1
|
Hinata D, Fukuda R, Ishiguro H, Kamada Y, Okiyoneda T. Enhanced CFTR modulator efficacy in ΔF508 CFTR mouse organoids by ablation of RFFL ubiquitin ligase. Biochem Biophys Res Commun 2024; 733:150433. [PMID: 39047427 DOI: 10.1016/j.bbrc.2024.150433] [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: 07/15/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
The most common CFTR mutant in cystic fibrosis (CF), ΔF508 CFTR, is eliminated by ubiquitination even in the presence of CF drugs, reducing their therapeutic efficacy. RFFL is one of the ubiquitin ligases that remove ΔF508 CFTR from the cell surface despite treatment with the triple combination of CFTR modulators (TEZ/ELX/IVA) used clinically. Although RFFL knockdown has been shown to enhance the efficacy of TEZ/ELX/IVA in cell culture models, its impact in mouse models has not been evaluated. Here, we demonstrate that RFFL ablation significantly improves the effect of TEZ/ELX/IVA, resulting in enhanced function of ΔF508 CFTR in mouse organoids. Since RFFL knockout mice showed no significant abnormalities, our findings support RFFL inhibition as a promising strategy to improve CFtreatment.
Collapse
Affiliation(s)
- Daichi Hinata
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, 669-1330, Hyogo, Japan
| | - Ryosuke Fukuda
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, 669-1330, Hyogo, Japan
| | - Hiroshi Ishiguro
- Department of Human Nutrition, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuka Kamada
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, 669-1330, Hyogo, Japan
| | - Tsukasa Okiyoneda
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, 669-1330, Hyogo, Japan.
| |
Collapse
|
2
|
Barabas AJ, Conlon RA, Hodges CA. Think Beyond the Room: Measuring Relative Humidity in the Home Cage and Its Impact on Reproduction in Laboratory Mice, Mus musculus. Animals (Basel) 2024; 14:3164. [PMID: 39595217 PMCID: PMC11591041 DOI: 10.3390/ani14223164] [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: 10/04/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Relative humidity (RH) is measured in vivaria with a broad range to accommodate seasonal fluctuations. It is assumed that measurements in the room (macroenvironment) reflect those in the cage (microenvironment). However, there is limited data comparing RH in the macroenvironment to the microenvironment and how the mice may be affected by variations in RH that fall within husbandry recommendations. This study aimed to compare RH in the macroenvironment to that of the microenvironment in various group sizes of laboratory mice; and examine how variation in microenvironmental RH impacts pup survival. Temperature and RH were measured using a temperature/humidity data logger attached to a solid top cage lid. The lid was rotated across N = 48 breeding trios and N = 33 same sex cages on a C57BL/6J background. Further, once a week, a single breeding trio was selected (N = 23) to compare RH readings to weekly rates of pup loss in a larger breeding colony. Across all cages, RH was higher in the microenvironment than the macroenvironment. RH was universally higher in the summer than in the winter, and increased with group size. For breeding cages, as microenvironmental RH increased, the proportion of pups lost each week decreased in a linear relationship. No threshold of decreased mortality could be identified. These data highlight RH as a potential extrinsic factor. While these patterns are correlational, they warrant further research focused on the causative role of RH on mouse welfare.
Collapse
Affiliation(s)
- Amanda J. Barabas
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA; (R.A.C.); (C.A.H.)
| | | | | |
Collapse
|
3
|
Gookin JL, Holmes J, Clarke LL, Stauffer SH, Meredith B, Vandewege MW, Torres-Machado N, Friedenberg SG, Seiler GS, Mathews KG, Meurs K. Acquired dysfunction of CFTR underlies cystic fibrosis-like disease of the canine gallbladder. Am J Physiol Gastrointest Liver Physiol 2024; 327:G513-G530. [PMID: 39041675 PMCID: PMC11482251 DOI: 10.1152/ajpgi.00145.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Mucocele formation in dogs is a unique and enigmatic muco-obstructive disease of the gallbladder caused by the amassment of abnormal mucus that bears striking pathological similarity to cystic fibrosis. We investigated the role of cystic fibrosis transmembrane conductance regulatory protein (CFTR) in the pathogenesis of this disease. The location and frequency of disease-associated variants in the coding region of CFTR were compared using whole genome sequence data from 2,642 dogs representing breeds at low-risk, high-risk, or with confirmed disease. Expression, localization, and ion transport activity of CFTR were quantified in control and mucocele gallbladders by NanoString, Western blotting, immunofluorescence imaging, and studies in Ussing chambers. Our results establish a significant loss of CFTR-dependent anion secretion by mucocele gallbladder mucosa. A significantly lower quantity of CFTR protein was demonstrated relative to E-cadherin in mucocele compared with control gallbladder mucosa. Immunofluorescence identified CFTR along the apical membrane of epithelial cells in control gallbladders but not in mucocele gallbladder epithelium. Decreases in mRNA copy number for CFTR were accompanied by decreases in mRNA for the Cl-/[Formula: see text] exchanger SLC26A3, K+ channels (KCNQ1, KCNN4), and vasoactive intestinal polypeptide receptor (VIPR1), which suggest a driving force for change in secretory function of gallbladder epithelial cells in the pathogenesis of mucocele formation. There were no significant differences in CFTR gene variant frequency, type, or predicted impact comparing low-risk, high-risk, and definitively diagnosed groups of dogs. This study describes a unique, naturally occurring muco-obstructive disease of the canine gallbladder, with uncanny similarity to cystic fibrosis, and driven by the underlying failure of CFTR function.NEW & NOTEWORTHY Cystic fibrosis transmembrane conductance regulatory protein (CFTR) genomic variants and expression of mRNA, protein, and electrogenic anion secretory activity of CFTR were characterized in dog gallbladder. Acquired inhibition of CFTR expression by gallbladder epithelium was identified as underpinning a naturally occurring muco-obstructive disease of the dog gallbladder that bears striking pathological similarity to animal models of cystic fibrosis.
Collapse
Affiliation(s)
- Jody L Gookin
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Jenny Holmes
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Lane L Clarke
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Stephen H Stauffer
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Bryanna Meredith
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Michael W Vandewege
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Nicole Torres-Machado
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, Minnesota, United States
| | - Gabriela S Seiler
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Kyle G Mathews
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| | - Kathryn Meurs
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States
| |
Collapse
|
4
|
Duncan RP, Moustafa DA, Lewin GR, Diggle FL, Bomberger JM, Whiteley M, Goldberg JB. Improvement of a mouse infection model to capture Pseudomonas aeruginosa chronic physiology in cystic fibrosis. Proc Natl Acad Sci U S A 2024; 121:e2406234121. [PMID: 39102545 PMCID: PMC11331117 DOI: 10.1073/pnas.2406234121] [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: 03/26/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Laboratory models are central to microbiology research, advancing the understanding of bacterial physiology by mimicking natural environments, from soil to the human microbiome. When studying host-bacteria interactions, animal models enable investigators to examine bacterial dynamics associated with a host, and in the case of human infections, animal models are necessary to translate basic research into clinical treatments. Efforts toward improving animal infection models are typically based on reproducing host genotypes/phenotypes and disease manifestations, leaving a gap in how well the physiology of microbes reflects their behavior in a human host. Understanding bacterial physiology is vital because it dictates host response and bacterial interactions with antimicrobials. Thus, our goal was to develop an animal model that accurately recapitulates bacterial physiology in human infection. The system we chose to model was a chronic Pseudomonas aeruginosa respiratory infection in cystic fibrosis (CF). To accomplish this goal, we leveraged a framework that we recently developed to evaluate model accuracy by calculating the percentage of bacterial genes that are expressed similarly in a model to how they are expressed in their infection environment. We combined two complementary models of P. aeruginosa infection-an in vitro synthetic CF sputum model (SCFM2) and a mouse acute pneumonia model. This combined model captured the chronic physiology of P. aeruginosa in CF better than the standard mouse infection model, showing the power of a data-driven approach to refining animal models. In addition, the results of this work challenge the assumption that a chronic infection model requires long-term colonization.
Collapse
Affiliation(s)
- Rebecca P. Duncan
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA30322
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
| | - Dina A. Moustafa
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA30322
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
| | - Gina R. Lewin
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
- School of Biological Sciences and Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA30322
| | - Frances L. Diggle
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
- School of Biological Sciences and Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA30322
| | - Jennifer M. Bomberger
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA15219
| | - Marvin Whiteley
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
- School of Biological Sciences and Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA30322
| | - Joanna B. Goldberg
- Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA30322
- Emory-Children’s Cystic Fibrosis Center, Atlanta, GA30322
| |
Collapse
|
5
|
Fröhlich E. Animals in Respiratory Research. Int J Mol Sci 2024; 25:2903. [PMID: 38474149 DOI: 10.3390/ijms25052903] [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: 01/17/2024] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The respiratory barrier, a thin epithelial barrier that separates the interior of the human body from the environment, is easily damaged by toxicants, and chronic respiratory diseases are common. It also allows the permeation of drugs for topical treatment. Animal experimentation is used to train medical technicians, evaluate toxicants, and develop inhaled formulations. Species differences in the architecture of the respiratory tract explain why some species are better at predicting human toxicity than others. Some species are useful as disease models. This review describes the anatomical differences between the human and mammalian lungs and lists the characteristics of currently used mammalian models for the most relevant chronic respiratory diseases (asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary hypertension, pulmonary fibrosis, and tuberculosis). The generation of animal models is not easy because they do not develop these diseases spontaneously. Mouse models are common, but other species are more appropriate for some diseases. Zebrafish and fruit flies can help study immunological aspects. It is expected that combinations of in silico, in vitro, and in vivo (mammalian and invertebrate) models will be used in the future for drug development.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
- Research Center Pharmaceutical Engineering GmbH, 8010 Graz, Austria
| |
Collapse
|
6
|
Manzor M, Koutsogiannaki S, DiBlasi M, Schaefers M, Priebe G, Yuki K. Cystic Fibrosis Mice Are Highly Susceptible to Repeated Acute Pseudomonas aeruginosa Pneumonia after Intranasal Inoculation. BIOMED RESEARCH INTERNATIONAL 2024; 2024:4769779. [PMID: 38347907 PMCID: PMC10861279 DOI: 10.1155/2024/4769779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/15/2024]
Abstract
Cystic fibrosis (CF) is a genetic disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) that controls chloride current. A number of different CFTR transgenic mouse lines have been developed and subjected to both acute and chronic infection models. However, prior studies showed no substantial differences in bacterial clearance between CF and non-CF mice after single inoculations. Here, using F508del transgenic CF mice, we examined the role of repeated acute Pseudomonas aeruginosa (PA) infection, with the second inoculation 7 days after the first. We found that CF mice were more susceptible to PA infection than non-CF mice following the second inoculation, with non-CF mice showing better neutrophil recruitment and effector functions. We further investigated the characteristics of lung immune cells using single-cell RNA sequencing, finding that non-CF lung neutrophils had more prominent upregulation of adhesion molecules including intercellular adhesion molecule-1 (ICAM-1) compared to CF lung neutrophils. Although people with CF are often colonized with bacteria and have high numbers of neutrophils in the airways during chronic infection, these data suggest that CF neutrophils have deficient effector functions in the setting of repeated acute infection.
