1
|
Lee E, Orton K. Cystic fibrosis carrier screening in Australia: comparing sequencing and targeted panels across diverse ancestries. J Med Genet 2025; 62:219-226. [PMID: 39740802 DOI: 10.1136/jmg-2024-110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Targeted cystic fibrosis (CF) carrier screening panels may lack sensitivity in non-European ancestry groups. This study aims to evaluate the sensitivity of various panels in Australian CF carriers identified through sequencing. METHODS The following panels were evaluated in 869 CF carriers: Asuragen, Elucigene, Devyser, American College of Medical Genetics and Genomics and Victorian Clinical Genetics Services. Ancestry-specific CF carrier frequencies from population databases and Bayesian analysis were used to estimate post-test residual carrier risks. RESULTS When variants with varying clinical consequences (VCC) were not considered, mean test sensitivity was highest in the Northern Europe group (95.6%) and lowest in the Southern Asia group (64.0%). The post-test residual carrier risk in the Northern Europe group was approximately 1 in 546, with only the Southern Asia group having a higher residual carrier risk of 1 in 179. CONCLUSION The Southern Asia group exhibited the lowest test sensitivity and the highest post-test residual carrier risk, surpassing that of the Northern Europe group. The inclusion or exclusion of VCC significantly impacted the calculated test sensitivities. Further research is suggested to better characterise CFTR variants in non-European ancestry groups and to determine which VCC, if any, should be included in carrier screening reports.
Collapse
Affiliation(s)
- Eric Lee
- Molecular Genetics, Virtus Diagnostics, Revesby, New South Wales, Australia
| | - Kaylee Orton
- Molecular Genetics, Virtus Diagnostics, Revesby, New South Wales, Australia
| |
Collapse
|
2
|
Sousa AA, Hemez C, Lei L, Traore S, Kulhankova K, Newby GA, Doman JL, Oye K, Pandey S, Karp PH, McCray PB, Liu DR. Systematic optimization of prime editing for the efficient functional correction of CFTR F508del in human airway epithelial cells. Nat Biomed Eng 2025; 9:7-21. [PMID: 38987629 PMCID: PMC11754097 DOI: 10.1038/s41551-024-01233-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
Prime editing (PE) enables precise and versatile genome editing without requiring double-stranded DNA breaks. Here we describe the systematic optimization of PE systems to efficiently correct human cystic fibrosis (CF) transmembrane conductance regulator (CFTR) F508del, a three-nucleotide deletion that is the predominant cause of CF. By combining six efficiency optimizations for PE-engineered PE guide RNAs, the PEmax architecture, the transient expression of a dominant-negative mismatch repair protein, strategic silent edits, PE6 variants and proximal 'dead' single-guide RNAs-we increased correction efficiencies for CFTR F508del from less than 0.5% in HEK293T cells to 58% in immortalized bronchial epithelial cells (a 140-fold improvement) and to 25% in patient-derived airway epithelial cells. The optimizations also resulted in minimal off-target editing, in edit-to-indel ratios 3.5-fold greater than those achieved by nuclease-mediated homology-directed repair, and in the functional restoration of CFTR ion channels to over 50% of wild-type levels (similar to those achieved via combination treatment with elexacaftor, tezacaftor and ivacaftor) in primary airway cells. Our findings support the feasibility of a durable one-time treatment for CF.
Collapse
Affiliation(s)
- Alexander A Sousa
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Colin Hemez
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Lei Lei
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Soumba Traore
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Katarina Kulhankova
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan L Doman
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Keyede Oye
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Smriti Pandey
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Philip H Karp
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, IA, USA
| | - Paul B McCray
- Stead Family Department of Pediatrics and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
3
|
Xu W, Zhang S, Qin H, Yao K. From bench to bedside: cutting-edge applications of base editing and prime editing in precision medicine. J Transl Med 2024; 22:1133. [PMID: 39707395 DOI: 10.1186/s12967-024-05957-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024] Open
Abstract
CRISPR-based gene editing technology theoretically allows for precise manipulation of any genetic target within living cells, achieving the desired sequence modifications. This revolutionary advancement has fundamentally transformed the field of biomedicine, offering immense clinical potential for treating and correcting genetic disorders. In the treatment of most genetic diseases, precise genome editing that avoids the generation of mixed editing byproducts is considered the ideal approach. This article reviews the current progress of base editors and prime editors, elaborating on specific examples of their applications in the therapeutic field, and highlights opportunities for improvement. Furthermore, we discuss the specific performance of these technologies in terms of safety and efficacy in clinical applications, and analyze the latest advancements and potential directions that could influence the future development of genome editing technologies. Our goal is to outline the clinical relevance of this rapidly evolving scientific field and preview a roadmap for successful DNA base editing therapies for the treatment of hereditary or idiopathic diseases.
Collapse
Affiliation(s)
- Weihui Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Shiyao Zhang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| |
Collapse
|
4
|
O'Brien M, Whyte S, Doyle S, McAuliffe FM. Genetic disorders in maternal medicine. Best Pract Res Clin Obstet Gynaecol 2024; 97:102546. [PMID: 39265229 DOI: 10.1016/j.bpobgyn.2024.102546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/22/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024]
Abstract
The role of genetic testing within maternal medicine is expanding. Advancing technology and the increasing availability of genetic testing have seen more patients receiving a genetic diagnosis than ever before. Improved healthcare and understanding of these rare diseases means that many patients are living well into their reproductive years and starting families. Individual diseases are considered by their patterns of inheritance i.e. autosomal recessive, autosomal dominant and chromosomal diseases. This chapter specifically addresses the following examples and outlines an approach to pre-conceptual and pregnancy management; autosomal recessive (cystic fibrosis, phenylketonuria), autosomal dominant (osteogenesis imperfecta, vascular Ehlers-Danlos syndrome) and chromosomal (Turner syndrome). For many rare and ultrarare genetic diseases, there may be no clear guidelines or consensus on the correct management in pregnancy. This chapter seeks to provide a framework for the clinician to use to address the unique needs and risk profile of these patients in pregnancy and pre-conceptually and plan accordingly. The role of pharmacogenetics in maternal medicine, the future of education in genetics for patients and clinicians and the important role of genetic counselling are all considered in this chapter. This overview highlights the important role of genetics in maternal medicine and how this can inform management and planning for the safe care of mother and baby.
Collapse
Affiliation(s)
- Maggie O'Brien
- UCD Perinatal Research Centre, University College Dublin, National Maternity Hospital, Dublin, Ireland.
| | - Sinead Whyte
- The Department of Perinatal Genetics, National Maternity Hospital, Dublin, Ireland
| | - Sam Doyle
- UCD Perinatal Research Centre, University College Dublin, National Maternity Hospital, Dublin, Ireland; The Department of Perinatal Genetics, National Maternity Hospital, Dublin, Ireland
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, University College Dublin, National Maternity Hospital, Dublin, Ireland
| |
Collapse
|
5
|
Conto JD, Dalcin PTR, Ziegler B. Factors associated with cystic fibrosis mortality before the age of 30: retrospective analysis of a cohort in southern Brazil. Braz J Med Biol Res 2024; 57:e13476. [PMID: 39194031 DOI: 10.1590/1414-431x2024e13476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/26/2024] [Indexed: 08/29/2024] Open
Abstract
The aim of this study was to retrospectively evaluate the factors associated with mortality before the age of 30 in adults with cystic fibrosis (CF) followed up at a referral center in southern Brazil. This study included individuals over 18 years of age. Clinical data related to childhood and the period of transition to an adult healthcare of individuals with CF were recorded, as well as spirometric and mortality data of individuals between 18 and 30 years of age. A total of 48 patients were included in this study, of which 28 (58.3%) were male. Comparing groups, we observed a higher prevalence of homozygosis for the F508del mutation (P=0.028), massive hemoptysis before the age of 18 (P=0.027), and lower values of pulmonary function, forced expiratory volume in the first second (FEV1) (%) (P=0.002), forced vital capacity (FVC) (%) (P=0.01), and FEV1/FVC (%) (P=0.001) in the group that died before age 30. F508del homozygosis, episodes of massive hemoptysis in childhood, and lower FEV1 values at age 18 were related to mortality before age 30 in a cohort of individuals with CF in southern Brazil.
Collapse
Affiliation(s)
- J De Conto
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - P T R Dalcin
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - B Ziegler
- Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| |
Collapse
|
6
|
Zeng C, Schlueter DJ, Tran TC, Babbar A, Cassini T, Bastarache LA, Denny JC. Comparison of phenomic profiles in the All of Us Research Program against the US general population and the UK Biobank. J Am Med Inform Assoc 2024; 31:846-854. [PMID: 38263490 PMCID: PMC10990551 DOI: 10.1093/jamia/ocad260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/05/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
IMPORTANCE Knowledge gained from cohort studies has dramatically advanced both public and precision health. The All of Us Research Program seeks to enroll 1 million diverse participants who share multiple sources of data, providing unique opportunities for research. It is important to understand the phenomic profiles of its participants to conduct research in this cohort. OBJECTIVES More than 280 000 participants have shared their electronic health records (EHRs) in the All of Us Research Program. We aim to understand the phenomic profiles of this cohort through comparisons with those in the US general population and a well-established nation-wide cohort, UK Biobank, and to test whether association results of selected commonly studied diseases in the All of Us cohort were comparable to those in UK Biobank. MATERIALS AND METHODS We included participants with EHRs in All of Us and participants with health records from UK Biobank. The estimates of prevalence of diseases in the US general population were obtained from the Global Burden of Diseases (GBD) study. We conducted phenome-wide association studies (PheWAS) of 9 commonly studied diseases in both cohorts. RESULTS This study included 287 012 participants from the All of Us EHR cohort and 502 477 participants from the UK Biobank. A total of 314 diseases curated by the GBD were evaluated in All of Us, 80.9% (N = 254) of which were more common in All of Us than in the US general population [prevalence ratio (PR) >1.1, P < 2 × 10-5]. Among 2515 diseases and phenotypes evaluated in both All of Us and UK Biobank, 85.6% (N = 2152) were more common in All of Us (PR >1.1, P < 2 × 10-5). The Pearson correlation coefficients of effect sizes from PheWAS between All of Us and UK Biobank were 0.61, 0.50, 0.60, 0.57, 0.40, 0.53, 0.46, 0.47, and 0.24 for ischemic heart diseases, lung cancer, chronic obstructive pulmonary disease, dementia, colorectal cancer, lower back pain, multiple sclerosis, lupus, and cystic fibrosis, respectively. DISCUSSION Despite the differences in prevalence of diseases in All of Us compared to the US general population or the UK Biobank, our study supports that All of Us can facilitate rapid investigation of a broad range of diseases. CONCLUSION Most diseases were more common in All of Us than in the general US population or the UK Biobank. Results of disease-disease association tests from All of Us are comparable to those estimated in another well-studied national cohort.
