1
|
Du J, Yang Y, Wei D, Wu J, Tian C, Hu Q, Bian H, Cheng C, Zhai X. The role of DUOXA2 in the clinical diagnosis of paediatric congenital hypothyroidism. Ann Med 2025; 57:2440121. [PMID: 39673194 PMCID: PMC11648137 DOI: 10.1080/07853890.2024.2440121] [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: 08/02/2024] [Revised: 10/03/2024] [Accepted: 11/07/2024] [Indexed: 12/16/2024] Open
Abstract
Background: Congenital hypothyroidism (CH) is a common metabolic disorder in children that can impact growth and neurodevelopment, particularly during infancy and early childhood. DUOXA2, a DUOX maturation factor, plays a crucial role in the maturation and activation of dual oxidase DUOX2 (a member of the NADPH oxidase family). DUOX2 can correctly migrate to the plasma membrane from the endoplasmic reticulum (ER) with the help of DUOXA2, and the two proteins together form a stable complex that promotes hydrogen peroxide (H2O2) generation in the synthesis of thyroid hormones. Genetic alterations in DUOXA2 lead to defects function of DUOX2 protein causing inherited CH. Objectives: This review discusses the relationship between DUOXA2 and CH, including the pathogenic mechanisms of CH in children caused by DUOXA2 mutations and the possibility or promise of DUOXA2 gene screening as a diagnostic marker for CH in the clinic. Methods: The review synthesizes current research on the biological role of DUOXA2 and DUOX2 in thyroid hormone synthesis, the molecular impact of DUOXA2 mutations, and the clinical implications of genetic screening for CH. Results: Mutations in DUOXA2 disrupt this process of H2O2 generation in the synthesis of thyroid hormones , leading to inherited CH. Early identification through DUOXA2 gene screening could improve diagnostic accuracy, which facilitates early intervention and personalized treatment. Conclusions: DUOXA2 gene screening holds promise for enhancing diagnostic accuracy in CH. However, it cannot be used as a sole diagnostic indicator, and to optimize diagnostic sensitivity, it should be combined with the screening of other relevant genetic mutations and diagnostic tools. Further research is needed to refine screening protocols and explore therapeutic options.
Collapse
Affiliation(s)
- Jiani Du
- Medical School of Yan’an University, Shaanxi, China
| | - Yanling Yang
- Medical School of Yan’an University, Shaanxi, China
| | - Ding Wei
- Medical School of Yan’an University, Shaanxi, China
| | - Jiajun Wu
- Medical School of Yan’an University, Shaanxi, China
| | | | - Qianqian Hu
- Medical School of Yan’an University, Shaanxi, China
| | - Hongyan Bian
- Medical School of Yan’an University, Shaanxi, China
| | - Chen Cheng
- Medical School of Yan’an University, Shaanxi, China
| | - Xiaoyan Zhai
- Medical School of Yan’an University, Shaanxi, China
| |
Collapse
|
2
|
Huang X, Shao Q, Weng S, Chen W, Yuan W, Tan J, Yang X, Su X. Mutation spectra and genotype‑phenotype analysis of congenital hypothyroidism in a neonatal population. Biomed Rep 2025; 22:30. [PMID: 39720292 PMCID: PMC11668128 DOI: 10.3892/br.2024.1908] [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: 06/13/2024] [Accepted: 10/07/2024] [Indexed: 12/26/2024] Open
Abstract
Congenital hypothyroidism (CH) is a common neonatal endocrine disorder that is characterized by irreversible neurodevelopmental and growth retardation due to insufficient biosynthesis of thyroid hormones at birth. Determining the causative genetic variants in infants is important for neonatal management. It was aimed to evaluate the variant frequencies and spectrum of CH in the neonatal population of Foshan, China. A total of 105 unrelated patients with CH and 138 controls from a neonatal screening program in Foshan, China were selected. A multiplex PCR amplification-based capture panel was performed which targeted the exon regions of 30 CH-related genes. Next-generation sequencing data were processed using an in-house bioinformatics system. A total of 91 variants distributed across 16 genes were identified in 74.29% (78/105) of the patients, of which 16 were novel variants and 75 were known variants. The most frequently mutated gene was DOUX2, followed by TG, TSHR and TPO. Specifically, DUOX2 variants p.Lys530Ter, p.Arg683Leu, p.Arg1110Gln, and IVS28 + 1G>T were highly recurrent in the cohort of the present study. Bi-allelic variants in DUOX2, TSHR and TPO were identified in 24.76% (26/105) of the patients. Monoallelic variants were identified in 28.57% (30/105) of the patients. Oligogenic variants were identified in 19.05% (20/105) of the patients. The most common variant combinations of oligogenic variants were DUOX2 and TG, and DUOX2 and SLC26A4. In addition, 2 patients harbored tri-allelic and tetra-allelic variants in DUOX2, respectively. In conclusion, DUOX2, TG, TSHR and TPO variants were the most common genetic defects in patients with CH in the neonatal population of Foshan. Specifically, biallelic DUOX2 variants were highly prevalent in the cohort. Further, the investigation provided a variant spectrum of CH-related genes and identified novel variants, which may allow for an improved understanding of the underlying genetic etiology of CH and provide evidence for further molecular epidemiological investigations that can guide preventive and therapeutic programs.
Collapse
Affiliation(s)
- Xiang Huang
- Newborn Screening Center, Foshan Women and Children Hospital, Foshan, Guangdong 528000, P.R. China
- Prenatal Diagnosis Center, Foshan Women and Children Hospital, Foshan, Guangdong 528000, P.R. China
| | - Qiaoyi Shao
- Newborn Screening Center, Foshan Women and Children Hospital, Foshan, Guangdong 528000, P.R. China
| | - Shi Weng
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wenfang Chen
- Newborn Screening Center, Foshan Women and Children Hospital, Foshan, Guangdong 528000, P.R. China
| | - Weixi Yuan
- Newborn Screening Center, Foshan Women and Children Hospital, Foshan, Guangdong 528000, P.R. China
| | - Jiayu Tan
- Emergency Department, Foshan Women and Children Hospital, Foshan, Guangdong 528000, P.R. China
| | - Xuexi Yang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xi Su
- Newborn Screening Center, Foshan Women and Children Hospital, Foshan, Guangdong 528000, P.R. China
| |
Collapse
|
3
|
Kahssay M, Ngwiri T. High yield of congenital hypothyroidism among infants attending Children Hospital, Nairobi, Kenya. Facility based study in the absence of newborn screening. J Pediatr Endocrinol Metab 2025; 38:51-57. [PMID: 39570906 DOI: 10.1515/jpem-2024-0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVES Congenital hypothyroidism (CHT) is a treatable cause of intellectual disability. Late diagnosis and delayed initiation of treatment leads to irreversible neurodevelopmental and intellectual disability. Thus, newborn screening is crucial. However, 71 % of babies are born in an area with no established newborn screening program and Kenya is not an exception. We aimed to determine the incidence of CHT, developmental outcomes of patients in the absence of newborn screening. METHODS A retrospective data of subjects who met the inclusion criteria, newborn and infants from 3 days to 2 years whose thyroid function test (TFT) was undertaken during well baby visit or clinical suspicion of CHT were collected. Laboratory reference range for age was used to interpret the result and TSH>10 Uiu/mL after 6 weeks of life is considered abnormal according to ESPE guideline. Developmental outcome of children was collected from patient file documented by primary physician and parental concern. RESULTS Of 1,426 children met inclusion criteria, 90 had elevated TSH. Out of which 70 repeat TFT showed normal TSH and free T4. The incidence of abnormal TSH across the different age groups was 2.4 , 7.2 and 10.5 % for ages 0-29 days, 1-11 months, and 1-2 years, respectively with p-value of =0.0002. While 20 cases with CHT identified with incidence of 14 per 1,000 children (1.4 %; 95 % CI: 0.9-2.1 %). Out this 12 (60 %) had poor developmental outcomes. Down's syndrome was the common associated condition 9/20 (45 %). All cases were primary CHT. CONCLUSIONS This study shows high incidence of CHT in a small cohort of patients over 5-year period with poor development outcome.
Collapse
Affiliation(s)
- Menbere Kahssay
- Department of Paediatrics and Child Health, 156412 Aga Khan University , Nairobi, Kenya
| | | |
Collapse
|
4
|
Ye L, Zhang Y, Feng J, Huang C, Wang X, Han L, Huang Y, Zou H, Zhu B, Miao J. Newborn Genetic Screening Improves the Screening Efficiency for Congenital Hypothyroidism: A Prospective Multicenter Study in China. Int J Neonatal Screen 2024; 10:78. [PMID: 39728398 DOI: 10.3390/ijns10040078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024] Open
Abstract
Newborn congenital hypothyroidism (CH) screening has been widely used worldwide. The objective of this study was to evaluate the effectiveness of applying biochemical and gene panel sequencing as screening tests for CH and to analyze the mutation spectrum of CH in China. Newborns were prospectively recruited from eight hospitals in China between February and December 2021. Clinical characteristics were collected. Second-generation sequencing was used to detect four CH-related genes, and the genetic patterns of the pathogenic genes were analyzed. We analyzed the relationship between genotype and biochemical phenotype. A total of 29,601 newborns were screened for CH. Gene panel sequencing identified 18 patients, including 10 patients affected by biochemically and genetically screened disorders and 8 patients affected by solely genetically screened disorders. The predictive positive value of genetic screening was 34.62%, which was much greater than that of biochemical screening alone (17.99%). A total of 94 cases of congenital thyroid dysfunction were confirmed by biochemical and genetic screening, including 30 CHs and 64 isolated hyperthyrotropinemia (HTT), with an incidence of 1/987 for CH and 1/463 for HTT, and a total incidence of 1/315 for hypothyroidism. The incidence rate and number of patients in Jinan were the highest, and the incidence rates in Shijiazhuang and Shanghai were the lowest. The gene mutation rate in this study was 19.1%, mainly DUOX2 mutation. The most common variant of DUOX2 was c.1588A>T(p.Lys530*). There was only a difference in sFT4 between groups with gene mutations and those without mutations. Genetic screening is a supplement to biochemical screening. Combining biochemical screening with genetic screening is useful for improving screening efficiency. The incidence of CH in China according to a multicenter study of nearly 30,000 NBS surveys was 1/315. DUOX2 gene mutations are commonly detected in these patients.
Collapse
Affiliation(s)
- Liang Ye
- Department of Pediatrics, Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Yinhong Zhang
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health in Western China, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650100, China
| | - Jizhen Feng
- Department of Genetic, Shijiazhuang Maternal and Child Health Hospital, Shijiazhuang 050006, China
| | - Cidan Huang
- Neonatal Disease Screening Center, Hainan Women and Children's Medical Center, Haikou 570206, China
| | - Xiaohua Wang
- Department of Genetics, Inner Mongolia Maternity and Child Health Care Hospital, Hohhot 750306, China
| | - Lianshu Han
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yonglan Huang
- Department of Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Hui Zou
- Neonatal Disease Screening Center, Jinan Maternity and Child Health Hospital Affiliated to Shandong First Medical University, Jinan 250001, China
| | - Baosheng Zhu
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health in Western China, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming 650100, China
| | - Jingkun Miao
- Department of Pediatrics, Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
5
|
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
|
6
|
Kurnaz E, Türkyılmaz A, Yaralı O, Dönmez AS, Çayır A. Genetic Analyses in a Cohort of Pediatric Patients with Congenital Hypothyroidism Based on Congenital Hypothyroidism Consensus Guideline. Horm Res Paediatr 2024:1-15. [PMID: 39378853 DOI: 10.1159/000541898] [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: 05/15/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION Pathogenic variants in the genes involved in the formation of thyroid tissue and thyroid hormone secretion have been reported to cause congenital hypothyroidism (CH) in some cases. This study aimed to evaluate the clinical and genetic findings of CH cases thought to be due to genetic variants. METHODS The study included cases whose genetic analysis was performed in accordance with the Congenital Hypothyroidism: A 2020-2021 Consensus Guidelines Update Guidelines recommendations criteria and analyzed them using the next-generation sequencing panel. RESULTS Sixty one Turkish patients from 45 families were included in the study. The overall frequency of variant detection was 37.7% (out of 45 families, 17 had a positive mutation). Segregation was carried out for all families with positive variants. Variants in the TPO gene are the most frequently encountered, and this situation was identified in 10 families. Variants followed this in the TSHR gene in 7 families, variants in the DUOX2 gene in 5 families, and two variants in the TG and NKX2-1 genes in 2 families each, which are six novel variants. Furthermore, among the NKX2-1 cases, one had thyroid involvement only, while the other had chorea only. We did not find differences between cases with detected mutations and mutation-negative cases regarding gender, neonatal/perinatal parameters, initial thyroid function values, and thyroid morphology. CONCLUSION In the current investigation, rare new variations in genes known to be related to CH were discovered, adding to the molecular genetic spectrum. When we compare the overall variant detection frequency, the selection criterion for genetic analysis based on the current guidelines is quite rational, considering the benefits and costs, on the other hand, present in new genes awaiting discovery. Also, TSHR mutations are likely to be common and may account for more than 5% of thyroid dysgenesis cases if we include nonfamilial thyroid dysgenesis.
