1
|
Sund KL, Liu J, Lee J, Garbe J, Abdelhamed Z, Maag C, Hallinan B, Wu SW, Sperry E, Deshpande A, Stottmann R, Smolarek TA, Dyer LM, Hestand MS. Long-read sequencing and optical genome mapping identify causative gene disruptions in noncoding sequence in two patients with neurologic disease and known chromosome abnormalities. Am J Med Genet A 2024; 194:e63818. [PMID: 39041659 DOI: 10.1002/ajmg.a.63818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/12/2024] [Accepted: 07/07/2024] [Indexed: 07/24/2024]
Abstract
Despite advances in next generation sequencing (NGS), genetic diagnoses remain elusive for many patients with neurologic syndromes. Long-read sequencing (LRS) and optical genome mapping (OGM) technologies improve upon existing capabilities in the detection and interpretation of structural variation in repetitive DNA, on a single haplotype, while also providing enhanced breakpoint resolution. We performed LRS and OGM on two patients with known chromosomal rearrangements and inconclusive Sanger or NGS. The first patient, who had epilepsy and developmental delay, had a complex translocation between two chromosomes that included insertion and inversion events. The second patient, who had a movement disorder, had an inversion on a single chromosome disrupted by multiple smaller inversions and insertions. Sequence level resolution of the rearrangements identified pathogenic breaks in noncoding sequence in or near known disease-causing genes with relevant neurologic phenotypes (MBD5, NKX2-1). These specific variants have not been reported previously, but expected molecular consequences are consistent with previously reported cases. As the use of LRS and OGM technologies for clinical testing increases and data analyses become more standardized, these methods along with multiomic data to validate noncoding variation effects will improve diagnostic yield and increase the proportion of probands with detectable pathogenic variants for known genes implicated in neurogenetic disease.
Collapse
Affiliation(s)
- Kristen L Sund
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jie Liu
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Joyce Lee
- Bionano Genomics, San Diego, California, USA
| | - John Garbe
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zakia Abdelhamed
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Chelsey Maag
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Barbara Hallinan
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Steven W Wu
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ethan Sperry
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Archana Deshpande
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rolf Stottmann
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Teresa A Smolarek
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lisa M Dyer
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Matthew S Hestand
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Carmona-Hidalgo B, Herrera-Ramos E, Rodríguez-López R, Nou-Fontanet L, C. Moreno J, Blasco-Amaro JA, Léger J, Ortigoza-Escobar JD. Systematic review of thyroid function in NKX2-1-related disorders: Treatment and follow-up. PLoS One 2024; 19:e0309064. [PMID: 39466809 PMCID: PMC11515955 DOI: 10.1371/journal.pone.0309064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/05/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND NKX2-1, a crucial transcription factor in thyroid, lung, and brain development, is associated with rare disorders featuring thyroid dysfunction, neurological abnormalities, and respiratory symptoms. The primary challenge in managing NKX2-1-related disorders (NKX2-1-RD) is early diagnosis of the genetic defect and treating specific endocrine disorders. Levothyroxine (LT4) serves as the standard hypothyroidism treatment, with required dosages influenced by the severity of the individual's disorder, which varies widely among affected individuals. OBJECTIVES This systematic review aims to assess the effectiveness of LT4 treatment in NKX2-1-RD and explore optimal dosing strategies. The primary focus is on the challenges associated with the prompt diagnosis of genetic defects, rather than the established treatment protocols for individual endocrine failures. METHODS Adhering to PRISMA guidelines, the review includes 42 studies involving 110 genetically confirmed NKX2-1-RD patients with hypothyroidism. The study investigates congenital hypothyroidism as the most prevalent endocrine alteration, along with gestational and overt hypothyroidism. The administration of LT4 treatment, dosages, and patient responses are analyzed. RESULTS Among the findings, congenital hypothyroidism emerges as the predominant endocrine alteration in 41% of patients. Notably, LT4 treatment is administered in only 10% of cases, with a mean dose of 52 μg/day. The variability in initiation and dosage is likely influenced by the age at diagnosis. Positive responses, characterized by TSH adjustments within normal ranges, are observed in 11 monitored patients. CONCLUSIONS Early detection of congenital hypothyroidism is emphasized for timely LT4 initiation. Challenges in standardization are highlighted due to the variability in clinical manifestations and diagnostic procedures across NKX2-1-RD cases. While this review provides valuable insights into thyroid and pituitary disease treatment, limited details on LT4 treatment represent a significant study limitation. Key reporting points for future case studies are proposed to enhance the understanding and management of NKX2-1-RD hypothyroidism.
Collapse
Affiliation(s)
- Beatriz Carmona-Hidalgo
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | - Estefanía Herrera-Ramos
- Evaluation Unit (SESCS), Canary Islands Health Service (SCS), Santa Cruz de Tenerife, Spain
- Canary Islands Health Research Institute Foundation (FIISC), Las Palmas de Gran Canaria, Spain
- Network for Research on Chronicity, Primary Care, and Health Promotion (RICAPPS), Madrid, Spain
| | - Rocío Rodríguez-López
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | - Laia Nou-Fontanet
- Department of Child Neurology, Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - José C. Moreno
- Thyroid Molecular Laboratory, Institute for Medical and Molecular Genetics (INGEMM), Research Institute of Paz University Hospital (IdiPAZ), Madrid, Spain
- U-753 The Rare Diseases Networking Biomedical Research Centre (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Antonio Blasco-Amaro
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | - Juliane Léger
- European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Endocrinology-Diabetology Department, Assistance Publique-Hôpitaux de Paris, Robert Debre´ University Hospital, Reference Center for Growth and Development Endocrine Diseases, Paris, France
- Université Paris Cité, NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM 1141), Paris, France
| | - Juan Darío Ortigoza-Escobar
- Department of Child Neurology, Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Barcelona, Spain
- European Reference Network for Rare Neurological Diseases (ERN-RND), Tübingen, Germany
| | | |
Collapse
|
3
|
Ognean ML, Anciuc-Crauciuc M, Galiș R, Stepan AE, Stepan MD, Bănescu C, Grosu F, Kramer BW, Cucerea M. ABCA3 c.838C>T (p.Arg280Cys, R280C) and c.697C>T (p.Gln233Ter, Q233X, Q233*) as Causative Variants for RDS: A Family Case Study and Literature Review. Biomedicines 2024; 12:2390. [PMID: 39457702 PMCID: PMC11505159 DOI: 10.3390/biomedicines12102390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Respiratory distress syndrome (RDS) is the primary cause of respiratory failure in preterm infants, but it also affects 5-7% of term infants. Dysfunctions in pulmonary surfactant metabolism, resulting from mutations of the lung surfactant genes, are rare diseases, ranging from fatal neonatal RDS to interstitial lung disease, associated with increased morbidity and mortality. This study aims to clarify the clinical significance of ABCA3 variants found in a specific family case, as existing data in the literature are inconsistent. Material and Methods: A family case report was conducted; targeted panel genetic testing identified a variant of the SFTPB gene and two variants of ABCA3 genes. Comprehensive research involving a systematic review of PubMed, Google Scholar databases, and genome browsers was used to clarify the pathogenicity of the two ABCA3 variants found in the index patient. Advanced prediction tools were employed to assess the pathogenicity of the two ABCA3 variants, ensuring the validity and reliability of our findings. Results: The index case exhibited fatal neonatal RDS. Genetic testing revealed the presence of the SFTPB p.Val267Ile variant, which was not previously reported but is a benign variant based on family genetic testing and history. Additionally, two ABCA3 gene variants were identified: c.697C>T, not yet reported, and c.838C>T. These variants were found to affect ABCA3 protein function and were likely associated with neonatal RDS. Prediction tools and data from nine other cases in the literature supported this conclusion. Conclusions: Based on in silico predictors, an analysis of the presented family, and cases described in the literature, it is reasonable to consider reclassifying the two ABCA3 variants identified in the index case as pathogenic/pathogenic. Reclassification will improve genetic counseling accuracy and facilitate correct diagnosis.
Collapse
Affiliation(s)
- Maria Livia Ognean
- Faculty of Medicine, Lucian Blaga University, 550169 Sibiu, Romania; (M.L.O.)
- Neonatology Department, Clinical County Emergency Hospital, 550245 Sibiu, Romania
| | - Mădălina Anciuc-Crauciuc
- Department of Neonatology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540142 Targu Mures, Romania;
| | - Radu Galiș
- Department of Neonatology, Emergency County Hospital Bihor, Oradea University, 410087 Oradea, Romania;
- Department of Neonatology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Alex-Emilian Stepan
- Department of Pathology, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania
| | - Mioara Desdemona Stepan
- Department of Infant Care-Pediatrics-Neonatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Claudia Bănescu
- Genetic Department, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Gheorghe Marinescu Street No. 38, 540136 Targu Mures, Romania
| | - Florin Grosu
- Faculty of Medicine, Lucian Blaga University, 550169 Sibiu, Romania; (M.L.O.)
- Imaging Department, Lucian Blaga University, 550169 Sibiu, Romania
| | - Boris W. Kramer
- Department of Neonatology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Manuela Cucerea
- Department of Neonatology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology, 540142 Targu Mures, Romania;
| |
Collapse
|
4
|
Griese M, Kurland G, Cidon M, Deterding RR, Epaud R, Nathan N, Schwerk N, Warburton D, Weinman JP, Young LR, Deutsch GH. Pulmonary fibrosis may begin in infancy: from childhood to adult interstitial lung disease. Thorax 2024:thorax-2024-221772. [PMID: 39153860 DOI: 10.1136/thorax-2024-221772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/25/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Childhood interstitial lung disease (chILD) encompasses a group of rare heterogeneous respiratory conditions associated with significant morbidity and mortality. Reports suggest that many patients diagnosed with chILD continue to have potentially progressive or fibrosing disease into adulthood. Over the last decade, the spectrum of conditions within chILD has widened substantially, with the discovery of novel entities through advanced genetic testing. However, most evidence is often limited to small case series, with reports disseminated across an array of subspecialty, clinical and molecular journals. In particular, the frequency, management and outcome of paediatric pulmonary fibrosis is not well characterised, unlike in adults, where clear diagnosis and treatment guidelines are available. METHODS AND RESULTS This review assesses the current understanding of pulmonary fibrosis in chILD. Based on registry data, we have provisionally estimated the occurrence of fibrosis in various manifestations of chILD, with 47 different potentially fibrotic chILD entities identified. Published evidence for fibrosis in the spectrum of chILD entities is assessed, and current and future issues in management of pulmonary fibrosis in childhood, continuing into adulthood, are considered. CONCLUSIONS There is a need for improved knowledge of chILD among pulmonologists to optimise the transition of care from paediatric to adult facilities. Updated evidence-based guidelines are needed that incorporate recommendations for the diagnosis and management of immune-mediated disorders, as well as chILD in older children approaching adulthood.
Collapse
Affiliation(s)
- Matthias Griese
- German Center for Lung Research (DZL), University of Munich, LMU Hospital Department of Pediatrics at Dr von Hauner Children's Hospital, Munchen, Germany
| | - Geoffrey Kurland
- Division of Pediatric Pulmonology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Michal Cidon
- Children's Hospital Los Angeles, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Robin R Deterding
- Section of Pediatric Pulmonary and Sleep Medicine Department of Pediatrics, University of Colorado Denver, Denver, Colorado, USA
- Children's Hospital Colorado, Aurora, Colorado, USA
| | - Ralph Epaud
- Pediatric Pulmonology Department, Centre Hospitalier Intercommunal de Créteil; Centre des Maladies Respiratoires Rares (RESPIRARE®); University Paris Est Créteil, INSERM, IMRB, Créteil, France
| | - Nadia Nathan
- Paediatric Pulmonology Department and Reference Centre for Rare Lung Diseases RespiRare, Laboratory of Childhood Genetic Diseases, Inserm UMS_S933, Sorbonne Université and AP-HP, Hôpital Trousseau, Paris, France
| | - Nicolaus Schwerk
- Clinic for Paediatric Pneumology, Allergy and Neonatology, Hannover Medical School, German Center for Lung Research (DZL), Hannover, Germany
| | - David Warburton
- Children's Hospital Los Angeles, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Jason P Weinman
- Department of Radiology, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Lisa R Young
- Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Gail H Deutsch
- Department of Pathology, Seattle Children's Hospital and University of Washington Medical Center, Seattle, Washington, USA
| |
Collapse
|
5
|
Skwara J, Nowicki M, Sharif L, Milanowski Ł, Dulski J, Elert-Dobkowska E, Skrzypek K, Hoffman-Zacharska D, Koziorowski D, Sławek J. Differential diagnosis of Huntington's disease- neurological aspects of NKX2-1-related disorders. J Neural Transm (Vienna) 2024; 131:1013-1024. [PMID: 38916623 PMCID: PMC11365827 DOI: 10.1007/s00702-024-02800-3] [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: 03/15/2024] [Accepted: 06/12/2024] [Indexed: 06/26/2024]
Abstract
Benign hereditary chorea (BHC) is an inherited neurological disorder consisting of childhood-onset, nonprogressive chorea, generally without any other manifestations. In most reported cases, the inheritance of BHC is autosomal dominant but both incomplete penetrance and variable expressivity are observed and can be caused by NKX2-1 mutations. The spectrum contains choreoathetosis, congenital hypothyroidism, and neonatal respiratory distress syndrome. The neurological symptoms can be misdiagnosed as Huntington's disease (HD). The two Polish families were diagnosed with NKX2-1 gene mutations and a literature review concerning the NKX2-1-related disorders was conducted. All family members were examined by experienced movement disorders specialists. PubMed database was searched to obtain previously described NKX2-1 cases. Whole exome sequencing (WES) was performed in one proband (Family A) and direct NKX2-1 sequencing in the second (Family B). Two Polish families were diagnosed with NKX2-1 gene mutations (p.Trp208Leu and p.Cys117Alafs*8). In one family, the co-occurrence of HD was reported. Forty-nine publications were included in the literature review and symptoms of 195 patients with confirmed NKX2-1 mutation were analyzed. The most common symptoms were chorea and choreiform movements, and delayed motor milestones. The NKX2-1 mutation should always be considered as a potential diagnosis in families with chorea, even with a family history of HD. Lack of chorea does not exclude the NKX2-1-related disorders.
Collapse
Affiliation(s)
- Julia Skwara
- Student's Scientific Group, Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Nowicki
- Student's Scientific Group, Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Lucia Sharif
- Student's Scientific Group, Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Łukasz Milanowski
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Ludwika Kondratowicza 8, Warsaw, 03-242, Poland.
| | - Jarosław Dulski
- Department of Neurology and Stroke, St. Adalbert Hospital, Gdańsk, Poland
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Katarzyna Skrzypek
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | | | - Dariusz Koziorowski
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, Ludwika Kondratowicza 8, Warsaw, 03-242, Poland
| | - Jarosław Sławek
- Department of Neurology and Stroke, St. Adalbert Hospital, Gdańsk, Poland
- Division of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
6
|
Casey A, Fiorino EK, Wambach J. Innovations in Childhood Interstitial and Diffuse Lung Disease. Clin Chest Med 2024; 45:695-715. [PMID: 39069332 PMCID: PMC11366208 DOI: 10.1016/j.ccm.2024.04.002] [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] [Indexed: 07/30/2024]
Abstract
Children's interstitial and diffuse lung diseases (chILDs) are a heterogenous and diverse group of lung disorders presenting during childhood. Infants and children with chILD disorders present with respiratory signs and symptoms as well as diffuse lung imaging abnormalities. ChILD disorders are associated with significant health care resource utilization and high morbidity and mortality. The care of patients with chILD has been improved through multidisciplinary care, multicenter collaboration, and the establishment of patient research networks in the United Stated and abroad. This review details past and current innovations in the diagnosis and clinical care of children with chILD.