Collapse
Affiliation(s)
- Mariel Manzor
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, USA
| | - Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, USA
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Immunology, Harvard Medical School, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| | - Marco DiBlasi
- Department of Anesthesiology, Critical Care and Pain Medicine, Critical Care Division, Boston Children's Hospital, Boston, USA
| | - Matthew Schaefers
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Critical Care Division, Boston Children's Hospital, Boston, USA
| | - Gregory Priebe
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Critical Care Division, Boston Children's Hospital, Boston, USA
- Department of Pediatrics, Division of Infectious Diseases, Boston Children's Hospital, Boston, USA
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, USA
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Immunology, Harvard Medical School, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, USA
| |
Collapse
|
7
|
Kunzelmann K, Ousingsawat J, Kraus A, Park JH, Marquardt T, Schreiber R, Buchholz B. Pathogenic Relationships in Cystic Fibrosis and Renal Diseases: CFTR, SLC26A9 and Anoctamins. Int J Mol Sci 2023; 24:13278. [PMID: 37686084 PMCID: PMC10487509 DOI: 10.3390/ijms241713278] [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: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The Cl--transporting proteins CFTR, SLC26A9, and anoctamin (ANO1; ANO6) appear to have more in common than initially suspected, as they all participate in the pathogenic process and clinical outcomes of airway and renal diseases. In the present review, we will therefore concentrate on recent findings concerning electrolyte transport in the airways and kidneys, and the role of CFTR, SLC26A9, and the anoctamins ANO1 and ANO6. Special emphasis will be placed on cystic fibrosis and asthma, as well as renal alkalosis and polycystic kidney disease. In essence, we will summarize recent evidence indicating that CFTR is the only relevant secretory Cl- channel in airways under basal (nonstimulated) conditions and after stimulation by secretagogues. Information is provided on the expressions of ANO1 and ANO6, which are important for the correct expression and function of CFTR. In addition, there is evidence that the Cl- transporter SLC26A9 expressed in the airways may have a reabsorptive rather than a Cl--secretory function. In the renal collecting ducts, bicarbonate secretion occurs through a synergistic action of CFTR and the Cl-/HCO3- transporter SLC26A4 (pendrin), which is probably supported by ANO1. Finally, in autosomal dominant polycystic kidney disease (ADPKD), the secretory function of CFTR in renal cyst formation may have been overestimated, whereas ANO1 and ANO6 have now been shown to be crucial in ADPKD and therefore represent new pharmacological targets for the treatment of polycystic kidney disease.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Physiological Institute, University of Regensburg, University Street 31, 93053 Regensburg, Germany; (J.O.); (R.S.)
| | - Jiraporn Ousingsawat
- Physiological Institute, University of Regensburg, University Street 31, 93053 Regensburg, Germany; (J.O.); (R.S.)
| | - Andre Kraus
- Department of Nephrology and Hypertension, Friedrich Alexander University Erlangen Nuremberg, 91054 Erlangen, Germany; (A.K.); (B.B.)
| | - Julien H. Park
- Department of Pediatrics, University Hospital Münster, 48149 Münster, Germany; (J.H.P.); (T.M.)
| | - Thorsten Marquardt
- Department of Pediatrics, University Hospital Münster, 48149 Münster, Germany; (J.H.P.); (T.M.)
| | - Rainer Schreiber
- Physiological Institute, University of Regensburg, University Street 31, 93053 Regensburg, Germany; (J.O.); (R.S.)
| | - Björn Buchholz
- Department of Nephrology and Hypertension, Friedrich Alexander University Erlangen Nuremberg, 91054 Erlangen, Germany; (A.K.); (B.B.)
| |
Collapse
|
8
|
Blutt SE, Coarfa C, Neu J, Pammi M. Multiomic Investigations into Lung Health and Disease. Microorganisms 2023; 11:2116. [PMID: 37630676 PMCID: PMC10459661 DOI: 10.3390/microorganisms11082116] [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: 07/12/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Diseases of the lung account for more than 5 million deaths worldwide and are a healthcare burden. Improving clinical outcomes, including mortality and quality of life, involves a holistic understanding of the disease, which can be provided by the integration of lung multi-omics data. An enhanced understanding of comprehensive multiomic datasets provides opportunities to leverage those datasets to inform the treatment and prevention of lung diseases by classifying severity, prognostication, and discovery of biomarkers. The main objective of this review is to summarize the use of multiomics investigations in lung disease, including multiomics integration and the use of machine learning computational methods. This review also discusses lung disease models, including animal models, organoids, and single-cell lines, to study multiomics in lung health and disease. We provide examples of lung diseases where multi-omics investigations have provided deeper insight into etiopathogenesis and have resulted in improved preventative and therapeutic interventions.
Collapse
Affiliation(s)
- Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Josef Neu
- Department of Pediatrics, Section of Neonatology, University of Florida, Gainesville, FL 32611, USA;
| | - Mohan Pammi
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX 77030, USA
| |
Collapse
|
9
|
Madrahimov N, Mutsenko V, Natanov R, Radaković D, Klapproth A, Hassan M, Rosenfeldt M, Kleefeldt F, Aleksic I, Ergün S, Otto C, Leyh RG, Bening C. Multiorgan recovery in a cadaver body using mild hypothermic ECMO treatment in a murine model. Intensive Care Med Exp 2023; 11:46. [PMID: 37537415 PMCID: PMC10400742 DOI: 10.1186/s40635-023-00534-2] [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: 02/10/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Transplant candidates on the waiting list are increasingly challenged by the lack of organs. Most of the organs can only be kept viable within very limited timeframes (e.g., mere 4-6 h for heart and lungs exposed to refrigeration temperatures ex vivo). Donation after circulatory death (DCD) using extracorporeal membrane oxygenation (ECMO) can significantly enlarge the donor pool, organ yield per donor, and shelf life. Nevertheless, clinical attempts to recover organs for transplantation after uncontrolled DCD are extremely complex and hardly reproducible. Therefore, as a preliminary strategy to fulfill this task, experimental protocols using feasible animal models are highly warranted. The primary aim of the study was to develop a model of ECMO-based cadaver organ recovery in mice. Our model mimics uncontrolled organ donation after an "out-of-hospital" sudden unexpected death with subsequent "in-hospital" cadaver management post-mortem. The secondary aim was to assess blood gas parameters, cardiac activity as well as overall organ state. The study protocol included post-mortem heparin-streptokinase administration 10 min after confirmed death induced by cervical dislocation under full anesthesia. After cannulation, veno-arterial ECMO (V-A ECMO) was started 1 h after death and continued for 2 h under mild hypothermic conditions followed by organ harvest. Pressure- and flow-controlled oxygenated blood-based reperfusion of a cadaver body was accompanied by blood gas analysis (BGA), electrocardiography, and histological evaluation of ischemia-reperfusion injury. For the first time, we designed and implemented, a not yet reported, miniaturized murine hemodialysis circuit for the treatment of severe hyperkalemia and metabolic acidosis post-mortem. RESULTS BGA parameters confirmed profound ischemia typical for cadavers and incompatible with normal physiology, including extremely low blood pH, profound negative base excess, and enormously high levels of lactate. Two hours after ECMO implantation, blood pH values of a cadaver body restored from < 6.5 to 7.3 ± 0.05, pCO2 was lowered from > 130 to 41.7 ± 10.5 mmHg, sO2, base excess, and HCO3 were all elevated from below detection thresholds to 99.5 ± 0.6%, - 4 ± 6.2 and 22.0 ± 6.0 mmol/L, respectively (Student T test, p < 0.05). A substantial decrease in hyperlactatemia (from > 20 to 10.5 ± 1.7 mmol/L) and hyperkalemia (from > 9 to 6.9 ± 1.0 mmol/L) was observed when hemodialysis was implemented. On balance, the first signs of regained heart activity appeared on average 10 min after ECMO initiation without cardioplegia or any inotropic and vasopressor support. This was followed by restoration of myocardial contractility with a heart rate of up to 200 beats per minute (bpm) as detected by an electrocardiogram (ECG). Histological examinations revealed no evidence of heart injury 3 h post-mortem, whereas shock-specific morphological changes relevant to acute death and consequent cardiac/circulatory arrest were observed in the lungs, liver, and kidney of both control and ECMO-treated cadaver mice. CONCLUSIONS Thus, our model represents a promising approach to facilitate studying perspectives of cadaveric multiorgan recovery for transplantation. Moreover, it opens new possibilities for cadaver organ treatment to extend and potentiate donation and, hence, contribute to solving the organ shortage dilemma.
Collapse
Affiliation(s)
- Nodir Madrahimov
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany.
| | - Vitalii Mutsenko
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Ruslan Natanov
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Dejan Radaković
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany
| | - André Klapproth
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Mohamed Hassan
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Mathias Rosenfeldt
- Institute for Pathology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Florian Kleefeldt
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Ivan Aleksic
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Rainer G Leyh
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Constanze Bening
- Department of Thoracic and Cardiovascular Surgery, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
10
|
Januska MN, Walsh MJ. Single-Cell RNA Sequencing Reveals New Basic and Translational Insights in the Cystic Fibrosis Lung. Am J Respir Cell Mol Biol 2023; 68:131-139. [PMID: 36194688 PMCID: PMC9986558 DOI: 10.1165/rcmb.2022-0038tr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 10/04/2022] [Indexed: 02/03/2023] Open
Abstract
Cystic fibrosis (CF) is a multisystemic, autosomal recessive disorder caused by mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene, with the majority of morbidity and mortality extending from lung disease. Single-cell RNA sequencing (scRNA-seq) has been leveraged in the lung and elsewhere in the body to articulate discrete cell populations, describing cell types, states, and lineages as well as their roles in health and disease. In this translational review, we provide an overview of the current applications of scRNA-seq to the study of the normal and CF lungs, allowing the beginning of a new cellular and molecular narrative of CF lung disease, and we highlight some of the future opportunities to further leverage scRNA-seq and complementary single-cell technologies in the study of CF as we bridge from scientific understanding to clinical application.
Collapse
Affiliation(s)
- Megan N. Januska
- Department of Pediatrics
- Department of Genetics and Genomic Sciences, and
| | - Martin J. Walsh
- Department of Pediatrics
- Department of Genetics and Genomic Sciences, and
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and
- Mount Sinai Center for RNA Biology and Medicine, New York, New York
| |
Collapse
|
11
|
Secli V, Di Biagio C, Martini A, Michetti E, Pacello F, Ammendola S, Battistoni A. Localized Infections with P. aeruginosa Strains Defective in Zinc Uptake Reveal That Zebrafish Embryos Recapitulate Nutritional Immunity Responses of Higher Eukaryotes. Int J Mol Sci 2023; 24:ijms24020944. [PMID: 36674459 PMCID: PMC9862628 DOI: 10.3390/ijms24020944] [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: 11/09/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
The innate immune responses of mammals to microbial infections include strategies based on manipulating the local concentration of metals such as iron (Fe) and zinc (Zn), commonly described as nutritional immunity. To evaluate whether these strategies are also present in zebrafish embryos, we have conducted a series of heart cavity-localized infection experiments with Pseudomonas aeruginosa strains characterized by a different ability to acquire Zn. We have found that, 48 h after infection, the bacterial strains lacking critical components of the Zn importers ZnuABC and ZrmABCD have a reduced colonization capacity compared to the wild-type strain. This observation, together with the finding of a high level of expression of Zur-regulated genes, suggests the existence of antimicrobial mechanisms based on Zn sequestration. However, we have observed that strains lacking such Zn importers have a selective advantage over the wild-type strain in the early stages of infection. Analysis of the expression of the gene that encodes for a Zn efflux pump has revealed that at short times after infection, P. aeruginosa is exposed to high concentrations of Zn. At the same time, zebrafish respond to the infection by activating the expression of the Zn transporters Slc30a1 and Slc30a4, whose mammalian homologs mediate a redistribution of Zn in phagocytes aimed at intoxicating bacteria with a metal excess. These observations indicate that teleosts share similar nutritional immunity mechanisms with higher vertebrates, and confirm the usefulness of the zebrafish model for studying host-pathogen interactions.
Collapse
Affiliation(s)
- Valerio Secli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Claudia Di Biagio
- Laboratory of Experimental Ecology and Aquaculture, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Arianna Martini
- Laboratory of Experimental Ecology and Aquaculture, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
- Council for Agricultural Research and Economics, Research, Centre for Animal Production and Aquaculture, Via Salaria 31, 00015 Monterotondo, Italy
| | - Emma Michetti
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Francesca Pacello
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Serena Ammendola
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Andrea Battistoni
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
- Correspondence:
| |
Collapse
|
12
|
Wood SJ, Kuzel TM, Shafikhani SH. Pseudomonas aeruginosa: Infections, Animal Modeling, and Therapeutics. Cells 2023; 12:199. [PMID: 36611992 PMCID: PMC9818774 DOI: 10.3390/cells12010199] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Pseudomonas aeruginosa is an important Gram-negative opportunistic pathogen which causes many severe acute and chronic infections with high morbidity, and mortality rates as high as 40%. What makes P. aeruginosa a particularly challenging pathogen is its high intrinsic and acquired resistance to many of the available antibiotics. In this review, we review the important acute and chronic infections caused by this pathogen. We next discuss various animal models which have been developed to evaluate P. aeruginosa pathogenesis and assess therapeutics against this pathogen. Next, we review current treatments (antibiotics and vaccines) and provide an overview of their efficacies and their limitations. Finally, we highlight exciting literature on novel antibiotic-free strategies to control P. aeruginosa infections.