Collapse
Affiliation(s)
- Erdal Kurnaz
- Department of Pediatric Endocrinology, Etlik City Hospital, University of Health Sciences, Ankara, Turkey
| | - Ayberk Türkyılmaz
- Department of Medical Genetics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Oğuzhan Yaralı
- Department of Medical Genetics, Erzurum City Hospital, Erzurum, Turkey
| | - Ayşe Sena Dönmez
- Department of Pediatrics, Erzurum City Hospital, Erzurum, Turkey
| | - Atilla Çayır
- Department of Pediatric Endocrinology, Erzurum City Hospital, University of Health Sciences, Erzurum, Turkey
| |
Collapse
|
7
|
Cheetham T, Wood C. Paediatric thyroid disease. Clin Endocrinol (Oxf) 2024. [PMID: 39072866 DOI: 10.1111/cen.15110] [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: 08/23/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/30/2024]
Abstract
The spectrum of thyroid disorders presenting to paediatricians is different to that seen by adult physicians. Referrals reflect cases detected by the neonatal screening programme for congenital hypothyroidism and many of the inherited defects of thyroid hormone generation or action will be manifest in early life. Autoimmune thyroid disease can be particularly challenging to manage in the young and the potential impact of thyroid status on neurodevelopment and schooling are key considerations throughout childhood and adolescence.
Collapse
Affiliation(s)
- Timothy Cheetham
- Newcastle University and Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Claire Wood
- Newcastle University and Great North Children's Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
8
|
Castets S, Albarel F, Bachelot A, Brun G, Bouligand J, Briet C, Bui Quoc E, Cazabat L, Chabbert-Buffet N, Christin-Maitre S, Courtillot C, Cuny T, De Filippo G, Donadille B, Illouz F, Pellegrini I, Reznik Y, Saveanu A, Teissier N, Touraine P, Vantyghem MC, Vergier J, Léger J, Brue T, Reynaud R. Position statement on the diagnosis and management of congenital pituitary deficiency in adults: The French National Diagnosis and Treatment Protocol (NDTP). ANNALES D'ENDOCRINOLOGIE 2024; 85:327-339. [PMID: 38452869 DOI: 10.1016/j.ando.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Pituitary deficiency, or hypopituitarism, is a rare chronic disease. It is defined by insufficient synthesis of one or more pituitary hormones (growth hormone, TSH, ACTH, LH-FSH, prolactin), whether or not associated with arginine vasopressin deficiency (formerly known as diabetes insipidus). In adult patients, it is usually acquired (notably during childhood), but can also be congenital, due to abnormal pituitary development. The present study focuses on congenital pituitary deficiency in adults, from diagnosis to follow-up, including special situations such as pregnancy or the elderly. The clinical presentation is highly variable, ranging from isolated deficit to multiple deficits, which may be part of a syndromic form or not. Diagnosis is based on a combination of clinical, biological (assessment of all hormonal axes), radiological (brain and hypothalamic-pituitary MRI) and genetic factors. Treatment consists in hormonal replacement therapy, adapted according to the period of life and the deficits, which may be progressive. Comorbidities, risk of complications and acute decompensation, and the impact on fertility and quality of life all require adaptative multidisciplinary care and long-term monitoring.
Collapse
Affiliation(s)
- Sarah Castets
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France.
| | - Frédérique Albarel
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Conception, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Anne Bachelot
- IE3M, ICAN, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Centre de Référence des Pathologies Gynécologiques Rares, hôpital Pitié-Salpêtrière, AP-HP, Paris, France; Sorbonne université, Paris, France
| | - Gilles Brun
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Hôpital Européen, Pôle imagerie médicale, 13003, Marseille, France
| | - Jérôme Bouligand
- Molecular Genetic, Pharmacogenetic and Hormonology, Kremlin-Bicêtre Hospital, Paris-Saclay University, AP-HP, Le Kremlin-Bicêtre, France
| | - Claire Briet
- Département d'endocrinologie-diabétologie nutrition, Centre de référence des maladies rares de la Thyroïde et des Récepteurs Hormonaux, Endo-ERN centre for rare endocrine diseases, CHU d'Angers, 4, rue larrey, 49100 Angers, France; Laboratoire MITOVASC, UMR CNRS 6015, Inserm 1083, Université d'Angers, rue Roger Amsler, 49100 Angers, France
| | - Emmanuelle Bui Quoc
- Ophthalmology Department, Robert-Debré University Hospital, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Laure Cazabat
- Department of Endocrinology, Diabetology and Nutrition, Ambroise Paré Hospital, AP-HP, UVSQ, Boulogne-Billancourt, France
| | - Nathalie Chabbert-Buffet
- Department of Gynecology and Obstetrics, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, 75020 Paris, France
| | - Sophie Christin-Maitre
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Carine Courtillot
- IE3M, ICAN, Department of Endocrinology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Centre de Référence des Pathologies Gynécologiques Rares, hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Thomas Cuny
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Gianpaolo De Filippo
- Service d'endocrinologie et diabétologie pédiatrique, centre de référence des maladies endocriniennes de la croissance et du développement, hôpital universitaire Robert-Debré, université Paris Cité, Assistance publique-Hôpitaux de Paris, Paris, France
| | - Bruno Donadille
- Department of Endocrinology, Diabetology and Reproductive Medicine, Centre de Référence des Maladies Endocriniennes Rares de la Croissance et du Développement (CMERC), Centre de Compétence HYPO, Hôpital Saint-Antoine, Sorbonne University, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - Frédéric Illouz
- Département d'endocrinologie-diabétologie nutrition, Centre de référence des maladies rares de la Thyroïde et des Récepteurs Hormonaux, Endo-ERN centre for rare endocrine diseases, CHU d'Angers, 4, rue larrey, 49100 Angers, France; Laboratoire MITOVASC, UMR CNRS 6015, Inserm 1083, Université d'Angers, rue Roger Amsler, 49100 Angers, France
| | - Isabelle Pellegrini
- Service d'endocrinologie, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Conception, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Yves Reznik
- Endocrinology and Diabetes Department, CHU Côte de Nacre and Unicaen, Caen Cedex, France
| | - Alexandru Saveanu
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France
| | - Natacha Teissier
- Department of Pediatric Otolaryngology, Robert Debré Hospital, AP-HP Nord, Paris, France
| | - Philippe Touraine
- Service d'endocrinologie et médecine de la reproduction, centre de maladies endocrinennes rares de la croissance et du développement, médecine-hôpital Pitié-Salpêtrière, Sorbonne université, Paris, France
| | - Marie-Christine Vantyghem
- Service d'endocrinologie, diabétologie et maladies métaboliques, CHRU de Lille, rue Polonowski, Lille cedex, France
| | - Julia Vergier
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Julianne Léger
- Service d'endocrinologie et diabétologie pédiatrique, centre de référence des maladies endocriniennes de la croissance et du développement, hôpital universitaire Robert-Debré, université Paris Cité, Assistance publique-Hôpitaux de Paris, Paris, France; Université Paris Cité, NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1141, Paris, France
| | - Thierry Brue
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France; Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Inserm, MMG, Laboratory of Molecular Biology, Hospital La Conception, Aix-Marseille University, AP-HM, Marseille, France
| | - Rachel Reynaud
- Service de pédiatrie multidisciplinaire, centre de référence des maladies rares de l'hypophyse HYPO, hôpital de la Timone Enfants, Assistance publique-Hôpitaux de Marseille (AP-HM), 13005 Marseille, France; Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Assistance Publique Hôpitaux de Marseille, Reference Center for Rare Pituitary Diseases HYPO, Assistance-Publique des Hôpitaux de Marseille, Laboratory of Molecular Biology, Conception Hospital, Marseille, France; Inserm, MMG, Laboratory of Molecular Biology, Hospital La Conception, Aix-Marseille University, AP-HM, Marseille, France
| |
Collapse
|
9
|
Yang M, Cao Z, Zhu W, Feng X, Zhou J, Liu J, Zhong Y, Zhou Y, Mei H, Cai X, Hu L, Zhou A, Xiao H. Associations between OGTT results during pregnancy and offspring TSH levels: a birth cohort study. BMC Pregnancy Childbirth 2024; 24:375. [PMID: 38760653 PMCID: PMC11100047 DOI: 10.1186/s12884-024-06554-4] [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: 01/25/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Limited evidence exists regarding the association between gestational diabetes mellitus (GDM) and elevated levels of thyroid-stimulating hormone (TSH) in newborns. Therefore, this study aimed to investigate the potential risk of elevated TSH levels in infants exposed to maternal GDM, considering the type and number of abnormal values obtained from the 75-gram oral glucose tolerance test (OGTT). METHODS A population-based, prospective birth cohort study was conducted in Wuhan, China. The study included women who underwent GDM screening using a 75-g OGTT. Neonatal TSH levels were measured via a time-resolved immunofluorescence assay. We estimated and stratified the overall risk (adjusted Risk Ratio [RR]) of elevated TSH levels (defined as TSH > 10 mIU/L or > 20 mIU/L) in offspring based on the type and number of abnormal OGTT values. RESULTS Out of 15,236 eligible mother-offspring pairs, 11.5% (1,753) of mothers were diagnosed with GDM. Offspring born to women diagnosed with GDM demonstrated a statistically significant elevation in TSH levels when compared to offspring of non-GDM mothers, with a mean difference of 0.20 [95% CI: 0.04-0.36]. The incidence of elevated TSH levels (TSH > 10 mIU/L) in offspring of non-GDM women was 6.3 per 1,000 live births. Newborns exposed to mothers with three abnormal OGTT values displayed an almost five-fold increased risk of elevated TSH levels (adjusted RR 4.77 [95% CI 1.64-13.96]). Maternal fasting blood glucose was independently and positively correlated with neonatal TSH levels and elevated TSH status (TSH > 20 mIU/L). CONCLUSIONS For newborns of women with GDM, personalized risk assessment for elevated TSH levels can be predicated on the type and number of abnormal OGTT values. Furthermore, fasting blood glucose emerges as a critical predictive marker for elevated neonatal TSH status.