Collapse
Affiliation(s)
- Alicia Casey
- Department of Pediatrics, Division of Pulmonary Medicine, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA.
| | - Elizabeth K Fiorino
- Department of Science Education and Pediatrics, Donald and Barabara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Jennifer Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Carmona-Hidalgo B, Martín-Gómez C, Herrera-Ramos E, Rodríguez-López R, Fontanet LN, Moreno JC, Blasco-Amaro JA, Léger J, Dario-Ortigoza-Escobar J. Systematic review of thyroid function in NKX2-1-related disorders: Screening and diagnosis. PLoS One 2024; 19:e0303880. [PMID: 38990976 PMCID: PMC11238965 DOI: 10.1371/journal.pone.0303880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND NKX2-1-related disorders (NKX2-1-RD) are rare conditions affecting lung, thyroid, and brain development, primarily caused by pathogenic variants or deletions in the NKX2-1 gene. Congenital hypothyroidism (CH) is a common endocrine manifestation, leading to irreversible intellectual disability if left untreated. OBJECTIVES The aim was to evaluate the current evidence for the use of screening and diagnostic techniques for endocrine alterations in patients with NKX2-1-RD. METHODS This systematic review was reported following the PRISMA guidelines. Two separate research questions in PICO format were addressed to cover initial screening and diagnosis procedures for endocrine diseases in patients with NKX2-1-RD. Eligibility criteria focused on patients with genetic confirmation of the disease and hypothyroidism. Various databases were searched, and data were extracted and assessed independently by two reviewers. RESULTS Out of 1012 potentially relevant studies, 46 were included, for a total of 113 patients. CH was the most frequent endocrine alteration (45% of patients). Neonatal screening was reported in only 21% of patients based on blood TSH measurements. TSH thresholds varied widely across studies, making hypothyroidism detection ranges difficult to establish. Diagnostic tests using serum TSH were used to diagnose hypothyroidism or confirm its presence. 35% of patients were diagnosed at neonatal age, and 42% at adult age. Other hormonal dysfunctions identified due to clinical signs, such as anterior pituitary deficiencies, were detected later in life. Thyroid scintigraphy and ultrasonography allowed for the description of the thyroid gland in 30% of cases of hypothyroidism. Phenotypic variability was observed in individuals with the same variants, making genotype-phenotype correlations challenging. CONCLUSION This review highlights the need for standardized protocols in endocrine screening for NKX2-1-RD, emphasizing the importance of consistent methodology and hormone threshold levels. Variability in NKX2-1 gene variants further complicates diagnostic efforts. Future research should concentrate on optimizing early screening protocols and diagnostic strategies.
Collapse
Affiliation(s)
- Beatriz Carmona-Hidalgo
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | - Carmen Martín-Gómez
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
- Research Group HUM604: Lifestyle Development in the Life Cycle and Health Promotion, University of Huelva, Huelva, Spain
| | - Estefanía Herrera-Ramos
- Evaluation Unit of the Canary Islands Health Service (SESCS), Canary Islands Health Research Institute Foundation (FIISC), Santa Cruz of Tenerife, Spain
| | - Rocío Rodríguez-López
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | - Laia-Nou Fontanet
- Department of Child Neurology, Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - José C. Moreno
- Thyroid Molecular Laboratory, Institute for Medical and Molecular Genetics (INGEMM). Research Institute of Paz University Hospital (IdiPAZ), Madrid, Spain
- U-753 The Rare Diseases Networking Biomedical Research Centre (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Antonio Blasco-Amaro
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | - Juliane Léger
- European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Endocrinology-Diabetology Department, Assistance Publique-Hôpitaux de Paris, Robert Debre´ University Hospital, Reference Center for Growth and Development Endocrine Diseases, Paris, France
- Université Paris Cité, NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM 1141), Paris, France
| | - Juan Dario-Ortigoza-Escobar
- Department of Child Neurology, Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Barcelona, Spain
- European Reference Network for Rare Neurological Diseases (ERN-RND), Tübingen, Germany
| | | |
Collapse
|
8
|
Nou-Fontanet L, Nguyen QTR, Bachoud-Levi AC, Reinhard C, Ortigoza-Escobar JD. Insights from European Reference Network for rare neurological disorders study surveys on diagnosis, treatment, and management of NKX2-1-related disorders. Eur J Paediatr Neurol 2024; 51:110-117. [PMID: 38917695 DOI: 10.1016/j.ejpn.2024.06.007] [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: 09/21/2023] [Revised: 04/28/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND NKX2-1-related disorder (NKX2-1-RD) is a rare disease characterized by a triad of primary hypothyroidism, neonatal respiratory distress, and neurological features, including chorea. OBJECTIVE This study aimed to identify discrepancies in the management of NKX2-1-RD among European Union (EU) specialists. METHODS The ERN-RND Chorea & Huntington disease group designed a survey to conduct a cross-sectional multicenter study on the management of NKX2-1-RD. Descriptive analysis was performed, and total responses are presented for each item. RESULTS The study involved 23 experts from 13 EU countries with experience in evaluating hyperkinetic patients with NKX2-1-RD: 11 were adult specialists, and 12 were pediatric specialists. NKX2-1-RD diagnosis was made at different ages, with the most common initial symptoms being hypotonia and/or motor developmental delay (reported by 11 experts) and chorea (reported by 8 experts). Chorea involved various body parts and showed improvement as reported by 9 experts, stabilization by 12 experts, and worsening by 2 experts with age. The pharmacological treatment of chorea varied widely among the experts. Misdiagnosis was reported by 14 experts. NKX2-1 pathogenic variants or deletions were confirmed in >75 % of patients (reported by 12 experts). Pulmonary and endocrinology evaluations were requested by 7 and 12 experts, respectively. The management of psychiatric comorbidities also varied among the different experts. CONCLUSIONS This study highlights the need for a clinical practice guideline for the management of NKX2-1-RD to ensure that patients across the EU receive consistent and appropriate care. Such a guideline would benefit both doctors and healthcare practitioners.
Collapse
Affiliation(s)
- Laia Nou-Fontanet
- Department of Paediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Quang Tuan Rémy Nguyen
- Assistance Publique-Hôpitaux de Paris, Henri Mondor Hospital, National Center of Reference for Huntington's Disease, Créteil, France; Département D'Etudes Cognitives, École Normale Supérieure, PSL University, Paris, France; Univ Paris Est Creteil, INSERM, U955, Institut Mondor de Recherche Biomédicale, Laboratoire de Neuropsychologie Interventionnelle, Creteil, France
| | - Anne-Catherine Bachoud-Levi
- Assistance Publique-Hôpitaux de Paris, Henri Mondor Hospital, National Center of Reference for Huntington's Disease, Créteil, France; Département D'Etudes Cognitives, École Normale Supérieure, PSL University, Paris, France; Centre for Rare Diseases and Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Carola Reinhard
- Centre for Rare Diseases and Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Juan Darío Ortigoza-Escobar
- Department of Paediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain; Centre for Rare Diseases and Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, Calwerstr. 7, 72076, Tübingen, Germany; U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Barcelona, Spain.
| |
Collapse
|
9
|
Javadirad SM. NKX2-1 gene is targeted by H19 lncRNA and is found to be overexpressed in benign nodular goiter tissues. Braz J Otorhinolaryngol 2024; 90:101409. [PMID: 38537502 PMCID: PMC10987871 DOI: 10.1016/j.bjorl.2024.101409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/07/2024] [Accepted: 01/31/2024] [Indexed: 04/07/2024] Open
Abstract
OBJECTIVE Nodular goiter may increase the risk of thyroid cancer, but the genetic factors contributing to nodular goiter are not well understood. There is an overexpression of H19 lncRNA in goiter tissue and its target remains unknown. In this study, we attempted to identify a new target for H19 in the context of goiter development. METHODS Using interaction energy calculations, the interaction between NKX2-1 mRNA and H19 lncRNA was examined. Putative microRNAs were found at the H19 lncRNA target site with the highest affinity for NKX2-1. RNAseq data was analyzed to determine the tissue specificity of gene expression. Samples were taken from 18 goiter and 18 normal tissues during thyroidectomy. The expression of NKX2-1 was determined by RT-qPCR using specific primers. RESULTS The interaction between NKX2-1 and H19 was characterized by six local base-pairing connections, with a maximum energy of -20.56 kcal/moL. Specifically, the sequence that displayed the highest affinity for binding with H19 overlapped with the binding site of has-miR-1827 to NKX2-1. It was found that NKX2-1 is exclusively co-expressed with H19 in normal thyroid tissue. As compared to adjacent normal tissues, nodular goiter tissues have a significant overexpression of NKX2-1 (relative expression = 1.195, p = 0.038). CONCLUSION NKX2-1 has been identified as the putative target of H19 lncRNA, which is overexpressed in nodular goiter tissues significantly. LEVEL OF EVIDENCE: 4
Collapse
Affiliation(s)
- Seyed-Morteza Javadirad
- University of Isfahan, Faculty of Biological Science and Technology, Department of Cell and Molecular Biology and Microbiology, Isfahan, Iran.
| |
Collapse
|
10
|
Villafuerte B, Carrasco-López C, Herranz A, Garzón L, Simón R, Natera-de-Benito D, Alikhani P, Tenorio J, García-Santiago F, Solis M, Del-Pozo Á, Lapunzina P, Ortigoza-Escobar JD, Santisteban P, Moreno JC. A Novel Missense Variant in the NKX2-1 Homeodomain Prevents Transcriptional Rescue by TAZ. Thyroid 2024; 34:942-948. [PMID: 38757609 DOI: 10.1089/thy.2023.0593] [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] [Indexed: 05/18/2024]
Abstract
Background: Brain-lung-thyroid syndrome (BLTS) is caused by NKX2-1 haploinsufficiency, resulting in chorea/choreoathetosis, respiratory problems, and hypothyroidism. Genes interacting with NKX2-1 mutants influence its phenotypic variability. We report a novel NKX2-1 missense variant and the modifier function of TAZ/WWTR1 in BLTS. Methods: A child with BLTS underwent next-generation sequencing panel testing for thyroid disorders. His family was genotyped for NKX2-1 variants and screened for germline mosaicism. Mutant NKX2-1 was generated, and transactivation assays were performed on three NKX2-1 target gene promoters. DNA binding capacity and protein-protein interaction were analyzed. Results: The patient had severe BLTS and carried a novel missense variant c.632A>G (p.N211S) in NKX2-1, which failed to bind to specific DNA promoters, reducing their transactivation. TAZ cotransfection did not significantly increase transcription of these genes, although the variant retained its ability to bind to TAZ. Conclusions: We identify a novel pathogenic NKX2-1 variant that causes severe BLTS and is inherited through germline mosaicism. The mutant lacks DNA-binding capacity, impairing transactivation and suggesting that NKX2-1 binding to DNA is essential for TAZ-mediated transcriptional rescue.
Collapse
Affiliation(s)
- Beatriz Villafuerte
- Thyroid Molecular Laboratory, Institute for Medical and Molecular Genetics (INGEMM), La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain
| | - Carlos Carrasco-López
- "Sols-Morreale" Biomedical Research Institute, Higher Council for Scientific Research (CSIC), Autonomous University of Madrid, Ciberonc, Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Amanda Herranz
- Thyroid Molecular Laboratory, Institute for Medical and Molecular Genetics (INGEMM), La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain
| | - Lucía Garzón
- Pediatric Endocrinology Unit, Pediatrics Department, 12 de Octubre University Hospital, Madrid, Spain
| | - Rogelio Simón
- Pediatric Neurology Unit, Pediatrics Department, 12 de Octubre University Hospital, Madrid, Spain
| | - Daniel Natera-de-Benito
- Neuromuscular Diseases Unit, Pediatric Neurology, Sant Joan de Déu Hospital, Barcelona, Spain
| | - Pouya Alikhani
- Thyroid Molecular Laboratory, Institute for Medical and Molecular Genetics (INGEMM), La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain
| | - Jair Tenorio
- Institute for Medical and Molecular Genetics (INGEMM), IdiPAZ, Center for Biomedical Research on the Rare Diseases Network (CIBERER), Carlos III Health Institute (ISCIII), ITHACA-European Reference Network, La Paz University Hospital, Madrid, Spain
| | - Fe García-Santiago
- Cytogenetics Section, Institute for Medical and Molecular Genetics (INGEMM), Center for Biomedical Research on the Rare Diseases Network (CIBERER), Carlos III Health Institute (ISCIII), La Paz University Hospital, Madrid, Spain
| | - Mario Solis
- Bioinformatics Section, Institute for Medical and Molecular Genetics (INGEMM), IdiPAZ, Center for Biomedical Research on the Rare Diseases Network (CIBERER), Carlos III Health Institute (ISCIII), La Paz University Hospital, Madrid, Spain
| | - Ángela Del-Pozo
- Bioinformatics Section, Institute for Medical and Molecular Genetics (INGEMM), IdiPAZ, Center for Biomedical Research on the Rare Diseases Network (CIBERER), Carlos III Health Institute (ISCIII), La Paz University Hospital, Madrid, Spain
| | - Pablo Lapunzina
- Institute for Medical and Molecular Genetics (INGEMM), IdiPAZ, Center for Biomedical Research on the Rare Diseases Network (CIBERER), Carlos III Health Institute (ISCIII), ITHACA-European Reference Network, La Paz University Hospital, Madrid, Spain
| | | | - Pilar Santisteban
- "Sols-Morreale" Biomedical Research Institute, Higher Council for Scientific Research (CSIC), Autonomous University of Madrid, Ciberonc, Carlos III Health Institute (ISCIII), Madrid, Spain
| | - José C Moreno
- Thyroid Molecular Laboratory, Institute for Medical and Molecular Genetics (INGEMM), La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain
- Unit 735, Center for Biomedical Research on the Rare Diseases Network (CIBERER), Carlos III Health Institute (ISCIII), Madrid, Spain
| |
Collapse
|
11
|
Uehara E, Hori N, Tanase-Nakao K, Akiba K, Sueoka H, Matsubara K, Narumi S. Congenital Hypothyroidism with Thyroid in situ: A Case Report with NKX2-1 and DUOX2 Hypomorphic Variants. Horm Res Paediatr 2024:1-7. [PMID: 38636486 DOI: 10.1159/000538895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
INTRODUCTION NK2 homeobox 1 (NKX2-1) encodes a transcription factor, NKX2-1, that is expressed in the thyroid gland, lung, and brain. Dual oxidase 2 (DUOX2) encodes an enzyme which generates hydrogen peroxide and is involved in the thyroid hormone synthesis. Cases of congenital hypothyroidism (CH) with dyshormonogenesis showing two or more genetic variants are increasingly reported. We describe the first case of transient dyshormonogenesis who had experimentally verified a loss-of-function NKX2-1 variant and DUOX2 variants. CASE PRESENTATION The proband was a 15-year-old female patient with CH who was diagnosed in the frame of newborn screening for CH. She had a mildly elevated serum TSH level (14.56 mU/L), a low free thyroxine level (0.87 ng/dL), and a high thyroglobulin (Tg) level (>800 ng/mL). Ultrasonography revealed goiter. She was followed clinically without levothyroxine treatment and showed normal growth and development. She had slightly high Tg levels throughout the clinical course. Next-generation sequencing-based genetic analysis revealed that the patient was heterozygous for an NKX2-1 variant (p.Ile228Ser), a nonsense DUOX2 variant (p.[Lys530*;His678Arg]), and a functional DUOX2 polymorphism (p.His678Arg). NKX2-1 p.Ile228Ser showed about 50% reduced residual activity on the Tg promoter. CONCLUSION A partial loss-of-function NKX2-1 variant with a monoallelic nonsense DUOX2 variant and a DUOX2 functional polymorphism can cause transient CH with high serum Tg levels.
