schröttner
- Institute for Medical Microbiology and Virology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
9
|
Dunnion S, Elbanna K, Krishna S, Brien CO. Imaging of cystic fibrosis manifestations in the abdomen. Abdom Radiol (NY) 2024:10.1007/s00261-024-04636-7. [PMID: 39527257 DOI: 10.1007/s00261-024-04636-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/29/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Cystic fibrosis is a common inherited autosomal recessive disease affecting 35,000 persons in the United States. It is caused by mutations of the cystic fibrosis transmembrane regulator (CFTR) gene, located on the long arm of chromosome 7.This protein carries chlorine in the membranes of epithelial cells of exocrine glands. Mutations in the CFTR gene results in production of abnormally viscous mucus. Although it primarily affects the lungs, cystic fibrosis is a multisystem disease with involvement of extra thoracic organs including the liver, pancreas, kidneys and digestive tract.With advances in the management of cystic fibrosis resulting in improved life expectancy, cystic fibrosis patients are surviving into adulthood and extrapulmonary disease has become more commonplace. It is essential that radiologists are aware of the spectrum of potential manifestations of cystic fibrosis to allow accurate diagnosis.The purpose of this manuscript is to provide an overview of the pathophysiology and imaging findings of abdominal entities unique to patients with cystic fibrosis. We will present a wide spectrum of renal, pancreatic, gastrointestinal, hepatobiliary and post-transplant cases describing the typical findings that will assist radiologists in providing a timely diagnosis for patients with cystic fibrosis.
Collapse
|
10
|
O'Leary F, Coffey N, Burke FM, Roberts A, Plant B, Hayes M. Caries experience of people with cystic fibrosis: A systematic review. J Dent 2024; 150:105328. [PMID: 39197529 DOI: 10.1016/j.jdent.2024.105328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024] Open
Abstract
OBJECTIVES Cystic Fibrosis is a multi-system disease, arising from a mutation of the cystic fibrosis transmembrane conductance regulator gene (CFTR). There is a lack of information regarding oral disease levels among people with cystic fibrosis. As part of an ongoing study assessing oral health in adults with cystic fibrosis at Cork University Dental School & Hospital, a systematic review of available literature was conducted to ascertain the caries experience of people with cystic fibrosis. The objective was to systematically present and evaluate the literature comparing caries experience between people with cystic fibrosis and people without cystic fibrosis. METHODS Five online databases were searched; Embase, Scopus, Web of Science Core Collection, Medline Ebsco and Cochrane Library. Studies that reported caries experience in people with cystic fibrosis were included in this review. RESULTS The initial search identified 1199 publications from online databases. Twenty-one studies were included for qualitative analysis. Fourteen studies reported a lower caries experience in children with CF compared to children without CF, five studies reported a higher caries experience in adults with CF, and two studies found inconclusive evidence regarding the association between caries experience and CF status. All studies had a risk of bias that may influence results. CONCLUSION Despite a lack of complete unanimity between all studies, there is a general trend that children with cystic fibrosis have a lower caries experience than their healthy counterparts, whereas adults with cystic fibrosis have a higher caries experience. The review highlights the need for further studies involving adults with cystic fibrosis as the majority of studies primarily consist of paediatric populations. CLINICAL SIGNIFICANCE Dental practitioners should be aware that adults with cystic fibrosis have higher caries experience. Tailored approaches to dental care specific to cystic fibrosis individuals should be developed.
Collapse
Affiliation(s)
- Fiona O'Leary
- Cork University Dental School & Hospital, Wilton, Cork, Ireland.
| | - Niamh Coffey
- Cork University Dental School & Hospital, Wilton, Cork, Ireland.
| | - Francis M Burke
- Cork University Dental School & Hospital, Wilton, Cork, Ireland.
| | - Anthony Roberts
- Cork University Dental School & Hospital, Wilton, Cork, Ireland.
| | - Barry Plant
- Cork University Hospital, Wilton, Cork, Ireland.
| | | |
Collapse
|
11
|
Reix P. The day after. Rethinking the Cystic Fibrosis model of care and structure of the CF team in the era of triple combination therapy. J Cyst Fibros 2024; 23:1043-1044. [PMID: 39327196 DOI: 10.1016/j.jcf.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Philippe Reix
- Centre de ressources et de compétences pour la mucoviscidose. Hôpital Femme Mère Enfants. Hospices Civils de Lyon. Bron. France et UMR5558. Equipe EMET. Villeurbanne. France.
| |
Collapse
|
12
|
Alqasmi M. Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials. J Clin Med 2024; 13:6530. [PMID: 39518670 PMCID: PMC11547045 DOI: 10.3390/jcm13216530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudomonas aeruginosa (Pa) poses a significant threat to individuals with cystic fibrosis (CF), as this bacterium is highly adaptable and resistant to antibiotics. While early-stage Pa infections can often be eradicated with aggressive antibiotic therapy, chronic infections are nearly impossible to eliminate and require treatments that focus on long-term bacterial suppression. Without such suppression, these persistent infections can severely damage the lungs, leading to serious complications and a reduced life expectancy for CF patients. Evidence for a specific treatment regimen for managing Pa infections in CF patients remains limited. This narrative review provides a detailed analysis of antimicrobial therapies assessed in completed phase IV trials, focusing on their safety and efficacy, especially with prolonged use. Key antibiotics, including tobramycin, colistin, meropenem, aztreonam, ceftolozane/tazobactam, ciprofloxacin, and azithromycin, are discussed, emphasizing their use, side effects, and delivery methods. Inhaled antibiotics are preferred for their targeted action and minimal side effects, while systemic antibiotics offer potency but carry risks like nephrotoxicity. The review also explores emerging treatments, such as phage therapy and antibiofilm agents, which show promise in managing chronic infections. Nonetheless, further research is necessary to enhance the safety and effectiveness of existing therapies while investigating new approaches for better long-term outcomes.
Collapse
Affiliation(s)
- Mohammed Alqasmi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
13
|
Hu H, Zhou Q, Ma Y, Zhang L. Genetic diagnosis and outcomes of intracytoplasmic sperm injection in South Chinese patients with congenital bilateral aplasia of the vas deferens. Basic Clin Androl 2024; 34:17. [PMID: 39402445 PMCID: PMC11479560 DOI: 10.1186/s12610-024-00233-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/29/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Obstructive azoospermia commonly is caused by CBAVD(Congenital Bilateral Aplasia of the Vas Deferens), mainly due to the cystic fibrosis transmembrane conductance regulator (CFTR) and adhesion G protein-coupled receptor G2(ADGRG2) mutations. The genetic landscape for Chinese CBAVD patients is unclear, leading to debates over genetic screening, counseling, and assisted reproduction strategies. This study investigates the prevalence of CFTR and ADGRG2 mutations in a southern Chinese cohort of CBAVD patients and evaluates the impact of CFTR mutations on intracytoplasmic sperm injection (ICSI) outcomes. RESULTS CFTR mutations were identified in 71.4% (30/42) of CBAVD patients, with a total of 36 CFTR mutation sites across 13 types identified, including two novel mutations. A novel ADGRG2 mutation was also detected. Betweenthe CFTR mutation-CBAVD group and the non-CBAVD OA group, a significant difference was observed only in the 2 Pronuclei(2PN) rate (79.5% vs 86.2%, P = 0.0065), while fertilization rates, pregnancy rates, miscarriage rates, and live birth rates showed no significant differences. Between the CFTR mutation-CBAVD group and the CBAVD group without CFTR mutation, there were no significant differences in fertilization rates, 2PN rates, pregnancy rates, miscarriage rates, or live birth rates. CONCLUSION Chinese CBAVD patients primarily exhibit mutations in the CFTR and ADGRG2 genes. Therefore, targeted gene testing for CFTR and ADGRG2 is more suitable compared to WES for CBAVD patients. Considering that the genetic factors of approximately 30% of CBAVD patients remain unknown, it is recommended to perform massive parallel sequencing for patients who test negative for CFTR and ADGRG2 gene screening. Despite these genetic factors, ICSI outcomes were not adversely affected, except for the 2PN rate. However, genetic counseling remains crucial for Chinese CBAVD patients before undergoing assisted reproduction.
Collapse
Affiliation(s)
- Haishan Hu
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
- Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research&Key Laboratory of Reproductive Health Diseases Research and Translation, Ministry of Education&Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571101, China
| | - Qing Zhou
- Department of Cerebrovascular Diseases, Hainan Provincial Peoples Hospital, Haikou, 570102, China
| | - Yanlin Ma
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
- Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research&Key Laboratory of Reproductive Health Diseases Research and Translation, Ministry of Education&Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571101, China.
| | - Lingxiao Zhang
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
- Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research&Key Laboratory of Reproductive Health Diseases Research and Translation, Ministry of Education&Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571101, China.
| |
Collapse
|
14
|
Stewart CG, Hilkin BM, Gansemer ND, Adam RJ, Dick DW, Sunderland JJ, Stoltz DA, Zabner J, Abou Alaiwa MH. Mucociliary clearance is impaired in small airways of cystic fibrosis pigs. Am J Physiol Lung Cell Mol Physiol 2024; 327:L415-L422. [PMID: 39104314 PMCID: PMC11482522 DOI: 10.1152/ajplung.00010.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/10/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder characterized by recurrent airway infections, inflammation, impaired mucociliary clearance, and progressive decline in lung function. The disease may start in the small airways; however, this is difficult to prove due to the limited accessibility of the small airways with the current single-photon mucociliary clearance assay. Here, we developed a dynamic positron emission tomography assay with high spatial and temporal resolution. We tested that mucociliary clearance is abnormal in the small airways of newborn cystic fibrosis pigs. Clearance of [68Ga]-tagged macroaggregated albumin from small airways started immediately after delivery and continued for the duration of the study. Initial clearance was fast but slowed down a few minutes after delivery. Cystic fibrosis pigs' small airways cleared significantly less than non-CF pigs' small airways (non-CF 25.1 ± 3.1% vs. CF 14.6 ± 0.1%). Stimulation of the cystic fibrosis airways with the purinergic secretagogue uridine-5'-triphosphate (UTP) further impaired clearance (non-CF with UTP 20.9 ± 0.3% vs. CF with UTP 13.0 ± 1.8%). None of the cystic fibrosis pigs treated with UTP (n = 6) cleared more than 20% of the delivered dose. These data indicate that mucociliary clearance in the small airways is fast and can easily be missed if the assay is not sensitive enough. The data also indicate that mucociliary clearance is impaired in the small airways of cystic fibrosis pigs. This defect is exacerbated by stimulation of mucus secretions with purinergic agonists.NEW & NOTEWORTHY We developed a novel positron emission tomography scan assay with unprecedented temporal and spatial resolution to measure mucociliary clearance in the small airways. We proved a long-standing but unproven assertion that mucociliary clearance is inherently abnormal in the small airways of newborn cystic fibrosis piglets that are otherwise free of infection or inflammation. This technique can be easily extended to other airway diseases such as asthma, idiopathic pulmonary fibrosis, or chronic obstructive pulmonary disease.