Collapse
Affiliation(s)
- Meng Yang
- Institute of Maternal and Child Health, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Zhongqiang Cao
- Institute of Maternal and Child Health, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Wanting Zhu
- Department of Obstetrics, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyuan Feng
- Department of echocardiography, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jieqiong Zhou
- Department of Obstetrics, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jiuying Liu
- Department of Obstetrics, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Zhong
- Department of Obstetrics, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhou
- Department of Obstetrics, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Mei
- Institute of Maternal and Child Health, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xiaonan Cai
- Institute of Maternal and Child Health, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Liqin Hu
- Institute of Maternal and Child Health, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Aifen Zhou
- Institute of Maternal and Child Health, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China.
| | - Han Xiao
- Institute of Maternal and Child Health, Tongji Medical College, Wuhan Children's Hospital (Wuhan Maternal and Child Health care Hospital, Huazhong University of Science and Technology, Wuhan, 430000, China.
| |
Collapse
|
10
|
Cortese A, Vegezzi E, Houlden H. Contraction or sequence variant of an intergenic repeat-Alu element leads to inherited thyroid disease. Nat Genet 2024; 56:738-739. [PMID: 38714867 DOI: 10.1038/s41588-024-01723-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Affiliation(s)
- Andrea Cortese
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, UK.
- Department of Brain and Behaviour Sciences, University of Pavia, Pavia, Italy.
| | | | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| |
Collapse
|
11
|
Grasberger H, Dumitrescu AM, Liao XH, Swanson EG, Weiss RE, Srichomkwun P, Pappa T, Chen J, Yoshimura T, Hoffmann P, França MM, Tagett R, Onigata K, Costagliola S, Ranchalis J, Vollger MR, Stergachis AB, Chong JX, Bamshad MJ, Smits G, Vassart G, Refetoff S. STR mutations on chromosome 15q cause thyrotropin resistance by activating a primate-specific enhancer of MIR7-2/MIR1179. Nat Genet 2024; 56:877-888. [PMID: 38714869 PMCID: PMC11472772 DOI: 10.1038/s41588-024-01717-7] [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: 07/30/2023] [Accepted: 03/14/2024] [Indexed: 05/22/2024]
Abstract
Thyrotropin (TSH) is the master regulator of thyroid gland growth and function. Resistance to TSH (RTSH) describes conditions with reduced sensitivity to TSH. Dominantly inherited RTSH has been linked to a locus on chromosome 15q, but its genetic basis has remained elusive. Here we show that non-coding mutations in a (TTTG)4 short tandem repeat (STR) underlie dominantly inherited RTSH in all 82 affected participants from 12 unrelated families. The STR is contained in a primate-specific Alu retrotransposon with thyroid-specific cis-regulatory chromatin features. Fiber-seq and RNA-seq studies revealed that the mutant STR activates a thyroid-specific enhancer cluster, leading to haplotype-specific upregulation of the bicistronic MIR7-2/MIR1179 locus 35 kb downstream and overexpression of its microRNA products in the participants' thyrocytes. An imbalance in signaling pathways targeted by these micro-RNAs provides a working model for this cause of RTSH. This finding broadens our current knowledge of genetic defects altering pituitary-thyroid feedback regulation.
Collapse
Affiliation(s)
- Helmut Grasberger
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Alexandra M Dumitrescu
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Elliott G Swanson
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Roy E Weiss
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Theodora Pappa
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Junfeng Chen
- Institute of Transformative Bio-Molecules (WPI-ITbM) and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takashi Yoshimura
- Institute of Transformative Bio-Molecules (WPI-ITbM) and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Phillip Hoffmann
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Rebecca Tagett
- Michigan Medicine BRCF Bioinformatics Core, University of Michigan, Ann Arbor, MI, USA
| | | | - Sabine Costagliola
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Jane Ranchalis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mitchell R Vollger
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrew B Stergachis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Jessica X Chong
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Michael J Bamshad
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Guillaume Smits
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, Brussels, Belgium
- Center of Human Genetics, Hôpital Erasme, Hôpital Universitaire de Bruxelles, and Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Gilbert Vassart
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, IL, USA.
- Committee on Genetics, The University of Chicago, Chicago, IL, USA.
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Danner E, Jääskeläinen J, Niuro L, Huopio H, Niinikoski H, Viikari L, Kero J, Sund R. Comorbidity in Congenital Hypothyroidism-A Nationwide, Population-based Cohort Study. J Clin Endocrinol Metab 2023; 108:e1695-e1701. [PMID: 37279943 PMCID: PMC10655533 DOI: 10.1210/clinem/dgad334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023]
Abstract
CONTEXT Patients with congenital hypothyroidism (CH) are affected more often than the general population by other chronic diseases and neurological difficulties. OBJECTIVE The aim of this nationwide population-based register study was to investigate the incidence of congenital malformations, comorbidities, and the use of prescribed drugs in patients with primary CH. METHODS The study cohort and matched controls were identified from national population-based registers in Finland. All diagnoses from birth until the end of 2018 were collected from the Care Register, and subject-specific prescription drug purchases were identified from The Prescription Register from birth until the end of 2017. RESULTS Diagnoses of neonatal and chronic diseases were collected for 438 full-term patients and 835 controls (median follow-up time 11.6 years; range, 0-23 years). Newborns with CH were more often found to have neonatal jaundice (11.2% and 2.0%; P < .001), hypoglycemia (8.9% and 2.8%; P < .001), metabolic acidemia (3.2% and 1.1%; P = .007), and respiratory distress (3.9% and 1.3%; P < .003) as compared to their matched controls.Congenital malformations were diagnosed in 66 of 438 (15.1%) CH patients and in 62 of 835 (7.4%) controls (P < .001). The most commonly affected extrathyroidal systems were the circulatory and musculoskeletal systems. The cumulative incidence of hearing loss and specific developmental disorders was higher among CH patients than controls. The use of antidepressant and antipsychotic drugs was similar in CH patients and their controls. CONCLUSION CH patients have more neonatal morbidity and congenital malformations than their matched controls. The cumulative incidence of neurological disorders is higher in CH patients. However, our results do not support the existence of severe psychiatric comorbidity.
Collapse
Affiliation(s)
- Emmi Danner
- Department of Pediatrics, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jarmo Jääskeläinen
- Department of Pediatrics, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Laura Niuro
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Hanna Huopio
- Department of Pediatrics, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Harri Niinikoski
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Liisa Viikari
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Jukka Kero
- Department of Pediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Reijo Sund
- School of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
13
|
Năstase L, Cristea O, Diaconu A, Stoicescu SM, Mohora R, Pascu BM, Tala ST, Roșca I. Two Cases of Congenital Hypothyroidism Revealing Thyroid Agenesis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1887. [PMID: 37893606 PMCID: PMC10608129 DOI: 10.3390/medicina59101887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023]
Abstract
Congenital hypothyroidism (CH) may have major detrimental effects on growth and neurological development, but early intervention leads to excellent outcomes. CH is classified as transient or permanent, primary or secondary, with primary CH being the most common neonatal endocrine disorder. Most patients with CH do not present any typical signs and symptoms of hypothyroidism shortly after birth, partly due to transplacental maternal thyroid hormone transfer and residual neonatal thyroid function. This paper reports on two CH cases. During the initial Neonatal Intensive Care Unit (NICU) admission phase, CH was not suspected due to nonspecific signs. The distinct characteristics of our cases are as follows: both infants were admitted to the NICU for respiratory distress syndrome, requiring invasive mechanical ventilation, and both were born to diabetic mothers. Following extubation, they both showed similar neurological issues, including reduced muscle tone and feeding difficulties. Initially, those symptoms were attributed to delayed clearance of analgesic and sedative medication. However, symptoms progressively worsened over time. Subsequent tests revealed both meeting CH diagnostic criteria: an unusual ultrasound indicating thyroid agenesis and abnormal hormone levels. Guided by the pediatric endocrinology team, prompt hormonal treatment was started with improvements in neurocognitive function and feeding. Usually, CH screening involves blood samples from healthy newborns at 2-3 days of life. Abnormal results require confirmation, prompting treatment within two weeks. Certain NICU-admitted infants face higher diagnosis delays, as seen in those two cases where CH screening was postponed. Thus, for all neonates with persistent pathologies unresponsive to standard etiological treatment, conducting a comprehensive anamnestic evaluation of the medical history, along with maternal preconceptional and prenatal nutrition, is recommended.
Collapse
Affiliation(s)
- Leonard Năstase
- Neonatology Department, National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 011061 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Octaviana Cristea
- Neonatology Department, National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 011061 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alexandra Diaconu
- Neonatology Department, National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 011061 Bucharest, Romania
| | - Silvia-Maria Stoicescu
- Neonatology Department, National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 011061 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ramona Mohora
- Neonatology Department, National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 011061 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Mihai Pascu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Endocrinology Department, National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 020395 Bucharest, Romania
| | - Simona Tania Tala
- Endocrinology Department, National Institute for Mother and Child Health “Alessandrescu-Rusescu”, 020395 Bucharest, Romania
| | - Ioana Roșca
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Neonatology Department, Clinical Hospital of Obstetrics and Gynecology “Prof. Dr. P. Sârbu”, 060251 Bucharest, Romania
| |
Collapse
|
14
|
Darrat M, Kayes L, Woodside JV, Mullan K, Abid N. Congenital hypothyroidism in Northern Ireland: 40 years' experience of national screening programme. Clin Endocrinol (Oxf) 2023; 99:409-416. [PMID: 37278153 DOI: 10.1111/cen.14940] [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: 02/16/2023] [Revised: 04/19/2023] [Accepted: 05/25/2023] [Indexed: 06/07/2023]
Abstract
OBJECTIVE The incidence of congenital hypothyroidism (CHT) has progressively increased in several regions around the world but has yet to be evaluated in Northern Ireland (NI). CHT screening programme was introduced in NI in 1980 and has had a relatively unchanged protocol since its inception. The purpose of the study was to evaluate the incidence of CHT in NI from 1981 to 2020 and to explore possible contributing factors to any changes seen over the 40-year period. DESIGN This was a retrospective database review of children diagnosed with CHT in NI between 1981 and 2020. Data was collected from the patients' medical (paper and electronic) records, including epidemiological, clinical, laboratory, and radiological features as well as outcomes at 3 years. RESULTS Of 800,404 new-borns who were screened for CHT in NI between January 1981 and March 2020, 471 were diagnosed with CHT. There was a steady and significant increase in incidence of CHT over time with an incidence of 26 cases per 100,000 livebirths in 1981 versus 71 cases per 100,000 in 2019 (p < .001). Of these 471, 77 new-borns (16%) were born preterm. The incidence of CHT was observed twice as much in female compared to male new-borns. Diagnostic imaging including radioisotope uptake and thyroid ultrasound scans were performed in 143 cases (30%). Of these, 101 (70%) cases had thyroid dysgenesis and 42 (30%) cases had thyroid dyshormonogenesis. There were 293 (62%) of 471 patients had confirmed permanent CHT, and 90 patients (19%) had transient CHT. Over that period at least 95% of the population were recorded as having United Kingdom/Ireland as country of birth. CONCLUSION Our findings demonstrate a nearly tripling of the CHT incidence observed over the last 40 years. This is against a background of a relatively stable population demographics. Future research should focus on the underlying cause(s) of this condition which may include changing environmental exposures in utero.