Collapse
Affiliation(s)
- Erika Uehara
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan,
- Department of Advanced Pediatric Medicine, Tohoku University School of Medicine, Tokyo, Japan,
| | - Naoaki Hori
- Department of Pediatrics, Ota Memorial Hospital, Ota, Japan
| | - Kanako Tanase-Nakao
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kazuhisa Akiba
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
- Division of Endocrinology and Metabolism, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Hidefumi Sueoka
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keiko Matsubara
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
- Division of Diversity Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Wang H, Jiang G, Dai D, Hong D, Zhou W, Qian L. Functional characterization of two novel NKX2-1 frameshift variants that cause pulmonary surfactant dysfunction. Pediatr Res 2024; 95:744-751. [PMID: 37935886 DOI: 10.1038/s41390-023-02882-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND We aim to report two unrelated patients with pulmonary surfactant dysfunction (PSD) that carried two novel NKX2-1 frameshift variants, and evaluated the impact of these variants in vitro. METHODS We enrolled children with PSD and NKX2-1 variants, and collected their clinical information and follow-up data. We constructed wild-type (WT) and variant NKX2-1 plasmids and transfected them into A549 and HEK293T cells. The functional characterization of variants was then evaluated by qRT-PCR, western blot, immunofluorescence, electrophoretic mobility shift assay, and dual-luciferase reporter assay. RESULTS Two novel heterozygous frameshift variants of NKX2-1, i.e., c.705delC (Gly236Alafs*29) and c.313_316 dup (Asn106Lysfs*304), were identified in children from two unrelated families. We discerned attenuated mRNA and protein expression in the Asn106Lysfs*304 variant, and reduced DNA -binding as well as transcriptional activation capabilities in both variants. While the Asn106Lysfs*304 variant lost its synergistic interactions with PAX8 and TAZ, the Gly236Alafs*29 variant partially retained its residual transcriptional activation capabilities and synergistic interactions with PAX8 and TAZ. CONCLUSIONS We reported on two children with two novel NKX2-1 frameshift variants. In vitro experiments revealed that the two frameshift variants have common and different mechanisms based on the loss or conservation of domains, which partially explained the phenotypical heterogeneity. IMPACT Pulmonary surfactant dysfunction is a rare heterogeneous disease that exhibits a great burden on children's quality of life. We reported two novel NKX2-1 frameshift variants carried by two children with different clinical phenotypes, thus broadening our knowledge base of gene variations and phenotypes in NKX2-1. We performed an in vitro study and uncovered different pathogenic mechanisms underlying the actions of two novel variants, and thereby partially explained the mechanisms of phenotypical heterogeneity caused by NKX2-1 variants.
Collapse
Affiliation(s)
- Huixian Wang
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Gaoli Jiang
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Dan Dai
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Da Hong
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Weitao Zhou
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Liling Qian
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China.
- Fujian Provincial Key Laboratory of Neonatal Diseases, Fujian, China.
| |
Collapse
|
13
|
Machida O, Sakamoto H, Yamamoto KS, Hasegawa Y, Nii S, Okada H, Nishikawa K, Sumimoto SI, Nishi E, Okamoto N, Yamamoto T. Haploinsufficiency of NKX2-1 is likely to contribute to developmental delay involving 14q13 microdeletions. Intractable Rare Dis Res 2024; 13:36-41. [PMID: 38404736 PMCID: PMC10883847 DOI: 10.5582/irdr.2023.01119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 02/27/2024] Open
Abstract
Nucleotide variations or deletions in the NK2 homeobox 1 gene (NKX2-1), located at 14q13.3, lead to symptoms associated with the brain, lungs, and thyroid, and the combination of these phenotypes is clinically recognized as the brain-lung-thyroid syndrome. Many types of nucleotide variants of NKX2-1 have been identified, and phenotypic variability has been reported. Chromosomal deletions involving NKX2-1 have also been reported; however, phenotypic differences between patients with nucleotide variants of NKX2-1 and patients with chromosomal deletions involving NKX2-1 have not been well established. Recently, we identified seven patients with 14q13 microdeletions involving the NKX2-1. Most patients exhibited developmental delay. This inquiry arises regarding the potential existence of haploinsufficiency effects beyond those attributed to NKX2-1 within the 14q13 microdeletion. However, a literature review has shown that developmental delay is not rare in patients with nucleotide alterations in NKX2-1. Rather, motor function impairment may have affected the total developmental assessment, and the haploinsufficiency of genes contiguous to NKX2-1 is unlikely to contribute to developmental delay.
Collapse
Affiliation(s)
- Osamu Machida
- Division of Gene Medicine, Graduate School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
- Department of Pediatrics, Tokyo Women's Medical University, Tokyo, Japan
| | - Haruko Sakamoto
- Department of Pediatrics, Japanese Red Cross Osaka Hospital, Osaka, Japan
- Otemae Rehabilitation Center for Children, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Keiko Shimojima Yamamoto
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Yuiko Hasegawa
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Satoi Nii
- Otemae Rehabilitation Center for Children, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Hidenori Okada
- Department of Pediatrics, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Kazuki Nishikawa
- Department of Pediatrics, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Shin-Ichi Sumimoto
- Otemae Rehabilitation Center for Children, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Eriko Nishi
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Toshiyuki Yamamoto
- Division of Gene Medicine, Graduate School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
- Department of Transfusion Medicine and Cell Processing, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
14
|
Huth EA, Zhao X, Owen N, Luna PN, Vogel I, Dorf ILH, Joss S, Clayton-Smith J, Parker MJ, Louw JJ, Gewillig M, Breckpot J, Kraus A, Sasaki E, Kini U, Burgess T, Tan TY, Armstrong R, Neas K, Ferrero GB, Brusco A, Kerstjens-Frederikse WS, Rankin J, Helvaty LR, Landis BJ, Geddes GC, McBride KL, Ware SM, Shaw CA, Lalani SR, Rosenfeld JA, Scott DA. Clinical exome sequencing efficacy and phenotypic expansions involving anomalous pulmonary venous return. Eur J Hum Genet 2023; 31:1430-1439. [PMID: 37673932 PMCID: PMC10689790 DOI: 10.1038/s41431-023-01451-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/08/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
Anomalous pulmonary venous return (APVR) frequently occurs with other congenital heart defects (CHDs) or extra-cardiac anomalies. While some genetic causes have been identified, the optimal approach to genetic testing in individuals with APVR remains uncertain, and the etiology of most cases of APVR is unclear. Here, we analyzed molecular data from 49 individuals to determine the diagnostic yield of clinical exome sequencing (ES) for non-isolated APVR. A definitive or probable diagnosis was made for 8 of those individuals yielding a diagnostic efficacy rate of 16.3%. We then analyzed molecular data from 62 individuals with APVR accrued from three databases to identify novel APVR genes. Based on data from this analysis, published case reports, mouse models, and/or similarity to known APVR genes as revealed by a machine learning algorithm, we identified 3 genes-EFTUD2, NAA15, and NKX2-1-for which there is sufficient evidence to support phenotypic expansion to include APVR. We also provide evidence that 3 recurrent copy number variants contribute to the development of APVR: proximal 1q21.1 microdeletions involving RBM8A and PDZK1, recurrent BP1-BP2 15q11.2 deletions, and central 22q11.2 deletions involving CRKL. Our results suggest that ES and chromosomal microarray analysis (or genome sequencing) should be considered for individuals with non-isolated APVR for whom a genetic etiology has not been identified, and that genetic testing to identify an independent genetic etiology of APVR is not warranted in individuals with EFTUD2-, NAA15-, and NKX2-1-related disorders.
Collapse
Affiliation(s)
- Emily A Huth
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaonan Zhao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Baylor Genetics, Houston, TX, USA
| | - Nichole Owen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Baylor Genetics, Houston, TX, USA
| | - Pamela N Luna
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ida Vogel
- Department of Clinical Medicine, Aarhus University, 8000, Aarhus, C, Denmark
| | - Inger L H Dorf
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Shelagh Joss
- West of Scotland Genomics Service, Queen Elizabeth University Hospital, Glasgow, G51 4TF, UK
| | - Jill Clayton-Smith
- Manchester Centre For Genomic Medicine, Manchester University Hospitals, Manchester, M13 9WL, UK
- University of Manchester, Manchester, M13 9PL, UK
| | - Michael J Parker
- Department of Clinical Genetics, Sheffield, Children's Hospital, UK
| | - Jacoba J Louw
- Pediatric Cardiology Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marc Gewillig
- Department of Cardiovascular Sciences KU Leuven, Leuven, Belgium
- Pediatric Cardiology University Hospitals Leuven, Leuven, Belgium
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospitals Leuven, Catholic University, Leuven, Belgium
| | - Alison Kraus
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds, LS7 4SA, UK
| | - Erina Sasaki
- Oxford Centre for Genomic Medicine, Oxford University Hospital, Oxford, OX3 7HE, UK
| | - Usha Kini
- Oxford Centre for Genomic Medicine, Oxford University Hospital, Oxford, OX3 7HE, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Trent Burgess
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Tiong Y Tan
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Ruth Armstrong
- East Anglian Medical Genetics Service, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | | | - Giovanni B Ferrero
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, Torino, Italy
- Città della Salute e della Scienza University Hospital, Torino, Italy
| | | | | | | | | | - Gabrielle C Geddes
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kim L McBride
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Stephanie M Ware
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Chad A Shaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Nou-Fontanet L, Martín-Gómez C, Isabel-Gómez R, Bachoud-Lévi AC, Zorzi G, Capuano A, Blasco-Amaro JA, Ortigoza-Escobar JD. Systematic review of drug therapy for chorea in NXK2-1-related disorders: Efficacy and safety evidence from case studies and series. Eur J Neurol 2023; 30:3928-3948. [PMID: 37694681 DOI: 10.1111/ene.16038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/25/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND The NKX2-1-related disorders (NKX2-1-RD) is a rare disorder characterized by choreiform movements along with respiratory and endocrine abnormalities. The European Reference Network of Rare Neurological Disorders funded by the European Commission conducted a systematic review to assess drug treatment of chorea in NKX2-1-RD, aiming to provide clinical recommendations for its management. METHODS A systematic pairwise review using various databases, including MEDLINE, Embase, Cochrane, CINAHL, and PsycInfo, was conducted. The review included patients diagnosed with chorea and NKX2-1-RD genetic diagnosis, drug therapy as intervention, no comparator, and outcomes of chorea improvement and adverse events. The methodological quality of the studies was assessed, and the study protocol was registered in PROSPERO. RESULTS Of the 1417 studies examined, 28 studies met the selection criteria, consisting of 68 patients. The studies reported 22 different treatments for chorea, including carbidopa/levodopa, tetrabenazine, clonazepam, methylphenidate, carbamazepine, topiramate, trihexyphenidyl, haloperidol, propranolol, risperidone, and valproate. No clinical improvements were observed with carbidopa/levodopa, tetrabenazine, or clonazepam, and various adverse effects were reported. However, most patients treated with methylphenidate experienced improvements in chorea and reported only a few negative effects. The quality of evidence was determined to be low. CONCLUSIONS The management of chorea in individuals with NKX2-1-RD presents significant heterogeneity and lack of clarity. While the available evidence suggests that methylphenidate may be effective in improving chorea symptoms, the findings should be interpreted with caution due to the limitations of the studies reviewed. Nonetheless, more rigorous and comprehensive studies are necessary to provide sufficient evidence for clinical recommendations.
Collapse
Affiliation(s)
- Laia Nou-Fontanet
- Department of Paediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Carmen Martín-Gómez
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health ("Fundación Progreso y Salud"-"FPS"), Seville, Spain
- Research Group HUM604: Lifestyle Development in the Life Cycle and Health Promotion, University of Huelva, Huelva, Spain
| | - Rebeca Isabel-Gómez
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health ("Fundación Progreso y Salud"-"FPS"), Seville, Spain
| | - Anne-Catherine Bachoud-Lévi
- Assistance Publique-Hôpitaux de Paris, Henri Mondor Hospital, National Center of Reference for Huntington's Disease, Créteil, France
- Département d'Etudes Cognitives, École Normale Supérieure, PSL University, Paris, France
- European Reference Network for Rare Neurological Diseases (ERN-RND), Tübingen, Germany
| | - Giovanna Zorzi
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alessandro Capuano
- Movement Disorders Clinic, Department of Neurosciences, Bambino Gesù Children's Hospital, Rome, Italy
| | - Juan Antonio Blasco-Amaro
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health ("Fundación Progreso y Salud"-"FPS"), Seville, Spain
| | - Juan Darío Ortigoza-Escobar
- Department of Paediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
- European Reference Network for Rare Neurological Diseases (ERN-RND), Tübingen, Germany
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
16
|
Peers de Nieuwburgh M, Wambach JA, Griese M, Danhaive O. Towards personalized therapies for genetic disorders of surfactant dysfunction. Semin Fetal Neonatal Med 2023; 28:101500. [PMID: 38036307 PMCID: PMC10753445 DOI: 10.1016/j.siny.2023.101500] [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] [Indexed: 12/02/2023]
Abstract
Genetic disorders of surfactant dysfunction are a rare cause of chronic, progressive or refractory respiratory failure in term and preterm infants. This review explores genetic mechanisms underpinning surfactant dysfunction, highlighting specific surfactant-associated genes including SFTPB, SFTPC, ABCA3, and NKX2.1. Pathogenic variants in these genes contribute to a range of clinical presentations and courses, from neonatal hypoxemic respiratory failure to childhood interstitial lung disease and even adult-onset pulmonary fibrosis. This review emphasizes the importance of early recognition, thorough phenotype assessment, and assessment of variant functionality as essential prerequisites for treatments including lung transplantation. We explore emerging treatment options, including personalized pharmacological approaches and gene therapy strategies. In conclusion, this comprehensive review offers valuable insights into the pathogenic mechanisms of genetic disorders of surfactant dysfunction, genetic fundamentals, available and emerging therapeutic options, and underscores the need for further research to develop personalized therapies for affected infants and children.