Collapse
Affiliation(s)
- Chenjie Zeng
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - David J Schlueter
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Health and Society, University of Toronto, Scarborough, Toronto, ON, Canada
| | - Tam C Tran
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Anav Babbar
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Thomas Cassini
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Lisa A Bastarache
- Center for Precision Medicine, Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Josh C Denny
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
7
|
Walker AJ, Graham C, Greenwood M, Woodall M, Maeshima R, O’Hara-Wright M, Sanz DJ, Guerrini I, Aldossary AM, O’Callaghan C, Baines DL, Harrison PT, Hart SL. Molecular and functional correction of a deep intronic splicing mutation in CFTR by CRISPR-Cas9 gene editing. Mol Ther Methods Clin Dev 2023; 31:101140. [PMID: 38027060 PMCID: PMC10661860 DOI: 10.1016/j.omtm.2023.101140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CFTR gene. The 10th most common mutation, c.3178-2477C>T (3849+10kb C>T), involves a cryptic, intronic splice site. This mutation was corrected in CF primary cells homozygous for this mutation by delivering pairs of guide RNAs (gRNAs) with Cas9 protein in ribonucleoprotein (RNP) complexes that introduce double-strand breaks to flanking sites to excise the 3849+10kb C>T mutation, followed by DNA repair by the non-homologous end-joining pathway, which functions in all cells of the airway epithelium. RNP complexes were delivered to CF basal epithelial cell by a non-viral, receptor-targeted nanocomplex comprising a formulation of targeting peptides and lipids. Canonical CFTR mRNA splicing was, thus, restored leading to the restoration of CFTR protein expression with concomitant restoration of electrophysiological function in airway epithelial air-liquid interface cultures. Off-target editing was not detected by Sanger sequencing of in silico-selected genomic sites with the highest sequence similarities to the gRNAs, although more sensitive unbiased whole genome sequencing methods would be required for possible translational developments. This approach could potentially be used to correct aberrant splicing signals in several other CF mutations and other genetic disorders where deep-intronic mutations are pathogenic.
Collapse
Affiliation(s)
- Amy J. Walker
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Carina Graham
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Miriam Greenwood
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Maximillian Woodall
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Ruhina Maeshima
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Michelle O’Hara-Wright
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - David J. Sanz
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Ileana Guerrini
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Ahmad M. Aldossary
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Christopher O’Callaghan
- Infection, Immunity & Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Deborah L. Baines
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Patrick T. Harrison
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Stephen L. Hart
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
8
|
Mention K, Cavusoglu-Doran K, Joynt AT, Santos L, Sanz D, Eastman AC, Merlo C, Langfelder-Schwind E, Scallan MF, Farinha CM, Cutting GR, Sharma N, Harrison PT. Use of adenine base editing and homology-independent targeted integration strategies to correct the cystic fibrosis causing variant, W1282X. Hum Mol Genet 2023; 32:3237-3248. [PMID: 37649273 PMCID: PMC10656707 DOI: 10.1093/hmg/ddad143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/21/2023] [Accepted: 08/12/2023] [Indexed: 09/01/2023] Open
Abstract
Small molecule drugs known as modulators can treat ~90% of people with cystic fibrosis (CF), but do not work for premature termination codon variants such as W1282X (c.3846G>A). Here we evaluated two gene editing strategies, Adenine Base Editing (ABE) to correct W1282X, and Homology-Independent Targeted Integration (HITI) of a CFTR superexon comprising exons 23-27 (SE23-27) to enable expression of a CFTR mRNA without W1282X. In Flp-In-293 cells stably expressing a CFTR expression minigene bearing W1282X, ABE corrected 24% of W1282X alleles, rescued CFTR mRNA from nonsense mediated decay and restored protein expression. However, bystander editing at the adjacent adenine (c.3847A>G), caused an amino acid change (R1283G) that affects CFTR maturation and ablates ion channel activity. In primary human nasal epithelial cells homozygous for W1282X, ABE corrected 27% of alleles, but with a notably lower level of bystander editing, and CFTR channel function was restored to 16% of wild-type levels. Using the HITI approach, correct integration of a SE23-27 in intron 22 of the CFTR locus in 16HBEge W1282X cells was detected in 5.8% of alleles, resulting in 7.8% of CFTR transcripts containing the SE23-27 sequence. Analysis of a clonal line homozygous for the HITI-SE23-27 produced full-length mature protein and restored CFTR anion channel activity to 10% of wild-type levels, which could be increased three-fold upon treatment with the triple combination of CF modulators. Overall, these data demonstrate two different editing strategies can successfully correct W1282X, the second most common class I variant, with a concomitant restoration of CFTR function.
Collapse
Affiliation(s)
- Karen Mention
- Department of Physiology, University College Cork, College Road, Cork, T12 K8AF, Ireland
- School of Microbiology, University College Cork, College Road, Cork, T12 K8AF, Ireland
| | - Kader Cavusoglu-Doran
- Department of Physiology, University College Cork, College Road, Cork, T12 K8AF, Ireland
| | - Anya T Joynt
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, United States
| | - Lúcia Santos
- Department of Physiology, University College Cork, College Road, Cork, T12 K8AF, Ireland
- Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisboa, Campo Grande, C8 bdg, Lisboa 1749-016, Portugal
| | - David Sanz
- Department of Physiology, University College Cork, College Road, Cork, T12 K8AF, Ireland
| | - Alice C Eastman
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, United States
| | - Christian Merlo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins Hospital, 1800 Orleans St, Baltimore, MD 21287, United States
| | - Elinor Langfelder-Schwind
- The Cystic Fibrosis Center, Lenox Hill Hospital, 100 E. 77th Street, 4E, New York, NY 10075, United States
| | - Martina F Scallan
- School of Microbiology, University College Cork, College Road, Cork, T12 K8AF, Ireland
| | - Carlos M Farinha
- Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute, University of Lisboa, Campo Grande, C8 bdg, Lisboa 1749-016, Portugal
| | - Garry R Cutting
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, United States
| | - Neeraj Sharma
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, United States
| | - Patrick T Harrison
- Department of Physiology, University College Cork, College Road, Cork, T12 K8AF, Ireland
| |
Collapse
|
9
|
Sepe A, Romano C, Landi I, Castaldo A, Cimbalo C, Farina F, Scorza M, Salvadori L, Raia V, Tosco A. Pseudo-Bartter syndrome in infant with cystic fibrosis screen positive, inconclusive diagnosis: A case report. Clin Case Rep 2023; 11:e8046. [PMID: 38028053 PMCID: PMC10659916 DOI: 10.1002/ccr3.8046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
The introduction of newborn screening for cystic fibrosis (CF) increased diagnosis of cystic fibrosis screen positive inconclusive diagnosis (CFSPID). We described the case of a 12-month-old boy with CFSPID who, during summer, presented Pseudo-Bartter syndrome with no diagnostic criteria for CF.
Collapse
Affiliation(s)
- Angela Sepe
- Department of Maternal and Child health, Paediatric Unit, Cystic Fibrosis Regional Reference CenterA.O.U. Federico IINaplesItaly
| | - Camilla Romano
- Paediatric Unit, Department of Translational Medical Sciences, Cystic Fibrosis Regional Reference CenterUniversity of Naples Federico IINaplesItaly
| | - Ivana Landi
- Paediatric Unit, Department of Translational Medical Sciences, Cystic Fibrosis Regional Reference CenterUniversity of Naples Federico IINaplesItaly
| | - Alice Castaldo
- Paediatric Unit, Department of Translational Medical Sciences, Cystic Fibrosis Regional Reference CenterUniversity of Naples Federico IINaplesItaly
| | - Chiara Cimbalo
- Paediatric Unit, Department of Translational Medical Sciences, Cystic Fibrosis Regional Reference CenterUniversity of Naples Federico IINaplesItaly
| | - Federica Farina
- CEINGE Biotecnologie Avanzate ScarlFranco SalvatoreNaplesItaly
| | - Manuela Scorza
- CEINGE Biotecnologie Avanzate ScarlFranco SalvatoreNaplesItaly
| | - Laura Salvadori
- Department of Maternal and Child health, Paediatric Unit, Cystic Fibrosis Regional Reference CenterA.O.U. Federico IINaplesItaly
| | - Valeria Raia
- Paediatric Unit, Department of Translational Medical Sciences, Cystic Fibrosis Regional Reference CenterUniversity of Naples Federico IINaplesItaly
| | - Antonella Tosco
- Department of Maternal and Child health, Paediatric Unit, Cystic Fibrosis Regional Reference CenterA.O.U. Federico IINaplesItaly
| |
Collapse
|
10
|
Andreou SH, Davies JC. Cystic fibrosis to CFSPID: Burden of care vs need and rational approach to weaning therapies. Paediatr Respir Rev 2023; 47:27-29. [PMID: 37487798 DOI: 10.1016/j.prrv.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023]
Abstract
We present a case of a 10-year-old boy initially diagnosed with CF based on NBS guidelines. However, as CF genetics knowledge has advanced, he has been reclassified as CFSPID based on normal investigations and excellent general clinical status, in line with updated CFSPID guidelines. This case highlights the significance of reviewing CF diagnoses according to the latest understanding of CFTR mutation phenotypes, as well as the patient's clinical status. In order to identify opportunities to save patients from burdensome CF treatment and management, we review current CFSPID guidelines, emphasizing care tailored to each individual case.
Collapse
Affiliation(s)
| | - J C Davies
- Royal Brompton Hospital, Sydney St, London SW3 6NP, United Kingdom
| |
Collapse
|
11
|
Salazar-Barragan M, Taub DR. The Effects of Elexacaftor, Tezacaftor, and Ivacaftor (ETI) on Blood Glucose in Patients With Cystic Fibrosis: A Systematic Review. Cureus 2023; 15:e41697. [PMID: 37575762 PMCID: PMC10413995 DOI: 10.7759/cureus.41697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2023] [Indexed: 08/15/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder resulting from defects in the cystic fibrosis transmembrane conductance regulator (CFTR) protein, which in turn results in a multi-systemic disorder. There are numerous known CF alleles associated with different mutations of the CFTR gene, with the most common CF allele being a three-base-pair deletion known as ΔF508. One common manifestation of CF is glycemic dysregulation associated with decreased insulin secretion, often progressing into a distinct form of diabetes known as cystic fibrosis-related diabetes (CFRD). In the past decade, a class of drugs known as CFTR modulators has entered clinical practice. These drugs interact with the CFTR protein to restore its function, with different modulators (or combinations of modulators) suitable for patients with different CFTR mutations. Previous research has established that the modulator ivacaftor is effective in decreasing blood glucose and sometimes resolving CFRD in patients with certain CFTR mutations (class III mutations). However, early modulator therapies for individuals with the common ΔF508 mutation (e.g., a combination of the modulators lumacaftor and ivacaftor) have largely proven ineffective in improving glucose regulation. More recently, a combination therapy of three modulators, namely elexacaftor, tezacaftor, and ivacaftor (ETI), has entered clinical practice for patients with the ΔF508 mutation. However, it is not clear whether this therapy is effective in treating dysglycemia. We searched for studies of any design that examined the effects of ETI on measures of blood glucose. All available studies were observational studies comparing patients before and after initiating ETI therapy. Measures of daily-life blood glucose (those obtained with continuous glucose monitoring systems or by measuring glycated hemoglobin (HbA1c)) and post-prandial glucose spikes from oral glucose tolerance tests showed significant improvements in at least some studies. The majority of studies showed significant improvements from pre- to post-ETI in one or more blood glucose measures. While the interpretation of this evidence is complicated by the lack of randomized controlled trials, it appears that ETI therapy is associated with improved glucose regulation for at least some patients with the ΔF508 mutation.