Collapse
Grants
- HL136813 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08 HL135433 NHLBI NIH HHS
- P30 CA086862 NCI NIH HHS
- R56 HL147073 NHLBI NIH HHS
- HL051670 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL091842 NHLBI NIH HHS
- R01 HL136813 NHLBI NIH HHS
- HL167025 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL051670 NHLBI NIH HHS
- R01 HL167025 NHLBI NIH HHS
- HL135433 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- ABOU20A0-KB Cystic Fibrosis Foundation (CFF)
- HL091842 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Carley G Stewart
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, United States
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Brieanna M Hilkin
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Nicholas D Gansemer
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ryan J Adam
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - David W Dick
- Department of Radiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - John J Sunderland
- Department of Radiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - David A Stoltz
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, United States
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
- Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Joseph Zabner
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, United States
- Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Mahmoud H Abou Alaiwa
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, United States
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
15
|
Yildirim T, Bali A, Koch M, Paul P, Latta L, Schneider-Daum N, Gallei M, Lehr CM. A New Class of Polyion Complex Vesicles (PIC-somes) to Improve Antimicrobial Activity of Tobramycin in Pseudomonas Aeruginosa Biofilms. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401926. [PMID: 38829185 DOI: 10.1002/smll.202401926] [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: 03/11/2024] [Revised: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Pseudomonas aeruginosa (PA) is a major healthcare concern due to its tolerance to antibiotics when enclosed in biofilms. Tobramycin (Tob), an effective cationic aminoglycoside antibiotic against planktonic PA, loses potency within PA biofilms due to hindered diffusion caused by interactions with anionic biofilm components. Loading Tob into nano-carriers can enhance its biofilm efficacy by shielding its charge. Polyion complex vesicles (PIC-somes) are promising nano-carriers for charged drugs, allowing higher drug loadings than liposomes and polymersomes. In this study, a new class of nano-sized PIC-somes, formed by Tob-diblock copolymer complexation is presented. This approach replaces conventional linear PEG with brush-like poly[ethylene glycol (methyl ether methacrylate)] (PEGMA) in the shell-forming block, distinguishing it from past methods. Tob paired with a block copolymer containing hydrophilic PEGMA induces micelle formation (PIC-micelles), while incorporating hydrophobic pyridyldisulfide ethyl methacrylate (PDSMA) monomer into PEGMA chains reduces shell hydrophilicity, leads to the formation of vesicles (PIC-somes). PDSMA unit incorporation enables unprecedented dynamic disulfide bond-based shell cross-linking, significantly enhancing stability under saline conditions. Neither PIC-somes nor PIC-micelles show any relevant cytotoxicity on A549, Calu-3, and dTHP-1 cells. Tob's antimicrobial efficacy against planktonic PA remains unaffected after encapsulation into PIC-somes and PIC-micelles, but its potency within PA biofilms significantly increases.
Collapse
Affiliation(s)
- Turgay Yildirim
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Aghiad Bali
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Pascal Paul
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Lorenz Latta
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Nicole Schneider-Daum
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
| | - Markus Gallei
- Polymer Chemistry, Saarland University, Campus C4 2, 66123, Saarbrücken, Germany
- Saarene - Saarland Center for Energy Materials and Sustainability, Campus C4 2, 66123, Saarbrücken, Germany
| | - Claus-Michael Lehr
- HIPS - Helmholtz Institute for Pharmaceutical Research Saarland, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
16
|
Lakhani A, Clementina R, Siddiqua Z, Shroff S, Bhavanam S, Pandya MH, Bagadia B, Khan U, Koyappathodi Machingal MM, Kimmatkar A, Chunawala P, Singh H, Reza H, Katta M. Exploring Therapeutic Strategies for Pediatric Cystic Fibrosis: An In-Depth Comparative Review. Cureus 2024; 16:e71913. [PMID: 39564025 PMCID: PMC11576056 DOI: 10.7759/cureus.71913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 11/21/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder that profoundly affects the respiratory and digestive systems, particularly in pediatric patients. This comprehensive review aims to conduct a comparative analysis of various treatment modalities employed in the management of CF in children. We systematically evaluated current literature focusing on pharmacological interventions, airway clearance techniques, nutritional management, and emerging therapies, including gene therapy and personalized medicine. Our analysis highlights the efficacy, safety, and accessibility of these treatments through a comparative lens, examining performance across diverse patient populations. Key comparisons include standard therapies, such as CF transmembrane conductance regulator (CFTR) modulators, versus traditional treatments and the effectiveness of airway clearance techniques in relation to lung function outcomes. We explore variations in treatment adherence and outcomes based on socioeconomic factors and healthcare systems. The review underscores the importance of individualized care plans tailored to the unique needs of pediatric patients. By synthesizing findings from clinical studies, meta-analyses, and expert guidelines, this review serves as a valuable resource for healthcare providers and researchers. Our goal is to optimize therapeutic strategies for pediatric patients with CF and ultimately improve their clinical outcomes.
Collapse
Affiliation(s)
- Alisha Lakhani
- Research, Research MD, Vadodara, IND
- Medicine, Shantabaa Medical College, Amreli, IND
| | | | | | - Shailee Shroff
- Internal Medicine, Gujarat Medical Education and Research Society (GMERS) Medical College, Himmatnagar, IND
| | | | | | - Bhoomi Bagadia
- Medicine, KLE Academy of Higher Education and Research (KAHER) Jawaharlal Nehru Medical College, Belagavi, IND
| | - Usman Khan
- Medicine, Akhtar Saeed Medical and Dental College, Lahore, PAK
| | | | | | - Prachi Chunawala
- Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | | | - Hasim Reza
- Medicine, Central America Health Sciences University, Belize City, BLZ
| | - Madhavi Katta
- Medicine, Maharashtra Institute of Medical Education and Research, Pune, IND
| |
Collapse
|
17
|
Frings M, Welsner M, Mousa C, Zensen S, Salhöfer L, Meetschen M, Beck N, Bos D, Westhölter D, Wienker J, Taube C, Umutlu L, Schaarschmidt BM, Forsting M, Haubold J, Sutharsan S, Opitz M. Low-dose high-resolution chest CT in adults with cystic fibrosis: intraindividual comparison between photon-counting and energy-integrating detector CT. Eur Radiol Exp 2024; 8:105. [PMID: 39298080 DOI: 10.1186/s41747-024-00502-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/02/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Regular disease monitoring with low-dose high-resolution (LD-HR) computed tomography (CT) scans is necessary for the clinical management of people with cystic fibrosis (pwCF). The aim of this study was to compare the image quality and radiation dose of LD-HR protocols between photon-counting CT (PCCT) and energy-integrating detector system CT (EID-CT) in pwCF. METHODS This retrospective study included 23 pwCF undergoing LD-HR chest CT with PCCT who had previously undergone LD-HR chest CT with EID-CT. An intraindividual comparison of radiation dose and image quality was conducted. The study measured the dose-length product, volumetric CT dose index, effective dose and signal-to-noise ratio (SNR). Three blinded radiologists assessed the overall image quality, image sharpness, and image noise using a 5-point Likert scale ranging from 1 (deficient) to 5 (very good) for image quality and image sharpness and from 1 (very high) to 5 (very low) for image noise. RESULTS PCCT used approximately 42% less radiation dose than EID-CT (median effective dose 0.54 versus 0.93 mSv, p < 0.001). PCCT was consistently rated higher than EID-CT for overall image quality and image sharpness. Additionally, image noise was lower with PCCT compared to EID-CT. The average SNR of the lung parenchyma was lower with PCCT compared to EID-CT (p < 0.001). CONCLUSION In pwCF, LD-HR chest CT protocols using PCCT scans provided significantly better image quality and reduced radiation exposure compared to EID-CT. RELEVANCE STATEMENT In pwCF, regular follow-up could be performed through photon-counting CT instead of EID-CT, with substantial advantages in terms of both lower radiation exposure and increased image quality. KEY POINTS Photon-counting CT (PCCT) and energy-integrating detector system CT (EID-CT) were compared in 23 people with cystic fibrosis (pwCF). Image quality was rated higher for PCCT than for EID-CT. PCCT used approximately 42% less radiation dose and offered superior image quality than EID-CT.
Collapse
Affiliation(s)
- Marko Frings
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany.
| | - Matthias Welsner
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
- Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
| | - Christin Mousa
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Sebastian Zensen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Luca Salhöfer
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Mathias Meetschen
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Nikolas Beck
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Denise Bos
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Dirk Westhölter
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
| | - Johannes Wienker
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
| | - Lale Umutlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Benedikt M Schaarschmidt
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Michael Forsting
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Johannes Haubold
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Sivagurunathan Sutharsan
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
- Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany
| | - Marcel Opitz
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| |
Collapse
|
18
|
Carbone A, Vitullo P, Di Gioia S, Castellani S, Conese M. A New Frontier in Cystic Fibrosis Pathophysiology: How and When Clock Genes Can Affect the Inflammatory/Immune Response in a Genetic Disease Model. Curr Issues Mol Biol 2024; 46:10396-10410. [PMID: 39329970 PMCID: PMC11430433 DOI: 10.3390/cimb46090618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Cystic fibrosis (CF) is a monogenic syndrome caused by variants in the CF Transmembrane Conductance Regulator (CFTR) gene, affecting various organ and systems, in particular the lung, pancreas, sweat glands, liver, gastrointestinal tract, vas deferens, and vascular system. While for some organs, e.g., the pancreas, a strict genotype-phenotype occurs, others, such as the lung, display a different pathophysiologic outcome in the presence of the same mutational asset, arguing for genetic and environmental modifiers influencing severity and clinical trajectory. CFTR variants trigger a pathophysiological cascade of events responsible for chronic inflammatory responses, many aspects of which, especially related to immunity, are not ascertained yet. Although clock genes expression and function are known modulators of the innate and adaptive immunity, their involvement in CF has been only observed in relation to sleep abnormalities. The aim of this review is to present current evidence on the clock genes role in immune-inflammatory responses at the lung level. While information on this topic is known in other chronic airway diseases (chronic obstructive pulmonary disease and asthma), CF lung disease (CFLD) is lacking in this knowledge. We will present the bidirectional effect between clock genes and inflammatory factors that could possibly be implicated in the CFLD. It must be stressed that besides sleep disturbance and its mechanisms, there are not studies directly addressing the exact nature of clock genes' involvement in inflammation and immunity in CF, pointing out the directions of new and deepened studies in this monogenic affection. Importantly, clock genes have been found to be druggable by means of genetic tools or pharmacological agents, and this could have therapeutic implications in CFLD.
Collapse
Affiliation(s)
- Annalucia Carbone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Pamela Vitullo
- Cystic Fibrosis Support Center, Ospedale “G. Tatarella”, 71042 Cerignola, Italy;
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Stefano Castellani
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| |
Collapse
|
19
|
Slimmen LJ, Broos JY, Manaï BH, Estevão SC, Giera M, Kooij G, Unger WW, Janssens HM. The Omega-6 Lipid pathway shift is associated with neutrophil influx and structural lung damage in early cystic fibrosis lung disease. Clin Transl Immunology 2024; 13:e70000. [PMID: 39286529 PMCID: PMC11403467 DOI: 10.1002/cti2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/24/2024] [Accepted: 08/06/2024] [Indexed: 09/19/2024] Open
Abstract
Objectives In cystic fibrosis (CF), an imbalanced lipid metabolism is associated with lung inflammation. Little is known about the role that specific lipid mediators (LMs) exert in CF lung inflammation, and whether their levels change during early disease progression. Therefore, we measured airway LM profiles of young CF patients, correlating these with disease-associated parameters. Methods Levels of omega (ω)-3/6 PUFAs and their LM derivatives were determined in bronchoalveolar lavage fluid (BALF) of children with CF ages 1-5 using a targeted high-performance liquid chromatography-tandem mass spectrometry approach. Hierarchical clustering analysis was performed on relative LM levels. Individual relative LM levels were correlated with neutrophilic inflammation (BALF %Neu) and structural lung damage (PRAGMA-CF %Disease). Significant correlations were included in a backward multivariate linear regression model to identify the LMs that are best related to disease progression. Results A total of 65 BALF samples were analysed for ω-3/6 lipid content. LM profiles clustered into an arachidonic acid (AA)-enriched and a linoleic acid (LA)-enriched sample cluster. AA derivatives like 17-OH-DH-HETE, 5-HETE, 5,15-diHETE, 15-HETE, 15-KETE, LTB4 and 6-trans-LTB4 positively correlated with BALF %Neu and/or PRAGMA %Dis. Contrastingly, 9-HoTrE and the LA derivatives 9-HoDE, 9(10)-EpOME, 9(10)-DiHOME, 13-HoDE, 13-oxoODE and 12(13)-EpOME negatively correlated with BALF %Neu and/or PRAGMA %Dis. 6-trans-LTB4 was the strongest predictor for BALF %Neu. 5-HETE and 15-KETE contributed most to PRAGMA %Dis prediction. Conclusions Our data provide more insight into the lung lipidome of infants with CF, and show that a shift from LA derivatives to AA derivatives in BALF associates with early CF lung disease progression.