Collapse
Affiliation(s)
- Stephen J. Wood
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Timothy M. Kuzel
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sasha H. Shafikhani
- Department of Medicine, Division of Hematology, Oncology, & Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Cancer Center, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Sui H, Xu X, Su Y, Gong Z, Yao M, Liu X, Zhang T, Jiang Z, Bai T, Wang J, Zhang J, Xu C, Luo M. Gene therapy for cystic fibrosis: Challenges and prospects. Front Pharmacol 2022; 13:1015926. [PMID: 36304167 PMCID: PMC9592762 DOI: 10.3389/fphar.2022.1015926] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/29/2022] [Indexed: 11/25/2022] Open
Abstract
Cystic fibrosis (CF) is a life-threatening autosomal-recessive disease caused by mutations in a single gene encoding cystic fibrosis transmembrane conductance regulator (CFTR). CF effects multiple organs, and lung disease is the primary cause of mortality. The median age at death from CF is in the early forties. CF was one of the first diseases to be considered for gene therapy, and efforts focused on treating CF lung disease began shortly after the CFTR gene was identified in 1989. However, despite the quickly established proof-of-concept for CFTR gene transfer in vitro and in clinical trials in 1990s, to date, 36 CF gene therapy clinical trials involving ∼600 patients with CF have yet to achieve their desired outcomes. The long journey to pursue gene therapy as a cure for CF encountered more difficulties than originally anticipated, but immense progress has been made in the past decade in the developments of next generation airway transduction viral vectors and CF animal models that reproduced human CF disease phenotypes. In this review, we look back at the history for the lessons learned from previous clinical trials and summarize the recent advances in the research for CF gene therapy, including the emerging CRISPR-based gene editing strategies. We also discuss the airway transduction vectors, large animal CF models, the complexity of CF pathogenesis and heterogeneity of CFTR expression in airway epithelium, which are the major challenges to the implementation of a successful CF gene therapy, and highlight the future opportunities and prospects.
Collapse
Affiliation(s)
- Hongshu Sui
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| | - Xinghua Xu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Yanping Su
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Zhaoqing Gong
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Minhua Yao
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Xiaocui Liu
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Ting Zhang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Ziyao Jiang
- Department of Histology and Embryology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Tianhao Bai
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Junzuo Wang
- The Affiliated Tai’an City Central Hospital of Qingdao University, Tai’an, Shandong, China
| | - Jingjun Zhang
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Changlong Xu
- The Reproductive Medical Center of Nanning Second People’s Hospital, Nanning, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| | - Mingjiu Luo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
- *Correspondence: Hongshu Sui, ; Changlong Xu, ; Mingjiu Luo,
| |
Collapse
|
14
|
Darwish T, Al-Khulaifi A, Ali M, Mowafy R, Arredouani A, Doi SA, Emara MM. Assessing the consistency of iPSC and animal models in cystic fibrosis modelling: A meta-analysis. PLoS One 2022; 17:e0272091. [PMID: 35944004 PMCID: PMC9362911 DOI: 10.1371/journal.pone.0272091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 07/12/2022] [Indexed: 12/09/2022] Open
Abstract
INTRODUCTION Cystic fibrosis (CF) is a hereditary autosomal recessive disorder caused by a range of mutations in the CF Transmembrane Conductance Regulator (CFTR) gene. This gene encodes the CFTR protein, which acts as a chloride channel activated by cyclic AMP (cAMP). This meta-analysis aimed to compare the responsiveness of induced pluripotent stem cells (iPSCs) to cAMP analogues to that of commonly used animal models. METHODS Databases searched included PubMed, Scopus, and Medline from inception to January 2020. A total of 8 and 3 studies, respectively, for animal models and iPSCs, were analyzed. Studies were extracted for investigating cAMP-stimulated anion transport by measuring the short circuit current (Isc) of chloride channels in different animal models and iPSC systems We utilized an inverse variance heterogeneity model for synthesis. RESULTS Our analysis showed considerable heterogeneity in the mean Isc value in both animal models and iPSCs studies (compared to their WT counterparts), and both suffer from variable responsiveness based on the nature of the underlying model. There was no clear advantage of one over the other. CONCLUSIONS Studies on both animal and iPSCs models generated considerable heterogeneity. Given the potential of iPSC-derived models to study different diseases, we recommend paying more attention to developing reproducible models of iPSC as it has potential if adequately developed.
Collapse
Affiliation(s)
- Toqa Darwish
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Azhar Al-Khulaifi
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Menatalla Ali
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Rana Mowafy
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| | - Abdelilah Arredouani
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, Doha, Qatar
| | - Suhail A. Doi
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mohamed M. Emara
- Basic Medical Sciences Department, College of Medicine, Qatar University, Doha, Qatar
| |
Collapse
|
15
|
Philp AR, Miranda F, Gianotti A, Mansilla A, Scudieri P, Musante I, Vega G, Figueroa CD, Galietta LJV, Sarmiento JM, Flores CA. KCa3.1 differentially regulates trachea and bronchi epithelial gene expression in a chronic-asthma mouse model. Physiol Genomics 2022; 54:273-282. [PMID: 35658672 DOI: 10.1152/physiolgenomics.00134.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ion channels are potentially exploitable as pharmacological targets to treat asthma. This study evaluated the role of KCa3.1 channels, encoded by Kcnn4, in regulating the gene expression of mouse airway epithelium and the development of asthma traits. We used the ovalbumin (OVA) challenge as an asthma model in wild type and Kcnn4-/- mice, performed histological analysis, and measured serum IgE to evaluate asthma traits. We analyzed gene expression of isolated epithelial cells of trachea or bronchi using mRNA sequencing and gene ontology and performed Ussing chamber experiments in mouse trachea to evaluate anion secretion. Gene expression of epithelial cells from mouse airways differed between trachea and bronchi, indicating regional differences in the inflammatory and transepithelial transport properties of proximal and distal airways. We found that Kcnn4 silencing reduced mast cell numbers, mucus, and collagen in the airways, and reduced the amount of epithelial anion secretion in the OVA-challenged animals. Additionally, gene expression was differentially modified in the trachea and bronchi, with Kcnn4 genetic silencing significantly altering the expression of genes involved in the TNF pathway, supporting the potential of KCa3.1 as a therapeutic target for asthma.
Collapse
Affiliation(s)
- Amber R Philp
- Centro de Estudios Científicos, Valdivia, Los Rios, Chile.,Austral University of Chile, Valdivia, Chile
| | - Fernando Miranda
- Departamento de Fisiología, Austral University of Chile, Valdivia, Chile
| | | | - Agustín Mansilla
- Centro de Estudios Científicos, Valdivia, Los Rios, Chile.,Austral University of Chile, Valdivia, Chile
| | | | | | - Génesis Vega
- Centro de Estudios Científicos, Valdivia, Los Rios, Chile
| | | | - Luis J V Galietta
- TIGEM, Pozzuoli, Italia.,Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - José M Sarmiento
- Departamento de Fisiología, Austral University of Chile, Valdivia, Chile
| | - Carlos A Flores
- Centro de Estudios Científicos, Valdivia, Los Rios, Chile.,Universidad San Sebastián, Valdivia, Chile
| |
Collapse
|
16
|
Blayac M, Coll P, Urbach V, Fanen P, Epaud R, Lanone S. The Impact of Air Pollution on the Course of Cystic Fibrosis: A Review. Front Physiol 2022; 13:908230. [PMID: 35721541 PMCID: PMC9202997 DOI: 10.3389/fphys.2022.908230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) is a lethal and widespread autosomal recessive disorder affecting over 80,000 people worldwide. It is caused by mutations of the CFTR gene, which encodes an epithelial anion channel. CF is characterized by a great phenotypic variability which is currently not fully understood. Although CF is genetically determined, the course of the disease might also depend on multiple other factors. Air pollution, whose effects on health and contribution to respiratory diseases are well established, is one environmental factor suspected to modulate the disease severity and influence the lung phenotype of CF patients. This is of particular interest as pulmonary failure is the primary cause of death in CF. The present review discusses current knowledge on the impact of air pollution on CF pathogenesis and aims to explore the underlying cellular and biological mechanisms involved in these effects.
Collapse
Affiliation(s)
- Marion Blayac
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | - Patrice Coll
- Université Paris Cité and Univ Paris Est Créteil, CNRS, LISA, Paris, France
| | | | - Pascale Fanen
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- AP-HP, Hopital Henri-Mondor, Service Génétique, Creteil, France
| | - Ralph Epaud
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
- Centre Hospitalier Intercommunal, Centre des Maladies Respiratoires Rares (RespiRare®)-CRCM, Creteil, France
| | - Sophie Lanone
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| |
Collapse
|
17
|
Grubb BR, Livraghi-Butrico A. Animal models of cystic fibrosis in the era of highly effective modulator therapies. Curr Opin Pharmacol 2022; 64:102235. [DOI: 10.1016/j.coph.2022.102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/17/2022]
|
18
|
Centorame A, Dumut DC, Youssef M, Ondra M, Kianicka I, Shah J, Paun RA, Ozdian T, Hanrahan JW, Gusev E, Petrof B, Hajduch M, Pislariu R, De Sanctis JB, Radzioch D. Treatment With LAU-7b Complements CFTR Modulator Therapy by Improving Lung Physiology and Normalizing Lipid Imbalance Associated With CF Lung Disease. Front Pharmacol 2022; 13:876842. [PMID: 35668939 PMCID: PMC9163687 DOI: 10.3389/fphar.2022.876842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic fibrosis (CF) is the most common autosomal recessive genetic disease in Caucasians, affecting more than 100,000 individuals worldwide. It is caused by pathogenic variants in the gene encoding CFTR, an anion channel at the plasma membrane of epithelial and other cells. Many CF pathogenic variants disrupt the biosynthesis and trafficking of CFTR or reduce its ion channel function. The most frequent mutation, loss of a phenylalanine at position 508 (F508del), leads to misfolding, retention in the endoplasmic reticulum, and premature degradation of the protein. The therapeutics available for treating CF lung disease include antibiotics, mucolytics, bronchodilators, physiotherapy, and most recently CFTR modulators. To date, no cure for this life shortening disease has been found. Treatment with the Triple combination drug therapy, TRIKAFTA®, is composed of three drugs: Elexacaftor (VX-445), Tezacaftor (VX-661) and Ivacaftor (VX-770). This therapy, benefits persons with CF, improving their weight, lung function, energy levels (as defined by reduced fatigue), and overall quality of life. We examined the effect of combining LAU-7b oral treatment and Triple therapy combination on lung function in a F508deltm1EUR mouse model that displays lung abnormalities relevant to human CF. We assessed lung function, lung histopathology, protein oxidation, lipid oxidation, and fatty acid and lipid profiles in F508deltm1EUR mice.
Collapse
Affiliation(s)
- Amanda Centorame
- Faculty of Medicine, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Daciana Catalina Dumut
- Faculty of Medicine, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Mina Youssef
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Martin Ondra
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University, Olomouc, Czechia
| | | | - Juhi Shah
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Radu Alexandru Paun
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Tomas Ozdian
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - John W. Hanrahan
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Ekaterina Gusev
- Meakins-Christie Laboratories, The Centre for Respiratory Research at McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Basil Petrof
- Meakins-Christie Laboratories, The Centre for Respiratory Research at McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University, Olomouc, Czechia
| | | | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University, Olomouc, Czechia
| | - Danuta Radzioch
- Faculty of Medicine, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Laurent Pharmaceuticals, Montreal, QC, Canada
| |
Collapse
|
19
|
Kelly J, Al-Rammahi M, Daly K, Flanagan PK, Urs A, Cohen MC, di Stefano G, Bijvelds MJC, Sheppard DN, de Jonge HR, Seidler UE, Shirazi-Beechey SP. Alterations of mucosa-attached microbiome and epithelial cell numbers in the cystic fibrosis small intestine with implications for intestinal disease. Sci Rep 2022; 12:6593. [PMID: 35449374 PMCID: PMC9023491 DOI: 10.1038/s41598-022-10328-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Defective CFTR leads to accumulation of dehydrated viscous mucus within the small intestine, luminal acidification and altered intestinal motility, resulting in blockage. These changes promote gut microbial dysbiosis, adversely influencing the normal proliferation and differentiation of intestinal epithelial cells. Using Illumina 16S rRNA gene sequencing and immunohistochemistry, we assessed changes in mucosa-attached microbiome and epithelial cell profile in the small intestine of CF mice and a CF patient compared to wild-type mice and non-CF humans. We found increased abundance of pro-inflammatory Escherichia and depletion of beneficial secondary bile-acid producing bacteria in the ileal mucosa-attached microbiome of CFTR-null mice. The ileal mucosa in a CF patient was dominated by a non-aeruginosa Pseudomonas species and lacked numerous beneficial anti-inflammatory and short-chain fatty acid-producing bacteria. In the ileum of both CF mice and a CF patient, the number of absorptive enterocytes, Paneth and glucagon-like peptide 1 and 2 secreting L-type enteroendocrine cells were decreased, whereas stem and goblet cell numbers were increased. These changes in mucosa-attached microbiome and epithelial cell profile suggest that microbiota-host interactions may contribute to intestinal CF disease development with implications for therapy.