Collapse
Affiliation(s)
| | - Daniel R Taub
- Biology, Southwestern University, Georgetown, TX, USA
| |
Collapse
|
12
|
Development of novel therapeutics for all individuals with CF (the future goes on). J Cyst Fibros 2023; 22 Suppl 1:S45-S49. [PMID: 36319570 DOI: 10.1016/j.jcf.2022.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 11/08/2022]
Abstract
Despite the major advances and successes in finding and establishing new treatments that tackle the basic defect in Cystic Fibrosis (CF), there is still an unmet need to bring these potentially curative therapies to all individuals with CF. Here, we review aspects of what is still missing to treat all individuals with CF by such approaches. On the one hand, we discuss novel holistic (high-throughput) approaches to elucidate mechanistic defects caused by distinct classes of mutations to identify novel drug targets. On the other hand, we examine therapeutic approaches to correct the gene in its own environment, i.e., in the genome.
Collapse
|
13
|
Bergougnoux A, Billet A, Ka C, Heller M, Degrugillier F, Vuillaume ML, Thoreau V, Sasorith S, Bareil C, Thèze C, Ferec C, Gac GL, Bienvenu T, Bieth E, Gaston V, Lalau G, Pagin A, Malinge MC, Dufernez F, Lemonnier L, Koenig M, Fergelot P, Claustres M, Taulan-Cadars M, Kitzis A, Reboul MP, Becq F, Fanen P, Mekki C, Audrezet MP, Girodon E, Raynal C. The multi-faceted nature of 15 CFTR exonic variations: Impact on their functional classification and perspectives for therapy. J Cyst Fibros 2022:S1569-1993(22)01423-0. [PMID: 36567205 DOI: 10.1016/j.jcf.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/30/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND The majority of variants of unknown clinical significance (VUCS) in the CFTR gene are missense variants. While change on the CFTR protein structure or function is often suspected, impact on splicing may be neglected. Such undetected splicing default of variants may complicate the interpretation of genetic analyses and the use of an appropriate pharmacotherapy. METHODS We selected 15 variants suspected to impact CFTR splicing after in silico predictions on 319 missense variants (214 VUCS), reported in the CFTR-France database. Six specialized laboratories assessed the impact of nucleotide substitutions on splicing (minigenes), mRNA expression levels (quantitative PCR), synthesis and maturation (western blot), cellular localization (immunofluorescence) and channel function (patch clamp) of the CFTR protein. We also studied maturation and function of the truncated protein, consecutive to in-frame aberrant splicing, on additional plasmid constructs. RESULTS Six of the 15 variants had a major impact on CFTR splicing by in-frame (n = 3) or out-of-frame (n = 3) exon skipping. We reclassified variants into: splicing variants; variants causing a splicing defect and the impairment of CFTR folding and/or function related to the amino acid substitution; deleterious missense variants that impair CFTR folding and/or function; and variants with no consequence on the different processes tested. CONCLUSION The 15 variants have been reclassified by our comprehensive approach of in vitro experiments that should be used to properly interpret very rare exonic variants of the CFTR gene. Targeted therapies may thus be adapted to the molecular defects regarding the results of laboratory experiments.
Collapse
Affiliation(s)
- A Bergougnoux
- Génétique Moléculaire, CHU Montpellier, Montpellier, France; PhyMedExp, INSERM, CNRS UMR, Montpellier, France; Université de Montpellier, Montpellier, France
| | - A Billet
- Laboratoire STIM, Université de Poitiers, Poitiers, France
| | - C Ka
- Service de génétique moléculaire, CHRU Brest, Brest, France; Université de Brest, Inserm, UMR 1078, GGB, Brest, France
| | - M Heller
- Service de Médecine Génomique des Maladies de Système et d'Organe, APHP Centre - Université de Paris, Hôpital Cochin, Paris, France
| | - F Degrugillier
- Université Paris-Est Créteil, INSERM, IMRB, Créteil F-94010, France
| | - M-L Vuillaume
- Génétique Moléculaire, CHU Bordeaux, Bordeaux, France
| | - V Thoreau
- Laboratoire NEUVACOD-3808, Université de Poitiers, Poitiers, France
| | - S Sasorith
- Génétique Moléculaire, CHU Montpellier, Montpellier, France; PhyMedExp, INSERM, CNRS UMR, Montpellier, France
| | - C Bareil
- Génétique Moléculaire, CHU Montpellier, Montpellier, France
| | - C Thèze
- Génétique Moléculaire, CHU Montpellier, Montpellier, France
| | - C Ferec
- Université de Brest, Inserm, UMR 1078, GGB, Brest, France
| | - G Le Gac
- Service de génétique moléculaire, CHRU Brest, Brest, France; Université de Brest, Inserm, UMR 1078, GGB, Brest, France
| | - T Bienvenu
- Service de Médecine Génomique des Maladies de Système et d'Organe, APHP Centre - Université de Paris, Hôpital Cochin, Paris, France
| | - E Bieth
- Génétique Médicale, CHU Toulouse, Toulouse, France
| | - V Gaston
- Génétique Médicale, CHU Toulouse, Toulouse, France
| | - G Lalau
- Biochimie et Biologie Moléculaire, CHU Lille, Lille, France
| | - A Pagin
- Biochimie et Biologie Moléculaire, CHU Lille, Lille, France
| | - M-C Malinge
- Biochimie et Génétique, CHU Angers, Angers, France
| | - F Dufernez
- Génétique, CHU Poitiers, Poitiers, France
| | - L Lemonnier
- Association Vaincre la Mucoviscidose, Paris, France
| | - M Koenig
- Génétique Moléculaire, CHU Montpellier, Montpellier, France; PhyMedExp, INSERM, CNRS UMR, Montpellier, France; Université de Montpellier, Montpellier, France
| | - P Fergelot
- MRGM, INSERM UMR 1211 Université de Bordeaux, Bordeaux, France
| | - M Claustres
- Université de Montpellier, Montpellier, France
| | - M Taulan-Cadars
- PhyMedExp, INSERM, CNRS UMR, Montpellier, France; Université de Montpellier, Montpellier, France
| | - A Kitzis
- Génétique, CHU Poitiers, Poitiers, France
| | - M-P Reboul
- Génétique Moléculaire, CHU Bordeaux, Bordeaux, France
| | - F Becq
- Laboratoire STIM, Université de Poitiers, Poitiers, France
| | - P Fanen
- AP-HP, Département de Biochimie-Biologie Moléculaire, Pharmacologie, Génétique Médicale, Hôpital Henri Mondor, Créteil F-94010, France
| | - C Mekki
- AP-HP, Département de Biochimie-Biologie Moléculaire, Pharmacologie, Génétique Médicale, Hôpital Henri Mondor, Créteil F-94010, France
| | - M-P Audrezet
- Service de génétique moléculaire, CHRU Brest, Brest, France; Université de Brest, Inserm, UMR 1078, GGB, Brest, France
| | - E Girodon
- Service de Médecine Génomique des Maladies de Système et d'Organe, APHP Centre - Université de Paris, Hôpital Cochin, Paris, France; INSERM U1151, Institut Necker Enfants Malades, Université de Paris, Paris, France
| | - C Raynal
- Génétique Moléculaire, CHU Montpellier, Montpellier, France; PhyMedExp, INSERM, CNRS UMR, Montpellier, France.
| |
Collapse
|
14
|
ECFS standards of care on CFTR-related disorders: Updated diagnostic criteria. J Cyst Fibros 2022; 21:908-921. [DOI: 10.1016/j.jcf.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022]
|
15
|
Ludovico A, Moran O, Baroni D. Modulator Combination Improves In Vitro the Microrheological Properties of the Airway Surface Liquid of Cystic Fibrosis Airway Epithelia. Int J Mol Sci 2022; 23:ijms231911396. [PMID: 36232697 PMCID: PMC9569604 DOI: 10.3390/ijms231911396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) protein, a plasma membrane protein expressed on the apical surface of secretory epithelia of the airways. In the airways, defective or absent function of the CFTR protein determines abnormalities of chloride and bicarbonate secretion and, in general, of the transepithelial homeostasis that lead to alterations of airway surface liquid (ASL) composition and properties. The reduction of ASL volume impairs ciliary beating with the consequent accumulation of a sticky mucus. This situation prevents normal mucociliary clearance, favoring the survival and proliferation of bacteria and contributing to the genesis of the CF pulmonary disease. We explored the potential of some CFTR modulators, namely ivacaftor, tezacaftor, elexacaftor and their combination KaftrioTM, capable of partially recovering the basic defects of the CFTR protein, to ameliorate the transepithelial fluid transport and the viscoelastic properties of the mucus when used singly or in combination. Primary human bronchial epithelial cells obtained from CF and non-CF patients were differentiated into a mucociliated epithelia in order to assess the effects of correctors tezacaftor, elexacaftor and their combination with potentiator ivacaftor on the key properties of ASL, such as fluid reabsorption, viscosity, protein content and pH. The treatment of airway epithelia bearing the deletion of a phenylalanine at position 508 (F508del) in the CFTR gene with tezacaftor and elexacaftor significantly improved the pericilial fluid composition, reducing the fluid reabsorption, correcting the ASL pH and reducing the viscosity of the mucus. KaftrioTM was more effective than single modulators in improving all the evaluated parameters, demonstrating once more that this combination recently approved for patients 6 years and older with cystic fibrosis who have at least one F508del mutation in the CFTR gene represents a valuable tool to defeat CF.
Collapse
Affiliation(s)
| | | | - Debora Baroni
- Correspondence: ; Tel.: +39-010-647-5559; Fax: +39-010-647-5500
| |
Collapse
|
16
|
Harrison PT. CFTR RNA- and DNA-based therapies. Curr Opin Pharmacol 2022; 65:102247. [PMID: 35709547 DOI: 10.1016/j.coph.2022.102247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/20/2022] [Accepted: 05/01/2022] [Indexed: 11/29/2022]
Abstract
This review provides an update on recent developments of RNA- and DNA-based methodologies and their intracellular targets in the context of cystic fibrosis (CF) lung disease. Ultimately, clinical success will require a suitable delivery system, but since the cargo for all these strategies is nucleic acid, it should hopefully be possible to exploit delivery breakthroughs from one study and apply these innovations to other experiments in order to identify the best strategy for everyone with CF. Ultimately, it may be the same approach for everyone, or possibly a number of different strategies tailored to particular mutations or classes/groups of mutations. And whilst the current focus is on CF lung disease, in the longer term the goal is to treat all affected organs in people with CF such as the pancreas, gut, and liver.