Collapse
Affiliation(s)
- Lisa Jm Slimmen
- Division of Respiratory Medicine and Allergology, Department of Paediatrics Erasmus MC Sophia Children's Hospital, Erasmus University Medical Centre Rotterdam The Netherlands
- Laboratory of Paediatrics, Infection and Immunity Group, Department of Paediatrics Erasmus University Medical Centre Rotterdam The Netherlands
| | - Jelle Y Broos
- Department of Molecular Cell Biology and Immunology, MS Centre Amsterdam Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Amsterdam The Netherlands
- Center for Proteomics and Metabolomics Leiden University Medical Centre Leiden The Netherlands
| | - Badies Han Manaï
- Division of Respiratory Medicine and Allergology, Department of Paediatrics Erasmus MC Sophia Children's Hospital, Erasmus University Medical Centre Rotterdam The Netherlands
| | - Silvia C Estevão
- Laboratory of Paediatrics, Infection and Immunity Group, Department of Paediatrics Erasmus University Medical Centre Rotterdam The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics Leiden University Medical Centre Leiden The Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, MS Centre Amsterdam Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC Amsterdam The Netherlands
| | - Wendy Wj Unger
- Laboratory of Paediatrics, Infection and Immunity Group, Department of Paediatrics Erasmus University Medical Centre Rotterdam The Netherlands
| | - Hettie M Janssens
- Division of Respiratory Medicine and Allergology, Department of Paediatrics Erasmus MC Sophia Children's Hospital, Erasmus University Medical Centre Rotterdam The Netherlands
| |
Collapse
|
20
|
Coca Barbado A, Zamarrón de Lucas E, Álvarez-Sala Walther R, Prados Sánchez MC. [Self-care capacity of adult cystic fibrosis patients during transition/transfer between services]. An Sist Sanit Navar 2024; 47:e1084. [PMID: 39185776 PMCID: PMC11408906 DOI: 10.23938/assn.1084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
BACKGROUND To analyze the knowledge, abilities, and emotional state of cystic fibrosis patients during a specific follow-up period and compare this with the recall they had of the transition (planned and gradual shift from the pediatric unit) / transfer (direct change skipping the steps recommended by the guidelines) to a specialized cystic fibrosis adult unit. METHODS Prospective cross-sectional study with cystic fibrosis adult patients under follow-up in a specialist consultation. Group 1 were patients who transitioned and Group 2 were transferred patients. The following information was collected: sociodemographic variables, degree of knowledge, skills, and emotional state using a survey designed for this purpose (as part of the internal consistency validation process). Participants also completed the emotional subscale of Cystic Fibrosis Questionnaire-Revised. Inter-group comparisons were made for the transition/transfer, at the follow-up, and during the evolution. RESULTS Thirty-five patients were analyzed; 65.8% male; mean age 31.9 years (SD =10.1). At the transition, Group 1 (n=19; 54.3%) had greater knowledge about their medication and reduced ability to manage appointments and making decisions in comparison to Group 2 at transfer. At follow-up, Group 1 made a better report on their emotional state and significantly improved their ability to manage appointments, communication, and decision-making. CONCLUSIONS Patients who were moved to an adult cystic fibrosis unit through transition were more knowledgeable about their medications. However, those who were transferred managed their appointments and decision-making better, but felt sadder.
Collapse
Affiliation(s)
- Amalia Coca Barbado
- Universidad Autónoma de Madrid. Facultad de Medicina y Cirugía. Departamento de Medicina. Madrid. España..
| | - Ester Zamarrón de Lucas
- Servicio Madrileño de Salud. Hospital Universitario la Paz. Servicio de Neumología. Madrid. España..
| | | | | |
Collapse
|
21
|
Katsoulis O, Pitts OR, Singanayagam A. The airway mycobiome and interactions with immunity in health and chronic lung disease. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae009. [PMID: 39206335 PMCID: PMC11357796 DOI: 10.1093/oxfimm/iqae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
The existence of commensal fungi that reside within the respiratory tract, termed the airway mycobiome, has only recently been discovered. Studies are beginning to characterize the spectrum of fungi that inhabit the human upper and lower respiratory tract but heterogeneous sampling and analysis techniques have limited the generalizability of findings to date. In this review, we discuss existing studies that have examined the respiratory mycobiota in healthy individuals and in those with inflammatory lung conditions such as asthma, chronic obstructive pulmonary disease and cystic fibrosis. Associations between specific fungi and features of disease pathogenesis are emerging but the precise functional consequences imparted by mycobiota upon the immune system remain poorly understood. It is imperative that further research is conducted in this important area as a more detailed understanding could facilitate the development of novel approaches to manipulating the mycobiome for therapeutic benefit.
Collapse
Affiliation(s)
- Orestis Katsoulis
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2DD, UK
| | - Oliver R Pitts
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2DD, UK
| | - Aran Singanayagam
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2DD, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2DD, UK
| |
Collapse
|
22
|
Mall MA, Burgel PR, Castellani C, Davies JC, Salathe M, Taylor-Cousar JL. Cystic fibrosis. Nat Rev Dis Primers 2024; 10:53. [PMID: 39117676 DOI: 10.1038/s41572-024-00538-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Cystic fibrosis is a rare genetic disease caused by mutations in CFTR, the gene encoding cystic fibrosis transmembrane conductance regulator (CFTR). The discovery of CFTR in 1989 has enabled the unravelling of disease mechanisms and, more recently, the development of CFTR-directed therapeutics that target the underlying molecular defect. The CFTR protein functions as an ion channel that is crucial for correct ion and fluid transport across epithelial cells lining the airways and other organs. Consequently, CFTR dysfunction causes a complex multi-organ disease but, to date, most of the morbidity and mortality in people with cystic fibrosis is due to muco-obstructive lung disease. Cystic fibrosis care has long been limited to treating symptoms using nutritional support, airway clearance techniques and antibiotics to suppress airway infection. The widespread implementation of newborn screening for cystic fibrosis and the introduction of a highly effective triple combination CFTR modulator therapy that has unprecedented clinical benefits in up to 90% of genetically eligible people with cystic fibrosis has fundamentally changed the therapeutic landscape and improved prognosis. However, people with cystic fibrosis who are not eligible based on their CFTR genotype or who live in countries where they do not have access to this breakthrough therapy remain with a high unmet medical need.
Collapse
Affiliation(s)
- Marcus A Mall
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany.
- German Centre for Lung Research (DZL), Associated Partner Site Berlin, Berlin, Germany.
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Berlin, Germany.
| | - Pierre-Régis Burgel
- Université Paris Cité and Institut Cochin, Inserm U1016, Paris, France
- Department of Respiratory Medicine and National Reference Center for Cystic Fibrosis, Cochin Hospital, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Carlo Castellani
- IRCCS Istituto Giannina Gaslini, Cystic Fibrosis Center, Genoa, Italy
| | - Jane C Davies
- National Heart & Lung Institute, Imperial College London, London, UK
- St Thomas' NHS Trust, London, UK
- Royal Brompton Hospital, Part of Guy's & St Thomas' Trust, London, UK
| | - Matthias Salathe
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Jennifer L Taylor-Cousar
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, USA
- Division of Paediatric Pulmonary Medicine, National Jewish Health, Denver, CO, USA
| |
Collapse
|
23
|
Oppelaar MC, Emond Y, Bannier MAGE, Reijers MHE, van der Vaart H, van der Meer R, Altenburg J, Conemans L, Rottier BL, Nuijsink M, van den Wijngaart LS, Merkus PJFM, Heinen M, Roukema J. Potential, Pitfalls, and Future Directions for Remote Monitoring of Chronic Respiratory Diseases: Multicenter Mixed Methods Study in Routine Cystic Fibrosis Care. J Med Internet Res 2024; 26:e54942. [PMID: 39106098 PMCID: PMC11336494 DOI: 10.2196/54942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/09/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND The current literature inadequately addresses the extent to which remote monitoring should be integrated into care models for chronic respiratory diseases (CRDs). OBJECTIVE This study examined a remote monitoring program (RMP) in cystic fibrosis (CF) by exploring experiences, future perspectives, and use behavior over 3 years, with the aim of developing future directions for remote monitoring in CRDs. METHODS This was a mixed methods, multicenter, observational study in 5 Dutch CF centers following a sequential explanatory design. Self-designed questionnaires using the technology acceptance model were sent out to people with CF who had a minimum of 12 months of experience with the RMP and local health care professionals (HCPs). Questionnaire outcomes were used to inform semistructured interviews with HCPs and people with CF. Qualitative findings were reported following the COREQ (Consolidated Criteria for Reporting Qualitative Research) checklist. Anonymous data on use frequency of all people with CF were analyzed. RESULTS Between the second quarter of 2020 and the end of 2022, a total of 608 people with CF were enrolled in the program, and a total of 9418 lung function tests and 2631 symptom surveys were conducted. In total, 65% (24/37) of HCPs and 89% (72/81) of people with CF responded to the questionnaire, and 7 HCPs and 12 people with CF participated in semistructured interviews. Both people with CF and HCPs were positive about remote monitoring in CF care and found the RMP a good addition to daily care (people with CF: 44/72, 61%; HCPs: 21/24, 88%). Benefits ranged from supporting individual patients to reducing health care consumption. The most valued monitoring tool was home spirometry by both people with CF (66/72, 92%) and HCPs (22/24, 92%). Downsides included the potential to lose sight of patients and negative psychosocial effects, as 17% (12/72) of people with CF experienced some form of stress due to the RMP. A large majority of people with CF (59/72, 82%) and HCPs (22/24, 92%) wanted to keep using the RMP in future, with 79% (19/24) of HCPs and 75% (54/72) of people with CF looking forward to more replacement of in-person care with digital care during periods of well-being. Future perspectives for the RMP were centered on creating hybrid care models, personalizing remote care, and balancing individual benefits with monitoring burden. CONCLUSIONS Remote monitoring has considerable potential in supporting people with CF and HCPs within the CF care model. We identified 4 practice-based future directions for remote monitoring in CF and CRD care. The strategies, ranging from patient driven to prediction driven, can help clinicians, researchers, and policy makers navigate the rapidly changing digital health field, integrate remote monitoring into local care models, and align remote care with patient and clinician needs.