Collapse
Affiliation(s)
- Jennifer Kelly
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Miran Al-Rammahi
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.,Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Al-Qadisiyah, Al Diwaniyah, 58002, Iraq
| | - Kristian Daly
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Paul K Flanagan
- Arrowe Park University Teaching Hospital NHS Trust, Wirral, CH49 5PE, UK.,Gastrointestinal and Liver Services, Aintree University Hospital, Lower Lane, Liverpool, Merseyside, L9 7AL, UK
| | - Arun Urs
- Sheffield Children's Hospital NHS Trust, Western Bank, Sheffield, S10 2TH, UK
| | - Marta C Cohen
- Histopathology Department, Sheffield Children's Hospital NHS Trust, Western Bank, Sheffield, S10 2TH, UK
| | - Gabriella di Stefano
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625, Hannover, Germany
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Ursula E Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625, Hannover, Germany
| | - Soraya P Shirazi-Beechey
- Department of Infection Biology and Microbiomes, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| |
Collapse
|
20
|
da Cunha MF, Pranke I, Sassi A, Schreiweis C, Moriceau S, Vidovic D, Hatton A, Carlon MS, Creste G, Berhal F, Prestat G, Freund R, Odolczyk N, Jais JP, Gravier-Pelletier C, Zielenkiewicz P, Jullien V, Hinzpeter A, Oury F, Edelman A, Sermet-Gaudelus I. Systemic bis-phosphinic acid derivative restores chloride transport in Cystic Fibrosis mice. Sci Rep 2022; 12:6132. [PMID: 35413967 PMCID: PMC9005718 DOI: 10.1038/s41598-022-09678-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/28/2022] [Indexed: 11/17/2022] Open
Abstract
Mutations in the Cystic Fibrosis Transmembrane Conductance Regulator gene (CFTR) are responsible for Cystic Fibrosis (CF). The most common CF-causing mutation is the deletion of the 508th amino-acid of CFTR (F508del), leading to dysregulation of the epithelial fluid transport in the airway’s epithelium and the production of a thickened mucus favoring chronic bacterial colonization, sustained inflammation and ultimately respiratory failure. c407 is a bis-phosphinic acid derivative which corrects CFTR dysfunction in epithelial cells carrying the F508del mutation. This study aimed to investigate c407 in vivo activity in the F508del Cftrtm1Eur murine model of CF. Using nasal potential difference measurement, we showed that in vivo administration of c407 by topical, short-term intraperitoneal and long-term subcutaneous route significantly increased the CFTR dependent chloride (Cl−) conductance in F508del Cftrtm1Eur mice. This functional improvement was correlated with a relocalization of F508del-cftr to the apical membrane in nasal epithelial cells. Importantly, c407 long-term administration was well tolerated and in vitro ADME toxicologic studies did not evidence any obvious issue. Our data provide the first in vivo preclinical evidence of c407 efficacy and absence of toxicity after systemic administration for the treatment of Cystic Fibrosis.
Collapse
Affiliation(s)
| | - Iwona Pranke
- INSERM U1151, équipe 11, Paris, France.,Université de Paris, Paris, France
| | - Ali Sassi
- INSERM U1151, équipe 11, Paris, France.,Université de Paris, Paris, France
| | | | | | - Dragana Vidovic
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Aurélie Hatton
- INSERM U1151, équipe 11, Paris, France.,Université de Paris, Paris, France
| | - Mariane Sylvia Carlon
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Geordie Creste
- Université de Paris, Paris, France.,UMR 8601, CNRS, LCBPT, Paris, France
| | - Farouk Berhal
- Université de Paris, Paris, France.,UMR 8601, CNRS, LCBPT, Paris, France
| | - Guillaume Prestat
- Université de Paris, Paris, France.,UMR 8601, CNRS, LCBPT, Paris, France
| | - Romain Freund
- Université de Paris, Paris, France.,Unité de Biostatistiques, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Norbert Odolczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.,Laboratory of Systems Biology, Institute of Experimental Plant Biology and Biotechnology, Warsaw, Poland
| | - Jean Philippe Jais
- Unité de Biostatistiques, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Piotr Zielenkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.,Laboratory of Systems Biology, Institute of Experimental Plant Biology and Biotechnology, Warsaw, Poland
| | - Vincent Jullien
- Laboratoire de Bactériologie-Virologie-Hygiène, Hôpital Avicenne, Bobigny, France
| | | | - Franck Oury
- Université de Paris, Paris, France.,INSERM U1151, équipe 8, Paris, France
| | - Aleksander Edelman
- INSERM U1151, équipe 11, Paris, France.,Université de Paris, Paris, France
| | - Isabelle Sermet-Gaudelus
- INSERM U1151, équipe 11, Paris, France. .,Université de Paris, Paris, France. .,Centre de Référence et de Compétence Maladies Rares, Mucoviscidose et maladies apparentées, Hôpital Necker Enfants malades, Paris, France. .,European Reference Network for Rare Respiratory Diseases (ERN-LUNG), Brussels, Belgium. .,Institut Necker Enfants Malades, 160 rue de Vaugirard, 75015, Paris, France.
| |
Collapse
|
21
|
Martinovich KM, Kicic A, Stick SM, Johnsen RD, Fletcher S, Wilton SD. Investigating the Implications of CFTR Exon Skipping Using a Cftr Exon 9 Deleted Mouse Model. Front Pharmacol 2022; 13:868863. [PMID: 35392567 PMCID: PMC8981082 DOI: 10.3389/fphar.2022.868863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Severity and disease progression in people with Cystic Fibrosis (CF) is typically dependent on their genotype. One potential therapeutic strategy for people with specific mutations is exon skipping with antisense oligonucleotides (AO). CFTR exon 9 is an in-frame exon and hence the exclusion of this exon would excise only 31 amino acids but not alter the reading frame of the remaining mRNA. Splice mutations 1209 + 1 G > C and 1209 + 2 T > G were documented to cause CFTR exon 9 skipping and these variants were reported to manifest as a milder CF disease, therefore exon 9 skipping could be beneficial for people with class I mutations that affect exon 9 such as p.Trp401X. While the impact of exon 9 skipping on gene expression and cellular pathways can be studied in cells in vitro, trace amount of full-length normal or mutated material could confound the evaluation. To overcome this limitation, the impact of CFTR exon 9 skipping on disease phenotype and severity is more effectively evaluated in a small animal model. It was hypothesised that antisense oligonucleotide-mediated skipping this particular exon could result in a "mild mouse CF phenotype". Methods: Cftr exon 9 deleted mice were generated using homologous recombination. Survival of homozygous (Cftr Δ9/Δ9 ) and heterozygous (Cftr Δ9/+ ) mice was compared to that of other CF mouse models, and lung and intestinal organ histology examined for any pathologies. Primary airway epithelial cells (pAECs) were harvested from Cftr Δ9/Δ9 mice and cultured at the Air Liquid Interface for CFTR functional assessment using Ussing Chamber analysis. Results: A Cftr Δ9/Δ9 mouse model presented with intestinal obstructions, and at time of weaning (21 days). Cftr Δ9/Δ9 mice had a survival rate of 83% that dropped to 38% by day 50. Histological sections of the small intestine from Cftr Δ9/Δ9 mice showed more goblet cells and mucus accumulation than samples from the Cftr Δ9/+ littermates. Airway epithelial cell cultures established from Cftr Δ9/Δ9 mice were not responsive to forskolin stimulation. Summary: The effect of Cftr exon 9 deletion on Cftr function was assessed and it was determined that the encoded Cftr isoform did not result in a milder "mouse CF disease phenotype," suggesting that Cftr exon 9 is not dispensable, although further investigation in human CF pAECs would be required to confirm this observation.
Collapse
Affiliation(s)
- Kelly M Martinovich
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Anthony Kicic
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Childrens Hospital, Nedlands, WA, Australia.,School of Population Health, Curtin University, Bentley, WA, Australia
| | - Stephen M Stick
- School of Medicine, The University of Western Australia, Perth, WA, Australia.,Telethon Kids Institute, Wal-yan Respiratory Research Centre, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Childrens Hospital, Nedlands, WA, Australia
| | - Russell D Johnsen
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia.,PYC Therapeutics, Perth, WA, Australia
| | - Steve D Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Perron Institute for Neurological and Translational Sciences, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
22
|
Immunoglobulin A Mucosal Immunity and Altered Respiratory Epithelium in Cystic Fibrosis. Cells 2021; 10:cells10123603. [PMID: 34944110 PMCID: PMC8700636 DOI: 10.3390/cells10123603] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022] Open
Abstract
The respiratory epithelium represents the first chemical, immune, and physical barrier against inhaled noxious materials, particularly pathogens in cystic fibrosis. Local mucus thickening, altered mucociliary clearance, and reduced pH due to CFTR protein dysfunction favor bacterial overgrowth and excessive inflammation. We aimed in this review to summarize respiratory mucosal alterations within the epithelium and current knowledge on local immunity linked to immunoglobulin A in patients with cystic fibrosis.
Collapse
|
23
|
Methodological tools to study species of the genus Burkholderia. Appl Microbiol Biotechnol 2021; 105:9019-9034. [PMID: 34755214 PMCID: PMC8578011 DOI: 10.1007/s00253-021-11667-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/26/2022]
Abstract
Bacteria belonging to the Burkholderia genus are extremely versatile and diverse. They can be environmental isolates, opportunistic pathogens in cystic fibrosis, immunocompromised or chronic granulomatous disease patients, or cause disease in healthy people (e.g., Burkholderia pseudomallei) or animals (as in the case of Burkholderia mallei). Since the genus was separated from the Pseudomonas one in the 1990s, the methodological tools to study and characterize these bacteria are evolving fast. Here we reviewed the techniques used in the last few years to update the taxonomy of the genus, to study gene functions and regulations, to deepen the knowledge on the drug resistance which characterizes these bacteria, and to elucidate their mechanisms to establish infections. The availability of these tools significantly impacts the quality of research on Burkholderia and the choice of the most appropriated is fundamental for a precise characterization of the species of interest. Key points • Updated techniques to study the genus Burkholderia were reviewed. • Taxonomy, genomics, assays, and animal models were described. • A comprehensive overview on recent advances in Burkholderia studies was made.
Collapse
|
24
|
Collin AM, Lecocq M, Detry B, Carlier FM, Bouzin C, de Sany P, Hoton D, Verleden S, Froidure A, Pilette C, Gohy S. Loss of ciliated cells and altered airway epithelial integrity in cystic fibrosis. J Cyst Fibros 2021; 20:e129-e139. [PMID: 34657818 DOI: 10.1016/j.jcf.2021.09.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 06/28/2021] [Accepted: 09/15/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND In cystic fibrosis, the respiratory epithelium is the target tissue of both the genetic abnormality of the disease and of external aggressions, notably by pathogens (Pseudomonas aeruginosa). A detailed characterisation of the cystic fibrosis bronchial epithelium is however lacking, as most previous studies focused on the nasal epithelium or on cell lines. This study aimed to characterise the abnormal phenotype and epithelial-to-mesenchymal transition in cystic fibrosis bronchial epithelium and to evaluate in cell cultures whether abnormalities persist ex vivo. METHODS Explant lung tissues (n = 44) were assessed for bronchial epithelial cell phenotyping by immunostaining. Human bronchial epithelial cells were derived from basal cells isolated from cystic fibrosis patients or control donors and cultured in air-liquid interface for 2, 4 or 6 weeks. RESULTS Enhanced mucin 5AC and decreased β-tubulin expression were observed in cystic fibrosis airways reflecting a decreased ciliated/goblet cell ratio, associated with increased number of vimentin-positive cells, indicating epithelial-to-mesenchymal transition process. These features were recapitulated in vitro, in cystic fibrosis-derived reconstituted epithelium. However, they were not induced by CFTR inhibition or Pseudomonas infection, and most abnormalities tended to disappear in long-term culture (6 weeks) except for increased fibronectin release, an epithelial-to-mesenchymal transition marker. CONCLUSIONS This study provides new insights into airway epithelial changes in cystic fibrosis, which are imprinted through an acquired mechanism that we could not relate to CFTR function.