Collapse
Affiliation(s)
- Patrick T Harrison
- Department of Physiology, BioSciences Institute, University College Cork, Ireland.
| |
Collapse
|
17
|
Strub MD, Gao L, Tan K, McCray PB. Analysis of multiple gene co-expression networks to discover interactions favoring CFTR biogenesis and ΔF508-CFTR rescue. BMC Med Genomics 2021; 14:258. [PMID: 34717611 PMCID: PMC8557508 DOI: 10.1186/s12920-021-01106-7] [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: 04/08/2021] [Accepted: 10/20/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND We previously reported that expression of a miR-138 mimic or knockdown of SIN3A in primary cultures of cystic fibrosis (CF) airway epithelia increased ΔF508-CFTR mRNA and protein levels, and partially restored CFTR-dependent chloride transport. Global mRNA transcript profiling in ΔF508-CFBE cells treated with miR-138 mimic or SIN3A siRNA identified two genes, SYVN1 and NEDD8, whose inhibition significantly increased ΔF508-CFTR trafficking, maturation, and function. Little is known regarding the dynamic changes in the CFTR gene network during such rescue events. We hypothesized that analysis of condition-specific gene networks from transcriptomic data characterizing ΔF508-CFTR rescue could help identify dynamic gene modules associated with CFTR biogenesis. METHODS We applied a computational method, termed M-module, to analyze multiple gene networks, each of which exhibited differential activity compared to a baseline condition. In doing so, we identified both unique and shared gene pathways across multiple differential networks. To construct differential networks, gene expression data from CFBE cells were divided into three groups: (1) siRNA inhibition of NEDD8 and SYVN1; (2) miR-138 mimic and SIN3A siRNA; and (3) temperature (27 °C for 24 h, 40 °C for 24 h, and 27 °C for 24 h followed by 40 °C for 24 h). RESULTS Interrogation of individual networks (e.g., NEDD8/SYVN1 network), combinations of two networks (e.g., NEDD8/SYVN1 + temperature networks), and all three networks yielded sets of 1-modules, 2-modules, and 3-modules, respectively. Gene ontology analysis revealed significant enrichment of dynamic modules in pathways including translation, protein metabolic/catabolic processes, protein complex assembly, and endocytosis. Candidate CFTR effectors identified in the analysis included CHURC1, GZF1, and RPL15, and siRNA-mediated knockdown of these genes partially restored CFTR-dependent transepithelial chloride current to ΔF508-CFBE cells. CONCLUSIONS The ability of the M-module to identify dynamic modules involved in ΔF508 rescue provides a novel approach for studying CFTR biogenesis and identifying candidate suppressors of ΔF508.
Collapse
Affiliation(s)
- Matthew D Strub
- Department of Pediatrics, University of Iowa, 6320 PBDB, 169 Newton Road, Iowa City, IA, 52242, USA.,Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52245, USA
| | - Long Gao
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kai Tan
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Paul B McCray
- Department of Pediatrics, University of Iowa, 6320 PBDB, 169 Newton Road, Iowa City, IA, 52242, USA. .,Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52245, USA.
| |
Collapse
|
18
|
Ruiz-Schultz N, Asay B, Rohrwasser A. Scalable Newborn Screening Solutions: Bioinformatics and Next-Generation Sequencing. Int J Neonatal Screen 2021; 7:ijns7040063. [PMID: 34698050 PMCID: PMC8544371 DOI: 10.3390/ijns7040063] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/25/2021] [Accepted: 09/23/2021] [Indexed: 11/20/2022] Open
Abstract
Expansion of the newborn disorder panel requires the incorporation of new testing modalities. This is especially true for disorders lacking robust biomarkers for detection in primary screening methods and for disorders requiring genotyping or sequencing as a second-tier and/or diagnostic test. In this commentary, we discuss how next-generation sequencing (NGS) methods can be used as a secondary testing method in NBS. Additionally, we elaborate on the importance of genomic variant repositories for the annotation and interpretation of variants. Barriers to the incorporation of NGS and bioinformatics within NBS are discussed, and ideas for a regional bioinformatics model and shared variant repository are presented as potential solutions.
Collapse
|
19
|
Sanders M, Lawlor JMJ, Li X, Schuen JN, Millard SL, Zhang X, Buck L, Grysko B, Uhl KL, Hinds D, Stenger CL, Morris M, Lamb N, Levy H, Bupp C, Prokop JW. Genomic, transcriptomic, and protein landscape profile of CFTR and cystic fibrosis. Hum Genet 2021; 140:423-439. [PMID: 32734384 PMCID: PMC7855842 DOI: 10.1007/s00439-020-02211-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/25/2020] [Indexed: 01/18/2023]
Abstract
Cystic Fibrosis (CF) is caused most often by removal of amino acid 508 (Phe508del, deltaF508) within CFTR, yet dozens of additional CFTR variants are known to give rise to CF and many variants in the genome are known to contribute to CF pathology. To address CFTR coding variants, we developed a sequence-to-structure-to-dynamic matrix for all amino acids of CFTR using 233 vertebrate species, CFTR structure within a lipid membrane, and 20 ns of molecular dynamic simulation to assess known variants from the CFTR1, CFTR2, ClinVar, TOPmed, gnomAD, and COSMIC databases. Surprisingly, we identify 18 variants of uncertain significance within CFTR from diverse populations that are heritable and a likely cause of CF that have been understudied due to nonexistence in Caucasian populations. In addition, 15 sites within the genome are known to modulate CF pathology, where we have identified one genome region (chr11:34754985-34836401) that contributes to CF through modulation of expression of a noncoding RNA in epithelial cells. These 15 sites are just the beginning of understanding comodifiers of CF, where utilization of eQTLs suggests many additional genomics of CFTR expressing cells that can be influenced by genomic background of CFTR variants. This work highlights that many additional insights of CF genetics are needed, particularly as pharmaceutical interventions increase in the coming years.
Collapse
Affiliation(s)
- Morgan Sanders
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - James M J Lawlor
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - Xiaopeng Li
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - John N Schuen
- Pediatric Pulmonology, Helen DeVos Children's Hospital, Grand Rapids, MI, 49503, USA
| | - Susan L Millard
- Pediatric Pulmonology, Helen DeVos Children's Hospital, Grand Rapids, MI, 49503, USA
| | - Xi Zhang
- Department of Pediatrics, Division of Pulmonary Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Leah Buck
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
- Department of Mathematics, University of North Alabama, Florence, AL, 35632, USA
| | - Bethany Grysko
- Spectrum Health Medical Genetics, Grand Rapids, MI, 49503, USA
| | - Katie L Uhl
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
| | - David Hinds
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - Cynthia L Stenger
- Department of Mathematics, University of North Alabama, Florence, AL, 35632, USA
| | - Michele Morris
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - Neil Lamb
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - Hara Levy
- Department of Pediatrics, Division of Pulmonary Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Caleb Bupp
- Spectrum Health Medical Genetics, Grand Rapids, MI, 49503, USA
| | - Jeremy W Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI, 49503, USA.
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
20
|
Fawcett LK, Wakefield CE, Sivam S, Middleton PG, Wark P, Widger J, Jaffe A, Waters SA. Avatar acceptability: views from the Australian Cystic Fibrosis community on the use of personalised organoid technology to guide treatment decisions. ERJ Open Res 2021; 7:00448-2020. [PMID: 33532470 PMCID: PMC7836586 DOI: 10.1183/23120541.00448-2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/18/2020] [Indexed: 12/22/2022] Open
Abstract
Background Patient-oriented research approaches that reflect the needs and priorities of those most affected by health research outcomes improves translation of research findings into practice. Targeted therapies for cystic fibrosis (CF) are now a viable treatment option for some eligible individuals despite the heterogeneous patient-specific therapeutic response. This has necessitated development of a clinical tool that predicts treatment response for individual patients. Patient-derived mini-organs (organoids) have been at the forefront of this development. However, little is known about their acceptability in CF patients and members of the public. Methods We used a cross-sectional observational design to conduct an online survey in people with CF, their carers and community comparisons. Acceptability was examined in five domains: 1) willingness to use organoids, 2) perceived advantages and disadvantages of organoids, 3) acceptable out-of-pocket costs, 4) turnaround time and 5) source of tissue. Results In total, 188 participants completed the questionnaire, including adults with CF and parents of children with CF (90 (48%)), and adults without CF and parents of children without CF (98 (52%)). Use of organoids to guide treatment decisions in CF was acceptable to 86 (95%) CF participants and 98 (100%) community participants. The most important advantage was that organoids may improve treatment selection, improving the patient's quality of life and life expectancy. The most important disadvantage was that the organoid recommended treatment might be unavailable or too expensive. Conclusions These findings indicate acceptance of patient-derived organoids as a tool to predict treatment response by the majority of people surveyed. This may indicate successful future implementation into healthcare systems. The perspective regarding clinical use of patient-derived organoid models to enable cystic fibrosis personalised therapeutic decision-making of 188 participating adults surveyedhttps://bit.ly/30nWDJ7
Collapse
Affiliation(s)
- Laura K Fawcett
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney and Sydney Children's Hospital, Sydney, NSW, Australia.,Dept of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Claire E Wakefield
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Sheila Sivam
- Dept of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Peter G Middleton
- Cystic Fibrosis Unit, Dept of Respiratory and Sleep Medicine, Westmead Hospital, Sydney, NSW, Australia
| | - Peter Wark
- Centre for Healthy Lungs, University of Newcastle, Newcastle, NSW, Australia
| | - John Widger
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney and Sydney Children's Hospital, Sydney, NSW, Australia.,Dept of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney and Sydney Children's Hospital, Sydney, NSW, Australia.,Dept of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Shafagh A Waters
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.,Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney and Sydney Children's Hospital, Sydney, NSW, Australia.,Dept of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW, Australia
| |
Collapse
|
21
|
Strub MD, McCray, Jr. PB. Transcriptomic and Proteostasis Networks of CFTR and the Development of Small Molecule Modulators for the Treatment of Cystic Fibrosis Lung Disease. Genes (Basel) 2020; 11:genes11050546. [PMID: 32414011 PMCID: PMC7288469 DOI: 10.3390/genes11050546] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/18/2022] Open
Abstract
Cystic fibrosis (CF) is a lethal autosomal recessive disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The diversity of mutations and the multiple ways by which the protein is affected present challenges for therapeutic development. The observation that the Phe508del-CFTR mutant protein is temperature sensitive provided proof of principle that mutant CFTR could escape proteosomal degradation and retain partial function. Several specific protein interactors and quality control checkpoints encountered by CFTR during its proteostasis have been investigated for therapeutic purposes, but remain incompletely understood. Furthermore, pharmacological manipulation of many CFTR interactors has not been thoroughly investigated for the rescue of Phe508del-CFTR. However, high-throughput screening technologies helped identify several small molecule modulators that rescue CFTR from proteosomal degradation and restore partial function to the protein. Here, we discuss the current state of CFTR transcriptomic and biogenesis research and small molecule therapy development. We also review recent progress in CFTR proteostasis modulators and discuss how such treatments could complement current FDA-approved small molecules.
Collapse
Affiliation(s)
- Matthew D. Strub
- Interdisciplinary Graduate Program in Genetics, The University of Iowa, Iowa City, IA 52242, USA;
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
| | - Paul B. McCray, Jr.
- Interdisciplinary Graduate Program in Genetics, The University of Iowa, Iowa City, IA 52242, USA;
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence: ; Tel.: +1-(319)-335-6844
| |
Collapse
|
22
|
Jiang T, Henderson JM, Coote K, Cheng Y, Valley HC, Zhang XO, Wang Q, Rhym LH, Cao Y, Newby GA, Bihler H, Mense M, Weng Z, Anderson DG, McCaffrey AP, Liu DR, Xue W. Chemical modifications of adenine base editor mRNA and guide RNA expand its application scope. Nat Commun 2020; 11:1979. [PMID: 32332735 PMCID: PMC7181807 DOI: 10.1038/s41467-020-15892-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
CRISPR-Cas9-associated base editing is a promising tool to correct pathogenic single nucleotide mutations in research or therapeutic settings. Efficient base editing requires cellular exposure to levels of base editors that can be difficult to attain in hard-to-transfect cells or in vivo. Here we engineer a chemically modified mRNA-encoded adenine base editor that mediates robust editing at various cellular genomic sites together with moderately modified guide RNA, and show its therapeutic potential in correcting pathogenic single nucleotide mutations in cell and animal models of diseases. The optimized chemical modifications of adenine base editor mRNA and guide RNA expand the applicability of CRISPR-associated gene editing tools in vitro and in vivo.