Collapse
Affiliation(s)
- Milad Darrat
- Regional centre for endocrinology and diabetes, Royal Victoria Hospital, Belfast, UK
| | - Lucy Kayes
- Regional centre for endocrinology and diabetes, Royal Victoria Hospital, Belfast, UK
- Centre for Public Health, Queen's University, Belfast, UK
| | | | - Karen Mullan
- Regional centre for endocrinology and diabetes, Royal Victoria Hospital, Belfast, UK
| | - Noina Abid
- Paediatric Endocrinology Department, Royal Belfast Hospital for Sick Children, Belfast, UK
| |
Collapse
|
15
|
Wu FY, Yang RM, Zhang HY, Zhan M, Tu PH, Fang Y, Zhang CX, Song SY, Dong M, Cui RJ, Liu XY, Yang L, Yan CY, Sun F, Zhang RJ, Wang Z, Liang J, Song HD, Cheng F, Zhao SX. Pathogenic variations in MAML2 and MAMLD1 contribute to congenital hypothyroidism due to dyshormonogenesis by regulating the Notch signalling pathway. J Med Genet 2023; 60:874-884. [PMID: 36898841 DOI: 10.1136/jmg-2022-108866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/25/2023] [Indexed: 03/12/2023]
Abstract
BACKGROUND In several countries, thyroid dyshormonogenesis is more common than thyroid dysgenesis in patients with congenital hypothyroidism (CH). However, known pathogenic genes are limited to those directly involved in hormone biosynthesis. The aetiology and pathogenesis of thyroid dyshormonogenesis remain unknown in many patients. METHODS To identify additional candidate pathogenetic genes, we performed next-generation sequencing in 538 patients with CH and then confirmed the functions of the identified genes in vitro using HEK293T and Nthy-ori 3.1 cells, and in vivo using zebrafish and mouse model organisms. RESULTS We identified one pathogenic MAML2 variant and two pathogenic MAMLD1 variants that downregulated canonical Notch signalling in three patients with CH. Zebrafish and mice treated with N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butylester, a γ-secretase inhibitor exhibited clinical manifestations of hypothyroidism and thyroid dyshormonogenesis. Through organoid culture of primary mouse thyroid cells and transcriptome sequencing, we demonstrated that Notch signalling within thyroid cells directly affects thyroid hormone biosynthesis rather than follicular formation. Additionally, these three variants blocked the expression of genes associated with thyroid hormone biosynthesis, which was restored by HES1 expression. The MAML2 variant exerted a dominant-negative effect on both the canonical pathway and thyroid hormone biosynthesis. MAMLD1 also regulated hormone biosynthesis through the expression of HES3, the target gene of the non-canonical pathway. CONCLUSIONS This study identified three mastermind-like family gene variants in CH and revealed that both canonical and non-canonical Notch signalling affected thyroid hormone biosynthesis.
Collapse
Affiliation(s)
- Feng-Yao Wu
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Meng Yang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Yang Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Zhan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Hui Tu
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Fang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cao-Xu Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Yang Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mei Dong
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren-Jie Cui
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Yu Liu
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Yang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Yan Yan
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Sun
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Jia Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liang
- Department of Endocrinology, The Central Hospital of Xuzhou Affiliated to Xuzhou Medical College, Xuzhou, China
| | - Huai-Dong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Cheng
- Department of Laboratory Medicine, Fujian Provincial Maternity and Children's Hospital, Fuzhou, China
| | - Shuang-Xia Zhao
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, State Key Laboratory of Medical Genomics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Nagendra L, Bhavani N, Pavithran PV, Shenoy M, Menon UV, Abraham N, Nair V, Kumar H. Etiological Profile, Targeted Levothyroxine Dosing and Impact of Partial Newborn Screening in Congenital Hypothyroidism-A Single Centre Experience. Indian J Endocrinol Metab 2023; 27:445-449. [PMID: 38107726 PMCID: PMC10723602 DOI: 10.4103/ijem.ijem_314_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/01/2023] [Accepted: 04/22/2023] [Indexed: 12/19/2023] Open
Abstract
Background Congenital hypothyroidism (CH) is the most common cause of preventable intellectual disability. Newborn screening (NBS) for CH has been in vogue in many parts of the world since 1970, but despite its well-known benefits, many developing countries including India have not been able to establish universal NBS for CH till date. Objective The aim of this study was to review the clinical aspects of congenital hypothyroidism in a tertiary care university referral teaching hospital, focusing on aetiology of CH, predictors of permanence, optimal targeted dose strategies based on aetiology and the effect of newborn screening on the time to diagnosis. Material and Methods The electronic medical records of 233 children with CH referred to our centre between January 2009 and December 2019 were analysed. A partial NBS was established in the state in 2012. Results Dyshormonogenesis (57.5%) was the most common aetiology of CH. The incidence of transient CH in children with a gland in situ (GIS) was 35%. Levothyroxine (LT-4) dose of >2.75 μg/kg/day (sensitivity 76.5, specificity 72), >2.15 μg/kg/day (sensitivity 82.4, specificity 61.9) and >1.85 μg/kg/day (sensitivity 76.5, specificity 61.9) at years 1, 2 and 3, respectively, were predictors of permanent CH. An initial LT-4 dose ≥8 μg/kg was sufficient and very seldom led to undertreatment in children with dyshormonogenesis. On the contrary, even doses ≥13 mcg/kg/day led to frequent undertreatment in children with thyroid dysgenesis. After the introduction of newborn screening, the median age at diagnosis came down from 45 days (IQR 14-180 days) to ten days (IQR 3-12 days). Conclusion Targeted dosing based on aetiology of CH may be more appropriate to optimise outcomes. The time to diagnosis of CH reduced significantly after the adoption of even a partial NBS program highlighting the urgent need for implementation of the same in resource poor settings.
Collapse
Affiliation(s)
- Lakshmi Nagendra
- Department of Endocrinology, JSS Medical College and Hospital, JSS Academy of Higher Education and Research, Cochin, Kerala, India
| | - Nisha Bhavani
- Department of Endocrinology, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| | - Praveen V. Pavithran
- Department of Endocrinology, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| | - Mohan Shenoy
- Department of Endocrinology, Sree Gokulam Medical College and Research Foundation, Thiruvananthapuram, Kerala, India
| | - Usha V. Menon
- Department of Endocrinology, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| | - Nithya Abraham
- Department of Endocrinology, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| | - Vasantha Nair
- Department of Endocrinology, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| | - Harish Kumar
- Department of Endocrinology, Amrita Institute of Medical Sciences, Cochin, Kerala, India
| |
Collapse
|
17
|
Levaillant L, Bouhours-Nouet N, Illouz F, Amsellem Jager J, Bachelot A, Barat P, Baron S, Bensignor C, Brac De La Perriere A, Braik Djellas Y, Caillot M, Caldagues E, Campas MN, Caquard M, Cartault A, Cheignon J, Decrequy A, Delemer B, Dieckmann K, Donzeau A, Doye E, Fradin M, Gaudillière M, Gatelais F, Gorce M, Hazart I, Houcinat N, Houdon L, Ister-Salome M, Jozwiak L, Jeannoel P, Labarthe F, Lacombe D, Lambert AS, Lefevre C, Leheup B, Leroy C, Maisonneuve B, Marchand I, Marquant E, Muszlak M, Pantalone L, Pochelu S, Quelin C, Radet C, Renoult-Pierre P, Reynaud R, Rouleau S, Teinturier C, Thevenon J, Turlotte C, Valle A, Vierge M, Villanueva C, Ziegler A, Dieu X, Bouzamondo N, Rodien P, Prunier-Mirebeau D, Coutant R. The Severity of Congenital Hypothyroidism With Gland-In-Situ Predicts Molecular Yield by Targeted Next-Generation Sequencing. J Clin Endocrinol Metab 2023; 108:e779-e788. [PMID: 36884306 PMCID: PMC10438870 DOI: 10.1210/clinem/dgad119] [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: 04/12/2022] [Revised: 02/07/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023]
Abstract
INTRODUCTION Congenital hypothyroidism with gland-in-situ (CH-GIS) is usually attributed to mutations in the genes involved in thyroid hormone production. The diagnostic yield of targeted next-generation sequencing (NGS) varied widely between studies. We hypothesized that the molecular yield of targeted NGS would depend on the severity of CH. METHODS Targeted NGS was performed in 103 CH-GIS patients from the French national screening program referred to the Reference Center for Rare Thyroid Diseases of Angers University Hospital. The custom targeted NGS panel contained 48 genes. Cases were classified as solved or probably solved depending on the known inheritance of the gene, the classification of the variants according to the American College of Medical Genetics and Genomics, the familial segregation, and published functional studies. Thyroid-stimulating hormone at CH screening and at diagnosis (TSHsc and TSHdg) and free T4 at diagnosis (FT4dg) were recorded. RESULTS NGS identified 95 variants in 10 genes in 73 of the 103 patients, resulting in 25 solved cases and 18 probably solved cases. They were mainly due to mutations in the TG (n = 20) and TPO (n = 15) genes. The molecular yield was, respectively, 73% and 25% if TSHsc was ≥ and < 80 mUI/L, 60% and 30% if TSHdg was ≥ and < 100 mUI/L, and 69% and 29% if FT4dg was ≤ and > 5 pmol/L. CONCLUSION NGS in patients with CH-GIS in France found a molecular explanation in 42% of the cases, increasing to 70% when TSHsc was ≥ 80 mUI/L or FT4dg was ≤ 5 pmol/L.