Collapse
Affiliation(s)
- Amandine M Collin
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marylène Lecocq
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bruno Detry
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - François M Carlier
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Philippe de Sany
- Pole of Microbiology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Delphine Hoton
- Department of Pathology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Stijn Verleden
- Lung Transplant Unit, Division of Respiratory Disease, Department of chronic disease, metabolism and aging, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Antoine Froidure
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Pneumology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Pneumology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Sophie Gohy
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental & Clinical Research, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Pneumology, Cliniques universitaires Saint-Luc, Brussels, Belgium; Centre de référence pour la mucoviscidose, Cliniques universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
25
|
Assessing the impact of exposome on the course of chronic obstructive pulmonary disease and cystc fibrosis: The REMEDIA European Project Approach. Environ Epidemiol 2021; 5:e165. [PMID: 34414348 PMCID: PMC8367060 DOI: 10.1097/ee9.0000000000000165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 06/28/2021] [Indexed: 11/26/2022] Open
Abstract
Because of the direct interaction of lungs with the environment, respiratory diseases are among the leading causes of environment-related deaths in the world. Chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF) are two highly debilitating diseases that are of particular interest in the context of environmental studies; they both are characterized by a similar progressive loss of lung function with small bronchi alterations, and a high phenotypic variability of unknown origin, which prevents a good therapeutic efficacy. In the last years, there has been an evolution in the apprehension of the study of diseases going from a restricted “one exposure, one disease” approach to a broader concept with other associating factors, the exposome. The overall objective of the REMEDIA project is to extend the understanding of the contribution of the exposome to COPD and CF diseases. To achieve our aim, we will (1) exploit data from existing cohorts and population registries to create a unified global database gathering phenotype and exposome information; (2) develop a flexible individual sensor device combining environmental and biomarker toolkits; (3) use a versatile atmospheric simulation chamber to simulate the health effects of complex exposomes; (4) use machine learning supervised analyses and causal inference models to identify relevant risk factors; and (5) develop econometric and cost-effectiveness models to assess the costs, performance, and cost-effectiveness of a selection of prevention strategies. The results will be used to develop guidelines to better predict disease risks and constitute the elements of the REMEDIA toolbox. The multidisciplinary approach carried out by the REMEDIA European project should represent a major breakthrough in reducing the morbidity and mortality associated with COPD and CF diseases.
Collapse
|
26
|
Bonfield TL, Sutton MT, Fletcher DR, Folz MA, Ragavapuram V, Somoza RA, Caplan AI. Donor-defined mesenchymal stem cell antimicrobial potency against nontuberculous mycobacterium. Stem Cells Transl Med 2021; 10:1202-1216. [PMID: 33943038 PMCID: PMC8284776 DOI: 10.1002/sctm.20-0521] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/02/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic nontuberculous mycobacterial infections with Mycobacterium avium and Mycobacterium intracellulare complicate bronchiectasis, chronic obstructive airway disease, and the health of aging individuals. These insidious intracellular pathogens cause considerable morbidity and eventual mortality in individuals colonized with these bacteria. Current treatment regimens with antibiotic macrolides are both toxic and often inefficient at providing infection resolution. In this article, we demonstrate that human marrow‐derived mesenchymal stem cells are antimicrobial and anti‐inflammatory in vitro and in the context of an in vivo sustained infection of either M. avium and/or M. intracellulare.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Morgan T Sutton
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA.,St. Jude Children's Research Hospital Graduate School of Biomedical Sciences, Memphis, Tennessee, USA
| | - David R Fletcher
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael A Folz
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Vaishnavi Ragavapuram
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Rodrigo A Somoza
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Arnold I Caplan
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
27
|
Ensinck M, Mottais A, Detry C, Leal T, Carlon MS. On the Corner of Models and Cure: Gene Editing in Cystic Fibrosis. Front Pharmacol 2021; 12:662110. [PMID: 33986686 PMCID: PMC8111007 DOI: 10.3389/fphar.2021.662110] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is a severe genetic disease for which curative treatment is still lacking. Next generation biotechnologies and more efficient cell-based and in vivo disease models are accelerating the development of novel therapies for CF. Gene editing tools, like CRISPR-based systems, can be used to make targeted modifications in the genome, allowing to correct mutations directly in the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) gene. Alternatively, with these tools more relevant disease models can be generated, which in turn will be invaluable to evaluate novel gene editing-based therapies for CF. This critical review offers a comprehensive description of currently available tools for genome editing, and the cell and animal models which are available to evaluate them. Next, we will give an extensive overview of proof-of-concept applications of gene editing in the field of CF. Finally, we will touch upon the challenges that need to be addressed before these proof-of-concept studies can be translated towards a therapy for people with CF.
Collapse
Affiliation(s)
- Marjolein Ensinck
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Angélique Mottais
- Institut de Recherche Expérimentale et Clinique, Louvain Centre for Toxicology and Applied Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Claire Detry
- Institut de Recherche Expérimentale et Clinique, Louvain Centre for Toxicology and Applied Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Teresinha Leal
- Institut de Recherche Expérimentale et Clinique, Louvain Centre for Toxicology and Applied Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Marianne S. Carlon
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Lopes-Pacheco M, Pedemonte N, Veit G. Discovery of CFTR modulators for the treatment of cystic fibrosis. Expert Opin Drug Discov 2021; 16:897-913. [PMID: 33823716 DOI: 10.1080/17460441.2021.1912732] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a life-threatening inherited disease caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR) protein, an anion channel expressed at the apical membrane of secretory epithelia. CF leads to multiorgan dysfunction with progressive deterioration of lung function being the major cause of untimely death. Conventional CF therapies target only symptoms and consequences downstream of the primary genetic defect and the current life expectancy and quality of life of these individuals are still very limited. AREA COVERED CFTR modulator drugs are novel-specialized therapies that enhance or even restore functional expression of CFTR mutants and have been approved for clinical use for individuals with specific CF genotypes. This review summarizes classical approaches used for the pre-clinical development of CFTR correctors and potentiators as well as emerging strategies aiming to accelerate modulator development and expand theratyping efforts. EXPERT OPINION Highly effective CFTR modulator drugs are expected to deeply modify the disease course for the majority of individuals with CF. A multitude of experimental approaches have been established to accelerate the development of novel modulators. CF patient-derived specimens are valuable cell models to predict therapeutic effectiveness of existing (and novel) modulators in a precision medicine approach.
Collapse
Affiliation(s)
| | | | - Guido Veit
- Department of Physiology, McGill University, Montréal, Canada
| |
Collapse
|
29
|
Leenaars CH, Vries RBD, Reijmer J, Holthaus D, Visser D, Heming A, Elzinga J, Kempkes RW, Beumer W, Punt C, Meijboom FL, Ritskes-Hoitinga M. Animal models for cystic fibrosis: a systematic search and mapping review of the literature. Part 2: nongenetic models. Lab Anim 2021; 55:307-316. [PMID: 33557683 DOI: 10.1177/0023677221990688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Various animal models are available to study cystic fibrosis (CF). These models may help to enhance our understanding of the pathology and contribute to the development of new treatments. We systematically searched all publications on CF animal models. Because of the large number of models retrieved, we split this mapping review into two parts. Previously, we presented the genetic CF animal models. In this paper we present the nongenetic CF animal models. While genetic animal models may, in theory, be preferable for genetic diseases, the phenotype of a genetic model does not automatically resemble human disease. Depending on the research question, other animal models may thus be more informative.We searched Pubmed and Embase and identified 12,303 unique publications (after duplicate removal). All references were screened for inclusion by two independent reviewers. The genetic animal models for CF (from 636 publications) were previously described. The non-genetic CF models (from 189 publications) are described in this paper, grouped by model type: infection-based, pharmacological, administration of human materials, xenografts and other. As before for the genetic models, an overview of basic model characteristics and outcome measures is provided. This CF animal model overview can be the basis for an objective, evidence-based model choice for specific research questions. Besides, it can help to retrieve relevant background literature on outcome measures of interest.
Collapse
Affiliation(s)
- Cathalijn Hc Leenaars
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands.,Faculty of Veterinary Medicine, Department of Animals in Science and Society, Utrecht University, The Netherlands.,Institute for Laboratory Animal Science, Hannover Medical School, Germany
| | - Rob Bm de Vries
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands
| | - Joey Reijmer
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands
| | - David Holthaus
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands
| | - Damian Visser
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands
| | - Anna Heming
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands
| | - Janneke Elzinga
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands
| | - Rosalie Wm Kempkes
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands
| | | | - Carine Punt
- ProQR Therapeutics NV,Leiden, the Netherlands; Present position: BunyaVax BV, Lelystad, The Netherlands
| | - Franck Lb Meijboom
- Faculty of Veterinary Medicine, Department of Animals in Science and Society, Utrecht University, The Netherlands
| | - Merel Ritskes-Hoitinga
- SYRCLE, Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, The Netherlands.,Department of Clinical Medicine, Aarhus University, Denmark
| |
Collapse
|
30
|
Tan Q, di Stefano G, Tan X, Renjie X, Römermann D, Talbot SR, Seidler UE. Inhibition of Na + /H + exchanger isoform 3 improves gut fluidity and alkalinity in cystic fibrosis transmembrane conductance regulator-deficient and F508del mutant mice. Br J Pharmacol 2021; 178:1018-1036. [PMID: 33179259 DOI: 10.1111/bph.15323] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Constipation and intestinal obstructive episodes are major health problems in cystic fibrosis (CF) patients. Three FDA-approved drugs against constipation-prone irritable bowel syndrome were tested for their ability to increase luminal fluidity and alkalinity in cystic fibrosis transmembrane conductance regulator (CFTR) null (cftr-/- ) and F508del mutant (F508delmut/mut ) murine intestine. EXPERIMENTAL APPROACH Guanylate cyclase C agonist linaclotide, PGE1 analogue lubiprostone and intestine-specific NHE3 inhibitor tenapanor were perfused through a ~3 cm jejunal, proximal or mid-distal colonic segment in anaesthetized cftr-/- , F508delmut/mut and WT mice. Net fluid balance was determined gravimetrically and alkaline output by pH-stat back titration. KEY RESULTS Basal jejunal fluid absorptive rates were significantly higher and basal HCO3 - output was significantly lower in cftr-/- and F508delmut/mut compared to WT mice. In cftr-/- and F508delmut/mut mice, all three drugs significantly inhibited the fluid absorptive rate and increased alkaline output in the jejunum and tenapanor and lubiprostone, but not linaclotide, in the colon. After tenapanor pre-incubation, linaclotide elicited a robust fluid secretory response in WT jejunum, while no further change in absorptive rates was observed in cftr-/- and F508delmut/mut jejunum, suggesting that the increase in gut fluidity and alkalinity by linaclotide in CF gut is mediated via NHE3 inhibition. Lubiprostone also inhibited fluid absorption in cftr-/- and F508delmut/mut jejunum via NHE3 inhibition but had a residual NHE3-independent effect. CONCLUSION AND IMPLICATIONS Linaclotide, lubiprostone and tenapanor reduced fluid absorption and increased alkaline output in the CF gut. Their application may ameliorate constipation and reduce obstructive episodes in CF patients.
Collapse
Affiliation(s)
- Qinghai Tan
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | | | - Xinjie Tan
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | - Xiu Renjie
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | - Dorothee Römermann
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| | - Steven R Talbot
- Institute of Veterinary Research, Hannover Medical School, Hanover, Germany
| | - Ursula E Seidler
- Department of Gastroenterology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
31
|
Panfoli I, Granata S, Candiano G, Verlato A, Lombardi G, Bruschi M, Zaza G. Analysis of urinary exosomes applications for rare kidney disorders. Expert Rev Proteomics 2021; 17:735-749. [PMID: 33395324 DOI: 10.1080/14789450.2020.1866993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Exosomes are nanovesicles that play important functions in a variety of physiological and pathological conditions. They are powerful cell-to-cell communication tool thanks to the protein, mRNA, miRNA, and lipid cargoes they carry. They are also emerging as valuable diagnostic and prognostic biomarker sources. Urinary exosomes carry information from all the cells of the urinary tract, downstream of the podocyte. Rare kidney diseases are a subset of an inherited diseases whose genetic diagnosis can be unclear, and presentation can vary due to genetic, epigenetic, and environmental factors. Areas covered: In this review, we focus on a group of rare and often neglected kidney diseases, for which we have sufficient available literature data on urinary exosomes. The analysis of their content can help to comprehend pathological mechanisms and to identify biomarkers for diagnosis, prognosis, and therapeutic targets. Expert opinion: The foreseeable large-scale application of system biology approach to the profiling of exosomal proteins as a source of renal disease biomarkers will be also useful to stratify patients with rare kidney diseases whose penetrance, phenotypic presentation, and age of onset vary sensibly. This can ameliorate the clinical management.