Collapse
Affiliation(s)
- Tingting Jiang
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | | | - Kevin Coote
- Cystic Fibrosis Foundation, CFFT Lab, Lexington, MA, 02421, USA
| | - Yi Cheng
- Cystic Fibrosis Foundation, CFFT Lab, Lexington, MA, 02421, USA
| | | | - Xiao-Ou Zhang
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Qin Wang
- School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China
| | - Luke H Rhym
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yueying Cao
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Hermann Bihler
- Cystic Fibrosis Foundation, CFFT Lab, Lexington, MA, 02421, USA
| | - Martin Mense
- Cystic Fibrosis Foundation, CFFT Lab, Lexington, MA, 02421, USA
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
23
|
Kutney K, Donnola SB, Flask CA, Gubitosi-Klug R, O’Riordan M, McBennett K, Sferra TJ, Kaminski B. Lumacaftor/ivacaftor therapy is associated with reduced hepatic steatosis in cystic fibrosis patients. World J Hepatol 2019; 11:761-772. [PMID: 31966908 PMCID: PMC6960296 DOI: 10.4254/wjh.v11.i12.761] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/28/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatic steatosis is a common form of cystic fibrosis associated liver disease (CFLD) seen in an estimated 15%-60% of patients with cystic fibrosis (CF). The pathophysiology and health implications of hepatic steatosis in cystic fibrosis remain largely unknown. In the general population, hepatic steatosis is strongly associated with insulin resistance and type 2 diabetes. Cystic fibrosis related diabetes (CFRD) impacts 40%-50% of CF adults and is characterized by both insulin insufficiency and insulin resistance. We hypothesized that patients with CFRD would have higher levels of hepatic steatosis than cystic fibrosis patients without diabetes.
AIM To determine whether CFRD is associated with hepatic steatosis and to explore the impact of lumacaftor/ivacaftor therapy on hepatic steatosis in CF.
METHODS Thirty patients with CF were recruited from a tertiary care medical center for this cross-sectional study. Only pancreatic insufficient patients with CFRD or normal glucose tolerance (NGT) were included. Patients with established CFLD, end stage lung disease, or persistently elevated liver enzymes were excluded. Mean magnetic resonance imaging (MRI) proton density fat fraction (PDFF) was obtained for all participants. Clinical characteristics [age, sex, body mass index, percent predicted forced expiratory volume at 1 s (FEV1), lumacaftor/ivacaftor use] and blood chemistries were assessed for possible association with hepatic steatosis. Hepatic steatosis was defined as a mean MRI PDFF > 5%. Patients were grouped by diabetes status (CFRD, NGT) and cystic fibrosis transmembrane conductance regulator (CFTR) modulator use (lumacaftor/ivacaftor, no lumacaftor/ivacaftor) to determine between group differences. Continuous variables were analyzed with a Wilcoxon rank sum test and discrete variables with a Chi square test or Fisher’s exact test.
RESULTS Twenty subjects were included in the final analysis. The median age was 22.3 years (11.3-39.0) and median FEV1 was 77% (33%-105%). Twelve subjects had CFRD and 8 had NGT. Nine subjects were receiving lumacaftor/ivacaftor. The median PDFF was 3.0% (0.0%-21.0%). Six subjects (30%) had hepatic steatosis defined as PDFF > 5%. Hepatic fat fraction was significantly lower in patients receiving lumacaftor/ivacaftor (median, range) (2.0%, 0.0%-6.4%) than in patients not receiving lumacaftor/ivacaftor (4.1%, 2.7-21.0%), P = 0.002. Though patients with CFRD had lower PDFF (2.2%, 0.0%-14.5%) than patients with NGT (4.9%, 2.4-21.0%) this did not reach statistical significance, P = 0.06. No other clinical characteristic was strongly associated with hepatic steatosis.
CONCLUSION Use of the CFTR modulator lumacaftor/ivacaftor was associated with significantly lower hepatic steatosis. No association between CFRD and hepatic steatosis was found in this cohort.
Collapse
Affiliation(s)
- Katherine Kutney
- Department of Pediatric Endocrinology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Shannon B Donnola
- Department of Radiology Case Western Reserve University, Cleveland, OH 44106, United States
| | - Chris A Flask
- Department of Radiology Case Western Reserve University, Cleveland, OH 44106, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Rose Gubitosi-Klug
- Department of Pediatric Endocrinology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States
| | - MaryAnn O’Riordan
- Department of Pediatric Endocrinology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Kimberly McBennett
- Department of Pediatric Endocrinology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Thomas J Sferra
- Department of Pediatric Endocrinology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Beth Kaminski
- Department of Pediatric Endocrinology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States
| |
Collapse
|
24
|
Ivanov M, Ivanov M, Kasianov A, Rozhavskaya E, Musienko S, Baranova A, Mileyko V. Novel bioinformatics quality control metric for next-generation sequencing experiments in the clinical context. Nucleic Acids Res 2019; 47:e135. [PMID: 31511888 PMCID: PMC6868350 DOI: 10.1093/nar/gkz775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 07/22/2019] [Accepted: 08/29/2019] [Indexed: 11/23/2022] Open
Abstract
As the use of next-generation sequencing (NGS) for the Mendelian diseases diagnosis is expanding, the performance of this method has to be improved in order to achieve higher quality. Typically, performance measures are considered to be designed in the context of each application and, therefore, account for a spectrum of clinically relevant variants. We present EphaGen, a new computational methodology for bioinformatics quality control (QC). Given a single NGS dataset in BAM format and a pre-compiled VCF-file of targeted clinically relevant variants it associates this dataset with a single arbiter parameter. Intrinsically, EphaGen estimates the probability to miss any variant from the defined spectrum within a particular NGS dataset. Such performance measure virtually resembles the diagnostic sensitivity of given NGS dataset. Here we present case studies of the use of EphaGen in context of BRCA1/2 and CFTR sequencing in a series of 14 runs across 43 blood samples and 504 publically available NGS datasets. EphaGen is superior to conventional bioinformatics metrics such as coverage depth and coverage uniformity. We recommend using this software as a QC step in NGS studies in the clinical context. Availability: https://github.com/m4merg/EphaGen or https://hub.docker.com/r/m4merg/ephagen.
Collapse
Affiliation(s)
- Maxim Ivanov
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Mikhail Ivanov
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation
| | - Artem Kasianov
- Vavilov Institute of General Genetics, Moscow, Russian Federation
| | - Ekaterina Rozhavskaya
- Vavilov Institute of General Genetics, Moscow, Russian Federation.,Atlas Oncology Diagnostics, Ltd, Moscow, Russian Federation
| | | | - Ancha Baranova
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation.,Atlas Oncology Diagnostics, Ltd, Moscow, Russian Federation.,Research Centre for Medical Genetics, Moscow, Russian Federation.,School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| | - Vladislav Mileyko
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141700, Russian Federation.,Atlas Oncology Diagnostics, Ltd, Moscow, Russian Federation
| |
Collapse
|
25
|
Mention K, Santos L, Harrison PT. Gene and Base Editing as a Therapeutic Option for Cystic Fibrosis-Learning from Other Diseases. Genes (Basel) 2019; 10:E387. [PMID: 31117296 PMCID: PMC6562706 DOI: 10.3390/genes10050387] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/11/2019] [Accepted: 05/15/2019] [Indexed: 12/21/2022] Open
Abstract
Cystic fibrosis (CF) is a monogenic autosomal recessive disorder caused by mutations in the CFTR gene. There are at least 346 disease-causing variants in the CFTR gene, but effective small-molecule therapies exist for only ~10% of them. One option to treat all mutations is CFTR cDNA-based therapy, but clinical trials to date have only been able to stabilise rather than improve lung function disease in patients. While cDNA-based therapy is already a clinical reality for a number of diseases, some animal studies have clearly established that precision genome editing can be significantly more effective than cDNA addition. These observations have led to a number of gene-editing clinical trials for a small number of such genetic disorders. To date, gene-editing strategies to correct CFTR mutations have been conducted exclusively in cell models, with no in vivo gene-editing studies yet described. Here, we highlight some of the key breakthroughs in in vivo and ex vivo gene and base editing in animal models for other diseases and discuss what might be learned from these studies in the development of editing strategies that may be applied to cystic fibrosis as a potential therapeutic approach. There are many hurdles that need to be overcome, including the in vivo delivery of editing machinery or successful engraftment of ex vivo-edited cells, as well as minimising potential off-target effects. However, a successful proof-of-concept study for gene or base editing in one or more of the available CF animal models could pave the way towards a long-term therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Karen Mention
- Department of Physiology, University College Cork, Cork T12 K8AF, Ireland.
| | - Lúcia Santos
- Department of Physiology, University College Cork, Cork T12 K8AF, Ireland.
- University of Lisboa Faculty of Sciences, BioISI-Biosystems & Integrative Sciences Institute, 1749-016 Lisboa, Portugal.
| | - Patrick T Harrison
- Department of Physiology, University College Cork, Cork T12 K8AF, Ireland.
| |
Collapse
|
26
|
Sequencing as a first-line methodology for cystic fibrosis carrier screening. Genet Med 2019; 21:2569-2576. [PMID: 31036917 PMCID: PMC6831513 DOI: 10.1038/s41436-019-0525-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose Medical society guidelines recommend offering genotyping-based
cystic fibrosis (CF) carrier screening to pregnant women or women considering
pregnancy. We assessed the performance of sequencing-based CF screening relative
to genotyping, in terms of analytical validity, clinical validity, clinical
impact, and clinical utility. Methods Analytical validity was assessed using orthogonal confirmation and
reference samples. Clinical validity was evaluated using the CFTR2 database.
Clinical impact was assessed using ~100,000 screened patients. Three screening
strategies were compared: genotyping 23 guideline-recommended variants (“CF23”),
sequencing all coding bases in CFTR (“NGS”),
and sequencing with large copy-number variant (CNV) identification
(“NGS + CNV”). Clinical utility was determined via self-reported actions of
at-risk couples (ARCs). Results Analytical accuracy of NGS + CNV was 100% for SNVs, indels, and
CNVs; interpretive clinical specificity relative to CFTR2 was 99.5%. NGS + CNV
detected 58 ARCs, 18 of whom would have gone undetected with CF23 alone. Most
ARCs (89% screened preconceptionally, 56% prenatally) altered pregnancy
management, and no significant differences were observed between ARCs with or
without at least one non-CF23 variant. Conclusion Modern NGS and variant interpretation enable accurate
sequencing-based CF screening. Limiting screening to 23 variants does not
improve analytical validity, clinical validity, or clinical utility, but does
fail to detect approximately 30% (18/58) of ARCs.