Collapse
Affiliation(s)
- Martinus C Oppelaar
- Department of Pediatric Pulmonology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Yvette Emond
- IQ Health Science Department, Radboud University Medical Center, Nijmegen, Netherlands
| | - Michiel A G E Bannier
- Department of Paediatric Pulmonology, MosaKids Children's Hospital, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Monique H E Reijers
- Department of Pulmonology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hester van der Vaart
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Josje Altenburg
- Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Lennart Conemans
- Department of Respiratory Medicine, Maastricht University Medical Centre+, Maastricht, Netherlands
- Division of Respiratory & Age-related Health, Department of Respiratory Medicine, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht, Netherlands
| | - Bart L Rottier
- Department of Pediatric Pulmonology and Pediatric Allergology, University Medical Center Groningen, Beatrix Children's Hospital, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marianne Nuijsink
- Haga Teaching Hospital, Juliana Children's Hospital, The Hague, Netherlands
| | - Lara S van den Wijngaart
- Department of Pediatric Pulmonology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter J F M Merkus
- Department of Pediatric Pulmonology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| | - Maud Heinen
- IQ Health Science Department, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jolt Roukema
- Department of Pediatric Pulmonology, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
24
|
Saegebrecht LS, Röhlig M, Schaub F, Ballmann M, Stachs O, Fischer DC. Glycemic Variability and the Thickness of Retinal Layers in Cystic Fibrosis Patients with and without Cystic Fibrosis Related Diabetes. Curr Eye Res 2024; 49:759-767. [PMID: 38557392 DOI: 10.1080/02713683.2024.2333770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Patients with cystic fibrosis (CF) are at risk to develop CF related diabetes (CFRD) and subsequently even diabetic neuro- and/or vasculopathy. We sought to determine if there are typical signs of diabetes-related retinal alterations present in CF patients with preserved and impaired glycemic control. METHODS During routine annual examination CF patients were offered an additional 7-day period of real time continuous glucose monitoring (rtCGM) and an ophthalmological examination including retinal optical coherence tomography (OCT). Patients were categorized according to the glycemic control, i.e. the results of an oral glucose tolerance test (OGTT) and rtCGM were taken into consideration. OCT data was analyzed by our previously published visual analysis software generating dedicated and spatially resolved deviation maps for visualization and quantification of differences in total retinal thickness and thickness of retinal nerve fiber layer (RNFL) as well as ganglion cell layer (GCL) in comparison to age-matched healthy controls and patients with either type 1 or type 2 diabetes mellitus. RESULTS Results of the rtCGM and/or OGTT enabled discrimination between patients with normal glycemic control (CFNG; n = 6), with abnormal glycemic control (CFAG; n = 6) and overt CFRD (n = 4). OCT data indicates gradually increasing retinal thinning in all 3 groups, depending on the degree of glucose metabolism disorder compared to healthy controls. At the foveal region total retinal thickness and GCL thickness were significantly thinner in CFRD patients compared to CFNG patients (total retinal thickness: 260.4 µm (239.3-270.8) vs. 275.4 µm (254.3-289.5); GCL: 11.82 µm (11.16-15.25) vs. 17.30 µm (13.95-19.82); each p < 0.05). CONCLUSION Although we investigated a rather small number of patients, we obtained evidence that intraretinal neurodegenerative changes occur in each of our subgroups (CFNG, CFAG, CFRD). Beyond this, our results favor the detrimental role of additional diabetes, as the deviations from healthy controls were most pronounced in the CFRD group and are similar to those seen in patients suffering from type 1 or type 2 diabetes.
Collapse
Affiliation(s)
- Luisa S Saegebrecht
- Department of Pediatrics, Rostock University Medical Centre, Rostock, Germany
- Department of Ophthalmology, Rostock University Medical Centre, Rostock, Germany
| | - Martin Röhlig
- Institute of Visual and Analytic Computing, University of Rostock, Rostock, Germany
| | - Friederike Schaub
- Department of Ophthalmology, Rostock University Medical Centre, Rostock, Germany
| | - Manfred Ballmann
- Department of Pediatrics, Rostock University Medical Centre, Rostock, Germany
| | - Oliver Stachs
- Department of Ophthalmology, Rostock University Medical Centre, Rostock, Germany
| | - Dagmar-C Fischer
- Department of Pediatrics, Rostock University Medical Centre, Rostock, Germany
| |
Collapse
|
25
|
Sims-Lucas S, Goetzman ES, Kleyman TR. Cystic fibrosis-related metabolic defects: crosstalk between ion channels and organs. J Clin Invest 2024; 134:e182329. [PMID: 38949023 PMCID: PMC11213462 DOI: 10.1172/jci182329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Cystic fibrosis is a debilitating disease characterized by a poor medical prognosis due to devastating lung injury. Recent medical advances targeting the major genetic mutation ΔF508 of the cystic fibrosis transmembrane conductance regulator (CFTR) protein have dramatically increased the lifespan of patients with this mutation. This development has led to major changes in the field and has pushed research beyond the ion transport nature of cystic fibrosis and toward multiorgan physiological reprogramming. In this issue of the JCI, Bae, Kim, and colleagues utilized a large animal pig model prior to the onset of disease. They revealed metabolic reprogramming and organ crosstalk that occurred prior to disease progression. These findings provide paradigm-shifting insight into this complex disease.
Collapse
Affiliation(s)
| | | | - Thomas R. Kleyman
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
Sutharsan S, Fischer R, Gleiber W, Horsley A, Crosby J, Guo S, Xia S, Yu R, Newman KB, Elborn JS. Randomised, phase 1/2a trial of ION-827359, an antisense oligonucleotide inhibitor of ENaC. ERJ Open Res 2024; 10:00986-2023. [PMID: 39286058 PMCID: PMC11403593 DOI: 10.1183/23120541.00986-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/11/2024] [Indexed: 09/19/2024] Open
Abstract
Background Hyperactivity of epithelial sodium channel (ENaC) with increased sodium absorption is a feature of cystic fibrosis (CF). ION-827359 is a 2.5-generation antisense oligonucleotide targeted to reduce ENaC protein. This study evaluated ION-827359 safety, pharmacokinetics and pharmacodynamics. Methods In this three-part phase 1/2a, double-blind, randomised study, healthy volunteers received single doses of placebo or ION-827359 (3, 10, 37.5 or 100 mg; Part 1) or multiple doses of placebo or ION-827359 (5×10 mg, 5×37.5 mg, 5×75 mg or 10×37.5 mg; Part 2). People with CF (pwCF) received multiple doses of placebo or ION-827359 (5×10 mg, 5×37.5 mg, 5×75 mg and 5×100 mg; Part 3). Treatments were administered via Pari eFlow© mesh nebuliser. The primary outcome was safety; pharmacokinetic and pharmacodynamic parameters were also assessed. Results 64 healthy volunteers and 34 pwCF were enrolled. ION-827359 was well tolerated with an acceptable safety profile. There were no clinically relevant changes in laboratory values, ECG or vital signs. Systemic drug exposure was low (plasma half-life ∼2 weeks). Multiple doses of ION-827359 were associated with dose-dependent reductions in ENaC mRNA in bronchial epithelium. After multiple dosing, forced expiratory volume in 1 s was slightly higher in pwCF receiving ION-827359 (+2.9% with ION-827359 100 mg versus placebo; p=0.27). Conclusions The tolerability and safety of ION-827359 appear favourable at this stage of investigation. Reduction in ENaC mRNA supports mechanistic efficacy at the doses and regimens tested, and supports further investigation of ION-827359 in pwCF.
Collapse
Affiliation(s)
- Sivagurunathan Sutharsan
- Division of Cystic Fibrosis, Department of Pulmonary Medicine, University Medicine Essen - Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | | | - Wolfgang Gleiber
- Schwerpunkt Pneumologie/Allergologie, Goethe University, Frankfurt, Germany
| | - Alex Horsley
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Jeff Crosby
- Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Shuling Guo
- Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Shuting Xia
- Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | - Rosie Yu
- Ionis Pharmaceuticals, Inc, Carlsbad, CA, USA
| | | | - J Stuart Elborn
- Centre for Infection and Immunity, Queen's University Belfast, Belfast, UK
| |
Collapse
|
27
|
Sheridan MB, Aksit MA, Pagel K, Hetrick K, Shultz-Lutwyche H, Myers B, Buckingham KJ, Pace RG, Ling H, Pugh E, O'Neal WK, Bamshad MJ, Gibson RL, Knowles MR, Blackman SM, Cutting GR, Raraigh KS. The clinical utility of sequencing the entirety of CFTR. J Cyst Fibros 2024; 23:707-715. [PMID: 38734509 DOI: 10.1016/j.jcf.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Cystic fibrosis (CF) is caused by deleterious variants in each CFTR gene. We investigated the utility of whole-gene CFTR sequencing when fewer than two pathogenic or likely pathogenic (P/LP) variants were detected by conventional testing (sequencing of exons and flanking introns) of CFTR. METHODS Individuals with features of CF and a CF-diagnostic sweat chloride concentration with zero or one P/LP variants identified by conventional testing enrolled in the CF Mutation Analysis Program (MAP) underwent whole-gene CFTR sequencing. Replication was performed on individuals enrolled in the CF Genome Project (CFGP), followed by phenotype review and interrogation of other genes. RESULTS Whole-gene sequencing identified a second P/LP variant in 20/43 MAP enrollees (47 %) and 10/22 CFGP enrollees (45 %) who had one P/LP variant after conventional testing. No P/LP variants were detected when conventional testing was negative (MAP: n = 43; CFGP: n = 13). Genome-wide analysis was unable to find an alternative etiology in CFGP participants with fewer than two P/LP CFTR variants and CF could not be confirmed in 91 % following phenotype re-review. CONCLUSIONS Whole-gene CFTR analysis is beneficial in individuals with one previously-identified P/LP variant and a CF-diagnostic sweat chloride. Negative conventional CFTR testing indicates that the phenotype should be re-evaluated.
Collapse
Affiliation(s)
- Molly B Sheridan
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Melis A Aksit
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kymberleigh Pagel
- The Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kurt Hetrick
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hannah Shultz-Lutwyche
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ben Myers
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kati J Buckingham
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Rhonda G Pace
- Department of Medicine, Marsico Lung Institute/UNC CF Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hua Ling
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth Pugh
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wanda K O'Neal
- Department of Medicine, Marsico Lung Institute/UNC CF Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute, Seattle, WA 98195, USA
| | - Ronald L Gibson
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Michael R Knowles
- Department of Medicine, Marsico Lung Institute/UNC CF Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Scott M Blackman
- Division of Pediatric Endocrinology and Diabetes, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Garry R Cutting
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Karen S Raraigh
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
28
|
Liu A, Chokshi M, Nguyen N, Powell RT, Stephan CC, Bao G. Cystic fibrosis cell models for high-throughput analysis and drug screening. J Cyst Fibros 2024; 23:716-724. [PMID: 39060183 DOI: 10.1016/j.jcf.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/24/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
Cystic fibrosis (CF) is a single-gene disorder that affects the lung, digestive system, and other organs. Mutations in the CF transmembrane conductance regulator (CFTR) gene are classified into several classes based on their pathogenic mechanism and clinical severity. The distinct and heterogeneous clinical behavior of each CF class and the respective CFTR mutations have made the development of a durable therapy for all CF patients extremely challenging. While the FDA-approved drug elexacaftor/tezacaftor/ivacaftor (Trikafta) benefits CF patients carrying at least one F508del mutation in CFTR, it's not effective for many CF patients carrying a variety of other CFTR mutations. To establish a better understanding of CF pathophysiology and aid the development of novel therapeutics for different classes of CF patients, we have created four CF-mutation-specific cell models that recapitulate respectively four distinct CF classes and disease phenotypes, as confirmed by sequencing, CFTR mRNA and protein quantification. The channel function of each cell model was first validated using a well-established FLIPR (Fluorescent Imaging Plate Reader) membrane potential assay and then assessed by the YFP-based functional assay. Integrated with a halide-sensitive fluorescent reporter, these CF cell models can be used for high-throughput drug screening, as demonstrated by a proof-of-concept study using Trikafta. These cell models have the potential to advance CFTR mutation-specific therapies thus addressing the unmet needs of CF patients with rare mutations.