Collapse
Affiliation(s)
- Lucie Levaillant
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
| | - Natacha Bouhours-Nouet
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
| | - Frédéric Illouz
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
- Department of Endocrinology, Diabetes and Nutrition, University Hospital of Angers, 49000 Angers, France
| | - Jessica Amsellem Jager
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
| | - Anne Bachelot
- Department of Endocrinology and Reproductive Medicine, Hôpital Pitié-Salpêtrière, ICAN, 75651 Paris, France
| | - Pascal Barat
- Pediatric Endocrinology, CHU de Bordeaux, 33000 Bordeaux, France
| | - Sabine Baron
- Pediatrics Department, CHU Nantes, 44000 Nantes, France
| | | | - Aude Brac De La Perriere
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, 69677 Bron, France
| | - Yasmine Braik Djellas
- Department of Endocrinology and Reproductive Medicine, Hôpital Pitié-Salpêtrière, ICAN, 75651 Paris, France
| | - Morgane Caillot
- Pediatrics Department, CH de Martigues, 13500 Martigues, France
| | | | | | | | - Audrey Cartault
- Endocrine, Genetics, Bone Diseases, and Paediatric Gynecology Unit, Children's Hospital, CHU Toulouse, 31059 Toulouse, France
| | - Julie Cheignon
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
| | - Anne Decrequy
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
| | - Brigitte Delemer
- Department of Endocrinology, Diabetes and Nutrition, CHU de Reims-Hôpital Robert-Debré, 51100 Reims, France
| | | | - Aurélie Donzeau
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
| | | | - Mélanie Fradin
- Service de Génétique, CLAD Ouest, CHU Rennes, 35200 Rennes, France
| | - Mélanie Gaudillière
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, 69677 Bron, France
| | | | - Magali Gorce
- Service de Génétique, 49000 Angers Cedex 9, France
| | | | - Nada Houcinat
- CHU Dijon, Centre de référence maladies rares Anomalies du Développement et Syndromes Malformatifs, Centre de Génétique, FHU TRANSLAD, CHU Dijon Bourgogne 21000, France
| | - Laure Houdon
- Pediatric Diabetology, University Hospital, St Pierre de la Reunion 97410, France
| | | | - Lucie Jozwiak
- Pediatrics Department, CH de Roubaix, 59100 Roubaix, France
| | | | - Francois Labarthe
- Reference Center for Inborn Errors of Metabolism, Tours University Hospital, 37044 Tours, France
| | - Didier Lacombe
- Department of Medical Genetics, CHU Bordeaux INSERM U1211, Université de Bordeaux, 33076 Bordeaux, France
| | - Anne-Sophie Lambert
- AP-HP, Bicêtre Paris Saclay Hospital, DMU SEA, Endocrinology and Diabetes for Children, Le Kremlin Bicêtre 94270, France
| | - Christine Lefevre
- Pediatric Endocrinology, Jeanne de Flandre Hospital, 59037 Lille, France
| | - Bruno Leheup
- Service de Génétique clinique, Höpital Brabois, Centre Hospitalier Universitaire de Nancy, Nancy, Lorraine 54500, France
| | - Clara Leroy
- Service d'Endocrinologie et Maladies Métaboliques, Centre Hospitalier Régional Universitaire de Lille, Hôpital Huriez, 59037 Lille, France
| | | | - Isis Marchand
- Pediatrics Department, CHI de Créteil, 94010 Créteil, France
| | - Emeline Marquant
- Assistance-Publique des Hôpitaux de Marseille, Department of Pediatrics, Hôpital de la Timone Enfants, 13005 Marseille, France
| | | | | | - Sandra Pochelu
- Pediatric Endocrinology, CHU de Bordeaux, 33000 Bordeaux, France
| | - Chloé Quelin
- Service de Génétique, CLAD Ouest, CHU Rennes, 35200 Rennes, France
| | | | - Peggy Renoult-Pierre
- Service de Médecine Interne, Unité d'Endocrinologie Diabétologie et Nutrition, Centre Hospitalier Universitaire et Faculté de Médecine, Université de Tours, 37044 Tours, France
| | - Rachel Reynaud
- Assistance-Publique des Hôpitaux de Marseille, Department of Pediatrics, Hôpital de la Timone Enfants, 13005 Marseille, France
| | - Stéphanie Rouleau
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
| | - Cécile Teinturier
- AP-HP, Bicêtre Paris Saclay Hospital, DMU SEA, Endocrinology and Diabetes for Children, Le Kremlin Bicêtre 94270, France
| | - Julien Thevenon
- Inserm UMR 1231 GAD Team, Genetics of Developmental Anomalies, and FHU-TRANSLAD, CHU/Université de Bourgogne-Franche Comté, 21000 Dijon, France
| | | | - Aline Valle
- Pediatrics Department, CH de Douai, 59187 Douai, France
| | - Melody Vierge
- Assistance-Publique des Hôpitaux de Marseille, Department of Pediatrics, Hôpital de la Timone Enfants, 13005 Marseille, France
| | - Carine Villanueva
- Hospices Civils de Lyon, Hôpital Femme Mère Enfant, Service d'Endocrinologie Pédiatrique, 69677 Bron, France
| | | | - Xavier Dieu
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
- Biochemistry and Molecular Biology Laboratory, University Hospital of Angers, 49000 Angers, France
| | - Nathalie Bouzamondo
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
- Biochemistry and Molecular Biology Laboratory, University Hospital of Angers, 49000 Angers, France
| | - Patrice Rodien
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
- Department of Endocrinology, Diabetes and Nutrition, University Hospital of Angers, 49000 Angers, France
| | - Delphine Prunier-Mirebeau
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
- Biochemistry and Molecular Biology Laboratory, University Hospital of Angers, 49000 Angers, France
| | - Régis Coutant
- Department of Pediatric Endocrinology and Diabetology, University Hospital of Angers, 49000 Angers, France
- Reference Center for Rare Diseases of Thyroid and Hormone Receptivity, University Hospital of Angers, 49000 Angers, France
| |
Collapse
|
18
|
Zhang T, Shen Y, Xu Y, Wu D, Chen C, Yang R. Clinical, biochemical characteristics and genotype-phenotype analysis of congenital hypothyroidism diagnosed by newborn screening in China. Clin Chim Acta 2023:117459. [PMID: 37390946 DOI: 10.1016/j.cca.2023.117459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND Congenital hypothyroidism (CH) is the most common neonatal endocrine disorder worldwide. However, the underlying etiology remains unclear in most patients. METHODS The newborn screening was performed for TSH in dried blood spots. Serum TSH, T3, T4, free T3(FT3) and free T4 (FT4) were detected for the recalled children. High-throughput sequencing were applied to detect 29 known CH genes. The statistical analyses were performed to analyze the differences between biochemical data, thyroid volume, clinical prognosis and genetic results for 97 patients who had one or more variants in CH related genes. RESULTS DUOX2 gene has the highest variant rate, followed by TG, TPO and TSHR gene. The "DUOX2 biallelic variants" group was associated with "Goiter", while "DUOX2 monoallelic variants" group was associated with "Agenesis". In addition, the TSH levels and initial L-T4 dose were significantly higher in "TPO biallelic variants" group than those in "DUOX2 and TSHR biallelic variants" groups. CONCLUSIONS Our study showed dyshormonogenesis (DH) might be the leading pathophysiology of CH in Chinese populations. DUOX2 gene mostly caused goiter, but also could be associated with hypoplasia. TPO might play a more irreplaceable role than DUOX2. The digenic variants combination indicated the complexity of genetic etiology in CH.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yaping Shen
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yanhua Xu
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Dingwen Wu
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chi Chen
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Rulai Yang
- Department of Genetics and Metabolism, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
19
|
Gevers EF, de Winter JP. New developments and therapies in pediatric endocrinology. Eur J Pediatr 2023; 182:1439-1443. [PMID: 36567374 DOI: 10.1007/s00431-022-04772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Evelien F Gevers
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
- Department of Paediatric Endocrinology and Diabetes, Barts Health NHS Trust - The Royal London Children's Hospital, London, United Kingdom.
| | - J Peter de Winter
- Department of Pediatrics, Spaarne Gasthuis, Haarlem/Hoofddorp, The Netherlands
- Leuven Child and Health Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Tuli G, Munarin J, Topalli K, Pavanello E, de Sanctis L. Neonatal Screening for Congenital Hypothyroidism in Preterm Infants: Is a Targeted Strategy Required? Thyroid 2023; 33:440-448. [PMID: 36802847 DOI: 10.1089/thy.2022.0495] [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] [Indexed: 02/20/2023]
Abstract
Background: Premature infants are at higher risk of developing congenital hypothyroidism (CH) but the neonatal screening strategy for this population is still debatable. The purpose of this retrospective study is to describe the results of a screening program for CH in a preterm infant cohort. Materials and Methods: All preterm newborns who underwent neonatal screening in the Italian region of Piedmont in the period January 2019-December 2021, were included in this retrospective cohort study. The first thyrotropin (TSH) measurement was performed at 72 hours, whereas the second at 15 days of life. Infants with TSH >20 mUI/L at first detection and >6 mUI/L at second were recalled for a full evaluation of thyroid function. Results: During the study period, 5930 preterm newborns were screened. Based on birthweight (BW), the mean TSH was 2.08 ± 0.15 for BW <1000 g, 2.01 ± 0.02 for BW 1001-1500 g, 2.28 ± 0.03 for BW 1501-2499 g, and 2.41 ± 0.03 mUI/L in normal-weight newborns (p < 0.005) at the first detection, with a significant difference observed at the second measurement (p < 0.005). Based on gestational age, the mean TSH at first detection was 1.71 ± 0.09 mUI/L for extremely preterm babies and 1.87 ± 0.06, 1.94 ± 0.05, and 2.42 ± 0.02 mUI/L for very preterm, moderately, and late preterm infants (p < 0.005), respectively. Significant between-group differences of TSH measurements were also at the second and third detections (p < 0.005 and p = 0.01). The 99% reference range in this cohort overlapped with the recommended TSH cutoffs for screening recall (8 mUI/L for first detection and 6 mUI/L for second detection). CH incidence was 1:156. Of the 38 patients diagnosed with CH, a eutopic gland was present in 30 (87.9%), with CH transient in 29 (76.8%). Conclusions: We observed no significant difference in the recall rate between preterm and at term infants screened in this study. Our current screening strategy therefore appears effective in avoiding misdiagnosis. CH screening approaches vary among countries. Development and testing of a uniform multinational screening strategy is needed.
Collapse
Affiliation(s)
- Gerdi Tuli
- Department of Pediatric Endocrinology, Regina Margherita Children's Hospital, Turin, Italy
- Postgraduate Program in Biomedical Sciences and Oncology, Department of Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Jessica Munarin
- Department of Pediatric Endocrinology, Regina Margherita Children's Hospital, Turin, Italy
- Postgraduate School of Pediatrics, Department of Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Kristela Topalli
- Department of Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Enza Pavanello
- Department of Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Luisa de Sanctis
- Department of Pediatric Endocrinology, Regina Margherita Children's Hospital, Turin, Italy
- Department of Health and Pediatric Sciences, University of Turin, Turin, Italy
| |
Collapse
|
21
|
Gumes-Felix HM, Ramalho RJR, Melo EV, Matos DM, Menezes NV, Oliveira CRP, Campos VC, Santos EG, da S Marques D, Vaz Dos Santos B, de Andrade BMR, Aguiar-Oliveira MH. Predictive factors for the diagnosis of permanent congenital hypothyroidism and its temporal changes in Sergipe, Brazil - A real-life retrospective study. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:189-196. [PMID: 36651708 PMCID: PMC10689040 DOI: 10.20945/2359-3997000000579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/03/2022] [Indexed: 01/19/2023]
Abstract
Objective Congenital hypothyroidism (CH) can be permanent (PCH) or transient (TCH). While the importance of thyroxine in myelination of the brain is undisputed, the benefits to neurodevelopmental outcomes of TCH treatment are controversial. Our objectives were to determine predictive factors for PCH and verify its prevalence changes over time. Subjects and methods A total of 165 children were evaluated at 3 years of age to verify the diagnosis of PCH. 130 were submitted to a two-step cluster analysis, with the aim of grouping them into homogeneous clusters. The mean incidence of PCH and TCH was calculated from 2004 to 2010 and 2011 to 2015. Results Sixty-six children were diagnosed with PCH, and 99 were diagnosed with TCH. Eighty-one percent of PCH children and all TCH children with thyroid imaging had glands in situ. Eighty children (61.5%) were in Cluster 1, 8 children (6.2%) were in Cluster 2 and 42 children (32.3%) were in Cluster 3. No children had PCH in Cluster 1, while 87.5% of children in Cluster 2 and all children in Cluster 3 had PCH. The most important predictor for PCH was the initial serum TSH, which was marginally higher in importance than the blood spot TSH, followed by the initial serum free T4. The mean incidence of PCH (odds ratio: 1.95, 95% CI 1.36 to 2.95, p < 0.0001) and TCH (odds ratio 1.33, 95%, CI 1.02 to 1.77, p = 0,038) increased over time. Conclusion The most important PCH predictors are the initial serum TSH and the blood spot TSH. The mean incidence of both PCH and TCH in our series increased.
Collapse
Affiliation(s)
- Hérika M Gumes-Felix
- Divisão de Endocrinologia, Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Roberto J R Ramalho
- Departamento de Medicina, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Enaldo V Melo
- Departamento de Medicina, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Diana M Matos
- Divisão de Endocrinologia, Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Nelmo V Menezes
- Departamento de Medicina, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Carla R P Oliveira
- Divisão de Endocrinologia, Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Viviane C Campos
- Divisão de Endocrinologia, Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Elenilde G Santos
- Divisão de Endocrinologia, Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | | | | | - Bruna M R de Andrade
- Departamento de Fonoaudiologia, Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, SE, Brasil
| | - Manuel H Aguiar-Oliveira
- Divisão de Endocrinologia, Programa de Pós-graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, SE, Brasil,
| |
Collapse
|
22
|
Yazici A, Kadioglu Simsek G, Elbayiyev S, Canpolat FE, Kanmaz Kutman HG. Thyroid Function in Neonates with Hypoxic Ischemic Encephalopathy. Ther Hypothermia Temp Manag 2023; 13:11-15. [PMID: 35696232 DOI: 10.1089/ther.2022.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We aimed to examine heel prick (capillary) and serum thyroid function test (TFT) results in neonates with hypoxic ischemic encephalopathy (HIE) to evaluate the effect of asphyxia and therapeutic hypothermia (TH) on thyroid functions. This retrospective chart review included infants who were born after 34 weeks of gestation, were diagnosed and treated for HIE. The patients were divided into those who did and did not undergo TH and the groups were compared in terms of demographic characteristics, laboratory results, capillary thyroid-stimulating hormone (cTSH) levels, and serum thyroid-stimulating hormone (TSH) and free thyroxine (fT4) levels. A total of 111 neonates were included in the study. There was no difference between the TH group (n = 90) and the nonhypothermia group (n = 21) in terms of median gestational age (38.3 ± 2.1 weeks vs. 38.6 ± 1.8 weeks, p = 0.42) or birth weight (3182 ± 509 g vs. 3174 ± 573 g, p = 0.72). Serum TFT was performed at a median of 10 days (range, 2-43) and capillary TSH analyzed at a median of 6 days (range, 1-26). Capillary TSH at 96 hours was analyzed in 36 patients in the TH group and 19 patients in the nonhypothermia group. Serum TSH and fT4 levels were similar in both groups (p = 0.29, p = 0.1). Overall cTSH and cTSH obtained in the first 4 days were 2.2 (0.5-10) and 4.3 (0.5-94), p = 0.059; 2 (0.5-22) and 5 (0.5-94), p = 0.04, respectively, whereas cTSH obtained after day 4 was similar in both groups (p = 0.058). Abnormal serum TSH (>5.5 mU/mL) was more frequent in the hypothermia group (44.4% vs. 19%, p = 0.026). Our results suggest that TH may cause some alterations on TFTs. Therefore, it may be reasonable to repeat TSH screening after TH.