Collapse
|
27
|
Guo X, Liu K, Liu Y, Situ Y, Tian X, Xu KF, Zhang X. Clinical and genetic characteristics of cystic fibrosis in CHINESE patients: a systemic review of reported cases. Orphanet J Rare Dis 2018; 13:224. [PMID: 30558651 PMCID: PMC6296146 DOI: 10.1186/s13023-018-0968-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023] Open
Abstract
Cystic fibrosis (CF) is a rare disease most commonly seen in Caucasians. Only a few Chinese CF patients have been described in literature, taking into account the large population of China. In this systematic review, we collected the clinical and genetic information of 71 Chinese CF patients based on all available data. Compared with Caucasians, Chinese CF patients often present atypical symptoms, mainly displaying symptoms of pulmonary infection with fewer digestive symptoms. An ethnicity-specific CFTR variant spectrum was also observed in CF patients of Chinese origin, with p.Gly970Asp as the most common mutation while p.Phe508del, the most common pathogenic mutation in CF patients of Caucasian origin, is rare, suggesting the necessity of a Chinese-specific CFTR variant screening panel. Besides, multiplex ligation-dependent probe amplification analysis should be routinely considered, especially for those with unidentified mutations. Potential under-diagnosis of CF in Chinese patients might be caused by a combination of atypical clinical features and genetic heterogeneity in Chinese CF patients, the inaccessibility of sweat and genetic testing facilities, and the one-child policy in China. With the approval of promising small molecule correctors and potentiators, molecular characterization of Chinese-specific CFTR mutations will help to realize more precise treatment for Chinese CF patients.
Collapse
Affiliation(s)
- Xiaobei Guo
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.,Emergency Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Keqiang Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Yaping Liu
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| | - Yusen Situ
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Xinlun Tian
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| | - Kai-Feng Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| |
Collapse
|
28
|
Pallin M. Cystic fibrosis vigilance in Arab countries: The role of genetic epidemiology. Respirology 2018; 24:93-94. [PMID: 30548951 DOI: 10.1111/resp.13461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Michael Pallin
- Monash Lung and Sleep, Monash Medical Centre, Melbourne, VIC, Australia
| |
Collapse
|
29
|
O'Neal WK, Knowles MR. Cystic Fibrosis Disease Modifiers: Complex Genetics Defines the Phenotypic Diversity in a Monogenic Disease. Annu Rev Genomics Hum Genet 2018; 19:201-222. [DOI: 10.1146/annurev-genom-083117-021329] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In many respects, genetic studies in cystic fibrosis (CF) serve as a paradigm for a human Mendelian genetic success story. From recognition of the condition as a heritable pathological entity to implementation of personalized treatments based on genetic findings, this multistep pathway of progress has focused on the genetic underpinnings of CF clinical disease. Along this path was the recognition that not all CFTR gene mutations produce the same disease and the recognition of the complex, multifactorial nature of CF genotype–phenotype relationships. The non- CFTR genetic components (gene modifiers) that contribute to variation in phenotype are the focus of this review. A multifaceted approach involving candidate gene studies, genome-wide association studies, and gene expression studies has revealed significant gene modifiers for multiple CF phenotypes. The bold challenges for the future are to integrate the findings into our understanding of CF pathogenesis and to use the knowledge to develop novel therapies.
Collapse
Affiliation(s)
- Wanda K. O'Neal
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;,
| | - Michael R. Knowles
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;,
| |
Collapse
|
30
|
Abstract
The majority of rare diseases affect children, most of whom have an underlying genetic cause for their condition. However, making a molecular diagnosis with current technologies and knowledge is often still a challenge. Paediatric genomics is an immature but rapidly evolving field that tackles this issue by incorporating next-generation sequencing technologies, especially whole-exome sequencing and whole-genome sequencing, into research and clinical workflows. This complex multidisciplinary approach, coupled with the increasing availability of population genetic variation data, has already resulted in an increased discovery rate of causative genes and in improved diagnosis of rare paediatric disease. Importantly, for affected families, a better understanding of the genetic basis of rare disease translates to more accurate prognosis, management, surveillance and genetic advice; stimulates research into new therapies; and enables provision of better support.
Collapse
|
31
|
Tordai H, Jakab K, Gyimesi G, András K, Brózik A, Sarkadi B, Hegedus T. ABCMdb reloaded: updates on mutations in ATP binding cassette proteins. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2017; 2017:3074791. [PMID: 28365738 PMCID: PMC5467578 DOI: 10.1093/database/bax023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/23/2017] [Indexed: 12/26/2022]
Abstract
ABC (ATP-Binding Cassette) proteins with altered function are responsible for numerous human diseases. To aid the selection of positions and amino acids for ABC structure/function studies we have generated a database, ABCMdb (Gyimesi et al., ABCMdb: a database for the comparative analysis of protein mutations in ABC transporters, and a potential framework for a general application. Hum Mutat 2012; 33:1547–1556.), with interactive tools. The database has been populated with mentions of mutations extracted from full text papers, alignments and structural models. In the new version of the database we aimed to collect the effect of mutations from databases including ClinVar. Because of the low number of available data, even in the case of the widely studied disease-causing ABC proteins, we also included the possible effects of mutations based on SNAP2 and PROVEAN predictions. To aid the interpretation of variations in non-coding regions, the database was supplemented with related DNA level information. Our results emphasize the importance of in silico predictions because of the sparse information available on variants and suggest that mutations at analogous positions in homologous ABC proteins have a strong predictive power for the effects of mutations. Our improved ABCMdb advances the design of both experimental studies and meta-analyses in order to understand drug interactions of ABC proteins and the effects of mutations on functional expression. Database URL:http://abcm2.hegelab.org
Collapse
Affiliation(s)
- Hedvig Tordai
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences and Department of Biophysics and Radiation Biology, Semmelweis University, Budapest 1094, Hungary
| | - Kristóf Jakab
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences and Department of Biophysics and Radiation Biology, Semmelweis University, Budapest 1094, Hungary
| | - Gergely Gyimesi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern 3012, Switzerland and
| | - Kinga András
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences and Department of Biophysics and Radiation Biology, Semmelweis University, Budapest 1094, Hungary
| | - Anna Brózik
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest 1117, Hungary
| | - Balázs Sarkadi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest 1117, Hungary
| | - Tamás Hegedus
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences and Department of Biophysics and Radiation Biology, Semmelweis University, Budapest 1094, Hungary
| |
Collapse
|
32
|
Sanz DJ, Hollywood JA, Scallan MF, Harrison PT. Cas9/gRNA targeted excision of cystic fibrosis-causing deep-intronic splicing mutations restores normal splicing of CFTR mRNA. PLoS One 2017; 12:e0184009. [PMID: 28863137 PMCID: PMC5581164 DOI: 10.1371/journal.pone.0184009] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/16/2017] [Indexed: 12/27/2022] Open
Abstract
Cystic Fibrosis is an autosomal recessive disorder caused by mutations in the CFTR gene. CRISPR mediated, template-dependent homology-directed gene editing has been used to correct the most common mutation, c.1521_1523delCTT / p.Phe508del (F508del) which affects ~70% of individuals, but the efficiency was relatively low. Here, we describe a high efficiency strategy for editing of three different rare CFTR mutations which together account for about 3% of individuals with Cystic Fibrosis. The mutations cause aberrant splicing of CFTR mRNA due to the creation of cryptic splice signals that result in the formation of pseudoexons containing premature stop codons c.1679+1634A>G (1811+1.6kbA>G) and c.3718-2477C>T (3849+10kbC>T), or an out-of-frame 5' extension to an existing exon c.3140-26A>G (3272-26A>G). We designed pairs of Cas9 guide RNAs to create targeted double-stranded breaks in CFTR either side of each mutation which resulted in high efficiency excision of the target genomic regions via non-homologous end-joining repair. When evaluated in a mini-gene splicing assay, we showed that targeted excision restored normal splicing for all three mutations. This approach could be used to correct aberrant splicing signals or remove disruptive transcription regulatory motifs caused by deep-intronic mutations in a range of other genetic disorders.
Collapse
Affiliation(s)
- David J. Sanz
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Jennifer A. Hollywood
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Patrick T. Harrison
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
33
|
De Wachter E, Thomas M, Wanyama SS, Seneca S, Malfroot A. What can the CF registry tell us about rare CFTR-mutations? A Belgian study. Orphanet J Rare Dis 2017; 12:142. [PMID: 28830496 PMCID: PMC5567473 DOI: 10.1186/s13023-017-0694-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/10/2017] [Indexed: 02/01/2023] Open
Abstract
Background CFTR2 provides clinical and functional information of the most common CFTR-mutations. Rare mutations (RMs) occur in only a few patients with limited reported clinical data. Their role in CF-disease liability is hardly documented. Methods Belgian CF-Registry 2013 data were analyzed to identify CF with at least 1 RM (CF+RM). Clinical data and sweat chloride of CF+RM were compared to CF-controls, carrying 2 class 1 to 3 mutations (CFclassic). Disease severity was compared between both groups. To avoid bias in the comparison, transplanted patients were excluded from each group. Results Seventy-seven CF+RM were identified (77/1183 = 6.5%). Sixty-four different RM were detected, of which 21 had not been previously reported. All RMs, corresponding to HGVS (Human Genome Variation Society) nomenclature, were listed in supplementary data. Seven transplanted CF+RM were excluded for further analysis. CF+RM had higher age at diagnosis [median (IQR)] [3.7 y (0.3–18.3) vs. 0.3y (0.1–2,0) (p < 0.0001)], lower sweat chloride [96 mmol/L (64–107) vs. 104 mmol/L (97–115) (p < 0.0001)], higher FEV1%pred [77%pred (58–96) vs. 68%pred (48–86) (p = 0.017)], were less frequently pancreatic insufficient [56% vs. 98% (p < 0.0001)], Pseudomonas aeruginosa colonized [24% vs. 44% (p = 0.0093)] and needed fewer IV antibiotics [36% vs. 51% (p = 0.041)] than CFclassic. However, a wide spectrum of disease severity was seen amongst CF+RM. Conclusions CF-patients with a RM cover 6.5% of the Belgian CF-population. Rare mutations can be found in severely ill patients, but more often in late diagnosed, pancreatic sufficient patients. Electronic supplementary material The online version of this article (doi:10.1186/s13023-017-0694-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- E De Wachter
- CF Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium.