Collapse
Affiliation(s)
- Aidi Liu
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| | - Mithil Chokshi
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| | - Nghi Nguyen
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, 77030, USA
| | - Reid T Powell
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, 77030, USA
| | - Clifford C Stephan
- Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA.
| |
Collapse
|
29
|
Sun Y, Chatterjee S, Lian X, Traylor Z, Sattiraju SR, Xiao Y, Dilliard SA, Sung YC, Kim M, Lee SM, Moore S, Wang X, Zhang D, Wu S, Basak P, Wang J, Liu J, Mann RJ, LePage DF, Jiang W, Abid S, Hennig M, Martinez A, Wustman BA, Lockhart DJ, Jain R, Conlon RA, Drumm ML, Hodges CA, Siegwart DJ. In vivo editing of lung stem cells for durable gene correction in mice. Science 2024; 384:1196-1202. [PMID: 38870301 DOI: 10.1126/science.adk9428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/17/2024] [Indexed: 06/15/2024]
Abstract
In vivo genome correction holds promise for generating durable disease cures; yet, effective stem cell editing remains challenging. In this work, we demonstrate that optimized lung-targeting lipid nanoparticles (LNPs) enable high levels of genome editing in stem cells, yielding durable responses. Intravenously administered gene-editing LNPs in activatable tdTomato mice achieved >70% lung stem cell editing, sustaining tdTomato expression in >80% of lung epithelial cells for 660 days. Addressing cystic fibrosis (CF), NG-ABE8e messenger RNA (mRNA)-sgR553X LNPs mediated >95% cystic fibrosis transmembrane conductance regulator (CFTR) DNA correction, restored CFTR function in primary patient-derived bronchial epithelial cells equivalent to Trikafta for F508del, corrected intestinal organoids and corrected R553X nonsense mutations in 50% of lung stem cells in CF mice. These findings introduce LNP-enabled tissue stem cell editing for disease-modifying genome correction.
Collapse
Affiliation(s)
- Yehui Sun
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sumanta Chatterjee
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xizhen Lian
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zachary Traylor
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - Yufen Xiao
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean A Dilliard
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yun-Chieh Sung
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Minjeong Kim
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sang M Lee
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Stephen Moore
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xu Wang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Di Zhang
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shiying Wu
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pratima Basak
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jialu Wang
- ReCode Therapeutics, Menlo Park, CA 94025, USA
| | - Jing Liu
- ReCode Therapeutics, Menlo Park, CA 94025, USA
| | - Rachel J Mann
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - David F LePage
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Weihong Jiang
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Shadaan Abid
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | - Raksha Jain
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ronald A Conlon
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mitchell L Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Craig A Hodges
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Daniel J Siegwart
- Department of Biomedical Engineering, Department of Biochemistry, Simmons Comprehensive Cancer Center, Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
30
|
Esposito A, Rossi A, Stabile M, Pinto G, De Fino I, Melessike M, Tamanini A, Cabrini G, Lippi G, Aureli M, Loberto N, Renda M, Galietta LJV, Amoresano A, Dechecchi MC, De Gregorio E, Bragonzi A, Guaragna A. Assessing the Potential of N-Butyl-l-deoxynojirimycin (l-NBDNJ) in Models of Cystic Fibrosis as a Promising Antibacterial Agent. ACS Pharmacol Transl Sci 2024; 7:1807-1822. [PMID: 38898954 PMCID: PMC11184606 DOI: 10.1021/acsptsci.4c00044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 06/21/2024]
Abstract
Over the past few years, l-iminosugars have revealed attractive pharmacological properties for managing rare diseases including Cystic Fibrosis (CF). The iminosugar N-butyl-l-deoxynojirimycin (l-NBDNJ, ent-1), prepared by a carbohydrate-based route, was herein evaluated for its anti-inflammatory and anti-infective potential in models of CF lung disease infection. A significant decrease in the bacterial load in the airways was observed in the murine model of Pseudomonas aeruginosa chronic infection in the presence of l-NBDNJ, also accompanied by a modest reduction of inflammatory cells. Mechanistic insights into the observed activity revealed that l-NBDNJ interferes with the expression of proteins regulating cytoskeleton assembly and organization of the host cell, downregulates the main virulence factors of P. aeruginosa involved in the host response, and affects pathogen adhesion to human cells. These findings along with the observation of the absence of an in vitro bacteriostatic/bactericidal action of l-NBDNJ suggest the potential use of this glycomimetic as an antivirulence agent in the management of CF lung disease.
Collapse
Affiliation(s)
- Anna Esposito
- Department
of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples I-80125, Italy
| | - Alice Rossi
- Infections
and Cystic Fibrosis Unit, Division of Immunology, Transplantation
and Infectious Diseases, IRCCS San Raffaele
Scientific Institute, Milan I-20132, Italy
| | - Maria Stabile
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples I-80131, Italy
| | - Gabriella Pinto
- Department
of Chemical Sciences, University of Naples
Federico II, Naples I-80126, Italy
| | - Ida De Fino
- Infections
and Cystic Fibrosis Unit, Division of Immunology, Transplantation
and Infectious Diseases, IRCCS San Raffaele
Scientific Institute, Milan I-20132, Italy
| | - Medede Melessike
- Infections
and Cystic Fibrosis Unit, Division of Immunology, Transplantation
and Infectious Diseases, IRCCS San Raffaele
Scientific Institute, Milan I-20132, Italy
| | - Anna Tamanini
- Section
of Clinical Biochemistry, Department of Engineering for Innovation
Medicine, University of Verona, Verona I-37134, Italy
| | - Giulio Cabrini
- Center on
Innovative Therapies for Cystic Fibrosis, Department of Life Sciences
and Biotechnology, University of Ferrara, Ferrara I-40121, Italy
| | - Giuseppe Lippi
- Section
of Clinical Biochemistry, Department of Engineering for Innovation
Medicine, University of Verona, Verona I-37134, Italy
| | - Massimo Aureli
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, Milan I-20054, Italy
| | - Nicoletta Loberto
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, Milan I-20054, Italy
| | - Mario Renda
- Telethon
Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples I-80078, Italy
| | - Luis J. V. Galietta
- Telethon
Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples I-80078, Italy
- Department
of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples I-80131, Italy
| | - Angela Amoresano
- Department
of Chemical Sciences, University of Naples
Federico II, Naples I-80126, Italy
- Istituto
Nazionale Biostrutture e Biosistemi, Consorzio Interuniversitario, Rome I-00136, Italy
| | - Maria Cristina Dechecchi
- Section
of Clinical Biochemistry, Department of Engineering for Innovation
Medicine, University of Verona, Verona I-37134, Italy
| | - Eliana De Gregorio
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples I-80131, Italy
| | - Alessandra Bragonzi
- Infections
and Cystic Fibrosis Unit, Division of Immunology, Transplantation
and Infectious Diseases, IRCCS San Raffaele
Scientific Institute, Milan I-20132, Italy
| | - Annalisa Guaragna
- Department
of Chemical Sciences, University of Naples
Federico II, Naples I-80126, Italy
| |
Collapse
|
31
|
Xiu W, Dong H, Chen X, Wan L, Lu L, Yang K, Yuwen L, Li Q, Ding M, Zhang Y, Mou Y, Wang L. Metabolic Modulation-Mediated Antibiotic and Immune Activation for Treatment of Chronic Lung Infections. ACS NANO 2024; 18:15204-15217. [PMID: 38803167 DOI: 10.1021/acsnano.4c03527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The Pseudomonas aeruginosa biofilm in recalcitrant chronic lung infections not only develops high antimicrobial tolerance but also induces an aberrant host inflammatory response. The metabolic condition plays a vital role in both the antimicrobial susceptibility of bacteria and the inflammatory response of immune cells, thereby offering a potential therapeutic target. Herein, we described a metabolic modulation strategy by using ultrasound-responsive liposomal nanoparticles containing a sonosensitizer and a hypoxia-activated prodrug against biofilm-associated chronic lung infections. Under ultrasound stimulation, the sonosensitizer generates antibacterial reactive oxygen species by oxygen consumption. Subsequently, the oxygen consumption-mediated hypoxia not only induces the anaerobic metabolism of bacteria for antibiotic activation but also triggers the glycolysis pathway of immune cells for inflammatory activation. Such metabolic modulation strategy demonstrated efficient therapeutic efficacy for P. aeruginosa biofilm-induced chronic lung infections in mice models and provides a promising way for combating biofilm-associated chronic infections.
Collapse
Affiliation(s)
- Weijun Xiu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Heng Dong
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaolong Chen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Ling Wan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Liang Lu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Kaili Yang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Lihui Yuwen
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| | - Qiang Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Meng Ding
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yu Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing210023, China
| |
Collapse
|
32
|
Upadhyay K, Nigam N, Gupta S, Tripathi SK, Jain A, Puri B. Current and future therapeutic approaches of CFTR and airway dysbiosis in an era of personalized medicine. J Family Med Prim Care 2024; 13:2200-2208. [PMID: 39027867 PMCID: PMC11254065 DOI: 10.4103/jfmpc.jfmpc_1085_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 12/06/2023] [Accepted: 01/17/2024] [Indexed: 07/20/2024] Open
Abstract
Cystic fibrosis (CF) is a life-threatening genetic disorder caused by mutations in the CFTR gene. This leads to a defective protein that impairs chloride transport, resulting in thick mucus buildup and chronic inflammation in the airways. The review discusses current and future therapeutic approaches for CFTR dysfunction and airway dysbiosis in the era of personalized medicine. Personalized medicine has revolutionized CF treatment with the advent of CFTR modulator therapies that target specific genetic mutations. These therapies have significantly improved patient outcomes, slowing disease progression, and enhancing quality of life. It also highlights the growing recognition of the airway microbiome's role in CF pathogenesis and discusses strategies to modulate the microbiome to further improve patient outcomes. This review discusses various therapeutic approaches for cystic fibrosis (CFTR) mutations, including adenovirus gene treatments, nonviral vectors, CRISPR/cas9 methods, RNA replacement, antisense-oligonucleotide-mediated DNA-based therapies, and cell-based therapies. It also introduces airway dysbiosis with CF and how microbes influence the lungs. The review highlights the importance of understanding the cellular and molecular causes of CF and the development of personalized medicine to improve quality of life and health outcomes.