Collapse
Affiliation(s)
- Isabella Panfoli
- Department of Pharmacy-DIFAR, University of Genoa , Genoa, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Gianmarco Lombardi
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| |
Collapse
|
32
|
Sharma J, Abbott J, Klaskala L, Zhao G, Birket SE, Rowe SM. A Novel G542X CFTR Rat Model of Cystic Fibrosis Is Sensitive to Nonsense Mediated Decay. Front Physiol 2020; 11:611294. [PMID: 33391025 PMCID: PMC7772197 DOI: 10.3389/fphys.2020.611294] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
Nonsense mutations that lead to the insertion of a premature termination codon (PTC) in the cystic fibrosis transmembrane conductance regulator (CFTR) transcript affect 11% of patients with cystic fibrosis (CF) worldwide and are associated with severe disease phenotype. While CF rat models have contributed significantly to our understanding of CF disease pathogenesis, there are currently no rat models available for studying CF nonsense mutations. Here we created and characterized the first homozygous CF rat model that bears the CFTR G542X nonsense mutation in the endogenous locus using CRISPR/Cas9 gene editing. In addition to displaying severe CF manifestations and developmental defects such as reduced growth, abnormal tooth enamel, and intestinal obstruction, CFTR G542X knockin rats demonstrated an absence of CFTR function in tracheal and intestinal sections as assessed by nasal potential difference and transepithelial short-circuit current measurements. Reduced CFTR mRNA levels in the model further suggested sensitivity to nonsense-mediated decay, a pathway elicited by the presence of PTCs that degrades the PTC-bearing transcripts and thus further diminishes the level of CFTR protein. Although functional restoration of CFTR was observed in G542X rat tracheal epithelial cells in response to single readthrough agent therapy, therapeutic efficacy was not observed in G542X knockin rats in vivo. The G542X rat model provides an invaluable tool for the identification and in vivo validation of potential therapies for CFTR nonsense mutations.
Collapse
Affiliation(s)
- Jyoti Sharma
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joseph Abbott
- Horizon Discovery Group, PLC, St. Louis, MO, United States
| | | | - Guojun Zhao
- Horizon Discovery Group, PLC, St. Louis, MO, United States
| | - Susan E. Birket
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Steven M. Rowe
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
33
|
McCarron A, Parsons D, Donnelley M. Animal and Cell Culture Models for Cystic Fibrosis: Which Model Is Right for Your Application? THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:228-242. [PMID: 33232694 DOI: 10.1016/j.ajpath.2020.10.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/01/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Over the past 30 years, a range of cystic fibrosis (CF) animal models have been generated for research purposes. Different species, including mice, rats, ferrets, rabbits, pigs, sheep, zebrafish, and fruit flies, have all been used to model CF disease. While access to such a variety of animal models is a luxury for any research field, it also complicates the decision-making process when it comes to selecting the right model for an investigation. The purpose of this review is to provide a guide for selecting the most appropriate CF animal model for any given application. In this review, the characteristics and phenotypes of each animal model are described, along with a discussion of the key considerations that must be taken into account when choosing a suitable animal model. Available in vitro systems of CF are also described and can offer a useful alternative to using animal models. Finally, the future of CF animal model generation and its use in research are speculated upon.
Collapse
Affiliation(s)
- Alexandra McCarron
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia.
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| |
Collapse
|
34
|
Lung immunoglobulin A immunity dysregulation in cystic fibrosis. EBioMedicine 2020; 60:102974. [PMID: 32927272 PMCID: PMC7495088 DOI: 10.1016/j.ebiom.2020.102974] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In cystic fibrosis (CF), recurrent infections suggest impaired mucosal immunity but whether production of secretory immunoglobulin A (S-IgA) is impaired remains elusive. S-IgA is generated following polymeric immunoglobulin receptor (pIgR)-mediated transepithelial transport of dimeric (d-)IgA and represents a major defence through neutralisation of inhaled pathogens like Pseudomonas aeruginosa (Pa). METHODS Human lung tissue (n = 74), human sputum (n = 118), primary human bronchial epithelial cells (HBEC) (cultured in air-liquid interface) (n = 19) and mouse lung tissue and bronchoalveolar lavage were studied for pIgR expression, IgA secretion and regulation. FINDINGS Increased epithelial pIgR immunostaining was observed in CF lung explants, associated with more IgA-producing plasma cells, sputum and serum IgA, especially Pa-specific IgA. In contrast, pIgR and IgA transport were downregulated in F508del mice, CFTR-inhibited HBEC, and CF HBEC. Moreover, the unfolded protein response (UPR) due to F508del mutation, inhibited IgA transport in Calu-3 cells. Conversely, pIgR expression and IgA secretion were strongly upregulated following Pa lung infection in control and F508del mice, through an inflammatory host response involving interleukin-17. INTERPRETATION A complex regulation of IgA secretion occurs in the CF lung, UPR induced by CFTR mutation/dysfunction inhibiting d-IgA transcytosis, and Pa infection unexpectedly unleashing this secretory defence mechanism. FUNDING This work was supported by the Forton's grant of the King Baudouin's Foundation, Belgium, the Fondazione Ricerca Fibrosi Cistica, Italy, and the Fonds National de la Recherche Scientifique, Belgium.
Collapse
|
35
|
Transport properties in CFTR-/- knockout piglets suggest normal airway surface liquid pH and enhanced amiloride-sensitive Na + absorption. Pflugers Arch 2020; 472:1507-1519. [PMID: 32712714 PMCID: PMC7476968 DOI: 10.1007/s00424-020-02440-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/12/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
Previous analysis of CFTR-knockout (CFTR-/-) in piglets has provided important insights into the pathology of cystic fibrosis. However, controversies exist as to the true contribution of CFTR to the pH balance in airways and intestine. We therefore compared ion transport properties in newborn wild-type (CFTR+/+) and CFTR-knockout (CFTR-/- piglets). Tracheas of CFTR-/- piglets demonstrated typical cartilage malformations and muscle cell bundles. CFTR-/- airway epithelial cells showed enhanced lipid peroxidation, suggesting inflammation early in life. CFTR was mainly expressed in airway submucosal glands and was absent in lungs of CFTR-/- piglets, while expression of TMEM16A was uncompromised. mRNA levels for TMEM16A, TMEM16F, and αβγENaC were unchanged in CFTR-/- airways, while mRNA for SLC26A9 appeared reduced. CFTR was undetectable in epithelial cells of CFTR-/- airways and intestine. Small intestinal epithelium of CFTR-/- piglets showed mucus accumulation. Secretion of both electrolytes and mucus was activated by stimulation with prostaglandin E2 and ATP in the intestine of CFTR+/+, but not of CFTR-/- animals. pH was measured inside small bronchi using a pH microelectrode and revealed no difference between CFTR+/+ and CFTR-/- piglets. Intracellular pH in porcine airway epithelial cells revealed only a small contribution of CFTR to bicarbonate secretion, which was absent in cells from CFTR-/- piglets. In contrast to earlier reports, our data suggest a minor impact of CFTR on ASL pH. In contrast, enhanced amiloride-sensitive Na+ absorption may contribute to lung pathology in CFTR-/- piglets, along with a compromised CFTR- and TMEM16A-dependent Cl- transport.
Collapse
|
36
|
Oliver KE, Rauscher R, Mijnders M, Wang W, Wolpert MJ, Maya J, Sabusap CM, Kesterson RA, Kirk KL, Rab A, Braakman I, Hong JS, Hartman JL, Ignatova Z, Sorscher EJ. Slowing ribosome velocity restores folding and function of mutant CFTR. J Clin Invest 2020; 129:5236-5253. [PMID: 31657788 DOI: 10.1172/jci124282] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/28/2019] [Indexed: 12/19/2022] Open
Abstract
Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR), with approximately 90% of patients harboring at least one copy of the disease-associated variant F508del. We utilized a yeast phenomic system to identify genetic modifiers of F508del-CFTR biogenesis, from which ribosomal protein L12 (RPL12/uL11) emerged as a molecular target. In the present study, we investigated mechanism(s) by which suppression of RPL12 rescues F508del protein synthesis and activity. Using ribosome profiling, we found that rates of translation initiation and elongation were markedly slowed by RPL12 silencing. However, proteolytic stability and patch-clamp assays revealed RPL12 depletion significantly increased F508del-CFTR steady-state expression, interdomain assembly, and baseline open-channel probability. We next evaluated whether Rpl12-corrected F508del-CFTR could be further enhanced with concomitant pharmacologic repair (e.g., using clinically approved modulators lumacaftor and tezacaftor) and demonstrated additivity of these treatments. Rpl12 knockdown also partially restored maturation of specific CFTR variants in addition to F508del, and WT Cftr biogenesis was enhanced in the pancreas, colon, and ileum of Rpl12 haplosufficient mice. Modulation of ribosome velocity therefore represents a robust method for understanding both CF pathogenesis and therapeutic response.
Collapse
Affiliation(s)
| | - Robert Rauscher
- Institute for Biochemistry & Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Marjolein Mijnders
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Wei Wang
- Gregory Fleming James Cystic Fibrosis Research Center and
| | | | - Jessica Maya
- Gregory Fleming James Cystic Fibrosis Research Center and
| | | | - Robert A Kesterson
- Department of Genetics, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Kevin L Kirk
- Gregory Fleming James Cystic Fibrosis Research Center and
| | - Andras Rab
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ineke Braakman
- Cellular Protein Chemistry, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Jeong S Hong
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - John L Hartman
- Gregory Fleming James Cystic Fibrosis Research Center and.,Department of Genetics, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Zoya Ignatova
- Institute for Biochemistry & Molecular Biology, University of Hamburg, Hamburg, Germany
| | | |
Collapse
|
37
|
Tang Y, Yan Z, Engelhardt JF. Viral Vectors, Animal Models, and Cellular Targets for Gene Therapy of Cystic Fibrosis Lung Disease. Hum Gene Ther 2020; 31:524-537. [PMID: 32138545 PMCID: PMC7232698 DOI: 10.1089/hum.2020.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
After more than two decades since clinical trials tested the first use of recombinant adeno-associated virus (rAAV) to treat cystic fibrosis (CF) lung disease, gene therapy for this disorder has undergone a tremendous resurgence. Fueling this enthusiasm has been an enhanced understanding of rAAV transduction biology and cellular processes that limit transduction of airway epithelia, the development of new rAAV serotypes and other vector systems with high-level tropism for airway epithelial cells, an improved understanding of CF lung pathogenesis and the cellular targets for gene therapy, and the development of new animal models that reproduce the human CF disease phenotype. These advances have created a preclinical path for both assessing the efficacy of gene therapies in the CF lung and interrogating the target cell types in the lung required for complementation of the CF disease state. Lessons learned from early gene therapy attempts with rAAV in the CF lung have guided thinking for the testing of next-generation vector systems. Although unknown questions still remain regarding the cellular targets in the lung that are required or sufficient to complement CF lung disease, the field is now well positioned to tackle these challenges. This review will highlight the role that next-generation CF animal models are playing in the preclinical development of gene therapies for CF lung disease and the knowledge gaps in disease pathophysiology that these models are attempting to fill.