| | - M Thomas
- Belgian CF Registry, Scientific Institute of Public Health (WIV-ISP), Brussels, Belgium
| | - S S Wanyama
- Belgian CF Registry, Scientific Institute of Public Health (WIV-ISP), Brussels, Belgium
| | - S Seneca
- Department for Reproduction and Genetics, Centre of Medical Genetics, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - A Malfroot
- CF Clinic, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| |
Collapse
|
34
|
Simhaev L, McCarty NA, Ford RC, Senderowitz H. Molecular Dynamics Flexible Fitting Simulations Identify New Models of the Closed State of the Cystic Fibrosis Transmembrane Conductance Regulator Protein. J Chem Inf Model 2017; 57:1932-1946. [DOI: 10.1021/acs.jcim.7b00091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Luba Simhaev
- Department
of Chemistry, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Nael A. McCarty
- Division
of Pulmonology, Allergy/Immunology, Cystic Fibrosis, and Sleep, Department
of Pediatrics, Emory + Children’s Center for Cystic Fibrosis
and Airways Disease Research, Emory University School of Medicine and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, Georgia 30322, United States
| | - Robert C. Ford
- Faculty
of Biology Medicine and Health, University of Manchester, Oxford
Road, Manchester, M13 9PL, U.K
| | | |
Collapse
|
35
|
Reisdorf WC, Chhugani N, Sanseau P, Agarwal P. Harnessing public domain data to discover and validate therapeutic targets. Expert Opin Drug Discov 2017; 12:687-693. [DOI: 10.1080/17460441.2017.1329296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- William C. Reisdorf
- Computational Biology, Target Sciences, GlaxoSmithKline R&D, King of Prussia, PA, USA
| | - Neha Chhugani
- Jacobs School of Engineering, University of California San Diego, Belle Mead, NJ, USA
| | - Philippe Sanseau
- Computational Biology, Target Sciences, GlaxoSmithKline R&D, Hertfordshire, UK
| | - Pankaj Agarwal
- Computational Biology, Target Sciences, GlaxoSmithKline R&D, King of Prussia, PA, USA
| |
Collapse
|
36
|
Genomic sequencing in cystic fibrosis newborn screening: what works best, two-tier predefined CFTR mutation panels or second-tier CFTR panel followed by third-tier sequencing? Genet Med 2017; 19:1159-1163. [PMID: 28471435 DOI: 10.1038/gim.2017.32] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 01/08/2023] Open
Abstract
PurposeThe purpose of this study was to model the performance of several known two-tier, predefined mutation panels and three-tier algorithms for cystic fibrosis (CF) screening utilizing the ethnically diverse California population.MethodsThe cystic fibrosis transmembrane conductance regulator (CFTR) mutations identified among the 317 CF cases in California screened between 12 August 2008 and 18 December 2012 were used to compare the expected CF detection rates for several two- and three-tier screening approaches, including the current California approach, which consists of a population-specific 40-mutation panel followed by third-tier sequencing when indicated.ResultsThe data show that the strategy of using third-tier sequencing improves CF detection following an initial elevated immunoreactive trypsinogen and detection of only one mutation on a second-tier panel.ConclusionIn a diverse population, the use of a second-tier panel followed by third-tier CFTR gene sequencing provides a better detection rate for CF, compared with the use of a second-tier approach alone, and is an effective way to minimize the referrals of CF carriers for sweat testing. Restricting screening to a second-tier testing to predefined mutation panels, even broad ones, results in some missed CF cases and demonstrates the limited utility of this approach in states that have diverse multiethnic populations.
Collapse
|
37
|
Hoskinson DC, Dubuc AM, Mason-Suares H. The current state of clinical interpretation of sequence variants. Curr Opin Genet Dev 2017; 42:33-39. [PMID: 28157586 DOI: 10.1016/j.gde.2017.01.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/20/2016] [Accepted: 01/09/2017] [Indexed: 01/19/2023]
Abstract
Accurate and consistent variant classification is required for Precision Medicine. But clinical variant classification remains in its infancy. While recent guidelines put forth jointly by the American College of Medical Genetics and Genomics (ACMG) and Association of Molecular Pathology (AMP) for the classification of Mendelian variants has advanced the field, the degree of subjectivity allowed by these guidelines can still lead to inconsistent classification across clinical molecular genetic laboratories. In addition, there are currently no such guidelines for somatic cancer variants, only published institutional practices. Additional variant classification guidelines, including disease- or gene-specific criteria, along with inter-laboratory data sharing is critical for accurate and consistent variant interpretation.
Collapse
Affiliation(s)
- Derick C Hoskinson
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, 65 Landsdowne Str., Cambridge, MA 02115 USA
| | - Adrian M Dubuc
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, 75 Francis Str., Boston, MA 02115 USA
| | - Heather Mason-Suares
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, 65 Landsdowne Str., Cambridge, MA 02115 USA; Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, 75 Francis Str., Boston, MA 02115 USA.
| |
Collapse
|
38
|
Collaco JM, Blackman SM, Raraigh KS, Corvol H, Rommens JM, Pace RG, Boelle PY, McGready J, Sosnay PR, Strug LJ, Knowles MR, Cutting GR. Sources of Variation in Sweat Chloride Measurements in Cystic Fibrosis. Am J Respir Crit Care Med 2016; 194:1375-1382. [PMID: 27258095 PMCID: PMC5148144 DOI: 10.1164/rccm.201603-0459oc] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/03/2016] [Indexed: 01/20/2023] Open
Abstract
RATIONALE Expanding the use of cystic fibrosis transmembrane conductance regulator (CFTR) potentiators and correctors for the treatment of cystic fibrosis (CF) requires precise and accurate biomarkers. Sweat chloride concentration provides an in vivo assessment of CFTR function, but it is unknown the degree to which CFTR mutations account for sweat chloride variation. OBJECTIVES To estimate potential sources of variation for sweat chloride measurements, including demographic factors, testing variability, recording biases, and CFTR genotype itself. METHODS A total of 2,639 sweat chloride measurements were obtained in 1,761 twins/siblings from the CF Twin-Sibling Study, French CF Modifier Gene Study, and Canadian Consortium for Genetic Studies. Variance component estimation was performed by nested mixed modeling. MEASUREMENTS AND MAIN RESULTS Across the tested CF population as a whole, CFTR gene mutations were found to be the primary determinant of sweat chloride variability (56.1% of variation) with contributions from variation over time (e.g., factors related to testing on different days; 13.8%), environmental factors (e.g., climate, family diet; 13.5%), other residual factors (e.g., test variability; 9.9%), and unique individual factors (e.g., modifier genes, unique exposures; 6.8%) (likelihood ratio test, P < 0.001). Twin analysis suggested that modifier genes did not play a significant role because the heritability estimate was negligible (H2 = 0; 95% confidence interval, 0.0-0.35). For an individual with CF, variation in sweat chloride was primarily caused by variation over time (58.1%) with the remainder attributable to residual/random factors (41.9%). CONCLUSIONS Variation in the CFTR gene is the predominant cause of sweat chloride variation; most of the non-CFTR variation is caused by testing variability and unique environmental factors. If test precision and accuracy can be improved, sweat chloride measurement could be a valuable biomarker for assessing response to therapies directed at mutant CFTR.
Collapse
Affiliation(s)
| | | | | | - Harriet Corvol
- Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, Paris, France
- Institut National de la Santé et la Recherche Médicale, Paris, France
| | - Johanna M. Rommens
- The Hospital for Sick Children and the University of Toronto, Toronto, Canada
| | - Rhonda G. Pace
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Pierre-Yves Boelle
- Institut National de la Santé et la Recherche Médicale, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie, Paris, France; and
- Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Paris, France
| | - John McGready
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | | | - Lisa J. Strug
- The Hospital for Sick Children and the University of Toronto, Toronto, Canada
| | - Michael R. Knowles
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | |
Collapse
|
39
|
Machogu E, Ren CL. Novel insights into the diagnostic and therapeutic challenges of the CFTR metabolic syndrome/CF screen positive indeterminate diagnosis. Pediatr Pulmonol 2016; 51:S45-S48. [PMID: 27662103 DOI: 10.1002/ppul.23478] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/01/2016] [Accepted: 05/02/2016] [Indexed: 11/09/2022]
Abstract
The growth of cystic fibrosis newborn screening (CF NBS) has led to an increased number of infants with a positive NBS test but inconclusive CF diagnostic testing. In the USA this condition is called CFTR related metabolic syndrome (CRMS), while in Europe the term CF screen positive, inconclusive diagnosis (CFSPID) is used. Recent advances in CF genetics and epidemiologic studies of CRMS/CFSPID have provided new insights into the prevalence and outcomes associated with this condition. Pediatr Pulmonol. 2016;51:S45-S48. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Evans Machogu
- Section of Pediatric Pulmonology, Allergy, and Sleep Medicine, Indiana University School of Medicine, Riley Hospital for Children, Indianapolis, Indiana
| | - Clement L Ren
- Section of Pediatric Pulmonology, Allergy, and Sleep Medicine, Indiana University School of Medicine, Riley Hospital for Children, Indianapolis, Indiana.
| |
Collapse
|
40
|
Hollywood JA, Lee CM, Scallan MF, Harrison PT. Analysis of gene repair tracts from Cas9/gRNA double-stranded breaks in the human CFTR gene. Sci Rep 2016; 6:32230. [PMID: 27557525 PMCID: PMC4997560 DOI: 10.1038/srep32230] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 08/04/2016] [Indexed: 12/18/2022] Open
Abstract
To maximise the efficiency of template-dependent gene editing, most studies describe programmable and/or RNA-guided endonucleases that make a double-stranded break at, or close to, the target sequence to be modified. The rationale for this design strategy is that most gene repair tracts will be very short. Here, we describe a CRISPR Cas9/gRNA selection-free strategy which uses deep sequencing to characterise repair tracts from a donor plasmid containing seven nucleotide differences across a 216 bp target region in the human CFTR gene. We found that 90% of the template-dependent repair tracts were >100 bp in length with equal numbers of uni-directional and bi-directional repair tracts. The occurrence of long repair tracts suggests that a single gRNA could be used with variants of the same template to create or correct specific mutations within a 200 bp range, the size of ~80% of human exons. The selection-free strategy used here also allowed detection of non-homologous end joining events in many of the homology-directed repair tracts. This indicates a need to modify the donor, possibly by silent changes in the PAM sequence, to prevent creation of a second double-stranded break in an allele that has already been correctly edited by homology-directed repair.
Collapse
Affiliation(s)
- Jennifer A. Hollywood
- Department of Physiology, University College Cork, Cork, Ireland
- Department of Microbiology, University College Cork, Cork, Ireland
| | - Ciaran M. Lee
- Department of Physiology, University College Cork, Cork, Ireland
- Department of Microbiology, University College Cork, Cork, Ireland
| | | | | |
Collapse
|
41
|
Harrison PT, Sanz DJ, Hollywood JA. Impact of gene editing on the study of cystic fibrosis. Hum Genet 2016; 135:983-92. [PMID: 27325484 DOI: 10.1007/s00439-016-1693-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/31/2016] [Indexed: 12/30/2022]
Abstract
Cystic fibrosis (CF) is a chronic and progressive autosomal recessive disorder of secretory epithelial cells, which causes obstructions in the lung airways and pancreatic ducts of 70,000 people worldwide (for recent review see Cutting Nat Rev Genet 16(1):45-56, 2015). The finding that mutations in the CFTR gene cause CF (Kerem et al. Science 245(4922):1073-1080, 1989; Riordan et al. Science 245(4922):1066-1073, 1989; Rommens et al. Science 245(4922):1059-1065, 1989), was hailed as the very happy middle of a story whose end is a cure for a fatal disease (Koshland Science 245(4922):1029, 1989). However, despite two licensed drugs (Ramsey et al. N Engl J Med 365(18):1663-1672, 2011; Wainwright et al. N Engl J Med 373(3):220-231, 2015), and a formal demonstration that repeated administration of CFTR cDNA to patients is safe and effects a modest but significant stabilisation of disease (Alton et al. Lancet Respir Med 3(9):684-691, 2015), we are still a long way from a cure, with many patients taking over 100 tablets per day, and a mean age at death of 28 years. The aim of this review is to discuss the impact on the study of CF of gene-editing techniques as they have developed over the last 30 years, up to and including the possibility of editing as a therapeutic approach.