Collapse
Affiliation(s)
- Kirti Upadhyay
- Cytogenetics Lab, Centre for Advance Research, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Nitu Nigam
- Cytogenetics Lab, Centre for Advance Research, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Surbhi Gupta
- Cytogenetics Lab, Centre for Advance Research, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Surya Kant Tripathi
- Department of Respiratory Medicine, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Amita Jain
- Department of Microbiology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Bipin Puri
- King George’s Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
33
|
Turuvekere Vittala Murthy N, Vlasova K, Renner J, Jozic A, Sahay G. A new era of targeting cystic fibrosis with non-viral delivery of genomic medicines. Adv Drug Deliv Rev 2024; 209:115305. [PMID: 38626860 DOI: 10.1016/j.addr.2024.115305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/21/2024]
Abstract
Cystic fibrosis (CF) is a complex genetic respiratory disorder that necessitates innovative gene delivery strategies to address the mutations in the gene. This review delves into the promises and challenges of non-viral gene delivery for CF therapy and explores strategies to overcome these hurdles. Several emerging technologies and nucleic acid cargos for CF gene therapy are discussed. Novel formulation approaches including lipid and polymeric nanoparticles promise enhanced delivery through the CF mucus barrier, augmenting the potential of non-viral strategies. Additionally, safety considerations and regulatory perspectives play a crucial role in navigating the path toward clinical translation of gene therapy.
Collapse
Affiliation(s)
| | - Kseniia Vlasova
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA
| | - Jonas Renner
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy at Oregon State University, Corvallis, OR 97331, USA; Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97201, USA; Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health & Science University, Portland, OR 97201, USA.
| |
Collapse
|
34
|
Wu X, Yang Y. Neutrophil extracellular traps (NETs) and fibrotic diseases. Int Immunopharmacol 2024; 133:112085. [PMID: 38626550 DOI: 10.1016/j.intimp.2024.112085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/18/2024]
Abstract
Fibrosis, a common cause and serious outcome of organ failure that can affect any organ, is responsible for up to 45% of all deaths in various clinical settings. Both preclinical models and clinical trials investigating various organ systems have shown that fibrosis is a highly dynamic process. Although many studies have sought to gain understanding of the mechanism of fibrosis progression, their findings have been mixed. In recent years, increasing evidence indicates that neutrophil extracellular traps (NETs) are involved in many inflammatory and autoimmune disorders and participate in the regulation of fibrotic processes in various organs and systems. In this review, we summarize the current understanding of the role of NETs in fibrosis development and progression and their possibility as therapeutic targets.
Collapse
Affiliation(s)
- Xiaojiao Wu
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
35
|
Stewart CG, Hilkin BM, Gansemer ND, Dick DW, Sunderland JJ, Stoltz DA, Abou Alaiwa MH, Zabner J. Mucociliary Clearance is Impaired in Small Airways of Cystic Fibrosis Pigs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595427. [PMID: 38826411 PMCID: PMC11142153 DOI: 10.1101/2024.05.22.595427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Rationale Cystic fibrosis is a genetic disorder characterized by recurrent airway infections, inflammation, and progressive decline in lung function. Autopsy and spirometry data suggest that cystic fibrosis may start in the small airways which, due to the fractal nature of the airways, account for most of the airway tree surface area. However, they are not easily accessible for testing. Objectives Here, we tested the hypothesis that mucociliary clearance is abnormal in the small airways of newborn cystic fibrosis pigs. Methods Current mucociliary clearance assays are limited therefore we developed a dynamic positron emission tomography scan assay with high spatial and temporal resolution. Each study was accompanied by a high-resolution computed tomography scan that helped identify the thin outer region of the lung that contained small airways. Measurements and Main Results Clearance of aerosolized [ 68 Ga]macro aggregated albumin from distal airways occurred within minutes after delivery and followed a two-phase process. In cystic fibrosis pigs, both early and late clearance rates were slower. Stimulation of the cystic fibrosis airways with the purinergic agonist UTP further impaired late clearance. Only 1 cystic fibrosis pig treated with UTP out of 6 cleared more than 20% of the delivered dose. Conclusions These data indicate that mucociliary transport in the small airways is fast and can easily be missed if the acquisition is not fast enough. The data also indicate that mucociliary transport is impaired in small airways of cystic fibrosis pigs. This defect is exacerbated by stimulation of mucus secretions with purinergic agonists.
Collapse
|
36
|
Bae H, Kim BR, Jung S, Le J, van der Heide D, Yu W, Park SH, Hilkin BM, Gansemer ND, Powers LS, Kang T, Meyerholz DK, Schuster VL, Jang C, Welsh MJ. Arteriovenous metabolomics in pigs reveals CFTR regulation of metabolism in multiple organs. J Clin Invest 2024; 134:e174500. [PMID: 38743489 PMCID: PMC11213515 DOI: 10.1172/jci174500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF), a multiorgan disease that is characterized by diverse metabolic defects. However, other than specific CFTR mutations, the factors that influence disease progression and severity remain poorly understood. Aberrant metabolite levels have been reported, but whether CFTR loss itself or secondary abnormalities (infection, inflammation, malnutrition, and various treatments) drive metabolic defects is uncertain. Here, we implemented comprehensive arteriovenous metabolomics in newborn CF pigs, and the results revealed CFTR as a bona fide regulator of metabolism. CFTR loss impaired metabolite exchange across organs, including disruption of lung uptake of fatty acids, yet enhancement of uptake of arachidonic acid, a precursor of proinflammatory cytokines. CFTR loss also impaired kidney reabsorption of amino acids and lactate and abolished renal glucose homeostasis. These and additional unexpected metabolic defects prior to disease manifestations reveal a fundamental role for CFTR in controlling multiorgan metabolism. Such discovery informs a basic understanding of CF, provides a foundation for future investigation, and has implications for developing therapies targeting only a single tissue.
Collapse
Affiliation(s)
- Hosung Bae
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Bo Ram Kim
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Sunhee Jung
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | | | - Wenjie Yu
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Sang Hee Park
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - Brieanna M. Hilkin
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Nicholas D. Gansemer
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Linda S. Powers
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Taekyung Kang
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
| | - David K. Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Victor L. Schuster
- Department of Internal Medicine, Albert Einstein College of Medicine, Bronx, New York, New York, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California – Irvine, Irvine, California, USA
- Center for Complex Biological Systems and
- Center for Epigenetics and Metabolism, University of California – Irvine, Irvine, California, USA
| | - Michael J. Welsh
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
- Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
37
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
38
|
Tręda C, Włodarczyk A, Rieske P. The hope, hype and obstacles surrounding cell therapy. J Cell Mol Med 2024; 28:e18359. [PMID: 38770886 PMCID: PMC11107145 DOI: 10.1111/jcmm.18359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 05/22/2024] Open
Abstract
Cell therapy offers hope, but it also presents challenges, most particularly the limited ability of human organs and tissues to regenerate. Since many diseases are associated with irreversible pathophysiological or traumatic changes, stem cells and their derivatives are unable to secure healing. Although regenerative medicine offers chances for improvements in many diseases, such as type one diabetes and Parkinson's disease, it cannot eliminate the primary cause of many of them. While successes can be expected for diseases such as sickle cell disease, this is not the case for hereditary diseases with varied mutation types or for ciliopathies, which start in embryogenesis. In this complicated medical environment, synthetic biology offers some solutions, but their implementation will take many years. Still, positive examples such as CAR-T therapy offer hope.
Collapse
Affiliation(s)
- Cezary Tręda
- Department of Tumor BiologyMedical University of LodzLodzPoland
| | | | - Piotr Rieske
- Department of Tumor BiologyMedical University of LodzLodzPoland
| |
Collapse
|
39
|
Boillot L, Costes F. [A systemic gene therapy for the treatment of cystic fibrosis]. Med Sci (Paris) 2024; 40:467-470. [PMID: 38819285 DOI: 10.1051/medsci/2024047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
Le master I2VB se propose de donner les bases conceptuelles et pratiques des différents aspects de l’infectiologie. Il s’appuie sur une coopération exemplaire entre les équipes de recherche en infectiologie et en immunologie de l’Université de Tours, et celles, entre autres, de l’Unité Infectiologie et Santé Publique (ISP) du Centre INRAE de Tours-Nouzilly, concrétisée par une profonde interaction entre chercheurs et enseignants-chercheurs.
Cette formation aborde aussi bien les aspects fondamentaux et appliqués de l’infectiologie et de l’immunologie allant de l’étude moléculaire des interactions entre le pathogène et son hôte, jusqu’à la conception et la mise sur le marché des produits de la vaccinologie, des biothérapies anti-infectieuses et des anticorps immuno-thérapeutiques.
Le master I2VB (niveau M1) donne lieu aux parcours ICM, I&B et AcT (niveau M2).
L’option Infectiologie Cellulaire et Moléculaire (ICM) (responsables : Françoise Debierre-Grockiego et Martine Braibant) a pour objectifs de :
former des scientifiques dotés d’une culture générale et technique spécialisée dans les biotechnologies, l’infectiologie, les interactions hôte-pathogène et les mécanismes de la réponse immunitaire anti-infectieuse, contribuant à l’avancée des connaissances scientifiques et à ses applications industrielles, demandes sociétales en forte progression.
former des pharmaciens, médecins, vétérinaires, ingénieurs agronomes aux enjeux actuels de l’infectiologie à la fois dans les domaines fondamentaux et appliqués.
L’option Immunité et biomédicaments (I&B) (responsables : Anne di Tommaso et Isabelle Dimier-Poisson) a pour objectifs de :
former des scientifiques dotés d’une culture générale et technique spécialisée dans les biotechnologies, l’infectiologie, la vaccinologie, les biomédicaments et les biothérapies anti-infectieuses contribuant à l’avancée des connaissances scientifiques et à ses applications industrielles, demandes sociétales en forte progression.
former de jeunes scientifiques, pharmaciens, médecins, vétérinaires, ingénieurs agronomes aux enjeux actuels de l’infectiologie et des biomédicaments à la fois dans les domaines fondamentaux et appliqués.
L’option Anticorps thérapeutiques (AcT) (responsables : Laurie Lajoie et Isabelle Dimier-Poisson) a pour objectifs de :
former des scientifiques dotés d’une culture générale et technique spécialisée dans les biotechnologies, l’immunologie, la cancérologie et les biomédicaments dont les anticorps thérapeutiques, contribuant à l’avancée des connaissances scientifiques et à ses application industrielles et juridiques, demandes sociétales en forte progression.
former de jeunes scientifiques, pharmaciens, médecins, vétérinaires, ingénieurs agronomes aux enjeux actuels de l’infectiologie et des biomédicaments à la fois dans les domaines fondamentaux et appliqués.
Collapse
Affiliation(s)
- Léa Boillot
- Université de Tours, Master 2 infectiologie, immunité, vaccinologie et biomédicaments, Tours, France
| | - Floriane Costes
- Université de Tours, Master 2 infectiologie, immunité, vaccinologie et biomédicaments, Tours, France
| |
Collapse
|
40
|
Bendoukha I, Boucherit-Otmani Z, Baba Ahmed-Kazi Tani ZZ, Seghir A, Madouni M, Radoui AK, Boucherit K. Initial characteristics of cystic fibrosis in Algeria: Description of 34 pediatric cases. Pediatr Pulmonol 2024; 59:1454-1461. [PMID: 38411325 DOI: 10.1002/ppul.26939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 01/28/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND Cystic fibrosis (CF) is a rare disease in Algeria, and its prognosis is poor in developing countries. The clinical and demographic knowledge of Algerian pediatric patients diagnosed with CF is incomplete due to the nonexistence of a national medical registry. Hence, the present study is the first Algerian multicentre study on CF. METHODS This retrospective study was conducted in western Algeria. Over 1 year, the study included all pediatric patients with a confirmed diagnosis of CF in the pediatric hospital of Oran. Patient characteristics, clinical manifestations, and the prescribed treatment were reported. RESULTS Thirty-four children (16 boys and 18 girls) participated in this study. Only 15 were diagnosed before the age of 6 months. The sweat chloride test was positive in all patients. Respiratory manifestations were found in all patients, chronic diarrhoea in 29 of them, and growth retardation in 10. Moreover, 25 (73.5%) had low to low intermediate socioeconomic levels. After diagnosis, respiratory complications marked the evolution of the 34 patients, with bronchial congestion observed in 33 of them, while 10 (29.4%) patients presented severe bronchopneumonia and 4 (11.8%) were affected by asthma. Consequently, three (8.8%) died at an average age of 9 years mainly because of respiratory failure. CONCLUSION The prognosis of CF is poor in Algeria compared to other developed countries due to the longer diagnostic delay and limited therapeutic alternatives. This representative subset of Algerian pediatric patients with CF will serve as a reference for future studies on CF in Algeria.