Collapse
Affiliation(s)
- Maureen Peers de Nieuwburgh
- Division of Neonatology, Department of Pediatrics, St-Luc University Hospital, Catholic University of Louvain, Brussels, Belgium.
| | - Jennifer A Wambach
- Washington University School of Medicine/St. Louis Children's Hospital, One Children's Place, St. Louis, Missouri, USA.
| | - Matthias Griese
- Pediatric Pulmonology, Dr von Hauner Children's Hospital, University-Hospital, German Center for Lung Research (DZL), Munich, Germany.
| | - Olivier Danhaive
- Division of Neonatology, Department of Pediatrics, St-Luc University Hospital, Catholic University of Louvain, Brussels, Belgium; Division of Neonatology, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Mio C, Baldan F, Damante G. NK2 homeobox gene cluster: Functions and roles in human diseases. Genes Dis 2023; 10:2038-2048. [PMID: 37492711 PMCID: PMC10363584 DOI: 10.1016/j.gendis.2022.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/15/2022] [Accepted: 10/01/2022] [Indexed: 07/27/2023] Open
Abstract
NK2 genes (NKX2 gene cluster in humans) encode for homeodomain-containing transcription factors that are conserved along the phylogeny. According to the most detailed classifications, vertebrate NKX2 genes are classified into two distinct families, NK2.1 and NK2.2. The former is constituted by NKX2-1 and NKX2-4 genes, which are homologous to the Drosophila scro gene; the latter includes NKX2-2 and NKX2-8 genes, which are homologous to the Drosophila vnd gene. Conservation of these genes is not only related to molecular structure and expression, but also to biological functions. In Drosophila and vertebrates, NK2 genes share roles in the development of ventral regions of the central nervous system. In vertebrates, NKX2 genes have a relevant role in the development of several other organs such as the thyroid, lung, and pancreas. Loss-of-function mutations in NKX2-1 and NKX2-2 are the monogenic cause of the brain-lung-thyroid syndrome and neonatal diabetes, respectively. Alterations in NKX2-4 and NKX2-8 genes may play a role in multifactorial diseases, autism spectrum disorder, and neural tube defects, respectively. NKX2-1, NKX2-2, and NKX2-8 are expressed in various cancer types as either oncogenes or tumor suppressor genes. Several data indicate that evaluation of their expression in tumors has diagnostic and/or prognostic value.
Collapse
Affiliation(s)
- Catia Mio
- Dipartimento di Area Medica, Università degli Studi di Udine, Udine 33100, Italy
| | - Federica Baldan
- Istituto di Genetica Medica, Azienda Sanitaria Universitaria Friuli Centrale, Udine 33100, Italy
| | - Giuseppe Damante
- Dipartimento di Area Medica, Università degli Studi di Udine, Udine 33100, Italy
- Istituto di Genetica Medica, Azienda Sanitaria Universitaria Friuli Centrale, Udine 33100, Italy
| |
Collapse
|
18
|
Soliani L, Alcalá San Martín A, Balsells S, Hernando‐Davalillo C, Ortigoza‐Escobar JD. Chromosome Microarray Analysis for the Investigation of Deletions in Pediatric Movement Disorders: A Systematic Review of the Literature. Mov Disord Clin Pract 2023; 10:547-557. [PMID: 37070051 PMCID: PMC10105116 DOI: 10.1002/mdc3.13711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/19/2023] [Accepted: 02/19/2023] [Indexed: 03/06/2023] Open
Abstract
Background Chromosome microarray analysis (CMA) can detect copy number variants (CNV) beyond the resolution of standard G-banded karyotyping. De novo or inherited microdeletions may cause autosomal dominant movement disorders. Objectives The purpose of this study was to analyze the clinical characteristics, associated features, and genetic information of children with deletions in known genes that cause movement disorders and to make recommendations regarding the diagnostic application of CMA. Methods Clinical cases published in English were identified in scientific databases (PubMed, ClinVar, and DECIPHER) from January 1998 to July 2019 following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Cases with deletions or microdeletions greater than 300 kb were selected. Information collected included age, sex, movement disorders, associated features, and the size and location of the deletion. Duplications or microduplications were not included. Results A total of 18.097 records were reviewed, and 171 individuals were identified. Ataxia (30.4%), stereotypies (23.9%), and dystonia (21%) were the most common movement disorders. A total of 16% of the patients demonstrated more than one movement disorder. The most common associated features were intellectual disability or developmental delay (78.9%) and facial dysmorphism (57.8%). The majority (77.7%) of microdeletions were smaller than 5 Mb. We find no correlation between movement disorders, their associated features, and the size of microdeletions. Conclusions Our results support the use of CMA as an investigational test in children with movement disorders. As the majority of identified articles were case reports and small case series (low quality), future efforts should focus on larger prospective studies to examine the causation of microdeletions in pediatric movement disorders.
Collapse
Affiliation(s)
- Luca Soliani
- IRCCS Istituto delle Scienze Neurologiche di Bologna UOC Neuropsichiatria dell'età PediatricaBolognaItaly
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC) Università di BolognaBolognaItaly
| | - Adrián Alcalá San Martín
- Department of Genetic and Molecular Medicine and Pediatric Institute of Rare DiseasesHospital Sant Joan de Déu BarcelonaBarcelonaSpain
| | - Sol Balsells
- Department of StatisticsInstitut de Recerca Sant Joan de DéuBarcelonaSpain
| | - Cristina Hernando‐Davalillo
- Department of Genetic and Molecular Medicine and Pediatric Institute of Rare DiseasesHospital Sant Joan de Déu BarcelonaBarcelonaSpain
| | - Juan Darío Ortigoza‐Escobar
- U‐703 Centre for Biomedical Research on Rare Diseases (CIBER‐ER)Instituto de Salud Carlos IIIBarcelonaSpain
- Movement Disorders Unit, Pediatric Neurology Department, Institut de RecercaHospital Sant Joan de Déu BarcelonaBarcelonaSpain
- European Reference Network for Rare Neurological Diseases (ERN‐RND)BarcelonaSpain
| |
Collapse
|
19
|
Borie R, Kannengiesser C, Antoniou K, Bonella F, Crestani B, Fabre A, Froidure A, Galvin L, Griese M, Grutters JC, Molina-Molina M, Poletti V, Prasse A, Renzoni E, van der Smagt J, van Moorsel CHM. European Respiratory Society statement on familial pulmonary fibrosis. Eur Respir J 2023; 61:13993003.01383-2022. [PMID: 36549714 DOI: 10.1183/13993003.01383-2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Genetic predisposition to pulmonary fibrosis has been confirmed by the discovery of several gene mutations that cause pulmonary fibrosis. Although genetic sequencing of familial pulmonary fibrosis (FPF) cases is embedded in routine clinical practice in several countries, many centres have yet to incorporate genetic sequencing within interstitial lung disease (ILD) services and proper international consensus has not yet been established. An international and multidisciplinary expert Task Force (pulmonologists, geneticists, paediatrician, pathologist, genetic counsellor, patient representative and librarian) reviewed the literature between 1945 and 2022, and reached consensus for all of the following questions: 1) Which patients may benefit from genetic sequencing and clinical counselling? 2) What is known of the natural history of FPF? 3) Which genes are usually tested? 4) What is the evidence for telomere length measurement? 5) What is the role of common genetic variants (polymorphisms) in the diagnostic workup? 6) What are the optimal treatment options for FPF? 7) Which family members are eligible for genetic sequencing? 8) Which clinical screening and follow-up parameters may be considered in family members? Through a robust review of the literature, the Task Force offers a statement on genetic sequencing, clinical management and screening of patients with FPF and their relatives. This proposal may serve as a basis for a prospective evaluation and future international recommendations.
Collapse
Affiliation(s)
- Raphael Borie
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | | | - Katerina Antoniou
- Laboratory of Molecular and Cellular Pneumonology, Department of Respiratory Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik, University Hospital, University of Essen, European Reference Network (ERN)-LUNG, ILD Core Network, Essen, Germany
| | - Bruno Crestani
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | - Aurélie Fabre
- Department of Histopathology, St Vincent's University Hospital and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Antoine Froidure
- Pulmonology Department, Cliniques Universitaires Saint-Luc and Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Liam Galvin
- European Pulmonary Fibrosis Federation, Blackrock, Ireland
| | - Matthias Griese
- Dr von Haunersches Kinderspital, University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Jan C Grutters
- ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
- Division of Heart and Lungs, UMC Utrecht, Utrecht, The Netherlands
| | - Maria Molina-Molina
- Interstitial Lung Disease Unit, Respiratory Department, University Hospital of Bellvitge, IDIBELL, Hospitalet de Llobregat (Barcelona), CIBERES, Barcelona, Spain
| | - Venerino Poletti
- Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
- Department of Experimental, Diagnostics and Speciality Medicine, University of Bologna, Bologna, Italy
| | - Antje Prasse
- Department of Pulmonology, Hannover Medical School, German Center for Lung Research (DZL), BREATH, Hannover, Germany
- Fraunhofer ITEM, Hannover, Germany
| | - Elisabetta Renzoni
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jasper van der Smagt
- Division of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
20
|
Martín-Gómez C, Ortigoza-Escobar JD, Nou-Fontanet L, Molina-Linde JM, Bachoud-Lévi AC, Léger J, Blasco-Amaro JA. Exploring the values, preferences, and information needs of patients with NKX2-1-related disorders: A qualitative study protocol. PLoS One 2023; 18:e0281573. [PMID: 36758103 PMCID: PMC9910649 DOI: 10.1371/journal.pone.0281573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/13/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND NKX2-1-related disorders have a prevalence of 1:500,000 and are therefore considered a rare condition according to the European Commission's definition. The European Reference Network of Rare Neurological Disorders is developing the first clinical practice guideline on the management of this condition, with the support of the Andalusian Health Technology Assessment Area, Endo-ERN, ERN-Lung and Imegen, within the framework of the ERNs Guidelines programme (DG SANTE/2018/B3/030). Within the scope of this programme, it becomes necessary to explore the patient perspective in order to include it in the ongoing clinical practice guideline and accompanying patient information booklet. METHODS AND ANALYSIS This study will use qualitative methods to explore the values, preferences and information needs of patient with NKX2-1-related disorders and their caregivers. Participants will come from a variety of countries throughout Europe. One focus group and four semi-structured interviews will be conducted. Pairs will analyse the data using Grounded Theory. The Andalusian Regional Ministry of Health's Ethics Coordinating Committee for Biomedical Research (Sevilla, Andalucía, Spain) has approved this study protocol (29/03/2022). DISCUSSION This is the first study to explore the values, preferences, and information needs of patients with NKX2-1-related disorders. The proposed study's findings will contribute to the generation of useful knowledge that will provide guidance to improve the care given to patients with the studied condition. While this study will provide valuable insights into the perspectives of patients with NKX2-1-related disorders, the findings are unlikely to be generalizable to patients with other conditions.
Collapse
Affiliation(s)
- Carmen Martín-Gómez
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
- Research Group HUM604: Lifestyle Development in the Life Cycle and Health Promotion, University of Huelva, Huelva, Spain
- * E-mail:
| | - Juan Dario Ortigoza-Escobar
- Department of Child Neurology, Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, Barcelona, Spain
- European Reference Network for Rare Neurological Diseases (ERN-RND), Tübingen, Germany
| | - Laia Nou-Fontanet
- Department of Paediatric Neurology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Juan M. Molina-Linde
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | - Anne-Catherine Bachoud-Lévi
- European Reference Network for Rare Neurological Diseases (ERN-RND), Tübingen, Germany
- Assistance Publique-Hôpitaux de Paris, Henri Mondor Hospital, National Center of Reference for Huntington’s Disease, Créteil, France
- Département d’Etudes Cognitives, École Normale Supérieure, PSL University, Paris, France
| | - Juliane Léger
- University Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Equipe NeuroPsychologie Interventionnelle, Creteil, France
- Endocrinology-Diabetology Department, Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Reference Center for Growth and Development Endocrine Diseases, Paris, France
- Université Paris Cité, NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM 1141), Paris, France
- European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
| | - Juan Antonio Blasco-Amaro
- Health Technology Assessment Area-AETSA, Andalusian Public Foundation for Progress and Health (“Fundación Progreso y Salud”–“FPS”), Seville, Spain
| | | |
Collapse
|
21
|
Magrinelli F, Rocca C, Simone R, Zenezini Chiozzi R, Jaunmuktane Z, Mencacci NE, Tinazzi M, Jayawant S, Nemeth AH, Demidov G, Houlden H, Bhatia KP. Detection and Characterization of a De Novo Alu Retrotransposition Event Causing NKX2-1-Related Disorder. Mov Disord 2023; 38:347-353. [PMID: 36420574 DOI: 10.1002/mds.29280] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/17/2022] [Accepted: 11/06/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Heterozygous NKX2-1 loss-of-function variants cause combinations of hyperkinetic movement disorders (MDs, particularly childhood-onset chorea), pulmonary dysfunction, and hypothyroidism. Mobile element insertions (MEIs) are potential disease-causing structural variants whose detection in routine diagnostics remains challenging. OBJECTIVE To establish the molecular diagnosis of two first-degree relatives with clinically suspected NKX2-1-related disorder who had negative NKX2-1 Sanger (SS), whole-exome (WES), and whole-genome (WGS) sequencing. METHODS The proband's WES was analyzed for MEIs. A candidate MEI in NKX2-1 underwent optimized SS after plasmid cloning. Functional studies exploring NKX2-1 haploinsufficiency at RNA and protein levels were performed. RESULTS A 347-bp AluYa5 insertion with a 65-bp poly-A tail followed by a 16-bp duplication of the pre-insertion wild-type sequence in exon 3 of NKX2-1 (ENST00000354822.7:c.556_557insAlu541_556dup) segregated with the disease phenotype. CONCLUSIONS We identified a de novo exonic AluYa5 insertion causing NKX2-1-related disorder in SS/WES/WGS-negative cases, suggesting that MEI analysis of short-read sequencing data or targeted long-read sequencing could unmask the molecular diagnosis of unsolved MD cases. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Francesca Magrinelli
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Clarissa Rocca
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Roberto Simone
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | | | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Niccolò E Mencacci
- Ken and Ruth Davee Department of Neurology and Simpson Querrey Center for Neurogenetics, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michele Tinazzi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Sandeep Jayawant
- Paediatric Neurology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Andrea H Nemeth
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Genomic Medicine, Oxford University Hospitals National Health Service Foundation Trust, Oxford, United Kingdom
| | - German Demidov
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
22
|
Sutton RM, Bittar HT, Sullivan DI, Silva AG, Bahudhanapati H, Parikh AH, Zhang Y, Gibson K, McDyer JF, Kass DJ, Alder JK. Rare surfactant-related variants in familial and sporadic pulmonary fibrosis. Hum Mutat 2022; 43:2091-2101. [PMID: 36135709 PMCID: PMC9771972 DOI: 10.1002/humu.24476] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/01/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023]
Abstract
The role of constitutional genetic defects in idiopathic pulmonary fibrosis (IPF) is increasingly appreciated. Monogenic disorders associated with IPF affect two pathways: telomere maintenance, accounting for approximately 10% of all patients with IPF, and surfactant biology, responsible for 1%-3% of cases and often co-occurring with lung cancer. We examined the prevalence of rare variants in five surfactant-related genes, SFTPA1, SFPTA2, SFTPC, ABCA3, and NKX2-1, that were previously linked to lung disease in whole genome sequencing data from 431 patients with IPF. We identified functionally deleterious rare variants in SFTPA2 with a prevalence of 1.3% in individuals with and without a family history of IPF. All individuals had no personal history of lung cancer, but substantial bronchiolar metaplasia was noted on lung explants and biopsies. Five patients had novel missense variants in NKX2-1, but the contribution to disease is unclear. In general, patients were younger and had longer telomeres compared with the majority of patients with IPF suggesting that these features may be useful for identifying this subset of patients in the clinic. These data suggest that SFTPA2 variants may be more common in unselected IPF cohorts and may manifest in the absence of personal/family history of lung cancer or IPF.