Collapse
Affiliation(s)
| | | | - Jennifer A Hollywood
- University College Cork, Cork, Ireland.,The University of Auckland, Auckland, New Zealand
| |
Collapse
|
42
|
Dalgleish R. LSDBs and How They Have Evolved. Hum Mutat 2016; 37:532-9. [DOI: 10.1002/humu.22979] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/18/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Raymond Dalgleish
- Department of Genetics; University of Leicester; Leicester United Kingdom
| |
Collapse
|
43
|
Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S, Gu B, Hart J, Hoffman D, Hoover J, Jang W, Katz K, Ovetsky M, Riley G, Sethi A, Tully R, Villamarin-Salomon R, Rubinstein W, Maglott DR. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res 2015; 44:D862-8. [PMID: 26582918 PMCID: PMC4702865 DOI: 10.1093/nar/gkv1222] [Citation(s) in RCA: 1934] [Impact Index Per Article: 193.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/24/2015] [Indexed: 12/16/2022] Open
Abstract
ClinVar (https://www.ncbi.nlm.nih.gov/clinvar/) at the National Center for Biotechnology Information (NCBI) is a freely available archive for interpretations of clinical significance of variants for reported conditions. The database includes germline and somatic variants of any size, type or genomic location. Interpretations are submitted by clinical testing laboratories, research laboratories, locus-specific databases, OMIM®, GeneReviews™, UniProt, expert panels and practice guidelines. In NCBI's Variation submission portal, submitters upload batch submissions or use the Submission Wizard for single submissions. Each submitted interpretation is assigned an accession number prefixed with SCV. ClinVar staff review validation reports with data types such as HGVS (Human Genome Variation Society) expressions; however, clinical significance is reported directly from submitters. Interpretations are aggregated by variant-condition combination and assigned an accession number prefixed with RCV. Clinical significance is calculated for the aggregate record, indicating consensus or conflict in the submitted interpretations. ClinVar uses data standards, such as HGVS nomenclature for variants and MedGen identifiers for conditions. The data are available on the web as variant-specific views; the entire data set can be downloaded via ftp. Programmatic access for ClinVar records is available through NCBI's E-utilities. Future development includes providing a variant-centric XML archive and a web page for details of SCV submissions.
Collapse
Affiliation(s)
- Melissa J Landrum
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Jennifer M Lee
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Mark Benson
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Garth Brown
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Chen Chao
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Shanmuga Chitipiralla
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Baoshan Gu
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Jennifer Hart
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Douglas Hoffman
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Jeffrey Hoover
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Wonhee Jang
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Kenneth Katz
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Michael Ovetsky
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - George Riley
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Amanjeev Sethi
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Ray Tully
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Ricardo Villamarin-Salomon
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Wendy Rubinstein
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| | - Donna R Maglott
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20893, USA
| |
Collapse
|
44
|
Lenherr N, Lurà M, Trachsel D, Latzin P, Hammer J. Ivacaftor in a young boy with the rare gating mutation S549R--use of lung clearance index to track progress: a case report. BMC Pulm Med 2015; 15:123. [PMID: 26474553 PMCID: PMC4609097 DOI: 10.1186/s12890-015-0120-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 10/08/2015] [Indexed: 11/23/2022] Open
Abstract
Background Ivacaftor acts as a potentiator of the cystic fibrosis transmembrane conductance regulator (CFTR) and increases the transepithelial chloride transport of CFTR in 9 of 10 known gating mutations causing cystic fibrosis. S549R is a rare gating mutation considered to be less sensitive to potentiators than all other gating mutations. Case presentation We report our first experience with ivacaftor in an 8-year-old boy with the rare S549R gating mutation. Besides subjective clinical improvements, the sweat chloride level and the lung clearance index decreased impressively within a few weeks of treatment while forced expiratory volume in the first second values remained in normal range. Conclusion We emphasize the value of measuring small airway function by lung clearance index as an outcome measure for new interventions targeting the correction of the CFTR defect at an age before traditional lung function parameters start to deteriorate.
Collapse
Affiliation(s)
- Nina Lenherr
- Division of Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland.
| | - Marco Lurà
- Division of Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland.
| | - Daniel Trachsel
- Division of Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland.
| | - Philipp Latzin
- Division of Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland.
| | - Juerg Hammer
- Division of Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, CH-4056, Basel, Switzerland.
| |
Collapse
|
45
|
Abstract
The successful application of precision genomic medicine requires an understanding of how a person's genome can influence his or her disease phenotype and how medical therapies can provide personalized therapy to one's genotype. In this review, we highlight advances in precision genomic medicine in cystic fibrosis (CF), a classic autosomal recessive genetic disorder. We discuss genotype-phenotype correlations in CF, genetic and environmental modifiers of disease, and pharmacogenetic therapies that target specific genetic mutations thereby addressing the primary defect of cystic fibrosis.
Collapse
Affiliation(s)
- Eugene H Chang
- Department of Otolaryngology-Head and Neck Surgery, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Joseph Zabner
- Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
46
|
Griesenbach U, Alton EW. Recent advances in understanding and managing cystic fibrosis transmembrane conductance regulator dysfunction. F1000PRIME REPORTS 2015; 7:64. [PMID: 26097737 PMCID: PMC4447040 DOI: 10.12703/p7-64] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cystic fibrosis is the most common autosomal recessive genetic disease in Caucasians and has been extensively studied for many decades. The cystic fibrosis transmembrane conductance regulator gene was identified in 1989. It encodes a complex protein which has numerous cellular functions. Our understanding of cystic fibrosis pathophysiology and genetics is constantly expanding and being refined, leading to improved management of the disease and increased life expectancy in affected individuals.
Collapse
|
47
|
Targeted mutation screening panels expose systematic population bias in detection of cystic fibrosis risk. Genet Med 2015; 18:174-9. [PMID: 25880441 DOI: 10.1038/gim.2015.52] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/04/2015] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Carrier screening for mutations contributing to cystic fibrosis (CF) is typically accomplished with panels composed of variants that are clinically validated primarily in patients of European descent. This approach has created a static genetic and phenotypic profile for CF. An opportunity now exists to reevaluate the disease profile of CFTR at a global population level. METHODS CFTR allele and genotype frequencies were obtained from a nonpatient cohort with more than 60,000 unrelated personal genomes collected by the Exome Aggregation Consortium. Likely disease-contributing mutations were identified with the use of public database annotations and computational tools. RESULTS We identified 131 previously described and likely pathogenic variants and another 210 untested variants with a high probability of causing protein damage. None of the current genetic screening panels or existing CFTR mutation databases covered a majority of deleterious variants in any geographical population outside of Europe. CONCLUSIONS Both clinical annotation and mutation coverage by commercially available targeted screening panels for CF are strongly biased toward detection of reproductive risk in persons of European descent. South and East Asian populations are severely underrepresented, in part because of a definition of disease that preferences the phenotype associated with European-typical CFTR alleles.
Collapse
|
48
|
Loukas YL, Thodi G, Molou E, Georgiou V, Dotsikas Y, Schulpis KH. Clinical diagnostic Next-Generation sequencing: the case of CFTR carrier screening. Scandinavian Journal of Clinical and Laboratory Investigation 2015; 75:374-81. [PMID: 25874479 DOI: 10.3109/00365513.2015.1031689] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A 23-mutation panel for CFTR carrier screening is recommended to women of reproductive age by the American College of Obstetricians and Gynecologists. In the present study the optimized efficiency regarding the carrier rate of Next-Generation sequencing (NGS) technology is compared to the one of limited mutation detection panels. A total of 824 consequent cases were subjected to the commercial Cystic Fibrosis Genotyping Assay. Some 188 negative samples randomly selected from the initial group of probands were further subjected to an extended mutation panel characterized by 92% detection rate, as well as to massive parallel sequencing. Twenty-two probands subjected to the commercial assay proved to carry one mutation included in the ACOG panel (carrier rate 0.0267). The latter panels revealed the presence of mutations not included in the ACOG panel in four probands, resulting to an increase of carrier rate of 0.0106 in the case of in-house panel and an increase of rate of 0.0213 if NGS was used. The above data seem to support the implementation of NGS in the routine CFTR carrier screening.
Collapse
Affiliation(s)
- Yannis L Loukas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Athens , Athens
| | | | | | | | | | | |
Collapse
|
49
|
Incidental Finding of a Homozygous p.M348K Asymptomatic Italian Patient Confirms the Many Faces of Cystic Fibrosis. Case Rep Genet 2015; 2015:289627. [PMID: 25922769 PMCID: PMC4397493 DOI: 10.1155/2015/289627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/22/2015] [Indexed: 11/17/2022] Open
Abstract
Cystic fibrosis (CF; OMIM number 219700) is an autosomal recessive disease caused by mutations in theCFTR(cystic fibrosis transmembrane conductance regulator) gene, which results in abnormal viscous mucoid secretions in multiple organs and whose main clinical features are pancreatic insufficiency, chronic endobronchial infection, and male infertility. We report the case of a 47-year-old apparently normal male resulting in homozygosity for the mutation p.M348K from nonconsanguineous parents. The proband was screened using a standard panel of 70 different tested on NanoChip 400 platform. The massive parallel pyrosequencing on 454 JS machine allowed the second level analysis. The patient was firstly screened with two different platforms available in our laboratory, obtaining an ambiguous signal for the p.R347P mutation. For this reason we decided to clarify the discordant result ofCFTRstatus by Next Generation Sequencing (NGS) using 454 Junior instrument. The patient is resulted no carrier of the p.R347P mutation, but NGS highlighted a homozygous substitution from T>A at position 1043 in the coding region, causing an amino acid substitution from methionine to lysine (p.M348K). Casual finding of p.M348K homozygote mutation in an individual, without any feature of classical or nonclassical CF form, allowed us to confirm that p.M348K is a benign rare polymorphism.
Collapse
|
50
|
Martiniano SL, Hoppe JE, Sagel SD, Zemanick ET. Advances in the diagnosis and treatment of cystic fibrosis. Adv Pediatr 2014; 61:225-43. [PMID: 25037130 DOI: 10.1016/j.yapd.2014.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CF is a genetic, life-shortening, multisystem disease that is most commonly diagnosed through newborn screen performed in all 50 states in the United States. In the past, therapies for CF lung disease have primarily targeted the downstream effects of a dysfunctional CFTR protein. Newer CFTR modulator therapies, targeting the basic defect in CF, are available for a limited group of people with CF, and offer the hope of improved treatment options for many more people with CF in the near future. Best practice is directed by consensus clinical care guidelines from the CFF and is provided with a multidisciplinary approach by the team at the CF care center and the primary care office.
Collapse
Affiliation(s)
- Stacey L Martiniano
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Denver, 13123 East 16th Avenue, B-395, Aurora, CO 80045, USA.
| | - Jordana E Hoppe
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Denver, 13123 East 16th Avenue, B-395, Aurora, CO 80045, USA
| | - Scott D Sagel
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Denver, 13123 East 16th Avenue, B-395, Aurora, CO 80045, USA
| | - Edith T Zemanick
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado Denver, 13123 East 16th Avenue, B-395, Aurora, CO 80045, USA
| |
Collapse
|