Collapse
Affiliation(s)
- Aybuke Yazici
- Division of Neonatology, Ankara City Hospital University of Health Sciences, Ankara, Turkey
| | - Gulsum Kadioglu Simsek
- Division of Neonatology, Ankara City Hospital University of Health Sciences, Ankara, Turkey
| | - Serhan Elbayiyev
- Division of Neonatology, Ankara City Hospital University of Health Sciences, Ankara, Turkey
| | - Fuat Emre Canpolat
- Division of Neonatology, Ankara City Hospital University of Health Sciences, Ankara, Turkey
| | | |
Collapse
|
23
|
Bontemps SH, Legagneur C, Guéant-Rodriguez RM, Remen T, Luc A, Renard E. Congenital hypothyroidism in children with eutopic gland or thyroid hemiagenesis: prognostic factors for transient vs. permanent hypothyroidism. J Pediatr Endocrinol Metab 2023; 36:353-363. [PMID: 36798001 DOI: 10.1515/jpem-2022-0101] [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: 02/23/2022] [Accepted: 01/29/2023] [Indexed: 02/18/2023]
Abstract
OBJECTIVES More than one third of children with congenital hypothyroidism (CH) and thyroid gland in situ (or eutopic gland) have transient hypothyroidism. It remains difficult to determine early on whether hypothyroidism will be transient which may cause overtreatment and its complications in these children. Our primary aim was to determine prognostic factors for transient hypothyroidism in children with congenital hypothyroidism and eutopic gland or thyroid hemiagenesis. METHODS We retrospectively reviewed medical records of 111 children, born between 1996 and 2017, diagnosed with congenital hypothyroidism and eutopic gland or hemiagenesis and treated at the Nancy Regional and University Hospital. RESULTS Fifty four infants (48.6%) had permanent congenital hypothyroidism (PCH) and 57 (51.4%) transient congenital hypothyroidism (TCH). Prognostic factors for TCH included prematurity, twin pregnancy, low birth weight and Apgar score <7, while low FT3 at diagnosis, maternal levothyroxine treatment, a family history of thyroid dysfunction and TSH ≥10 mUI/L while receiving treatment were associated with PCH. Knee epiphyses on X-ray at diagnosis were absent only in children with PCH. The median levothyroxine dose during follow-up was significantly lower in the TCH group compared to the PCH group. A levothyroxine dose of ≤3.95, ≤2.56, ≤2.19 and ≤2.12 μg/kg/day at 6 months, 1, 2 and 3 years of follow-up, respectively, had the best sensitivity-to-specificity ratio for predicting TCH. CONCLUSIONS Even though it remains difficult to predict the course of hypothyroidism at diagnosis, we were able to identify several prognostic factors for TCH including perinatal problems and lower levothyroxine requirements that can guide the physician on the evolution of hypothyroidism. Clinical Trial Registration Number: NCT04712760.
Collapse
Affiliation(s)
| | - Carole Legagneur
- Department of Pediatrics, University Hospital of Nancy, Vandoeuvre-Lès-Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- Department of Biochemistry, University Hospital of Nancy, Vandoeuvre-Lès-Nancy, France.,Institut national de la santé et de la recherche médicale 1256 [National Institute of Health and Medical Research], University of Lorraine, N-GERE Nutrition Genetics and Environmental Risks, Vandoeuvre-Lès-Nancy, France
| | - Thomas Remen
- MPI Department, Methodology, Data Management and Statistic Unit, Délégation à la Recherche Clinique et à l'Innovation [Delegation for Clinical Research and Innovation], University Hospital of Nancy, Vandœuvre-Lès-Nancy, France
| | - Amandine Luc
- MPI Department, Methodology, Data Management and Statistic Unit, Délégation à la Recherche Clinique et à l'Innovation [Delegation for Clinical Research and Innovation], University Hospital of Nancy, Vandœuvre-Lès-Nancy, France
| | - Emeline Renard
- Department of Pediatrics, University Hospital of Nancy, Vandoeuvre-Lès-Nancy, France.,Institut national de la santé et de la recherche médicale 1256 [National Institute of Health and Medical Research], University of Lorraine, N-GERE Nutrition Genetics and Environmental Risks, Vandoeuvre-Lès-Nancy, France
| |
Collapse
|
24
|
Büyükavcı MA, Dundar I. Comparison of developmental outcomes in children with permanent and transient congenital hypothyroidism. J Pediatr Endocrinol Metab 2023; 36:364-370. [PMID: 36794655 DOI: 10.1515/jpem-2022-0539] [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: 10/22/2022] [Accepted: 01/29/2023] [Indexed: 02/17/2023]
Abstract
OBJECTIVES Congenital hypothyroidism (CH) is still one of the most common causes of preventable cognitive impairment in children, and its early detection and treatment prevent irreversible neurodevelopmental delay. Depending on the underlying cause, cases with CH may be transient or permanent. This study aimed to compare the developmental evaluation results of transient and permanent CH patients and to reveal any differences. METHODS A total of 118 patients with CH, who were followed up jointly in pediatric endocrinology and developmental pediatrics clinics, were included. The patients' progress was evaluated per the International Guide for Monitoring Child Development (GMCD). RESULTS Of the cases, 52 (44.1%) were female, and 66 (55.9%) were male. While 20 (16.9%) cases were diagnosed with permanent CH, 98 (83.1%) were diagnosed with transient CH. According to the results of the developmental evaluation made with GMCD, the development of 101 (85.6%) children was compatible with their age, while 17 (14.4%) children had delays in at least one developmental area. All 17 patients had a delay in expressive language. Developmental delay was detected in 13 (13.3%) of those with transient CH and 4 (20%) with permanent CH. CONCLUSIONS There is difficulty in expressive language in all cases of CH with developmental delay. No significant difference was found between the developmental evaluations of permanent and transient CH cases. The results revealed the importance of developmental follow-up, early diagnosis and interventions in those children. GMCD is thought to be an important guide to help monitoring the development of patients with CH.
Collapse
Affiliation(s)
- Mehmet Akif Büyükavcı
- Department of Developmental Pediatrics, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Ismail Dundar
- Department of Pediatric Endocrinology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| |
Collapse
|
25
|
Gentilini D, Muzza M, de Filippis T, Vigone MC, Weber G, Calzari L, Cassio A, Di Frenna M, Bartolucci M, Grassi ES, Carbone E, Olivieri A, Persani L. Stochastic epigenetic mutations as possible explanation for phenotypical discordance among twins with congenital hypothyroidism. J Endocrinol Invest 2023; 46:393-404. [PMID: 36071330 PMCID: PMC9859866 DOI: 10.1007/s40618-022-01915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/29/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE The elevated frequency of discordance for congenital hypothyroidism (CH) phenotype between monozygotic twins suggests the involvement of non-mendelian mechanisms. The aim of the study was to investigate the role of epigenetics in CH pathogenesis. METHODS A genome-wide DNA methylation analysis was performed on the peripheral blood of 23 twin pairs (10 monozygotic and 13 dizygotic), 4 concordant and 19 discordant pairs for CH at birth. RESULTS Differential methylation analysis did not show significant differences in methylation levels between CH cases and controls, but a different methylation status of several genes may explain the CH discordance of a monozygotic twin couple carrying a monoallelic nonsense mutation of DUOX2. In addition, the median number of hypo-methylated Stochastic Epigenetic Mutations (SEMs) resulted significantly increased in cases compared to controls. The prioritization analysis for CH performed on the genes epimutated exclusively in the cases identified SLC26A4, FOXI1, NKX2-5 and TSHB as the genes with the highest score. The analysis of significantly SEMs-enriched regions led to the identification of two genes (FAM50B and MEG8) that resulted epigenetically dysregulated in cases. CONCLUSION Epigenetic modifications may potentially account for CH pathogenesis and explain discordance among monozygotic twins.
Collapse
Affiliation(s)
- D Gentilini
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095, Milan, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - M Muzza
- Laboratory of Endocrine and Metabolic Research, Department of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, Piazzale Brescia 20, 20149, Milan, Italy
| | - T de Filippis
- Laboratory of Endocrine and Metabolic Research, Department of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, Piazzale Brescia 20, 20149, Milan, Italy
| | - M C Vigone
- Department of Pediatrics, Endocrine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - G Weber
- Department of Pediatrics, Endocrine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - L Calzari
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095, Milan, Italy
| | - A Cassio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - M Di Frenna
- Department of Pediatrics, Endocrine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - M Bartolucci
- Department of Maternal and Child Sciences and Urology, University "La Sapienza", Rome, Italy
| | - E S Grassi
- Department of Medical Biotechnology and Experimental Medicine, University of Milan, 20122, Milan, Italy
| | - E Carbone
- Laboratory of Endocrine and Metabolic Research, Department of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, Piazzale Brescia 20, 20149, Milan, Italy
| | - A Olivieri
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Italian National Institute of Health, 00161, Rome, Italy
| | - L Persani
- Laboratory of Endocrine and Metabolic Research, Department of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano IRCCS, Piazzale Brescia 20, 20149, Milan, Italy.
- Department of Medical Biotechnology and Experimental Medicine, University of Milan, 20122, Milan, Italy.
| |
Collapse
|
26
|
Clinical and genetic investigation in patients with permanent congenital hypothyroidism. Clin Chim Acta 2023; 539:1-6. [PMID: 36423704 DOI: 10.1016/j.cca.2022.11.007] [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: 08/03/2022] [Revised: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Permanent congenital hypothyroidism (CH) is usually a more severe type of CH. However, the molecular etiology and clinical features of permanent CH remain unclear. METHODS We recruited 42 patients who were diagnosed with CH and followed-up after diagnosis. Demographic information and data at diagnosis and treatment were recorded. Genetic analyses were performed using whole exome sequencing. Based on the presence or absence of variants and differences in clinical features, we grouped the study participants and analyzed their characteristics. RESULTS A total of 29 patients (69.0 %) were identified as having variants potentially related to their disease. Among the 24 patients with normal-sized thyroid gland-in-situ (GIS) or goiter, 23 (95.8 %, P < 0.001) had variants. This is compared to 18 patients with thyroid dysgenesis (TD), of which six (33.3 %) had genetic variants. We detected 55 variants in six genes, the most frequently mutated gene being DUOX2 (70.9 %). Biallelic DUOX2 variants were detected in 14 of 24 (58.3 %) GIS or goiter patients. Compared to the cases with variants, the L-T4 dose at 2 and 3 years of age and current dose were higher in the unmutated cases. At 2 years of age, patients with TD required higher doses of L-T4 supplementation. Patients with DUOX2 variants showed lower doses of L-T4 being required at 2 and 3 years of age and current. Furthermore, patients with GIS or goiter with DUOX2 variants showed lower doses of L-T4. CONCLUSIONS Patients with CH, whether TD or GIS or goiter, are at risk of developing a permanent condition. Compared with patients with TD, the detection of variants was higher in patients with GIS or goiter. The most frequently mutated gene was DUOX2, with a biallelic type. Patients with TD required higher doses of L-T4 supplementation with age, whereas those patients with the DUOX2 variant required relatively lower doses.