Collapse
Affiliation(s)
- Rachel M Sutton
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Humberto Trejo Bittar
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Daniel I Sullivan
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Agustin Gil Silva
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Harinath Bahudhanapati
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Anishka H Parikh
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Yingze Zhang
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin Gibson
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - John F McDyer
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Daniel J Kass
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| | - Jonathan K Alder
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
23
|
Moran C, Schoenmakers N, Visser WE, Schoenmakers E, Agostini M, Chatterjee K. Genetic disorders of thyroid development, hormone biosynthesis and signalling. Clin Endocrinol (Oxf) 2022; 97:502-514. [PMID: 35999191 PMCID: PMC9544560 DOI: 10.1111/cen.14817] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/24/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
Development and differentiation of the thyroid gland is directed by expression of specific transcription factors in the thyroid follicular cell which mediates hormone biosynthesis. Membrane transporters are rate-limiting for cellular entry of thyroid hormones (TH) (T4 and T3) into some tissues, with selenocysteine-containing, deiodinase enzymes (DIO1 and DIO2) converting T4 to the biologically active hormone T3. TH regulate expression of target genes via hormone-inducible nuclear receptors (TRα and TRβ) to exert their physiological effects. Primary congenital hypothyroidism (CH) due to thyroid dysgenesis may be mediated by defects in thyroid transcription factors or impaired thyroid stimulating hormone receptor function. Dyshormonogenic CH is usually due to mutations in genes mediating thyroidal iodide transport, organification or iodotyrosine synthesis and recycling. Disorders of TH signalling encompass conditions due to defects in membrane TH transporters, impaired hormone metabolism due to deficiency of deiodinases and syndromes of Resistance to thyroid hormone due to pathogenic variants in either TRα or TRβ. Here, we review the genetic basis, pathogenesis and clinical features of congenital, dysgenetic or dyshormonogenic hypothyroidism and disorders of TH transport, metabolism and action.
Collapse
Affiliation(s)
- Carla Moran
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Present address:
Beacon Hospital and School of MedicineUniversity CollegeDublinIreland
| | - Nadia Schoenmakers
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - W. Edward Visser
- Department of Internal MedicineErasmus Medical Center, Academic Center for Thyroid DiseasesRotterdamThe Netherlands
| | - Erik Schoenmakers
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - Maura Agostini
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - Krishna Chatterjee
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| |
Collapse
|
24
|
Heidari MM, Madani Manshadi SA, Eshghi AR, Talebi F, Khatami M, Bragança J, Ordooei M, Chamani R, Ghasemi F. Mutational and bioinformatics analysis of the NKX2.1 gene in a cohort of Iranian pediatric patients with congenital hypothyroidism (CH). Physiol Int 2022; 109:261-277. [DOI: https:/doi.org/10.1556/2060.2022.00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Abstract
Congenital hypothyroidism (CH) occurs with a relatively alarming prevalence in infants, and if not diagnosed and treated in time, it can have devastating consequences for the development of the nervous system. CH is associated with genetic changes in several genes that encode transcription factors responsible for thyroid development, including mutations in the NK2 homeobox 1 (NKX2.1) gene, which encodes the thyroid transcription factor-1 (TTF-1). Although CH is frequently observed in pediatric populations, there is still a limited understanding of the genetic factors and molecular mechanisms contributing to this disease. The sequence of the NKX2.1 gene was investigated in 75 pediatric patients with CH by polymerase chain reaction (PCR), single-stranded conformation polymorphism (SSCP), and direct DNA sequencing. Four missense heterozygous variations were identified in exon 3 of the NKX2.1 gene, including three novel missense variations, namely c.708A>G, p.Gln202Arg; c.713T>G, p.Tyr204Asp; c.833T>G, p.Tyr244Asp, and a previously reported variant rs781133468 (c.772C>G, p.His223Gln). Importantly, these variations occur in highly conserved residues of the TTF-1 DNA-binding domain and were predicted by bioinformatics analysis to alter the protein structure, with a probable alteration in the protein function. These results indicate that nucleotide changes in the NKX2.1 gene may contribute to CH pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - José Bragança
- Faculty of Medicine and Biomedical Sciences, Algarve Biomedical Centre Research Institute, University of Algarve, Faro, Portugal
| | - Mahtab Ordooei
- Department of Pediatrics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | |
Collapse
|
25
|
Heidari MM, Madani Manshadi SA, Eshghi AR, Talebi F, Khatami M, Bragança J, Ordooei M, Chamani R, Ghasemi F. Mutational and bioinformatics analysis of the NKX2.1 gene in a cohort of Iranian pediatric patients with congenital hypothyroidism (CH). Physiol Int 2022; 109:261-277. [DOI: 10.1556/2060.2022.00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/01/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022]
Abstract
Abstract
Congenital hypothyroidism (CH) occurs with a relatively alarming prevalence in infants, and if not diagnosed and treated in time, it can have devastating consequences for the development of the nervous system. CH is associated with genetic changes in several genes that encode transcription factors responsible for thyroid development, including mutations in the NK2 homeobox 1 (NKX2.1) gene, which encodes the thyroid transcription factor-1 (TTF-1). Although CH is frequently observed in pediatric populations, there is still a limited understanding of the genetic factors and molecular mechanisms contributing to this disease. The sequence of the NKX2.1 gene was investigated in 75 pediatric patients with CH by polymerase chain reaction (PCR), single-stranded conformation polymorphism (SSCP), and direct DNA sequencing. Four missense heterozygous variations were identified in exon 3 of the NKX2.1 gene, including three novel missense variations, namely c.708A>G, p.Gln202Arg; c.713T>G, p.Tyr204Asp; c.833T>G, p.Tyr244Asp, and a previously reported variant rs781133468 (c.772C>G, p.His223Gln). Importantly, these variations occur in highly conserved residues of the TTF-1 DNA-binding domain and were predicted by bioinformatics analysis to alter the protein structure, with a probable alteration in the protein function. These results indicate that nucleotide changes in the NKX2.1 gene may contribute to CH pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - José Bragança
- Faculty of Medicine and Biomedical Sciences, Algarve Biomedical Centre Research Institute, University of Algarve, Faro, Portugal
| | - Mahtab Ordooei
- Department of Pediatrics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | |
Collapse
|
26
|
Leung RF, George AM, Roussel EM, Faux MC, Wigle JT, Eisenstat DD. Genetic Regulation of Vertebrate Forebrain Development by Homeobox Genes. Front Neurosci 2022; 16:843794. [PMID: 35546872 PMCID: PMC9081933 DOI: 10.3389/fnins.2022.843794] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/19/2023] Open
Abstract
Forebrain development in vertebrates is regulated by transcription factors encoded by homeobox, bHLH and forkhead gene families throughout the progressive and overlapping stages of neural induction and patterning, regional specification and generation of neurons and glia from central nervous system (CNS) progenitor cells. Moreover, cell fate decisions, differentiation and migration of these committed CNS progenitors are controlled by the gene regulatory networks that are regulated by various homeodomain-containing transcription factors, including but not limited to those of the Pax (paired), Nkx, Otx (orthodenticle), Gsx/Gsh (genetic screened), and Dlx (distal-less) homeobox gene families. This comprehensive review outlines the integral role of key homeobox transcription factors and their target genes on forebrain development, focused primarily on the telencephalon. Furthermore, links of these transcription factors to human diseases, such as neurodevelopmental disorders and brain tumors are provided.
Collapse
Affiliation(s)
- Ryan F. Leung
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Ankita M. George
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Enola M. Roussel
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Maree C. Faux
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jeffrey T. Wigle
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - David D. Eisenstat
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Delestrain C, Aissat A, Nattes E, Gibertini I, Lacroze V, Simon S, Decrouy X, de Becdelièvre A, Fanen P, Epaud R. Deciphering an isolated lung phenotype of NKX2-1 frameshift pathogenic variant. Front Pediatr 2022; 10:978598. [PMID: 36733766 PMCID: PMC9888430 DOI: 10.3389/fped.2022.978598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND to perform a functional analysis of a new NK2 homeobox 1 (NKX2-1) variant (c.85_86del denominated NKX2-1DEL) identified in a family presenting with isolated respiratory disease, in comparison to another frameshift variant (c.254dup denominated NKX2-1DUP) identified in a subject with classical brain-lung-thyroid syndrome. METHODS pathogenic variants were introduced into the pcDNA3-1(+)-wt-TTF1 plasmid. The proteins obtained were analyzed by western blot assay. Subcellular localization was assessed by confocal microscopy in A549 and Nthy cells. Transactivation of SFTPA, SFTPB, SFTPC, and ABCA3 promoters was assessed in A549 cells. Thyroglobulin promoter activity was measured with the paired box gene 8 (PAX8) cofactor in Nthy cells. RESULTS The two sequence variants were predicted to produce aberrant proteins identical from the 86th amino acid, with deletion of their functional homeodomain, including the nuclear localization signal. However, 3D conformation prediction of the conformation prediction of the mutant protein assumed the presence of a nuclear localization signal, a bipartite sequence, confirmed by confocal microscopy showing both mutant proteins localized in the nucleus and cytoplasm. Transcriptional activity with SFTPA, SFTPB, SFTPC, ABCA3 and thyroglobulin promoters was significantly decreased with both variants. However, with NKX2-1DEL, thyroglobulin transcriptional activity was maintained with the addition of PAX8. CONCLUSION These results provide novel insights into understanding the molecular mechanism of phenotypes associated with NKX2-1 pathogenic variants.
Collapse
Affiliation(s)
- Céline Delestrain
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Centre Hospitalier Intercommunal de Créteil, Service de Pédiatrie Générale, Créteil, France
| | - Abdel Aissat
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Département de Génétique, AP-HP, Hopital Henri Mondor, DMU de Biologie-Pathologie, Créteil, France
| | - Elodie Nattes
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Centre Hospitalier Intercommunal de Créteil, Service de Pédiatrie Générale, Créteil, France.,Département de Génétique, AP-HP, Hopital Henri Mondor, DMU de Biologie-Pathologie, Créteil, France
| | - Isabelle Gibertini
- Département de Pédiatrie, Centre Hospitalier Universitaire de Tours, Tours, France
| | - Valérie Lacroze
- AP-HM, Hôpital de la Conception, Service de Médecine Néonatale, Marseille, France
| | | | | | - Alix de Becdelièvre
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Département de Génétique, AP-HP, Hopital Henri Mondor, DMU de Biologie-Pathologie, Créteil, France
| | - Pascale Fanen
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Département de Génétique, AP-HP, Hopital Henri Mondor, DMU de Biologie-Pathologie, Créteil, France
| | - Ralph Epaud
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.,Centre Hospitalier Intercommunal de Créteil, Service de Pédiatrie Générale, Créteil, France
| |
Collapse
|
28
|
Karolak JA, Gambin T, Szafranski P, Maywald RL, Popek E, Heaney JD, Stankiewicz P. Perturbation of semaphorin and VEGF signaling in ACDMPV lungs due to FOXF1 deficiency. Respir Res 2021; 22:212. [PMID: 34315444 PMCID: PMC8314029 DOI: 10.1186/s12931-021-01797-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/01/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a rare lethal congenital lung disorder in neonates characterized by severe progressive respiratory failure and refractory pulmonary hypertension, resulting from underdevelopment of the peripheral pulmonary tree. Causative heterozygous single nucleotide variants (SNVs) or copy-number variant (CNV) deletions involving FOXF1 or its distant lung-specific enhancer on chromosome 16q24.1 have been identified in 80-90% of ACDMPV patients. FOXF1 maps closely to and regulates the oppositely oriented FENDRR, with which it also shares regulatory elements. METHODS To better understand the transcriptional networks downstream of FOXF1 that are relevant for lung organogenesis, using RNA-seq, we have examined lung transcriptomes in 12 histopathologically verified ACDMPV patients with or without pathogenic variants in the FOXF1 locus and analyzed gene expression profile in FENDRR-depleted fetal lung fibroblasts, IMR-90. RESULTS RNA-seq analyses in ACDMPV neonates revealed changes in the expression of several genes, including semaphorins (SEMAs), neuropilin 1 (NRP1), and plexins (PLXNs), essential for both epithelial branching and vascular patterning. In addition, we have found deregulation of the vascular endothelial growth factor (VEGF) signaling that also controls pulmonary vasculogenesis and a lung-specific endothelial gene TMEM100 known to be essential in vascular morphogenesis. Interestingly, we have observed a substantial difference in gene expression profiles between the ACDMPV samples with different types of FOXF1 defect. Moreover, partial overlap between transcriptome profiles of ACDMPV lungs with FOXF1 SNVs and FENDRR-depleted IMR-90 cells suggests contribution of FENDRR to ACDMPV etiology. CONCLUSIONS Our transcriptomic data imply potential crosstalk between several lung developmental pathways, including interactions between FOXF1-SHH and SEMA-NRP or VEGF/VEGFR2 signaling, and provide further insight into complexity of lung organogenesis in humans.
Collapse
Affiliation(s)
- Justyna A Karolak
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-781, Poznań, Poland
| | - Tomasz Gambin
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Institute of Computer Science, Warsaw University of Technology, 00-665, Warsaw, Poland
| | - Przemyslaw Szafranski
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Rebecca L Maywald
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Edwina Popek
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Paweł Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Krude H, Mundlos S, Øien NC, Opitz R, Schuelke M. What can go wrong in the non-coding genome and how to interpret whole genome sequencing data. MED GENET-BERLIN 2021; 33:121-131. [PMID: 38836035 PMCID: PMC11007630 DOI: 10.1515/medgen-2021-2071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/24/2021] [Indexed: 06/06/2024]
Abstract
Whole exome sequencing discovers causative mutations in less than 50 % of rare disease patients, suggesting the presence of additional mutations in the non-coding genome. So far, non-coding mutations have been identified in less than 0.2 % of individuals with genetic diseases listed in the ClinVar database and exhibit highly diverse molecular mechanisms. In contrast to our capability to sequence the whole genome, our ability to discover and functionally confirm such non-coding mutations is lagging behind severely. We discuss the problems and present examples of confirmed mutations in deep intronic sequences, non-coding triplet repeats, enhancers, and larger structural variants and highlight their proposed disease mechanisms. Finally, we discuss the type of data that would be required to establish non-coding mutation detection in routine diagnostics.
Collapse
Affiliation(s)
- Heiko Krude
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Mundlos
- Institute for Medical and Human Genetics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nancy Christine Øien
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert Opitz
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
30
|
van Moorsel CHM, van der Vis JJ, Grutters JC. Genetic disorders of the surfactant system: focus on adult disease. Eur Respir Rev 2021; 30:30/159/200085. [PMID: 33597124 DOI: 10.1183/16000617.0085-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
Genes involved in the production of pulmonary surfactant are crucial for the development and maintenance of healthy lungs. Germline mutations in surfactant-related genes cause a spectrum of severe monogenic pulmonary diseases in patients of all ages. The majority of affected patients present at a very young age, however, a considerable portion of patients have adult-onset disease. Mutations in surfactant-related genes are present in up to 8% of adult patients with familial interstitial lung disease (ILD) and associate with the development of pulmonary fibrosis and lung cancer.High disease penetrance and variable expressivity underscore the potential value of genetic analysis for diagnostic purposes. However, scarce genotype-phenotype correlations and insufficient knowledge of mutation-specific pathogenic processes hamper the development of mutation-specific treatment options.This article describes the genetic origin of surfactant-related lung disease and presents spectra for gene, age, sex and pulmonary phenotype of adult carriers of germline mutations in surfactant-related genes.