Collapse
Affiliation(s)
- Yinghua Tang
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
38
|
Age-Dependent Progression in Lung Pathophysiology can be Prevented by Restoring Fatty Acid and Ceramide Imbalance in Cystic Fibrosis. Lung 2020; 198:459-469. [DOI: 10.1007/s00408-020-00353-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/02/2020] [Indexed: 11/29/2022]
|
39
|
McCarron A, Cmielewski P, Reyne N, McIntyre C, Finnie J, Craig F, Rout-Pitt N, Delhove J, Schjenken JE, Chan HY, Boog B, Knight E, Gilmore RC, O'Neal WK, Boucher RC, Parsons D, Donnelley M. Phenotypic Characterization and Comparison of Cystic Fibrosis Rat Models Generated Using CRISPR/Cas9 Gene Editing. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:977-993. [PMID: 32084371 DOI: 10.1016/j.ajpath.2020.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/20/2019] [Accepted: 01/21/2020] [Indexed: 01/10/2023]
Abstract
Animal models of cystic fibrosis (CF) are essential for investigating disease mechanisms and trialing potential therapeutics. This study generated two CF rat models using clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated protein 9 gene editing. One rat model carries the common human Phe508del (ΔF508) CF transmembrane conductance regulator (CFTR) mutation, whereas the second is a CFTR knockout model. Phenotype was characterized using a range of functional and histologic assessments, including nasal potential difference to measure electrophysiological function in the upper airways, RNAscope in situ hybridization and quantitative PCR to assess CFTR mRNA expression in the lungs, immunohistochemistry to localize CFTR protein in the airways, and histopathologic assessments in a range of tissues. Both rat models revealed a range of CF manifestations, including reduced survival, intestinal obstruction, bioelectric defects in the nasal epithelium, histopathologic changes in the trachea, large intestine, and pancreas, and abnormalities in the development of the male reproductive tract. The CF rat models presented herein will prove useful for longitudinal assessments of pathophysiology and therapeutics.
Collapse
Affiliation(s)
- Alexandra McCarron
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.
| | - Patricia Cmielewski
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicole Reyne
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Chantelle McIntyre
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - John Finnie
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Division of Anatomical Pathology, SA Pathology, Adelaide, South Australia, Australia
| | - Fiona Craig
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Nathan Rout-Pitt
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Juliette Delhove
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - John E Schjenken
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Hon Y Chan
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Bernadette Boog
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Emma Knight
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Rodney C Gilmore
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Wanda K O'Neal
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Richard C Boucher
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David Parsons
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Martin Donnelley
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
40
|
Hepatobiliary Involvement in Cystic Fibrosis. Respir Med 2020. [DOI: 10.1007/978-3-030-42382-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
41
|
Strong toll-like receptor responses in cystic fibrosis patients are associated with higher lung function. J Cyst Fibros 2019; 19:608-613. [PMID: 31813753 DOI: 10.1016/j.jcf.2019.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) airways disease varies widely among patients with identical cystic fibrosis transmembrane conductance regulator (CFTR) genotypes. Robust airway inflammation is thought to be deleterious in CF; inter-individual variation in Toll-like receptor (TLR)-mediated innate immune inflammatory responses (TMIIR) might account for a portion of the phenotypic variation. We tested if TMIIR in people with CF are different than those of healthy controls, and whether higher TMIIR in people with CF are associated with reduced lung function. METHODS We cultured whole blood from clinically stable subjects with CF (n = 76) and healthy controls (n = 45) with TLR agonists, and measured cytokine production and expression of TLR-associated genes. We tested for differences in TLR-stimulated cytokine levels between subjects with CF and healthy subjects, and for associations between cytokine and gene expression levels with baseline lung function (forced expiratory volume in one second percent predicted (FEV1%)) and decline in FEV1% over time. RESULTS TMIIR in blood from subjects with CF were lower than in healthy controls. Expression of TLR regulators SARM1, TOLLIP, and AKT1 were downregulated in CF. In subjects with CF we found that lower TLR4-agonist-induced IL-8 was associated with lower FEV1% at enrollment (p<0.001) and with greater five year FEV1% decline (p<0.001). CONCLUSIONS TMIIR were lower in people with CF relative to healthy controls; however, unexpectedly, greater whole blood TMIIR were positively associated with lung function in people with CF. These findings suggest a complex interaction between inflammation and disease in people with CF.
Collapse
|
42
|
Lidington D, Fares JC, Uhl FE, Dinh DD, Kroetsch JT, Sauvé M, Malik FA, Matthes F, Vanherle L, Adel A, Momen A, Zhang H, Aschar-Sobbi R, Foltz WD, Wan H, Sumiyoshi M, Macdonald RL, Husain M, Backx PH, Heximer SP, Meissner A, Bolz SS. CFTR Therapeutics Normalize Cerebral Perfusion Deficits in Mouse Models of Heart Failure and Subarachnoid Hemorrhage. JACC Basic Transl Sci 2019; 4:940-958. [PMID: 31909302 PMCID: PMC6939007 DOI: 10.1016/j.jacbts.2019.07.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 01/01/2023]
Abstract
Heart failure (HF) and subarachnoid hemorrhage (SAH) chronically reduce cerebral perfusion, which negatively affects clinical outcome. This work demonstrates a strong relationship between cerebral artery cystic fibrosis transmembrane conductance regulator (CFTR) expression and altered cerebrovascular reactivity in HF and SAH. In HF and SAH, CFTR corrector compounds (C18 or lumacaftor) normalize pathological alterations in cerebral artery CFTR expression, vascular reactivity, and cerebral perfusion, without affecting systemic hemodynamic parameters. This normalization correlates with reduced neuronal injury. Therefore, CFTR therapeutics have emerged as valuable clinical tools to manage cerebrovascular dysfunction, impaired cerebral perfusion, and neuronal injury.
Collapse
Key Words
- CBF, cerebral blood flow
- CFTR, cystic fibrosis transmembrane conductance regulator
- HF, heart failure
- MAP, mean arterial pressure
- MOPS, 3-morpholinopropanesulfonic acid
- MRI, magnetic resonance imaging
- NIH, National Institutes of Health
- PCA, posterior cerebral artery
- S1P, sphingosine-1-phosphate
- SAH, subarachnoid hemorrhage
- TNF, tumor necrosis factor
- TPR, total peripheral resistance
- cognitive impairment
- corrector compounds
- cystic fibrosis transmembrane conductance regulator (CFTR)
- myogenic vasoconstriction
- sphingosine-1-phosphate
- tumor necrosis factor
- vascular smooth muscle cells
Collapse
Affiliation(s)
- Darcy Lidington
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Jessica C. Fares
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Franziska E. Uhl
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Danny D. Dinh
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Jeffrey T. Kroetsch
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Meghan Sauvé
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | - Firhan A. Malik
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Frank Matthes
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lotte Vanherle
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Arman Adel
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Abdul Momen
- Division of Cell & Molecular Biology, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Hangjun Zhang
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
| | | | - Warren D. Foltz
- STTARR Innovation Centre, Department of Radiation Oncology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Hoyee Wan
- Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Research Centre for Biomedical Research and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
- Division of Neurosurgery, St. Michael’s Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Sunnybrook Research Institute, Physical Sciences Platform and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Manabu Sumiyoshi
- Division of Neurosurgery, St. Michael’s Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Biosciences, Department of Neurosurgery, University of Tokushima Graduate School, Tokushima, Japan
| | - R. Loch Macdonald
- Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Research Centre for Biomedical Research and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
- Division of Neurosurgery, St. Michael’s Hospital, and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Mansoor Husain
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Division of Cell & Molecular Biology, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter H. Backx
- Division of Cardiology, University Health Network, Toronto, Ontario, Canada
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Scott P. Heximer
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
| | - Anja Meissner
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Wallenberg Center for Molecular Medicine and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Steffen-Sebastian Bolz
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at The Ted Rogers Centre for Heart Research Translational Biology and Engineering Program, University of Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence for Cardiovascular Research, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Dreano E, Bacchetta M, Simonin J, Galmiche L, Usal C, Slimani L, Sadoine J, Tesson L, Anegon I, Concordet J, Hatton A, Vignaud L, Tondelier D, Sermet‐Gaudelus I, Chanson M, Cottart C. Characterization of two rat models of cystic fibrosis-KO and F508del CFTR-Generated by Crispr-Cas9. Animal Model Exp Med 2019; 2:297-311. [PMID: 31942562 PMCID: PMC6930998 DOI: 10.1002/ame2.12091] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/22/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Genetically engineered animals are essential for gaining a proper understanding of the disease mechanisms of cystic fibrosis (CF). The rat is a relevant laboratory model for CF because of its zootechnical capacity, size, and airway characteristics, including the presence of submucosal glands. METHODS We describe the generation of a CF rat model (F508del) homozygous for the p.Phe508del mutation in the transmembrane conductance regulator (Cftr) gene. This model was compared to new Cftr -/- rats (CFTR KO). Target organs in CF were examined by histological staining of tissue sections and tooth enamel was quantified by micro-computed tomography. The activity of CFTR was evaluated by nasal potential difference (NPD) and short-circuit current measurements. The effect of VX-809 and VX-770 was analyzed on nasal epithelial primary cell cultures from F508del rats. RESULTS Both newborn F508del and Knock out (KO) animals developed intestinal obstruction that could be partly compensated by special diet combined with an osmotic laxative. The two rat models exhibited CF phenotypic anomalies such as vas deferens agenesis and tooth enamel defects. Histology of the intestine, pancreas, liver, and lungs was normal. Absence of CFTR function in KO rats was confirmed ex vivo by short-circuit current measurements on colon mucosae and in vivo by NPD, whereas residual CFTR activity was observed in F508del rats. Exposure of F508del CFTR nasal primary cultures to a combination of VX-809 and VX-770 improved CFTR-mediated Cl- transport. CONCLUSIONS The F508del rats reproduce the phenotypes observed in CFTR KO animals and represent a novel resource to advance the development of CF therapeutics.
Collapse
Affiliation(s)
| | - Marc Bacchetta
- Département de PédiatrieGynécologie & Obstétrique et Département de Physiologie Cellulaire & MétabolismeUniversité de GenèveGenèveSwitzerland
| | - Juliette Simonin
- Département de PédiatrieGynécologie & Obstétrique et Département de Physiologie Cellulaire & MétabolismeUniversité de GenèveGenèveSwitzerland
| | - Louise Galmiche
- Département de PathologieAPHPCHU Necker‐Enfants MaladesParisFrance
| | - Claire Usal
- Centre de Recherche en Transplantation & ImmunologieUMR 1064INSERMUniversité de NantesNantesFrance
- Plateforme Trangénèse Rat & ImmunoPhénomiqueINSERM 1064 & SFR François BonamyCNRS UMS3556NantesFrance
| | - Lotfi Slimani
- Pathologie, Imagerie & Biothérapies OrofacialesMontrougeFrance
- Plateforme Imageries du vivantFaculté de chirurgie dentaireUniversité de ParisParisFrance
| | - Jérémy Sadoine
- Pathologie, Imagerie & Biothérapies OrofacialesMontrougeFrance
| | - Laurent Tesson
- Centre de Recherche en Transplantation & ImmunologieUMR 1064INSERMUniversité de NantesNantesFrance
- Plateforme Trangénèse Rat & ImmunoPhénomiqueINSERM 1064 & SFR François BonamyCNRS UMS3556NantesFrance
| | - Ignacio Anegon
- Centre de Recherche en Transplantation & ImmunologieUMR 1064INSERMUniversité de NantesNantesFrance
- Plateforme Trangénèse Rat & ImmunoPhénomiqueINSERM 1064 & SFR François BonamyCNRS UMS3556NantesFrance
| | | | | | | | | | - Isabelle Sermet‐Gaudelus
- INSERM 1151INEMUniversité de ParisParisFrance
- AP‐HPCentre Maladie Rare Mucoviscidose et Maladies du CFTRAssistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
- Faculté de Médecine de ParisUniversité de ParisParisFrance
| | - Marc Chanson
- Département de PédiatrieGynécologie & Obstétrique et Département de Physiologie Cellulaire & MétabolismeUniversité de GenèveGenèveSwitzerland
| | - Charles‐Henry Cottart
- INSERM 1151INEMUniversité de ParisParisFrance
- AP‐HPCentre Maladie Rare Mucoviscidose et Maladies du CFTRAssistance Publique Hôpitaux de ParisHôpital Necker‐Enfants MaladesParisFrance
- Faculté de Pharmacie de ParisUniversité de ParisParisFrance
| |
Collapse
|
44
|
D'Antonio M, Reyna J, Jakubosky D, Donovan MKR, Bonder MJ, Matsui H, Stegle O, Nariai N, D'Antonio-Chronowska A, Frazer KA. Systematic genetic analysis of the MHC region reveals mechanistic underpinnings of HLA type associations with disease. eLife 2019; 8:e48476. [PMID: 31746734 PMCID: PMC6904215 DOI: 10.7554/elife.48476] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
The MHC region is highly associated with autoimmune and infectious diseases. Here we conduct an in-depth interrogation of associations between genetic variation, gene expression and disease. We create a comprehensive map of regulatory variation in the MHC region using WGS from 419 individuals to call eight-digit HLA types and RNA-seq data from matched iPSCs. Building on this regulatory map, we explored GWAS signals for 4083 traits, detecting colocalization for 180 disease loci with eQTLs. We show that eQTL analyses taking HLA type haplotypes into account have substantially greater power compared with only using single variants. We examined the association between the 8.1 ancestral haplotype and delayed colonization in Cystic Fibrosis, postulating that downregulation of RNF5 expression is the likely causal mechanism. Our study provides insights into the genetic architecture of the MHC region and pinpoints disease associations that are due to differential expression of HLA genes and non-HLA genes.