Collapse
Affiliation(s)
- Imene Bendoukha
- Laboratory Antibiotics Antifungals: Physico-chemical Synthesis and Biological Activity (LapSab), University of Abou Bekr Belkaid, Tlemcen, Algeria
| | - Zahia Boucherit-Otmani
- Laboratory Antibiotics Antifungals: Physico-chemical Synthesis and Biological Activity (LapSab), University of Abou Bekr Belkaid, Tlemcen, Algeria
| | - Zahira Zakia Baba Ahmed-Kazi Tani
- Laboratory Antibiotics Antifungals: Physico-chemical Synthesis and Biological Activity (LapSab), University of Abou Bekr Belkaid, Tlemcen, Algeria
| | - Abdelfettah Seghir
- Laboratory Antibiotics Antifungals: Physico-chemical Synthesis and Biological Activity (LapSab), University of Abou Bekr Belkaid, Tlemcen, Algeria
| | - Mourad Madouni
- Faculties of Economics and Business and Management, University of Tahar Moulay, Saida, Algeria
| | - Abdel Karim Radoui
- Department of Pediatric Pneumology and Allergology, Specialised Hospital Establishment of Canastel, Oran, Algeria
| | - Kebir Boucherit
- Laboratory Antibiotics Antifungals: Physico-chemical Synthesis and Biological Activity (LapSab), University of Abou Bekr Belkaid, Tlemcen, Algeria
| |
Collapse
|
41
|
Keens T, Hoffman V, Topuria I, Elder K, Cerf S, Mulder K, Roberts J, Lysinger J, Del Carmen Reyes M, Berdella M, Cairns AM, Jain M, Ganapathy V, Lou Y, Morcos B, Wu C, Sass L. Real-world effectiveness of elexacaftor/tezacaftor/ivacaftor on the burden of illness in adolescents and adults with cystic fibrosis. Heliyon 2024; 10:e28508. [PMID: 38586424 PMCID: PMC10998118 DOI: 10.1016/j.heliyon.2024.e28508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Background Elexacaftor/tezacaftor/ivacaftor (ELX/TEZ/IVA) has been shown to be safe and efficacious in people with cystic fibrosis (CF) aged ≥2 years. Here, we describe results from an observational study assessing change in burden of illness following initiating ELX/TEZ/IVA in real-world settings. Methods This US-based, multicenter, observational study used data from electronic medical records to evaluate real-world burden of illness before and after ELX/TEZ/IVA initiation in people with CF aged ≥12 years heterozygous for F508del and a minimal function mutation (F/MF) or an uncharacterized CFTR mutation. Endpoints included absolute change from baseline in percent predicted forced expiratory volume in 1 s (ppFEV1), body mass index (BMI) and BMI-for-age z-score, glycated hemoglobin (HbA1c), and numbers of pulmonary exacerbations (PEx). Results Overall, 206 people with CF were enrolled (mean [SD] age 22.5 [11.1] years; 192 [93.2%] with F/MF genotype). Mean follow-up was 15.6 (SD, 1.6) months. Improvements in ppFEV1 (7.3 [95% CI: 5.7, 8.8] percentage points) were observed from baseline through follow-up. Increases in BMI (1.40 [95% CI: 1.07, 1.77] kg/m2) and BMI-for-age z-score (0.14 [95% CI: 0.00, 0.28]) were also observed from baseline at 12 months. The estimated annualized rate of any PEx was 1.31 at baseline and 0.61 over follow-up (rate ratio 0.47 [95% CI: 0.39, 0.55]), with annualized rates of PEx requiring antibiotics and hospitalizations of 0.55 and 0.88 in the baseline period and 0.12 and 0.36 over follow-up (rate ratios 0.22 [95% CI: 0.15, 0.31] and 0.41 [95% CI: 0.32, 0.51]), respectively. Absolute change in HbA1c was -0.22 (95% CI: -0.38, -0.06) from baseline through follow-up. Conclusions ELX/TEZ/IVA treatment was associated with improved lung function, increased BMI, reduced frequency of PEx, and improved (i.e., reduced) HbA1c. These results confirm the broad clinical benefits of ELX/TEZ/IVA seen in clinical trials and show the potential for ELX/TEZ/IVA to improve markers of glucose metabolism.
Collapse
Affiliation(s)
- Thomas Keens
- Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | - Jon Roberts
- Driscoll Children's Hospital, Corpus Christi, TX, USA
| | | | | | | | | | - Manu Jain
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Yiyue Lou
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Bassem Morcos
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Chuntao Wu
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Laura Sass
- Children's Hospital of The King's Daughter, Norfolk, VA, USA
| | - for the VX19-CFD-003 Study Group
- Children's Hospital of Los Angeles, Los Angeles, CA, USA
- OM1 Incorporated, Boston, MA, USA
- Driscoll Children's Hospital, Corpus Christi, TX, USA
- Billings Clinic Hospital, Billings, MT, USA
- Northwell Health, New York, NY, USA
- Maine Medical Center, Portland, ME, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
- Children's Hospital of The King's Daughter, Norfolk, VA, USA
| |
Collapse
|
42
|
Han X, Li D, Zhu Y, Schneider-Futschik EK. Recommended Tool Compounds for Modifying the Cystic Fibrosis Transmembrane Conductance Regulator Channel Variants. ACS Pharmacol Transl Sci 2024; 7:933-950. [PMID: 38633590 PMCID: PMC11019735 DOI: 10.1021/acsptsci.3c00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 04/19/2024]
Abstract
Cystic fibrosis (CF) is a genetic disorder arising from variations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, leading to multiple organ system defects. CFTR tool compounds are molecules that can modify the activity of the CFTR channel. Especially, patients that are currently not able to benefit from approved CFTR modulators, such as patients with rare CFTR variants, benefit from further research in discovering novel tools to modulate CFTR. This Review explores the development and classification of CFTR tool compounds, including CFTR blockers (CFTRinh-172, GlyH-101), potentiators (VRT-532, Genistein), correctors (VRT-325, Corr-4a), and other approved and unapproved modulators, with detailed descriptions and discussions for each compound. The challenges and future directions in targeting rare variants and optimizing drug delivery, and the potential synergistic effects in combination therapies are outlined. CFTR modulation holds promise not only for CF treatment but also for generating CF models that contribute to CF research and potentially treating other diseases such as secretory diarrhea. Therefore, continued research on CFTR tool compounds is critical.
Collapse
Affiliation(s)
- XiaoXuan Han
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Danni Li
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yimin Zhu
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elena K. Schneider-Futschik
- Department of Biochemistry & Pharmacology,
School of Biomedical Sciences, Faculty of Medicine, Dentistry and
Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
43
|
Halperin SJ, Dhodapkar MM, Radford ZJ, Kaszuba SV, Rubin LE, Grauer JN. Patients With Cystic Fibrosis Undergoing Total Hip and Total Knee Arthroplasty Are at Increased Risk for Perioperative Complications. J Am Acad Orthop Surg 2024; 32:309-315. [PMID: 38165956 DOI: 10.5435/jaaos-d-23-00783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/16/2023] [Indexed: 01/04/2024] Open
Abstract
INTRODUCTION Patients with cystic fibrosis (CF) are living longer and may be considered for total hip arthroplasty (THA) or total knee arthroplasty (TKA). Perioperative outcomes and implant survival after these procedures performed for those with CF have not been previously described. METHODS Using the M151 PearlDiver database, a large, national, administrative database, THA and TKA patients with and without CF were identified and matched 1:10 based on age, sex, and Elixhauser Comorbidity Index. Ninety-day perioperative outcomes and 2-year revision rates were assessed and compared with multivariable logistic regression. RESULTS For THA, 185 patients with CF were matched with 1,846 control subjects without CF. Patients with CF were at significantly increased odds of 90-day postoperative events including sepsis (odd radio [OR] 4.15), pneumonia (OR 3.40), pleural effusion (OR 2.77), minor events (OR 1.73), any adverse event (OR 1.64), urinary tract infection (UTI) (OR 1.63), and severe events (OR 1.60) ( P < 0.05 for each). For TKA, 505 patients with CF were matched with 5,047 control subjects without CF. Patients with CF were at significantly increased odds of 90-day postoperative events including pneumonia (OR 4.95), respiratory failure (OR 4.31), cardiac event (OR 2.29), minor events (OR 2.16), pleural effusion (OR 2.35), severe events (OR 2.06), urinary tract infection (OR 2.06), any adverse event (OR 1.96), atelectasis (OR 1.94), and acute kidney injury (OR 1.61) ( P < 0.05 for each). For both THA and TKA, those with CF were not at greater odds of 2-year rates of revision. DISCUSSION After THA and TKA, those with CF were found to be at increased odds of multiple defined postoperative events (predominantly infectious/pulmonary), but not 2-year revision rates. These findings help define areas in need of focused optimization and are reassuring regarding risks of surgery.
Collapse
Affiliation(s)
- Scott J Halperin
- From the Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, CT
| | | | | | | | | | | |
Collapse
|
44
|
Patrick SC, Beer PD, Davis JJ. Solvent effects in anion recognition. Nat Rev Chem 2024; 8:256-276. [PMID: 38448686 DOI: 10.1038/s41570-024-00584-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
Anion recognition is pertinent to a range of environmental, medicinal and industrial applications. Recent progress in the field has relied on advances in synthetic host design to afford a broad range of potent recognition motifs and novel supramolecular structures capable of effective binding both in solution and at derived molecular films. However, performance in aqueous media remains a critical challenge. Understanding the effects of bulk and local solvent on anion recognition by host scaffolds is imperative if effective and selective detection in real-world media is to be viable. This Review seeks to provide a framework within which these effects can be considered both experimentally and theoretically. We highlight proposed models for solvation effects on anion binding and discuss approaches to retain strong anion binding in highly competitive (polar) solvents. The synthetic design principles for exploiting the aforementioned solvent effects are explored.
Collapse
Affiliation(s)
| | - Paul D Beer
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Jason J Davis
- Department of Chemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
45
|
Baker PJ, Bohrer AC, Castro E, Amaral EP, Snow-Smith M, Torres-Juárez F, Gould ST, Queiroz ATL, Fukutani ER, Jordan CM, Khillan JS, Cho K, Barber DL, Andrade BB, Johnson RF, Hilligan KL, Mayer-Barber KD. The inflammatory microenvironment of the lung at the time of infection governs innate control of SARS-CoV-2 replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586885. [PMID: 38585846 PMCID: PMC10996686 DOI: 10.1101/2024.03.27.586885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
SARS-CoV-2 infection leads to vastly divergent clinical outcomes ranging from asymptomatic infection to fatal disease. Co-morbidities, sex, age, host genetics and vaccine status are known to affect disease severity. Yet, how the inflammatory milieu of the lung at the time of SARS-CoV-2 exposure impacts the control of viral replication remains poorly understood. We demonstrate here that immune events in the mouse lung closely preceding SARS-CoV-2 infection significantly impact viral control and we identify key innate immune pathways required to limit viral replication. A diverse set of pulmonary inflammatory stimuli, including resolved antecedent respiratory infections with S. aureus or influenza, ongoing pulmonary M. tuberculosis infection, ovalbumin/alum-induced asthma or airway administration of defined TLR ligands and recombinant cytokines, all establish an antiviral state in the lung that restricts SARS-CoV-2 replication upon infection. In addition to antiviral type I interferons, the broadly inducible inflammatory cytokines TNFα and IL-1 precondition the lung for enhanced viral control. Collectively, our work shows that SARS-CoV-2 may benefit from an immunologically quiescent lung microenvironment and suggests that heterogeneity in pulmonary inflammation that precedes or accompanies SARS-CoV-2 exposure may be a significant factor contributing to the population-wide variability in COVID-19 disease outcomes.