Collapse
Affiliation(s)
- Coline H M van Moorsel
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Hearts and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joanne J van der Vis
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Dept of Clinical Chemistry, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Jan C Grutters
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Hearts and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Liao J, Coffman KA, Locker J, Padiath QS, Nmezi B, Filipink RA, Hu J, Sathanoori M, Madan-Khetarpal S, McGuire M, Schreiber A, Moran R, Friedman N, Hoffner L, Rajkovic A, Yatsenko SA, Surti U. Deletion of conserved non-coding sequences downstream from NKX2-1: A novel disease-causing mechanism for benign hereditary chorea. Mol Genet Genomic Med 2021; 9:e1647. [PMID: 33666368 PMCID: PMC8123744 DOI: 10.1002/mgg3.1647] [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: 10/05/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 11/17/2022] Open
Abstract
Background Benign hereditary chorea (BHC) is an autosomal dominant disorder characterized by early‐onset non‐progressive involuntary movements. Although NKX2‐1 mutations or deletions are the cause of BHC, some BHC families do not have pathogenic alterations in the NKX2‐1 gene, indicating that mutations of non‐coding regulatory elements of NKX2‐1 may also play a role. Methods and Results By using whole‐genome microarray analysis, we identified a 117 Kb founder deletion in three apparently unrelated BHC families that were negative for NKX2‐1 sequence variants. Targeted next generation sequencing analysis confirmed the deletion and showed that it was part of a complex local genomic rearrangement. In addition, we also detected a 648 Kb de novo deletion in an isolated BHC case. Both deletions are located downstream from NKX2‐1 on chromosome 14q13.2‐q13.3 and share a 33 Kb smallest region of overlap with six previously reported cases. This region has no gene but contains multiple evolutionarily highly conserved non‐coding sequences. Conclusion We propose that the deletion of potential regulatory elements necessary for NKX2‐1 expression in this critical region is responsible for BHC phenotype in these patients, and this is a novel disease‐causing mechanism for BHC.
Collapse
Affiliation(s)
- Jun Liao
- Pittsburgh Cytogenetics Laboratory, Magee-Womens Hospital of UPMC, Pittsburgh, PA, USA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Keith A Coffman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joseph Locker
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Quasar S Padiath
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce Nmezi
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robyn A Filipink
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jie Hu
- Pittsburgh Cytogenetics Laboratory, Magee-Womens Hospital of UPMC, Pittsburgh, PA, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Malini Sathanoori
- Pittsburgh Cytogenetics Laboratory, Magee-Womens Hospital of UPMC, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Marianne McGuire
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Rocio Moran
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Neil Friedman
- Center for Pediatric Neurology, Cleveland Clinic, Cleveland, OH, USA
| | - Lori Hoffner
- Magee Womens Research Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Aleksandar Rajkovic
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.,Magee Womens Research Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Svetlana A Yatsenko
- Pittsburgh Cytogenetics Laboratory, Magee-Womens Hospital of UPMC, Pittsburgh, PA, USA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.,Magee Womens Research Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Urvashi Surti
- Pittsburgh Cytogenetics Laboratory, Magee-Womens Hospital of UPMC, Pittsburgh, PA, USA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.,Magee Womens Research Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
32
|
van Trotsenburg P, Stoupa A, Léger J, Rohrer T, Peters C, Fugazzola L, Cassio A, Heinrichs C, Beauloye V, Pohlenz J, Rodien P, Coutant R, Szinnai G, Murray P, Bartés B, Luton D, Salerno M, de Sanctis L, Vigone M, Krude H, Persani L, Polak M. Congenital Hypothyroidism: A 2020-2021 Consensus Guidelines Update-An ENDO-European Reference Network Initiative Endorsed by the European Society for Pediatric Endocrinology and the European Society for Endocrinology. Thyroid 2021; 31:387-419. [PMID: 33272083 PMCID: PMC8001676 DOI: 10.1089/thy.2020.0333] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: An ENDO-European Reference Network (ERN) initiative was launched that was endorsed by the European Society for Pediatric Endocrinology and the European Society for Endocrinology with 22 participants from the ENDO-ERN and the two societies. The aim was to update the practice guidelines for the diagnosis and management of congenital hypothyroidism (CH). A systematic literature search was conducted to identify key articles on neonatal screening, diagnosis, and management of primary and central CH. The evidence-based guidelines were graded with the Grading of Recommendations, Assessment, Development and Evaluation system, describing both the strength of recommendations and the quality of evidence. In the absence of sufficient evidence, conclusions were based on expert opinion. Summary: The recommendations include the various neonatal screening approaches for CH as well as the etiology (also genetics), diagnostics, treatment, and prognosis of both primary and central CH. When CH is diagnosed, the expert panel recommends the immediate start of correctly dosed levothyroxine treatment and frequent follow-up including laboratory testing to keep thyroid hormone levels in their target ranges, timely assessment of the need to continue treatment, attention for neurodevelopment and neurosensory functions, and, if necessary, consulting other health professionals, and education of the child and family about CH. Harmonization of diagnostics, treatment, and follow-up will optimize patient outcomes. Lastly, all individuals with CH are entitled to a well-planned transition of care from pediatrics to adult medicine. Conclusions: This consensus guidelines update should be used to further optimize detection, diagnosis, treatment, and follow-up of children with all forms of CH in the light of the most recent evidence. It should be helpful in convincing health authorities of the benefits of neonatal screening for CH. Further epidemiological and experimental studies are needed to understand the increased incidence of this condition.
Collapse
Affiliation(s)
- Paul van Trotsenburg
- Department of Pediatric Endocrinology, Emma Children's Hospital, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Athanasia Stoupa
- Pediatric Endocrinology, Gynecology and Diabetology Department, Assistance Publique Hôpitaux de Paris (APHP), Hôpital Universitaire Necker Enfants Malades, Paris, France
- Université de Paris, Paris, France
- INSERM U1163, IMAGINE Institute, Paris, France
- INSERM U1016, Cochin Institute, Paris, France
| | - Juliane Léger
- Department of Pediatric Endocrinology and Diabetology, Reference Center for Growth and Development Endocrine Diseases, Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1141, Paris, France
| | - Tilman Rohrer
- Department of Pediatric Endocrinology, University Children's Hospital, Saarland University Medical Center, Homburg, Germany
| | - Catherine Peters
- Department of Pediatric Endocrinology, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Laura Fugazzola
- Department of Endocrinology and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Alessandra Cassio
- Department of Pediatric Endocrinology, Unit of Pediatrics, Department of Medical & Surgical Sciences, University of Bologna, Bologna Italy
| | - Claudine Heinrichs
- Pediatric Endocrinology Unit, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Veronique Beauloye
- Unité d'Endocrinologie Pédiatrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Joachim Pohlenz
- Department of Pediatrics, Johannes Gutenberg University Medical School, Mainz, Germany
| | - Patrice Rodien
- Centre de Référence des Maladies Rares de la Thyroïde et des Récepteurs Hormonaux, Service EDN, CHU d'Angers, Institut MITOVASC, Université d'Angers, Angers, France
| | - Regis Coutant
- Unité d' Endocrinologie Diabetologie Pédiatrique and Centre des Maladies Rares de la Réceptivité Hormonale, CHU-Angers, Angers, France
| | - Gabor Szinnai
- Department of Pediatric Endocrinology, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Philip Murray
- European Society for Pediatric Endocrinology
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Beate Bartés
- Thyroid Group, European Patient Advocacy Group Patient Representative (ePAG), Association Vivre sans Thyroide, Léguevin, France
| | - Dominique Luton
- Department of Obstetrics and Gynecology, University Hospitals Paris Nord Val de Seine (HUPNVS), Assistance Publique Hôpitaux de Paris (APHP), Bichat Hospital, Paris, France
- Department Risks and Pregnancy (DHU), Université de Paris, Inserm U1141, Paris, France
| | - Mariacarolina Salerno
- Pediatric Endocrine Unit, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Luisa de Sanctis
- Department of Public Health and Pediatrics, University of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Mariacristina Vigone
- Department of Pediatrics, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Heiko Krude
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Luca Persani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Michel Polak
- Pediatric Endocrinology, Gynecology and Diabetology Department, Assistance Publique Hôpitaux de Paris (APHP), Hôpital Universitaire Necker Enfants Malades, Paris, France
- Université de Paris, Paris, France
- INSERM U1163, IMAGINE Institute, Paris, France
- INSERM U1016, Cochin Institute, Paris, France
- Paris Regional Newborn Screening Program, Centre régional de dépistage néonatal, Paris, France
- Centre de Référence Maladies Endocriniennes de la Croissance et du Développement, INSERM U1016, IMAGINE Institute, Paris, France
- ENDO-European Reference Network, Main Thematic Group 8, Paris, France
| |
Collapse
|
33
|
Prasad R, Nicholas AK, Schoenmakers N, Barton J. Haploinsufficiency of NKX2-1 in Brain-Lung-Thyroid Syndrome with Additional Multiple Pituitary Dysfunction. Horm Res Paediatr 2020; 92:340-344. [PMID: 31707387 DOI: 10.1159/000503683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/25/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Heterozygous mutations or haploinsufficiency of NKX2-1 are associated with the brain-lung-thyroid syndrome incorporating primary hypothyroidism, respiratory distress, and neurological disturbances. CASE PRESENTATION We report a patient presenting in the neonatal period with multiple pituitary hormone deficiency including central hypothyroidism and hypoadrenalism, growth hormone deficiency, undetectable gonadotrophins, and a small anterior pituitary on MRI. CGH microarray revealed haploinsufficiency for NKX2.1 and during subsequent follow-up, she has exhibited the classic triad of brain-lung-thyroid syndrome with undetectable tissue on thyroid ultrasonography. Whilst the role of NKX2-1 is well described in murine pituitary development, this report constitutes the first description of multiple pituitary dysfunction in humans associated with the syndrome and haploinsufficiency NKX2-1. CONCLUSION The report highlights a potential need for pituitary screening in patients with established brain-lung-thyroid syndrome and implicates NKX2.1 in human pituitary disease.
Collapse
Affiliation(s)
- Rathi Prasad
- Department of Paediatric Endocrinology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom,
| | - Adeline K Nicholas
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Nadia Schoenmakers
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - John Barton
- Department of Paediatric Endocrinology, Bristol Royal Hospital for Children, University Hospitals Bristol NHS Foundation Trust, Bristol, United Kingdom
| |
Collapse
|
34
|
Hu X, Liu J, Guo R, Guo J, Zhao Z, Li W, Xu B, Hao C. A novel 14q13.1-21.1 deletion identified by CNV-Seq in a patient with brain-lung-thyroid syndrome, tooth agenesis and immunodeficiency. Mol Cytogenet 2019; 12:51. [PMID: 31890031 PMCID: PMC6924084 DOI: 10.1186/s13039-019-0463-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
Background Chromosome 14q11-q22 deletion syndrome (OMIM 613457) is a rare genomic disorder. The phenotype heterogeneity depends on the deletion size, breakpoints and genes deleted. Critical genes like FOXG1, NKX2–1, PAX9 were identified. Case presentation We performed whole exome sequencing (WES) and copy number variation sequencing (CNV-seq) for a patient with mild speech and motor developmental delay, short stature, recurrent pulmonary infections, tooth agenesis and triad of brain-lung-thyroid syndrome. By using CNV-seq, we identified a 3.1 Mb de novo interstitial deletion of the 14q13.2q21.1 region encompassing 17 OMIM genes including NKX2–1, PAX9 and NFKBIA. Our patient’s phenotype is consistent with other published 14q13 deletion patients. Conclusion Our results showed the combination of WES and CNV-seq is an effective diagnostic strategy for patients with genetic or genomic disorders. After reviewing published patients, we also proposed a new critical region for 14q13 deletion syndrome with is a more benign disorder compared to 14q11-q22 deletion syndrome.
Collapse
Affiliation(s)
- Xuyun Hu
- 1Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| | - Jun Liu
- 2China National Clinical Research Center of Respiratory Diseases, Respiratory Department of Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| | - Ruolan Guo
- 1Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| | - Jun Guo
- 1Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| | - Zhipeng Zhao
- 2China National Clinical Research Center of Respiratory Diseases, Respiratory Department of Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| | - Wei Li
- 1Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| | - Baoping Xu
- 2China National Clinical Research Center of Respiratory Diseases, Respiratory Department of Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| | - Chanjuan Hao
- 1Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute; MOE Key Laboratory of Major Diseases in Children; Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045 China
| |
Collapse
|
35
|
Santos-Silva R, Rosário M, Grangeia A, Costa C, Castro-Correia C, Alonso I, Leão M, Fontoura M. Genetic analyses in a cohort of Portuguese pediatric patients with congenital hypothyroidism. J Pediatr Endocrinol Metab 2019; 32:1265-1273. [PMID: 31430255 DOI: 10.1515/jpem-2019-0047] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/05/2019] [Indexed: 11/15/2022]
Abstract
Background Permanent primary congenital hypothyroidism (CH) can be caused by thyroid dysgenesis or dyshormonogenesis. A molecular genetic study is recommended in dyshormonogenesis, in syndromic hypothyroidism and when there is a family history of CH. The aim of this study was to identify a monogenic etiology for CH in selected individuals from a cohort of primary permanent CH. Methods From an initial cohort of 79 patients with permanent CH (3-19 years), 11 patients were selected for molecular analyses. Nine patients with dyshormonogenesis (normal in-situ gland or goiter) were screened for causative variants, by next-generation sequencing (NGS), in 28 genes known to be responsible for CH. One patient with a family history of CH was screened for the paired-box gene 8 (PAX8) gene and another patient with a syndromic CH was screened for the NKX2-1 gene. Results We found a monogenic basis of disease in eight patients, involving the thyroid peroxidase (TPO) gene (four patients), the thyroglobulin (TG) gene (two patients), and the PAX8 and NKX2-1 genes (one patient each). Two patients were heterozygotes, one harboring a variant in the TG gene and the other in the SLC5A5 gene. In one patient, we found no potential causative variants in any of the 28 genes screened. We described five novel variants: three in the TG gene, one in the NKX2-1 and one in the SLC5A5 gene, all of them classified as pathogenic. Conclusions In eight of the 11 screened patients, a monogenic disease was found. These results highlight the advantage of using an NGS panel and provide further data regarding the molecular basis of CH.
Collapse
Affiliation(s)
- Rita Santos-Silva
- Department of Pediatrics, Pediatric Endocrinology Unit, Centro Hospitalar S. João, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Marta Rosário
- Department of Pediatrics, Centro Hospitalar S. João, Porto, Portugal
| | - Ana Grangeia
- Department of Medical Genetics, Centro Hospitalar de S. João, Porto, Portugal
| | - Carla Costa
- Department of Pediatrics, Pediatric Endocrinology Unit, Centro Hospitalar S. João, Alameda Prof. Hernâni Monteiro, Porto, Portugal
| | - Cíntia Castro-Correia
- Department of Pediatrics, Pediatric Endocrinology Unit, Centro Hospitalar S. João, Alameda Prof. Hernâni Monteiro, Porto, Portugal
| | - Isabel Alonso
- UnIGENe and CGPP/Units at i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Miguel Leão
- Department of Medical Genetics, Centro Hospitalar de S. João, Porto, Portugal
| | - Manuel Fontoura
- Department of Pediatrics, Pediatric Endocrinology Unit, Centro Hospitalar S. João, Alameda Prof. Hernâni Monteiro, Porto, Portugal
| |
Collapse
|
36
|
Mio C, Grani G, Durante C, Damante G. Molecular defects in thyroid dysgenesis. Clin Genet 2019; 97:222-231. [PMID: 31432505 DOI: 10.1111/cge.13627] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022]
Abstract
Congenital hypothyroidism (CH) is a neonatal endocrine disorder that might occur as itself or be associated to congenital extra-thyroidal defects. About 85% of affected subjects experience thyroid dysgenesis (TD), characterized by defect in thyroid gland development. In vivo experiments on null mice paved the way for the identification of genes involved thyroid morphogenesis and development, whose mutation has been strongly associated to TD. Most of them are thyroid-specific transcription factors expressed during early thyroid development. Despite the arduous effort in unraveling the genetics of TD in animal models, up to now these data have been discontinuously confirmed in humans and only 5% of TD have associated with known null mice-related mutations (mainly PAX8 and TSHR). Notwithstanding, the advance in genetic testing represented by the next-generation sequencing (NGS) approach is steadily increasing the list of genes whose highly penetrant mutation predisposes to TD. In this review we intend to outline the molecular bases of TD, summarizing the current knowledge on thyroid development in both mice and humans and delineating the genetic features of its monogenetic forms. We will also highlight current strategies to enhance the insight into the non-Mendelian mechanisms of abnormal thyroid development.