Collapse
|
27
|
Li M, Li Z, Chen M, Hu Z, Zhou M, Wu L, Zhang C, Liang D. Novel Missense Variants in PAX8 and NKX2-1 Cause Congenital Hypothyroidism. Int J Mol Sci 2023; 24:ijms24010786. [PMID: 36614229 PMCID: PMC9821711 DOI: 10.3390/ijms24010786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Primary congenital hypothyroidism (CH) is a common neonatal endocrine disorder characterized by elevated concentrations of thyroid stimulating hormone (TSH) and low concentrations of free thyroxine (FT4). PAX8 and NKX2-1 are important transcription factors involved in thyroid development. In this study, we detected three novel variants in PAX8 (c.149A > C and c.329G > A) and NKX2-1 (c.706A > G) by whole exome sequencing (WES) in three unrelated CH patients with variable phenotypes. The results of Western blot and immunofluorescence analysis showed that the three variants had no effect on protein expression and subcellular localization. However, the results of the electrophoretic mobility shift assay (EMSA) and dual-luciferase reporter assay suggested that the three variants in PAX8 and NKX2-1 both affected their DNA-binding ability and reduced their transactivation capacity. Moreover, a dominant-negative effect in K236E−NKX2-1 was identified by dual-luciferase reporter assay. To sum up, our findings extend our knowledge of the current mutation spectrum of PAX8 and NKX2-1 and provide important information for diagnosing, treating, and preventing CH in these families.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Chunhua Zhang
- Correspondence: (C.Z.); (D.L.); Tel.: +86-871-65174598 (C.Z.); +86-731-84805252 (D.L.)
| | - Desheng Liang
- Correspondence: (C.Z.); (D.L.); Tel.: +86-871-65174598 (C.Z.); +86-731-84805252 (D.L.)
| |
Collapse
|
28
|
Grassi ES, Rurale G, de Filippis T, Gentilini D, Carbone E, Coscia F, Uraghi S, Bullock M, Clifton-Bligh RJ, Gupta AK, Persani L. The length of FOXE1 polyalanine tract in congenital hypothyroidism: Evidence for a pathogenic role from familial, molecular and cohort studies. Front Endocrinol (Lausanne) 2023; 14:1127312. [PMID: 37008944 PMCID: PMC10060985 DOI: 10.3389/fendo.2023.1127312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION FOXE1 is required for thyroid function and its homozygous mutations cause a rare syndromic form of congenital hypothyroidism (CH). FOXE1 has a polymorphic polyalanine tract whose involvement in thyroid pathology is controversial. Starting from genetic studies in a CH family, we explored the functional role and involvement of FOXE1 variations in a large CH population. METHODS We applied NGS screening to a large CH family and a cohort of 1752 individuals and validated these results by in silico modeling and in vitro experiments. RESULTS A new heterozygous FOXE1 variant segregated with 14-Alanine tract homozygosity in 5 CH siblings with athyreosis. The p.L107V variant demonstrated to significantly reduce the FOXE1 transcriptional activity. The 14-Alanine-FOXE1 displayed altered subcellular localization and significantly impaired synergy with other transcription factors, when compared with the more common 16-Alanine-FOXE1. The CH group with thyroid dysgenesis was largely and significantly enriched with the 14-Alanine-FOXE1 homozygosity. DISCUSSION We provide new evidence that disentangle the pathophysiological role of FOXE1 polyalanine tract, thereby significantly broadening the perspective on the role of FOXE1 in the complex pathogenesis of CH. FOXE1 should be therefore added to the group of polyalanine disease-associated transcription factors.
Collapse
Affiliation(s)
- Elisa Stellaria Grassi
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giuditta Rurale
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | - Tiziana de Filippis
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Bioinformatics and Statistical Genomics Unit, Milano, Italy
| | - Erika Carbone
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | | | - Sarah Uraghi
- Department of Health Science, University of Milan, Milan, Italy
| | - Martyn Bullock
- Cancer Genetics Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Roderick J. Clifton-Bligh
- Cancer Genetics Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Endocrinology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Abhinav K. Gupta
- Department of Diabetes and Endocrine Sciences, CK Birla Hospitals, Jaipur, Rajasthan, India
| | - Luca Persani
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Laboratory of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
- *Correspondence: Luca Persani,
| |
Collapse
|
29
|
Itonaga T, Hasegawa Y, Higuchi S, Satoh M, Sawada H, Shimura K, Takahashi I, Takubo N, Nagasaki K. Knowns and unknowns about congenital hypothyroidism: 2022 update. Clin Pediatr Endocrinol 2023; 32:11-25. [PMID: 36761498 PMCID: PMC9887299 DOI: 10.1297/cpe.2022-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/13/2022] [Indexed: 11/19/2022] Open
Abstract
Several excellent guidelines and expert opinions on congenital hypothyroidism (CH) are currently available. Nonetheless, these guidelines do not address several issues related to CH in detail. In this review, the authors chose the following seven clinical issues that they felt were especially deserving of closer scrutiny in the hope that drawing attention to them through discussion would help pediatric endocrinologists and promote further interest in the treatment of CH. 1. How high should the levothyroxine (L-T4) dose be for initial treatment of severe and permanent CH? 2. What is the optimal method for monitoring treatment of severe CH? 3. At what level does maternal iodine intake during pregnancy affect fetal and neonatal thyroid function? 4. Does serum thyroglobulin differ between patients with a dual oxidase 2 (DUOX2) variants and those with excess iodine? 5. Who qualifies for a genetic diagnosis? 6. What is the best index for distinguishing transient and permanent CH? 7. Is there any cancer risk associated with CH? The authors discussed these topics and jointly edited the manuscript to improve the understanding of CH and related issues.
Collapse
Affiliation(s)
- Tomoyo Itonaga
- Department of Pediatrics, Oita University Faculty of
Medicine, Oita, Japan
| | - Yukihiro Hasegawa
- Division of Endocrinology and Metabolism, Tokyo Metropolitan
Children’s Medical Center, Tokyo, Japan
| | - Shinji Higuchi
- Division of Pediatric Endocrinology and Metabolism,
Children’s Medical Center, Osaka City General Hospital, Osaka, Japan
| | - Mari Satoh
- Department of Pediatrics, Toho University Omori Medical
Center, Tokyo, Japan
| | - Hirotake Sawada
- Division of Pediatrics, Faculty of Medicine, University of
Miyazaki Hospital, Miyazaki, Japan
| | - Kazuhiro Shimura
- Department of Pediatrics, Keio University School of Medicine,
Tokyo, Japan
| | - Ikuko Takahashi
- Department of Pediatrics, Akita University Graduate School of
Medicine, Akita, Japan
| | - Noriyuki Takubo
- Department of Pediatrics and Adolescent Medicine, Juntendo
University Graduate School of Medicine, Tokyo, Japan
| | - Keisuke Nagasaki
- Department of Pediatrics, Niigata University Medical and
Dental Hospital, Niigata, Japan
| |
Collapse
|
30
|
Liu R, Tian JL, Huang XL, Song YZ. Genetic Factors Causing Thyroid Dyshormonogenesis as the Major Etiologies for Primary Congenital Hypothyroidism: Clinical and Genetic Characterization of 33 Patients. J Clin Med 2022; 11:7313. [PMID: 36555929 PMCID: PMC9786654 DOI: 10.3390/jcm11247313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND AIMS Although the significance of primary congenital hypothyroidism (CH) is supported by an increasing amount of evidence, the clinical and genetic characteristics of this condition are still poorly understood. This study aimed to explore the underlying genetic etiologies in a cohort of primary CH patients. SUBJECTS AND METHODS The clinical data of 33 patients with primary CH were collected and analyzed via a cross-sectional study. Genetic analysis was performed by high-throughput sequencing and Sanger verification, and the pathogenicity of the novel missense variants was predicted using a variety of comprehensive bioinformatic tools. RESULTS Among the 33 patients, 22 (22/33, 66.7%) harbored pathogenic variants in the causative genes of thyroid dysgenesis or dyshormonogenesis, with DUOX2 (15/33, 45.5%) topping the list, followed by TG, TPO, DUOXA2 and PAX8. Four novel genetic variants were detected, including a pathogenic frameshift and three likely pathogenic missense variants. Positive neonatal screening for TSH, neonatal jaundice and abnormal thyroid morphology were the main positive findings among all cases. Although 31 of the total 33 CH patients exhibited normal anthropometric and social performance, the other 2 had poor prognosis in this study. CONCLUSIONS This study reported 33 new CH patients bearing four novel genetic variants, which enriched the variant spectrum of CH genes. In this cohort, genetic factors causing thyroid dyshormonogenesis were the main etiologies of CH development. Most patients exhibited a favorable prognosis; however, systematic management remains a challenge in achieving improved clinical outcomes for CH patients.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Jing-Li Tian
- Department of Pediatrics, Huizhou No. 2 Women’s and Children’s Healthcare Hospital, Huizhou 516000, China
| | - Xiao-Ling Huang
- Neonatal Screening Center, Dongguan Maternal and Child Healthcare Hospital, Dongguan 523125, China
| | - Yuan-Zong Song
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| |
Collapse
|
31
|
Banerjee A, Biswas D, Barpanda A, Halder A, Sibal S, Kattimani R, Shah A, Mahadevan A, Goel A, Srivastava S. The First Pituitary Proteome Landscape From Matched Anterior and Posterior Lobes for a Better Understanding of the Pituitary Gland. Mol Cell Proteomics 2022; 22:100478. [PMID: 36470533 PMCID: PMC9877467 DOI: 10.1016/j.mcpro.2022.100478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
To date, very few mass spectrometry (MS)-based proteomics studies are available on the anterior and posterior lobes of the pituitary. In the past, MS-based investigations have focused exclusively on the whole pituitary gland or anterior pituitary lobe. In this study, for the first time, we performed a deep MS-based analysis of five anterior and five posterior matched lobes to build the first lobe-specific pituitary proteome map, which documented 4090 proteins with isoforms, mostly mapped into chromosomes 1, 2, and 11. About 1446 differentially expressed significant proteins were identified, which were studied for lobe specificity, biological pathway enrichment, protein-protein interaction, regions specific to comparison of human brain and other neuroendocrine glands from Human Protein Atlas to identify pituitary-enriched proteins. Hormones specific to each lobe were also identified and validated with parallel reaction monitoring-based target verification. The study identified and validated hormones, growth hormone and thyroid-stimulating hormone subunit beta, exclusively to the anterior lobe whereas oxytocin-neurophysin 1 and arginine vasopressin to the posterior lobe. The study also identified proteins POU1F1 (pituitary-specific positive transcription factor 1), POMC (pro-opiomelanocortin), PCOLCE2 (procollagen C-endopeptidase enhancer 2), and NPTX2 (neuronal pentraxin-2) as pituitary-enriched proteins and was validated for their lobe specificity using parallel reaction monitoring. In addition, three uPE1 proteins, namely THEM6 (mesenchymal stem cell protein DSCD75), FSD1L (coiled-coil domain-containing protein 10), and METTL26 (methyltransferase-like 26), were identified using the NeXtProt database, and depicted tumor markers S100 proteins having high expression in the posterior lobe. In summary, the study documents the first matched anterior and posterior pituitary proteome map acting as a reference control for a better understanding of functional and nonfunctional pituitary adenomas and extrapolating the aim of the Human Proteome Project towards the investigation of the proteome of life.