Collapse
Affiliation(s)
- Paul J. Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Current Address: Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Ehydel Castro
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Eduardo P. Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Maryonne Snow-Smith
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Human Eosinophil Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Flor Torres-Juárez
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Sydnee T. Gould
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
- Current Address: Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Artur T. L. Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Eduardo R. Fukutani
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Cassandra M. Jordan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Jaspal S. Khillan
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Kyoungin Cho
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Daniel L. Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Bruno B. Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Reed F. Johnson
- SCV2 Virology Core, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Kerry L. Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| |
Collapse
|
46
|
Lan PD, Nissley DA, Sitarik I, Vu QV, Jiang Y, To P, Xia Y, Fried SD, Li MS, O'Brien EP. Synonymous Mutations Can Alter Protein Dimerization Through Localized Interface Misfolding Involving Self-entanglements. J Mol Biol 2024; 436:168487. [PMID: 38341172 PMCID: PMC11260358 DOI: 10.1016/j.jmb.2024.168487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Synonymous mutations in messenger RNAs (mRNAs) can reduce protein-protein binding substantially without changing the protein's amino acid sequence. Here, we use coarse-grain simulations of protein synthesis, post-translational dynamics, and dimerization to understand how synonymous mutations can influence the dimerization of two E. coli homodimers, oligoribonuclease and ribonuclease T. We synthesize each protein from its wildtype, fastest- and slowest-translating synonymous mRNAs in silico and calculate the ensemble-averaged interaction energy between the resulting dimers. We find synonymous mutations alter oligoribonuclease's dimer properties. Relative to wildtype, the dimer interaction energy becomes 4% and 10% stronger, respectively, when translated from its fastest- and slowest-translating mRNAs. Ribonuclease T dimerization, however, is insensitive to synonymous mutations. The structural and kinetic origin of these changes are misfolded states containing non-covalent lasso-entanglements, many of which structurally perturb the dimer interface, and whose probability of occurrence depends on translation speed. These entangled states are kinetic traps that persist for long time scales. Entanglements cause altered dimerization energies for oligoribonuclease, as there is a large association (odds ratio: 52) between the co-occurrence of non-native self-entanglements and weak-binding dimer conformations. Simulated at all-atom resolution, these entangled structures persist for long timescales, indicating the conclusions are independent of model resolution. Finally, we show that regions of the protein we predict to have changes in entanglement are also structurally perturbed during refolding, as detected by limited-proteolysis mass spectrometry. Thus, non-native changes in entanglement at dimer interfaces is a mechanism through which oligomer structure and stability can be altered.
Collapse
Affiliation(s)
- Pham Dang Lan
- Institute for Computational Sciences and Technology, Ho Chi Minh City, Viet Nam; Faculty of Physics and Engineering Physics, VNUHCM-University of Science, 227, Nguyen Van Cu Street, District 5, Ho Chi Minh City, Viet Nam
| | - Daniel Allen Nissley
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Ian Sitarik
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Quyen V Vu
- Institute of Physics, Polish Academy of Sciences, 02-668 Warsaw, Poland
| | - Yang Jiang
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Philip To
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yingzi Xia
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Stephen D Fried
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA; Thomas C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Mai Suan Li
- Institute for Computational Sciences and Technology, Ho Chi Minh City, Viet Nam; Institute of Physics, Polish Academy of Sciences, 02-668 Warsaw, Poland
| | - Edward P O'Brien
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA; Bioinformatics and Genomics Graduate Program, The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA; Institute for Computational and Data Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
47
|
Díez Rodríguez GR, Figueredo Lago JE, Armas Cayarga A, González González YJ, García de la Rosa I, Collazo Mesa T, López Reyes I, Batista Lozada Y, Rodríguez Calá FR, García Sánchez JB. A novel high-resolution melting analysis strategy for detecting cystic fibrosis-causing variants. Lab Med 2024; 55:185-197. [PMID: 37417450 DOI: 10.1093/labmed/lmad058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023] Open
Abstract
Cystic fibrosis (CF), an autosomal recessive disease, is caused by variants in both alleles of the CF transmembrane conductance regulator (CFTR) gene. A new assay based on allele-specific polymerase chain reaction and high-resolution melting analysis was developed for the detection of 18 CF-causing CFTR variants previously identified in Cuba and Latin America. The assay is also useful for zygosity determination of mutated alleles and includes internal controls. The reaction mixtures were normalized and evaluated using blood samples collected on filter paper. The evaluation of analytical parameters demonstrated the specificity and sensitivity of the method to detect the included CFTR variants. Internal and external validations yielded a 100% agreement between the new assay and the used reference tests. This assay can complement CF newborn screening not only in Cuba but also in Latin America.
Collapse
Affiliation(s)
| | | | | | | | | | - Teresa Collazo Mesa
- National Center of Medical Genetics, Medical University of Havana, Playa, Havana, Cuba
| | - Ixchel López Reyes
- National Center of Medical Genetics, Medical University of Havana, Playa, Havana, Cuba
| | | | | | | |
Collapse
|
48
|
Lee RE, Mascenik TM, Major SC, Galiger JR, Bulik-Sullivan E, Siesser PF, Lewis CA, Bear JE, Le Suer JA, Hawkins FJ, Pickles RJ, Randell SH. Viral airway injury promotes cell engraftment in an in vitro model of cystic fibrosis cell therapy. Am J Physiol Lung Cell Mol Physiol 2024; 326:L226-L238. [PMID: 38150545 PMCID: PMC11280688 DOI: 10.1152/ajplung.00421.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023] Open
Abstract
Cell therapy is a potential treatment for cystic fibrosis (CF). However, cell engraftment into the airway epithelium is challenging. Here, we model cell engraftment in vitro using the air-liquid interface (ALI) culture system by injuring well-differentiated CF ALI cultures and delivering non-CF cells at the time of peak injury. Engraftment efficiency was quantified by measuring chimerism by droplet digital PCR and functional ion transport in Ussing chambers. Using this model, we found that human bronchial epithelial cells (HBECs) engraft more efficiently when they are cultured by conditionally reprogrammed cell (CRC) culture methods. Cell engraftment into the airway epithelium requires airway injury, but the extent of injury needed is unknown. We compared three injury models and determined that severe injury with partial epithelial denudation facilitates long-term cell engraftment and functional CFTR recovery up to 20% of wildtype function. The airway epithelium promptly regenerates in response to injury, creating competition for space and posing a barrier to effective engraftment. We examined competition dynamics by time-lapse confocal imaging and found that delivered cells accelerate airway regeneration by incorporating into the epithelium. Irradiating the repairing epithelium granted engrafting cells a competitive advantage by diminishing resident stem cell proliferation. Intentionally, causing severe injury to the lungs of people with CF would be dangerous. However, naturally occurring events like viral infection can induce similar epithelial damage with patches of denuded epithelium. We found that viral preconditioning promoted effective engraftment of cells primed for viral resistance.NEW & NOTEWORTHY Cell therapy is a potential treatment for cystic fibrosis (CF). Here, we model cell engraftment by injuring CF air-liquid interface cultures and delivering non-CF cells. Successful engraftment required severe epithelial injury. Intentionally injuring the lungs to this extent would be dangerous. However, naturally occurring events like viral infection induce similar epithelial damage. We found that viral preconditioning promoted the engraftment of cells primed for viral resistance leading to CFTR functional recovery to 20% of the wildtype.
Collapse
Affiliation(s)
- Rhianna E Lee
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Teresa M Mascenik
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Sidra C Major
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Jacob R Galiger
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Emily Bulik-Sullivan
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Priscila F Siesser
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Catherine A Lewis
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Jake A Le Suer
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts, United States
- Department of Medicine, The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Finn J Hawkins
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts, United States
- Department of Medicine, The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Raymond J Pickles
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
49
|
Shiratsuchi G, Konishi S, Yano T, Yanagihashi Y, Nakayama S, Katsuno T, Kashihara H, Tanaka H, Tsukita K, Suzuki K, Herawati E, Watanabe H, Hirai T, Yagi T, Kondoh G, Gotoh S, Tamura A, Tsukita S. Dual-color live imaging unveils stepwise organization of multiple basal body arrays by cytoskeletons. EMBO Rep 2024; 25:1176-1207. [PMID: 38316902 PMCID: PMC10933483 DOI: 10.1038/s44319-024-00066-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 02/07/2024] Open
Abstract
For mucociliary clearance of pathogens, tracheal multiciliated epithelial cells (MCCs) organize coordinated beating of cilia, which originate from basal bodies (BBs) with basal feet (BFs) on one side. To clarify the self-organizing mechanism of coordinated intracellular BB-arrays composed of a well-ordered BB-alignment and unidirectional BB-orientation, determined by the direction of BB to BF, we generated double transgenic mice with GFP-centrin2-labeled BBs and mRuby3-Cep128-labeled BFs for long-term, high-resolution, dual-color live-cell imaging in primary-cultured tracheal MCCs. At early timepoints of MCC differentiation, BB-orientation and BB-local alignment antecedently coordinated in an apical microtubule-dependent manner. Later during MCC differentiation, fluctuations in BB-orientation were restricted, and locally aligned BB-arrays were further coordinated to align across the entire cell (BB-global alignment), mainly in an apical intermediate-sized filament-lattice-dependent manner. Thus, the high coordination of the BB-array was established for efficient mucociliary clearance as the primary defense against pathogen infection, identifying apical cytoskeletons as potential therapeutic targets.
Collapse
Affiliation(s)
- Gen Shiratsuchi
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Satoshi Konishi
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Tomoki Yano
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | | | - Shogo Nakayama
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- RIKEN Center for Biosystems Dynamics Research, Hyogo, Japan
| | - Tatsuya Katsuno
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroka Kashihara
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Hiroo Tanaka
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- School of Medicine, Teikyo University, Tokyo, Japan
| | - Kazuto Tsukita
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koya Suzuki
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Elisa Herawati
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Faculty of Mathematics and Natural Sciences, Universitas Sebelas Maret, Surakarta, Central Java, Indonesia
| | - Hitomi Watanabe
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Yagi
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Gen Kondoh
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shimpei Gotoh
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Atsushi Tamura
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan.
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
- School of Medicine, Teikyo University, Tokyo, Japan.
| | - Sachiko Tsukita
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan.
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
50
|
Cicuta P. Mucociliary transport in cystic fibrosis. Proc Natl Acad Sci U S A 2024; 121:e2400674121. [PMID: 38377219 PMCID: PMC10907239 DOI: 10.1073/pnas.2400674121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024] Open
Affiliation(s)
- Pietro Cicuta
- Cavendish Laboratory, University of Cambridge, CambridgeCB3 0HE, United Kingdom
| |
Collapse
|