Collapse
Affiliation(s)
- Catia Mio
- Department of Medicine, University of Udine, Udine, Italy
| | - Giorgio Grani
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Cosimo Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Damante
- Department of Medicine, University of Udine, Udine, Italy.,Institute of Medical Genetics, Academic Hospital "Azienda Sanitaria Universitaria Integrata di Udine", Udine, Italy
| |
Collapse
|
37
|
Gonçalves D, Lourenço L, Guardiano M, Castro-Correia C, Sampaio M, Leão M. Chiari Malformation Type I in a Patient with a Novel NKX2-1 Mutation. J Pediatr Neurosci 2019; 14:169-172. [PMID: 31649781 PMCID: PMC6798275 DOI: 10.4103/jpn.jpn_108_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/09/2018] [Accepted: 08/12/2019] [Indexed: 11/04/2022] Open
Abstract
Chiari Malformation Type 1 is a congenital, condition characterized by abnormally shaped cerebellar tonsils that are displaced below the level of the foramen magnum. NKX2-1 gene encodes a transcription factor expressed during early development of thyroid, lung, and forebrain, and germline NKX2-1 mutations can lead to dysfunction in any of these three organs, resulting in brain-lung-thyroid syndrome. There have been few reports of structural brain anomalies in patients with an NKX2-1-related disorder. We report the first case of a girl with a genetically identified mutation in NKX2-1 that presents with a Chiari Malformation Type 1, eventually expanding the phenotypic spectrum of NKX2-1-related disorders while also highlighting a novel heterozygous pathogenic variant at exon 3 that disrupts the reading framework, originating an NKX2-1 protein with a different C-terminal.
Collapse
Affiliation(s)
- Daniel Gonçalves
- Serviço de Pediatria do, Centro Hospitalar de São João, Porto, Portugal
| | - Lara Lourenço
- Serviço de Pediatria do, Centro Hospitalar de São João, Porto, Portugal
| | - Micaela Guardiano
- Serviço de Pediatria do, Centro Hospitalar de São João, Porto, Portugal
| | | | - Mafalda Sampaio
- Serviço de Pediatria do, Centro Hospitalar de São João, Porto, Portugal
| | - Miguel Leão
- Serviço de Genética do, Centro Hospitalar de São João, Porto, Portugal
| |
Collapse
|
38
|
Novel non-synonymous mutations of PAX8 in a cohort of Chinese with congenital hypothyroidism. Chin Med J (Engl) 2019; 132:1322-1327. [PMID: 30888984 PMCID: PMC6629355 DOI: 10.1097/cm9.0000000000000213] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The transcription factor paired box 8 (PAX8) was associated with type 2 congenital non-goitrous hypothyroidism (CHNG2), a clinical phenotype of congenital hypothyroidism (CH). Though studied in a few regions with different ethnicities, the incidence of PAX8 mutations varied, even among Chinese cohorts in different regions. This study aimed to identify and characterize PAX8 mutations and explore the prevalence of its mutations in another cohort of CH. METHODS The 105 unrelated Chinese patients with CH were collected from four major hospitals. Exomes of the 105 samples were sequenced by Hiseq 2000 platform to identify mutations of PAX8 on genomic DNAs extracted from peripheral blood samples. Luciferase reporter assays were used to assess the effects of mutations on the transcription of thyroid peroxidase (TPO). RESULTS Three PAX8 mutations in four subjects were identified in 105 samples. One variant, rs189229644, was detected in two subjects, and categorized as uncertain significance. The other two missense mutations (275T>C/Ile92Thr and 398G>A/Arg133Gln) were not detected in three large-scale genotyping projects, namely 1000 Genome Project, Exome Aggregation Consortium and GO Exome Sequencing Project. Functional studies for the two mutations revealed that they could impair the transcription ability of PAX8 on one of its target genes, TPO. Therefore, the two mutations were causative for the pathogenesis of CHNG2. After combining the studies of PAX8 mutations, an average frequency of 1.74% (21/1209) could be obtained in Chinese patients with CH. CONCLUSION The study specifically demonstrates the role of two mutations in impairing the transcription ability of PAX8, which should be considered as pathogenic variants for CH.
Collapse
|
39
|
High-resolution computed tomography findings of thyroid transcription factor 1 deficiency (NKX2-1 mutations). Pediatr Radiol 2019; 49:869-875. [PMID: 30927038 DOI: 10.1007/s00247-019-04388-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/02/2019] [Accepted: 03/18/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND The expression of the NKX2-1 gene and its encoded protein, thyroid transcription factor 1 (TTF-1), plays a role in pulmonary surfactant homeostasis and lung development. NKX2-1 mutations have been associated with neonatal respiratory distress, hypotonia, choreoathetosis and congenital hypothyroidism. These clinical findings have been coined brain-lung-thyroid syndrome, although not all three organs are always involved. While many of these children develop interstitial lung disease, no systematic review of chest high-resolution CT (HRCT) findings has been reported. OBJECTIVE To summarize the clinical presentations, pathology and HRCT imaging findings of children with NKX2-1 mutations. MATERIALS AND METHODS We identified six children with NKX2-1 mutations, deletions or duplications confirmed via genetic testing at our institution. Three pediatric radiologists reviewed the children's HRCT imaging findings and ranked the dominant findings in order of prevalence via consensus. We then correlated the imaging findings with histopathology and clinical course. RESULTS All children in the study were heterozygous for NKX2-1 mutations, deletions or duplications. Ground-glass opacities were the most common imaging feature, present in all but one child. Consolidation was also a prevalent finding in 4/6 of the children. Architectural distortion was less common. CONCLUSION HRCT findings of TTF-1 deficiency are heterogeneous and evolve over time. There is significant overlap between the HRCT findings of TTF-1 deficiency, other surfactant dysfunction mutations, and pulmonary interstitial glycogenosis. TTF-1 deficiency should be considered in term infants presenting with interstitial lung disease, especially if hypotonia or hypothyroidism is present.
Collapse
|
40
|
Parnes M, Bashir H, Jankovic J. Is Benign Hereditary Chorea Really Benign? Brain-Lung-Thyroid Syndrome Caused by NKX2-1 Mutations. Mov Disord Clin Pract 2019; 6:34-39. [PMID: 30746413 PMCID: PMC6335533 DOI: 10.1002/mdc3.12690] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/20/2018] [Accepted: 09/09/2018] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Since its localization to the NKX2-1 gene in 2002, the phenotype of the disorder historically called "benign hereditary chorea" has been expanding beyond chorea. METHODS The phenomenology of movement disorders and other symptomatology associated with mutations in NKX2-1 were characterized after a detailed evaluation of consecutive patients evaluated in our clinic over the past 3 years. RESULTS We studied 5 patients (3 females), ages 2 to 31 years, with confirmed pathogenic variants in NKX2-1. All patients exhibited chorea, gross motor delay, and gait impairment. Other symptoms included neonatal respiratory failure (n = 4), cognitive deficits (n = 3), hypothyroidism (n = 4), joint laxity (n = 2), myoclonus (n = 1), hypotonia (n = 3), and seizures (n = 1). Chorea often proved refractory to medical therapies. CONCLUSIONS The phenotype associated with pathogenic variants in NKX2-1 frequently includes disabling and often medically refractory neurological and non-neurological abnormalities. We therefore suggest that the term benign hereditary chorea be abandoned in favor of its genetic designation as NKX2-1-related disorder.
Collapse
Affiliation(s)
- Mered Parnes
- Pediatric Movement Disorders Clinic, Blue Bird Circle Clinic for Pediatric Neurology, Section of Pediatric Neurology and Developmental NeuroscienceTexas Children's HospitalHoustonTexasUSA
- Parkinson's Disease Center and Movement Disorders Clinic, Department of NeurologyBaylor College of MedicineHoustonTexasUSA
| | - Hassaan Bashir
- Parkinson's Disease Center and Movement Disorders Clinic, Department of NeurologyBaylor College of MedicineHoustonTexasUSA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of NeurologyBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
41
|
Peters C, van Trotsenburg ASP, Schoenmakers N. DIAGNOSIS OF ENDOCRINE DISEASE: Congenital hypothyroidism: update and perspectives. Eur J Endocrinol 2018; 179:R297-R317. [PMID: 30324792 DOI: 10.1530/eje-18-0383] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Congenital hypothyroidism (CH) may be primary, due to a defect affecting the thyroid gland itself, or central, due to impaired thyroid-stimulating hormone (TSH)-mediated stimulation of the thyroid gland as a result of hypothalamic or pituitary pathology. Primary CH is the most common neonatal endocrine disorder, traditionally subdivided into thyroid dysgenesis (TD), referring to a spectrum of thyroid developmental abnormalities, and dyshormonogenesis, where a defective molecular pathway for thyroid hormonogenesis results in failure of hormone production by a structurally intact gland. Delayed treatment of neonatal hypothyroidism may result in profound neurodevelopmental delay; therefore, CH is screened for in developed countries to facilitate prompt diagnosis. Central congenital hypothyroidism (CCH) is a rarer entity which may occur in isolation, or (more frequently) in association with additional pituitary hormone deficits. CCH is most commonly defined biochemically by failure of appropriate TSH elevation despite subnormal thyroid hormone levels and will therefore evade diagnosis in primary, TSH-based CH-screening programmes. This review will discuss recent genetic aetiological advances in CH and summarize epidemiological data and clinical diagnostic challenges, focussing on primary CH and isolated CCH.
Collapse
Affiliation(s)
- C Peters
- Department of Endocrinology, Great Ormond Street Hospital for Children, London, UK
| | - A S P van Trotsenburg
- Department of Paediatric Endocrinology, Emma Children’s Hospital Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - N Schoenmakers
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research
Council Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| |
Collapse
|
42
|
Attarian SJ, Leibel SL, Yang P, Alfano DN, Hackett BP, Cole FS, Hamvas A. Mutations in the thyroid transcription factor gene NKX2-1 result in decreased expression of SFTPB and SFTPC. Pediatr Res 2018; 84. [PMID: 29538355 PMCID: PMC6599453 DOI: 10.1038/pr.2018.30] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Mutations in the NK2 homeobox 1 (NKX2-1) gene are associated with lung disease in infants and children. We hypothesize that disruption of normal surfactant gene expression with these mutations contributes to the respiratory phenotypes observed. METHODS To assess transactivational activity, cotransfection of luciferase reporter vectors containing surfactant protein B or C (SFTPB or SFTPC) promoters with NKX2-1 plasmids was performed and luciferase activity was measured. To assess the binding of mutated proteins to target DNA, electrophoretic mobility shift assays (EMSA) were performed using nuclear protein labeled with oligonucleotide probes representing NKX2-1 consensus binding sequences followed by gel electrophoresis. The effect of overexpression of wild-type (WT) and mutant NKX2-1 on SFTPB and SFTPC was evaluated with quantitative real-time PCR. RESULTS Decreased transactivation of the SFTPB promoter by both mutants and decreased transactivation of the SFTPC promoter by the L197P mutation was observed. EMSA demonstrated decreased DNA binding of both mutations to NKX2-1 consensus binding sequences. Transfection of A549 cells with NKX2-1 expression vectors demonstrated decreased stimulation of SFTPB and SFTPC expression by mutant proteins compared with that of WT. CONCLUSION Disruption of transcriptional activation of surfactant protein genes by these DNA-binding domain mutations is a plausible biological mechanism for disruption of surfactant function and subsequent respiratory distress.
Collapse
Affiliation(s)
- Stephanie J Attarian
- Edward Mallinckrodt Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine and St Louis Children's Hospital, St Louis, Missouri, USA.
| | - Sandra L Leibel
- Department of Pediatrics, University of California San Diego School of Medicine, San Diego, California
| | - Ping Yang
- Edward Mallinckrodt Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine and St Louis Children’s Hospital, St Louis, Missouri
| | - Danielle N Alfano
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Brian P Hackett
- Edward Mallinckrodt Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine and St Louis Children’s Hospital, St Louis, Missouri
| | - F Sessions Cole
- Edward Mallinckrodt Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine and St Louis Children’s Hospital, St Louis, Missouri
| | - Aaron Hamvas
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
43
|
Persani L, Rurale G, de Filippis T, Galazzi E, Muzza M, Fugazzola L. Genetics and management of congenital hypothyroidism. Best Pract Res Clin Endocrinol Metab 2018; 32:387-396. [PMID: 30086865 DOI: 10.1016/j.beem.2018.05.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Several evidences support a relevant genetic origin for Congenital Hypothyroidism (CH), however familial forms are uncommon. CH can be due to morphogenetic or functional defects and several genes have been originally associated either with thyroid dysgenesis or dyshormonogenesis, with a highly variable expressivity and a frequently incomplete penetrance of the genetic defects. The phenotype-driven genetic analyses rarely yielded positive results in more than 10% of cases, thus raising doubts on the genetic origin of CH. However, more recent unsupervised approaches with systematic Next Generation Sequencing (NGS) analysis revealed the existence of hypomorphic alleles of these candidate genes whose combination can explain a significant portion of CH cases. The co-segregation studies of the hypothyroid phenotype with multiple gene variants in pedigrees confirmed the potential oligogenic origin of CH, which finally represents a suitable explanation for the frequent sporadic occurrence of this disease.