Collapse
Affiliation(s)
- Matteo D'Antonio
- Institute for Genomic MedicineUniversity of California, San DiegoSan DiegoUnited States
- Department of PediatricsRady Children’s Hospital, University of California, San DiegoSan DiegoUnited States
| | - Joaquin Reyna
- Department of PediatricsRady Children’s Hospital, University of California, San DiegoSan DiegoUnited States
- Biomedical Sciences Graduate ProgramUniversity of California, San DiegoLa JollaUnited States
| | - David Jakubosky
- Biomedical Sciences Graduate ProgramUniversity of California, San DiegoLa JollaUnited States
- Bioinformatics and Systems Biology Graduate ProgramUniversity of California, San DiegoSan DiegoUnited States
| | - Margaret KR Donovan
- Bioinformatics and Systems Biology Graduate ProgramUniversity of California, San DiegoSan DiegoUnited States
- Department of Biomedical InformaticsUniversity of California, San DiegoSan DiegoUnited States
| | - Marc-Jan Bonder
- European Molecular Biology Laboratory, European Bioinformatics InstituteCambridgeUnited Kingdom
| | - Hiroko Matsui
- Institute for Genomic MedicineUniversity of California, San DiegoSan DiegoUnited States
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics InstituteCambridgeUnited Kingdom
| | - Naoki Nariai
- Department of PediatricsRady Children’s Hospital, University of California, San DiegoSan DiegoUnited States
| | - Agnieszka D'Antonio-Chronowska
- Institute for Genomic MedicineUniversity of California, San DiegoSan DiegoUnited States
- Department of PediatricsRady Children’s Hospital, University of California, San DiegoSan DiegoUnited States
| | - Kelly A Frazer
- Institute for Genomic MedicineUniversity of California, San DiegoSan DiegoUnited States
- Department of PediatricsRady Children’s Hospital, University of California, San DiegoSan DiegoUnited States
| |
Collapse
|
45
|
Webster JD, Santagostino SF, Foreman O. Applications and considerations for the use of genetically engineered mouse models in drug development. Cell Tissue Res 2019; 380:325-340. [DOI: 10.1007/s00441-019-03101-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
46
|
Breton S, Nair AV, Battistone MA. Epithelial dynamics in the epididymis: role in the maturation, protection, and storage of spermatozoa. Andrology 2019; 7:631-643. [PMID: 31044554 PMCID: PMC6688936 DOI: 10.1111/andr.12632] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/07/2019] [Accepted: 03/29/2019] [Indexed: 01/10/2023]
Abstract
Epithelial cells line the lumen of tubular organs and are key players in their respective functions. They establish a unique luminal environment by providing a protective barrier and by performing vectorial transport of ions, nutrients, solutes, proteins, and water. Complex intercellular communication networks, specific for each organ, ensure their interaction with adjacent epithelial and non-epithelial cells, allowing them to respond to and modulate their immediate environment. In the epididymis, several epithelial cell types work in a concerted manner to establish a luminal acidic milieu that is essential for the post-testicular maturation and storage of spermatozoa. The epididymis also prevents autoimmune responses against auto-antigenic spermatozoa, while ensuring protection against ascending and blood pathogens. This is achieved by a network of immune cells that are in close contact and interact with epithelial cells. This review highlights the coordinated interactions between spermatozoa, basal cells, principal cells, narrow cells, clear cells, and immune cells that contribute to the maturation, protection, selection, and storage of spermatozoa in the lumen of the epididymis.
Collapse
Affiliation(s)
- S Breton
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Harvard Medical School, Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - A V Nair
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Harvard Medical School, Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - M A Battistone
- Program in Membrane Biology, Nephrology Division, Department of Medicine, Harvard Medical School, Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
47
|
Leenaars CH, De Vries RB, Heming A, Visser D, Holthaus D, Reijmer J, Elzinga J, Kempkes RW, Punt C, Beumer W, Meijboom FL, Ritskes-Hoitinga M. Animal models for cystic fibrosis: A systematic search and mapping review of the literature - Part 1: genetic models. Lab Anim 2019; 54:330-340. [PMID: 31411127 DOI: 10.1177/0023677219868502] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Animal models for cystic fibrosis (CF) have enhanced our understanding of the pathology and contributed to the development of new treatments. In the field of CF, many animal models have been developed and described. To our knowledge, thus far, none of the reviews of CF animal models has used a systematic methodology. A systematic approach to creating model overviews can lead to an objective, evidence-based choice of an animal model for new research questions. We searched Pubmed and Embase for the currently available animal models for CF. Two independent reviewers screened the results. We included all primary studies describing an animal model for CF. After duplicate removal, 12,304 publications were left. Because of the large number of models, in the current paper, only the genetic models are presented. A total of 636 publications were identified describing genetic animal models for CF in mice, pigs, ferrets, rats and zebrafish. Most of these models have an altered Cftr gene. An overview of basic model characteristics and outcome measures for these genetic models is provided, together with advice on using these data. As far as the authors are aware, this is one of the largest systematic mapping reviews on genetic animal models for CF. It can aid in selecting a suitable model and outcome measures. In general, the reporting quality of the included publications was poor. Further systematic reviews are warranted to determine the quality and translational value of these models further.
Collapse
Affiliation(s)
- Cathalijn Hc Leenaars
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands.,Faculty of Veterinary Medicine, Department of Animals in Science and Society, Utrecht University, Utrecht, The Netherlands.,Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Rob Bm De Vries
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna Heming
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Damian Visser
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David Holthaus
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joey Reijmer
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janneke Elzinga
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rosalie Wm Kempkes
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carine Punt
- ProQR Therapeutics NV, Leiden, The Netherlands
| | | | - Franck Lb Meijboom
- Faculty of Veterinary Medicine, Department of Animals in Science and Society, Utrecht University, Utrecht, The Netherlands
| | - Merel Ritskes-Hoitinga
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Fiorotto R, Strazzabosco M. Pathophysiology of Cystic Fibrosis Liver Disease: A Channelopathy Leading to Alterations in Innate Immunity and in Microbiota. Cell Mol Gastroenterol Hepatol 2019; 8:197-207. [PMID: 31075352 PMCID: PMC6664222 DOI: 10.1016/j.jcmgh.2019.04.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022]
Abstract
Cystic fibrosis (CF) is a monogenic disease caused by mutation of Cftr. CF-associated liver disease (CFLD) is a common nonpulmonary cause of mortality in CF and accounts for approximately 2.5%-5% of overall CF mortality. The peak of the disease is in the pediatric population, but a second wave of liver disease in CF adults has been reported in the past decade in association with an increase in the life expectancy of these patients. New drugs are available to correct the basic defect in CF but their efficacy in CFLD is not known. The cystic fibrosis transmembrane conductance regulator, expressed in the apical membrane of cholangiocytes, is a major determinant for bile secretion and CFLD classically has been considered a channelopathy. However, the recent findings of the cystic fibrosis transmembrane conductance regulator as a regulator of epithelial innate immunity and the possible influence of the intestinal disease with an altered microbiota on the liver complication have opened new mechanistic insights on the pathogenesis of CFLD. This review provides an overview of the current understanding of the pathophysiology of the disease and discusses a potential target for intervention.
Collapse
Affiliation(s)
- Romina Fiorotto
- Section of Digestive Diseases, Yale Liver Center, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.
| | - Mario Strazzabosco
- Section of Digestive Diseases, Yale Liver Center, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
49
|
Bose SJ, Bijvelds MJC, Wang Y, Liu J, Cai Z, Bot AGM, de Jonge HR, Sheppard DN. Differential thermostability and response to cystic fibrosis transmembrane conductance regulator potentiators of human and mouse F508del-CFTR. Am J Physiol Lung Cell Mol Physiol 2019; 317:L71-L86. [PMID: 30969810 PMCID: PMC6689747 DOI: 10.1152/ajplung.00034.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cross-species comparative studies have highlighted differences between human and mouse cystic fibrosis transmembrane conductance regulator (CFTR), the epithelial Cl- channel defective in cystic fibrosis (CF). Here, we compare the impact of the most common CF mutation F508del on the function of human and mouse CFTR heterologously expressed in mammalian cells and their response to CFTR modulators using the iodide efflux and patch-clamp techniques. Once delivered to the plasma membrane, human F508del-CFTR exhibited a severe gating defect characterized by infrequent channel openings and was thermally unstable, deactivating within minutes at 37°C. By contrast, the F508del mutation was without effect on the gating pattern of mouse CFTR, and channel activity demonstrated thermostability at 37°C. Strikingly, at all concentrations tested, the clinically approved CFTR potentiator ivacaftor was without effect on the mouse F508del-CFTR Cl- channel. Moreover, eight CFTR potentiators, including ivacaftor, failed to generate CFTR-mediated iodide efflux from CHO cells expressing mouse F508del-CFTR. However, they all produced CFTR-mediated iodide efflux with human F508del-CFTR-expressing CHO cells, while fifteen CFTR correctors rescued the plasma membrane expression of both human and mouse F508del-CFTR. Interestingly, the CFTR potentiator genistein enhanced CFTR-mediated iodide efflux from CHO cells expressing either human or mouse F508del-CFTR, whereas it only potentiated human F508del-CFTR Cl- channels in cell-free membrane patches, suggesting that its action on mouse F508del-CFTR is indirect. Thus, the F508del mutation has distinct effects on human and mouse CFTR Cl- channels.
Collapse
Affiliation(s)
- Samuel J Bose
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - Yiting Wang
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Jia Liu
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Zhiwei Cai
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| | - Alice G M Bot
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - Hugo R de Jonge
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center , Rotterdam , The Netherlands
| | - David N Sheppard
- School of Physiology, Pharmacology and Neuroscience, University of Bristol , Bristol , United Kingdom
| |
Collapse
|
50
|
Villella VR, Tosco A, Esposito S, Ferrari E, Bona G, Kroemer G, Raia V, Maiuri L. Personalization of therapies in rare diseases: a translational approach for the treatment of cystic fibrosis. Minerva Pediatr 2019; 71:362-370. [PMID: 30761822 DOI: 10.23736/s0026-4946.19.05511-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
High variability in the response rates to treatments can make the interpretation of data from clinical trials very difficult, particularly in rare genetic diseases in which the enrolment of thousands of patients is problematic. Personalized medicine largely depends on the establishment of appropriate early detectors of drug efficacy that may guide the administration (or discontinuation) of specific treatments. Such biomarkers should be capable of predicting the therapeutic response of individual patients and of monitoring early benefits of candidate drugs before late clinical benefits become evident. The identification of these biomarkers implies a rigorous stepwise process of translation from preclinical evaluation in cultured cells, suitable animal models or patient-derived freshly isolated cells to clinical application. In this review, we will discuss how a process of research translation can lead to the implementation of functional and mechanistic disease-relevant biomarkers. Moreover, we will address how preclinical data can be translated into the clinic in a personalized medical approach that can provide the right drug to the right patient within the right timeframe.
Collapse
Affiliation(s)
- Valeria R Villella
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Tosco
- Regional Cystic Fibrosis Center, Unit of Pediatrics, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Speranza Esposito
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Ferrari
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy.,Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Gianni Bona
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Guido Kroemer
- Equipe11 Labellisée Ligue Nationale contre le Cancer, Cordeliers Research Center, Paris, France.,INSERM U1138, Cordeliers Research Center, Paris, France.,Paris Descartes University, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Institute, Villejuif, France.,Section of Biology, Georges Pompidou European Hospital, Assistance Publique, Hôpitaux de Paris (AP-HP), Paris, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Unit of Pediatrics, Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Luigi Maiuri
- European Institute for Research in Cystic Fibrosis, San Raffaele Scientific Institute, Milan, Italy - .,Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| |
Collapse
|