Collapse
Affiliation(s)
- Luca Persani
- Division of Endocrine and Metabolic Diseases & Labs of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, 20149, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy.
| | - Giuditta Rurale
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | - Tiziana de Filippis
- Division of Endocrine and Metabolic Diseases & Labs of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, 20149, Milan, Italy
| | - Elena Galazzi
- Division of Endocrine and Metabolic Diseases & Labs of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, 20149, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | - Marina Muzza
- Division of Endocrine and Metabolic Diseases & Labs of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, 20149, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, 20122, Milan, Italy
| | - Laura Fugazzola
- Division of Endocrine and Metabolic Diseases & Labs of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, 20149, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, 20122, Milan, Italy
| |
Collapse
|
44
|
Villafuerte B, Natera-de-Benito D, González A, Mori MA, Palomares M, Nevado J, García-Miñaur S, Lapunzina P, González-Granado LI, Allende LM, Moreno JC. The Brain-Lung-Thyroid syndrome (BLTS): A novel deletion in chromosome 14q13.2-q21.1 expands the phenotype to humoral immunodeficiency. Eur J Med Genet 2018; 61:393-398. [DOI: 10.1016/j.ejmg.2018.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/13/2017] [Accepted: 02/17/2018] [Indexed: 12/11/2022]
|
45
|
Puusepp S, Reinson K, Pajusalu S, Murumets Ü, Õiglane-Shlik E, Rein R, Talvik I, Rodenburg RJ, Õunap K. Effectiveness of whole exome sequencing in unsolved patients with a clinical suspicion of a mitochondrial disorder in Estonia. Mol Genet Metab Rep 2018; 15:80-89. [PMID: 30009132 PMCID: PMC6043467 DOI: 10.1016/j.ymgmr.2018.03.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Reaching a genetic diagnosis of mitochondrial disorders (MDs) is challenging due to their broad phenotypic and genotypic heterogeneity. However, there is growing evidence that the use of whole exome sequencing (WES) for diagnosing patients with a clinical suspicion of an MD is effective (39-60%). We aimed to study the effectiveness of WES in clinical practice in Estonia, in patients with an unsolved, but suspected MD. We also show our first results of mtDNA analysis obtained from standard WES reads. METHODS Retrospective cases were selected from a database of 181 patients whose fibroblast cell cultures had been stored from 2003 to 2013. Prospective cases were selected during the period of 2014-2016 from patients referred to a clinical geneticist in whom an MD was suspected. We scored each patient according to the mitochondrial disease criteria (MDC) (Morava et al., 2006) after re-evaluation of their clinical data, and then performed WES analysis. RESULTS A total of 28 patients were selected to the study group. A disease-causing variant was found in 16 patients (57%) using WES. An MD was diagnosed in four patients (14%), with variants in the SLC25A4, POLG, SPATA5, and NDUFB11 genes. Other variants found were associated with a neuromuscular disease (SMN1, MYH2, and LMNA genes), neurodegenerative disorder (TSPOAP1, CACNA1A, ALS2, and SCN2A genes), multisystemic disease (EPG5, NKX1-2, ATRX, and ABCC6 genes), and one in an isolated cardiomyopathy causing gene (MYBPC3). The mtDNA point mutation was found in the MT-ATP6 gene of one patient upon mtDNA analysis. CONCLUSIONS The diagnostic yield of WES in our cohort was 57%, proving to be a very good effectiveness. However, MDs were found in only 14% of the patients. We suggest WES analysis as a first-tier method in clinical genetic practice for children with any multisystem, neurological, and/or neuromuscular problem, as nuclear DNA variants are more common in children with MDs; a large number of patients harbor disease-causing variants in genes other than the mitochondria-related ones, and the clinical presentation might not always point towards an MD. We have also successfully conducted analysis of mtDNA from standard WES reads, providing further evidence that this method could be routinely used in the future.
Collapse
Affiliation(s)
- Sanna Puusepp
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, 2 L. Puusepa Street, Tartu 51014, Estonia
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, 2 L. Puusepa Street, Tartu 51014, Estonia
| | - Karit Reinson
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, 2 L. Puusepa Street, Tartu 51014, Estonia
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, 2 L. Puusepa Street, Tartu 51014, Estonia
| | - Sander Pajusalu
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, 2 L. Puusepa Street, Tartu 51014, Estonia
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, 2 L. Puusepa Street, Tartu 51014, Estonia
| | - Ülle Murumets
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, 2 L. Puusepa Street, Tartu 51014, Estonia
| | - Eve Õiglane-Shlik
- Children's Clinic, Tartu University Hospital, 6 Lunini Street, Tartu 51014, Estonia
- Department of Pediatrics, Institute of Clinical Medicine, University of Tartu, 6 Lunini Street, Tartu 51014, Estonia
| | - Reet Rein
- Children's Clinic, Tartu University Hospital, 6 Lunini Street, Tartu 51014, Estonia
| | - Inga Talvik
- Tallinn Children's Hospital, 28 Tervise Street, Tallinn 13419, Estonia
| | - Richard J. Rodenburg
- Radboud Center for Mitochondrial Medicine, 830 Translational Metabolic Laboratory, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Katrin Õunap
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, 2 L. Puusepa Street, Tartu 51014, Estonia
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, 2 L. Puusepa Street, Tartu 51014, Estonia
| |
Collapse
|
46
|
Löf C, Patyra K, Kero A, Kero J. Genetically modified mouse models to investigate thyroid development, function and growth. Best Pract Res Clin Endocrinol Metab 2018; 32:241-256. [PMID: 29779579 DOI: 10.1016/j.beem.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The thyroid gland produces thyroid hormones (TH), which are essential regulators for growth, development and metabolism. The thyroid is mainly controlled by the thyroid-stimulating hormone (TSH) that binds to its receptor (TSHR) on thyrocytes and mediates its action via different G protein-mediated signaling pathways. TSH primarily activates the Gs-pathway, and at higher concentrations also the Gq/11-pathway, leading to an increase of intracellular cAMP and Ca2+, respectively. To date, the physiological importance of other G protein-mediated signaling pathways in thyrocytes is unclear. Congenital hypothyroidism (CH) is defined as the lack of TH at birth. In familial cases, high-throughput sequencing methods have facilitated the identification of novel mutations. Nevertheless, the precise etiology of CH yet remains unraveled in a proportion of cases. Genetically modified mouse models can reveal new pathophysiological mechanisms of thyroid diseases. Here, we will present an overview of genetic mouse models for thyroid diseases, which have provided crucial insights into thyroid gland development, function, and growth with a special focus on TSHR and microRNA signaling.
Collapse
Affiliation(s)
- C Löf
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - K Patyra
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - A Kero
- Department of Pediatrics, Turku University Hospital, Kiinamyllynkatu 4-8, 20521, Turku, Finland
| | - J Kero
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland; Department of Pediatrics, Turku University Hospital, Kiinamyllynkatu 4-8, 20521, Turku, Finland.
| |
Collapse
|
47
|
Uusimaa J, Kaarteenaho R, Paakkola T, Tuominen H, Karjalainen MK, Nadaf J, Varilo T, Uusi-Mäkelä M, Suo-Palosaari M, Pietilä I, Hiltunen AE, Ruddock L, Alanen H, Biterova E, Miinalainen I, Salminen A, Soininen R, Manninen A, Sormunen R, Kaakinen M, Vuolteenaho R, Herva R, Vieira P, Dunder T, Kokkonen H, Moilanen JS, Rantala H, Nogee LM, Majewski J, Rämet M, Hallman M, Hinttala R. NHLRC2 variants identified in patients with fibrosis, neurodegeneration, and cerebral angiomatosis (FINCA): characterisation of a novel cerebropulmonary disease. Acta Neuropathol 2018; 135:727-742. [PMID: 29423877 DOI: 10.1007/s00401-018-1817-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 11/26/2022]
Abstract
A novel multi-organ disease that is fatal in early childhood was identified in three patients from two non-consanguineous families. These children were born asymptomatic but at the age of 2 months they manifested progressive multi-organ symptoms resembling no previously known disease. The main clinical features included progressive cerebropulmonary symptoms, malabsorption, progressive growth failure, recurrent infections, chronic haemolytic anaemia and transient liver dysfunction. In the affected children, neuropathology revealed increased angiomatosis-like leptomeningeal, cortical and superficial white matter vascularisation and congestion, vacuolar degeneration and myelin loss in white matter, as well as neuronal degeneration. Interstitial fibrosis and previously undescribed granuloma-like lesions were observed in the lungs. Hepatomegaly, steatosis and collagen accumulation were detected in the liver. A whole-exome sequencing of the two unrelated families with the affected children revealed the transmission of two heterozygous variants in the NHL repeat-containing protein 2 (NHLRC2); an amino acid substitution p.Asp148Tyr and a frameshift 2-bp deletion p.Arg201GlyfsTer6. NHLRC2 is highly conserved and expressed in multiple organs and its function is unknown. It contains a thioredoxin-like domain; however, an insulin turbidity assay on human recombinant NHLRC2 showed no thioredoxin activity. In patient-derived fibroblasts, NHLRC2 levels were low, and only p.Asp148Tyr was expressed. Therefore, the allele with the frameshift deletion is likely non-functional. Development of the Nhlrc2 null mouse strain stalled before the morula stage. Morpholino knockdown of nhlrc2 in zebrafish embryos affected the integrity of cells in the midbrain region. This is the first description of a fatal, early-onset disease; we have named it FINCA disease based on the combination of pathological features that include fibrosis, neurodegeneration, and cerebral angiomatosis.
Collapse
Affiliation(s)
- Johanna Uusimaa
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland.
- Department of Children and Adolescents, Oulu University Hospital, PO Box 23, 90029, Oulu, Finland.
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland.
| | - Riitta Kaarteenaho
- Research Unit of Internal Medicine, Respiratory Research, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu and Unit of Internal Medicine and Respiratory Medicine, Oulu University Hospital, PO Box 20, 90029, Oulu, Finland
| | - Teija Paakkola
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Hannu Tuominen
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Department of Pathology, Oulu University Hospital, PO Box 50, 90029, Oulu, Finland
| | - Minna K Karjalainen
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
| | - Javad Nadaf
- McGill University and Génome Québec Innovation Centre, Montreal, QC, H3A 0G1, Canada
- St. Jude Children's Research Hospital (SJCRH), 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Teppo Varilo
- Department of Medical Genetics, University of Helsinki, Haartmaninkatu 8, 00251, Helsinki, Finland
| | - Meri Uusi-Mäkelä
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Maria Suo-Palosaari
- Department of Diagnostic Radiology and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, PO Box 50, 90029, Oulu, Finland
| | - Ilkka Pietilä
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Anniina E Hiltunen
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Lloyd Ruddock
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Heli Alanen
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Ekaterina Biterova
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Ilkka Miinalainen
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Annamari Salminen
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
| | - Raija Soininen
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Aki Manninen
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Raija Sormunen
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Mika Kaakinen
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | | | - Riitta Herva
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Päivi Vieira
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, PO Box 23, 90029, Oulu, Finland
| | - Teija Dunder
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, PO Box 23, 90029, Oulu, Finland
| | - Hannaleena Kokkonen
- Northern Finland Laboratory Centre NordLab, Oulu University Hospital, PO Box 500, 90029, Oulu, Finland
- Department of Clinical Chemistry and Medical Research Center Oulu, University Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
| | - Jukka S Moilanen
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Department of Clinical Genetics, Oulu University Hospital, PO Box 23, 90029, Oulu, Finland
| | - Heikki Rantala
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, PO Box 23, 90029, Oulu, Finland
| | - Lawrence M Nogee
- Division of Neonatology, Johns Hopkins University School of Medicine, CMSC 6-104A, 600 N. Wolfe St., Baltimore, MD, 21287, USA
| | - Jacek Majewski
- McGill University and Génome Québec Innovation Centre, Montreal, QC, H3A 0G1, Canada
| | - Mika Rämet
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, PO Box 23, 90029, Oulu, Finland
- BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Mikko Hallman
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, PO Box 23, 90029, Oulu, Finland
| | - Reetta Hinttala
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, PO Box 5000, 90014, Oulu, Finland
- Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| |
Collapse
|
48
|
Brčić L, Gračan S, Barić A, Gunjača I, Torlak Lovrić V, Kolčić I, Zemunik T, Polašek O, Barbalić M, Punda A, Boraska Perica V. Association of Established Thyroid-stimulating Hormone and Free Thyroxine Genetic Variants with Hashimoto's Thyroiditis. Immunol Invest 2018; 46:625-638. [PMID: 28753406 DOI: 10.1080/08820139.2017.1337785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hashimoto's thyroiditis (HT), the most frequent autoimmune thyroid disease (AITD), is characterized by chronic inflammation of the thyroid gland that usually results in hypothyroidism. Thyroid-stimulating hormone (TSH) and free thyroxine (FT4) levels are used as clinical determinants of thyroid function. The main aim of this study was to explore the association of established TSH and FT4 genetic variants with HT. We performed a case-control analysis using 23 genetic markers in 200 HT patients and 304 controls. Additionally, we tested the association of selected variants with several thyroid-related quantitative traits in HT cases only. Two genetic variants showed nominal association with HT: rs11935941 near NR3C2 gene (p = 0.0034, OR = 0.57, 95% CI = 0.39-0.83) and rs1537424 near MBIP gene (p = 0.0169, OR = 0.72, 95% CI = 0.55-0.94). Additionally, three SNPs showed nominal association with thyroglobulin antibody (TgAb) levels: rs4804416 in INSR gene (p = 0.0073, β = -0.51), rs6435953 near IGFBP5 gene (p = 0.0081, β = 0.75), and rs1537424 near MBIP gene (p = 0.0117, β = 0.49). GLIS3 genetic variant rs10974423 showed nominal association with thyroid peroxidase antibody (TPOAb) levels (p = 0.0465, β = -0.56) and NRG1 genetic variant rs7825175 was nominally associated with thyroid gland volume (p = 0.0272, β = -0.18). All detected loci were previously related to thyroid function or pathology. Findings from our study suggest biological relevance of NR3C2 and MBIP with HT, although these loci require additional confirmation in a larger replication study.
Collapse
Affiliation(s)
- Luka Brčić
- a Department of Medical Biology , University of Split, School of Medicine , Split , Croatia
| | - Sanda Gračan
- b Department of Nuclear Medicine , University Hospital Split , Split , Croatia
| | - Ana Barić
- b Department of Nuclear Medicine , University Hospital Split , Split , Croatia
| | - Ivana Gunjača
- a Department of Medical Biology , University of Split, School of Medicine , Split , Croatia
| | | | - Ivana Kolčić
- c Department of Epidemiology , University of Split, School of Medicine , Split , Croatia
| | - Tatijana Zemunik
- a Department of Medical Biology , University of Split, School of Medicine , Split , Croatia
| | - Ozren Polašek
- c Department of Epidemiology , University of Split, School of Medicine , Split , Croatia
| | - Maja Barbalić
- a Department of Medical Biology , University of Split, School of Medicine , Split , Croatia
| | - Ante Punda
- b Department of Nuclear Medicine , University Hospital Split , Split , Croatia
| | - Vesna Boraska Perica
- a Department of Medical Biology , University of Split, School of Medicine , Split , Croatia
| |
Collapse
|
49
|
Benign hereditary chorea and deletions outside NKX2-1: What's the role of MBIP? Eur J Med Genet 2018; 61:581-584. [PMID: 29621620 DOI: 10.1016/j.ejmg.2018.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/09/2018] [Accepted: 03/27/2018] [Indexed: 11/22/2022]
Abstract
Heterozygous point mutations or deletions of the NKX2-1 gene cause benign hereditary chorea (BHC) or a various combinations of primary hypothyroidism, respiratory distress and neurological disorders. Deletions proximal to, but not encompassing, NKX2-1 have been described in few subjects with brain-lung-thyroid syndrome. We report on a three-generation Italian family, with 6 subjects presenting BHC and harboring a genomic deletion adjacent to NKX2-1 and including the gene MBIP, recently proposed to be relevant for the pathogenesis of brain-lung-thyroid syndrome. We observed a clear reduction of NKX2-1 transcript levels in fibroblasts from our patients compared to controls; this finding suggests that MBIP deletion affects NKX2-1 expression, mimicking haploinsufficiency caused by classical NKX2-1 related mutations.
Collapse
|
50
|
Abstract
Chorea is a symptom of a broad array of genetic, structural, and metabolic disorders. While chorea can result from systemic illness and damage to diverse brain structures, injury to the basal ganglia, especially the putamen or globus pallidus, appears to be a uniting features of these diverse neuropathologies. The timing of onset, rate of progression, and the associated neurological or systemic symptoms can often narrow the differential diagnosis to a few disorders. Recognizing the correct etiology for childhood chorea is critical, as numerous disorders in this category are potentially curable, or are remediable, with early treatment.
Collapse
Affiliation(s)
- Claudio M de Gusmao
- Department of Neurology, Boston Children's Hospital, Boston, MA; Department of Neurology, Brigham and Women's Hospital, Boston, MA
| | - Jeff L Waugh
- Department of Neurology, Boston Children's Hospital, Boston, MA; Department of Neurology, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|