Collapse
Affiliation(s)
- Arghya Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Deepatarup Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Abhilash Barpanda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ankit Halder
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Shamira Sibal
- Lokmanya Tilak Municipal Medical College, Mumbai, India
| | | | - Abhidha Shah
- Department of Neurosurgery at King Edward Memorial Hospital and Seth G. S. Medical College, Mumbai, India
| | - Anita Mahadevan
- Human Brain Bank, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Atul Goel
- Department of Neurosurgery at King Edward Memorial Hospital and Seth G. S. Medical College, Mumbai, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India.
| |
Collapse
|
32
|
Dündar İ, Büyükavcı MA, Çiftçi N. Etiological, clinical, and laboratory evaluation of congenital hypothyroidism and determination of levothyroxine (LT4) dose at treatment interruption in differentiating permanent vs. transient patients. Turk J Med Sci 2022; 52:1863-1871. [PMID: 36945978 PMCID: PMC10390198 DOI: 10.55730/1300-0144.5533] [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: 05/13/2022] [Accepted: 07/24/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Congenital hypothyroidism (CH) is the most common cause of preventable but irreversible mental retardation in children, although the risk has been widely abolished by national neonatal screening programs. The aim of this study was to determine, (a) the cause of CH, (b) the etiological cause of persistent CH and (c) to investigate the role of laboratory and clinical data in predicting persistent and transient CH. METHODS Patients diagnosed with CH, who started L-thyroxine treatment and were followed up for at least three years were included. Patient data were reviewed retrospectively. Serum thyroid hormones were measured four weeks after discontinuation of therapy at age three or earlier. Cases with a thyroid-stimulating hormone (TSH) value of >10 mIU/mL were accepted as permanent hypothyroidism, while cases with normal TSH values for six months after cessation were accepted as transient hypothyroidism. RESULTS There were 232 treated cases, of whom 108 (46.6%) were female, and 169 (72.8%) were eventually diagnosed with transient CH. The best cut-off point for predicting permanent status was determined as LT4 cut-off dose ≥1.45 mcg/kg/day. The median (range) duration of L-thyroxine treatment in transient hypothyroid cases was 24 (range: 6-36) months, and treatment was discontinued before the age of three years in 64%. DISCUSSION There were 232 treated cases, of whom 108 (46.6%) were female, and 169 (72.8%) were eventually diagnosed with transient CH. The best cut-off point for predicting permanent status was determined as LT4 cut-off dose ≥1.45 mcg/kg/day. The median (range) duration of L-thyroxine treatment in transient hypothyroid cases was 24 (range: 6-36) months, and treatment was discontinued before the age of three years in 64%.
Collapse
Affiliation(s)
- İsmail Dündar
- Department of Pediatric Endocrinology, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Mehmet Akif Büyükavcı
- Department of Developmental Pediatrics, Faculty of Medicine, İnönü University, Malatya, Turkey
| | - Nurdan Çiftçi
- Department of Pediatric Endocrinology, Malatya Training and Research Hospital, Malatya, Turkey
| |
Collapse
|
33
|
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: 17] [Impact Index Per Article: 5.7] [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
|
34
|
Fourneaux R, Castets S, Godefroy A, Grelet M, Abeillon-du Payrat J, Saveanu A, Castinetti F, Reynaud R. Congenital Central Hypothyroidism Caused by a Novel IGSF1 Variant Identified in a French Family. Horm Res Paediatr 2022; 95:296-303. [PMID: 35350016 DOI: 10.1159/000524233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/15/2022] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Congenital central hypothyroidism (CCH) is a rare disorder that can be caused by X-linked mutations in the immunoglobulin superfamily member 1 (IGSF1) gene. Here, we describe four familial cases with a variable presentation due to a novel IGSF1 pathogenic variant. CASE PRESENTATION In the index case, an investigation at birth of a suspected brain-lung-thyroid syndrome surprisingly revealed a central hypothyroidism. Next-generation sequencing uncovered a novel IGSF1 pathogenic variant: a hemizygous single base duplication (G) resulting in a premature stop codon (NM_001555.4: c.2485dup, p.Ala829Glyfs*15). Further family investigations revealed missed neonatal CCH for the older brother who presented with prolonged jaundice (thyroid stimulating hormone 3.06 mUI/L, FT4 9.4 pmol/L, FT3 4.2 pmol/L). It also led to the diagnosis of CCH at 11 months of age for the younger brother, whose thyroid function was considered normal at birth. Neuropsychological evaluations showed no cognitive impairment for the eldest two brothers, but a slightly reduced processing-speed index compared with the other parameters for the oldest. Furthermore, a maternal uncle was diagnosed with biochemical CCH at 34 years of age, despite having few symptoms, and a complete workup revealed prolactin deficiency and macroorchidism. DISCUSSION This report of a rare case of neonatal CCH caused by IGSF1 deficiency highlights the importance of recognizing the neonatal signs of hypothyroidism to diagnose CCH as early as possible. Our results also show the importance of performing family genetic screening if a pathogenic variant is identified, to properly monitor carriers as CCH may develop over time. We suggest that these families should be followed up in the long term to better understand the natural history of this syndrome and evaluate the need for hormone substitution.
Collapse
Affiliation(s)
- Rachel Fourneaux
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille Maladies Rares (MarMaRa), Aix-Marseille Université, Marseille, France.,Assistance Publique-Hôpitaux de Marseille (APHM), Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares de l'hypophyse HYPO, Marseille, France
| | - Sarah Castets
- Department of Multidisciplinary Peadiatrics, Centre de Référence des Maladies Rares d'origine hypophysaire HYPO, Hôpital Timone-Enfants, APHM, Marseille, France
| | - Alice Godefroy
- Department of General Peadiatrics, Hôpital Saint Joseph, Marseille, France
| | - Maude Grelet
- Department of Genetics, Centre Hospitalier Sainte Musse, Toulon, France
| | | | - Alexandru Saveanu
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille Maladies Rares (MarMaRa), Aix-Marseille Université, Marseille, France.,Laboratory of Molecular Biology, CHU Conception, Assistance Publique-Hôpitaux de Marseille (APHM), Marseille, France
| | - Frederic Castinetti
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille Maladies Rares (MarMaRa), Aix-Marseille Université, Marseille, France.,Assistance Publique-Hôpitaux de Marseille (APHM), Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares de l'hypophyse HYPO, Marseille, France
| | - Rachel Reynaud
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), Institut Marseille Maladies Rares (MarMaRa), Aix-Marseille Université, Marseille, France.,Department of Multidisciplinary Peadiatrics, Centre de Référence des Maladies Rares d'origine hypophysaire HYPO, Hôpital Timone-Enfants, APHM, Marseille, France
| |
Collapse
|
35
|
Fang Y, Wan JP, Zhang RJ, Sun F, Yang L, Zhao SX, Dong M, Song HD. Tpo knockout in zebrafish partially recapitulates clinical manifestations of congenital hypothyroidism and reveals the involvement of TH in proper development of glucose homeostasis. Gen Comp Endocrinol 2022; 323-324:114033. [PMID: 35367205 DOI: 10.1016/j.ygcen.2022.114033] [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: 01/27/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/26/2022]
Abstract
Congenital hypothyroidism (CH) is a highly prevalent but treatable neonatal endocrine disorder. Thyroid peroxidase (TPO) catalyzes key reactions in thyroid hormone (TH) synthesis. TPO mutations have been found to underlie approximately 5% of congenital hypothyroidism in Chinese patients with more severe phenotypes, the treatment of whom usually requires a higher dose of L-thyroxine. The Tpo gene of zebrafish has 66% homology with the human TPO gene, and synteny analysis has indicated that it is likely a human TPO ortholog. In this study, we generated a tpo-/- mutant zebrafish line through knockout of tpo with CRISPR/Cas9 and investigated the associated phenotypes. Tpo-/- mutant zebrafish displayed growth retardation; an increased number of thyroid follicular cells; and abnormal extrathyroidal phenotypes including pigmentation defects, erythema in the thoracic region, delayed scale development and failure of swim bladder secondary lobe formation. All these abnormal phenotypes were reversed by 30 nM thyroxine (T4) treatment starting at 1 month of age. Tpo-/- mutants also showed increased glucose levels during larval stages, and the increases were induced at least in part by increasing glucagon and decreasing insulin expression. Our work indicates that tpo-mutant zebrafish may serve as a human congenital hypothyroidism model for studying TPO- and TH-related disease mechanisms.
Collapse
Affiliation(s)
- Ya Fang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jia-Ping Wan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Rui-Jia Zhang
- Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Feng Sun
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Liu Yang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mei Dong
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
36
|
Peters C, Schoenmakers N. MECHANISMS IN ENDOCRINOLOGY: The pathophysiology of transient congenital hypothyroidism. Eur J Endocrinol 2022; 187:R1-R16. [PMID: 35588090 PMCID: PMC9254299 DOI: 10.1530/eje-21-1278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/19/2022] [Indexed: 11/08/2022]
Abstract
Transient congenital hypothyroidism (TCH) refers to congenital hypothyroidism which spontaneously resolves in the first few months or years of life. Currently, there is a paucity of reliable markers predicting TCH at diagnosis, and the diagnosis is established following the withdrawal of levothyroxine therapy around 3 years of age. The incidence of TCH is increasing, and it is a major contributor to the overall increase in the incidence of CH in recent studies. Both genetic factors, in particular mutations affecting DUOX2 and DUOXA2, and environmental factors, for example, iodine deficiency and excess, anti- TSHR antibodies and exposure to antithyroid or iodine-rich medications, may cause TCH. Resolution of TCH in childhood may reflect both normal thyroid physiology (decreased thyroid hormone biosynthesis requirements after the neonatal period) and clearance or cessation of environmental precipitants. The relative contributions and interactions of genetic and environmental factors to TCH, and the extent to which TCH may be prevented, require evaluation in future population-based studies.
Collapse
Affiliation(s)
- Catherine Peters
- Department of Endocrinology, Great Ormond Street Hospital for Children, London, UK
| | - Nadia Schoenmakers
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Correspondence should be addressed to N Schoenmakers;
| |
Collapse
|
37
|
Stern E, Schoenmakers N, Nicholas AK, Kassif E, Hamiel OP, Yeshayahu Y. A Novel Mutation in the Thyroglobulin Gene Resulting in Neonatal Goiter and Congenital Hypothyroidism in an Eritrean Infant. J Clin Res Pediatr Endocrinol 2022; 14:221-226. [PMID: 33832185 PMCID: PMC9176088 DOI: 10.4274/jcrpe.galenos.2021.2020.0278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Congenital hypothyroidism (CH) due to dyshormonogenesis may occur due to mutations in any of the key genes involved in thyroid hormone biosynthesis (TG, TPO, DUOX2, DUOXA2, SLC5A5, IYD, SLC26A4 and SLC26A7). Mutations in the thyroglobulin gene (TG) are frequently associated with goiter, which may present fetally or neonatally, although a spectrum of phenotypes is reported. We present the case of a woman of Eritrean origin who presented in the third trimester of pregnancy in the early stages of labor. Ultrasound at presentation revealed a fetal neck swelling consistent with a goiter. Following delivery by Caesarian section with minimal respiratory support, the infant was found to be hypothyroid with undetectable serum levels of thyroglobulin. Sequencing of the TG revealed a homozygous donor splice site pathogenic variant (c.5686+1delG) not previously described in the literature. Levothyroxine treatment resulted in normal growth and psychomotor development. Goitrous CH with inappropriately low thyroglobulin has previously been reported in patients harbouring homozygous single nucleotide substitutions at the same TG donor splice site, which result in exon skipping and retention of malformed thyroglobulin by the endoplasmic reticulum. We conclude that the TG c.5686+1delG pathogenic variant is the likely basis for our patient’s fetal goiter and CH, and that the clinical phenotype associated with TG c.5686+1delG is comparable to that seen with single nucleotide substitutions at the same site.
Collapse
Affiliation(s)
- Eve Stern
- Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Pediatric Endocrine and Diabetes Unit, Ramat-Gan, Israel,* Address for Correspondence: Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Pediatric Endocrine and Diabetes Unit, Ramat-Gan, Israel Phone: +97235305015 E-mail:
| | - Nadia Schoenmakers
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Adeline K. Nicholas
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Eran Kassif
- Sheba Medical Center, Department of Obstetrics and Gynecology, Tel Hashomer, Israel,Tel Aviv University, Sackler School of Medicine, Tel Aviv, Israel
| | - Orit Pinhas Hamiel
- Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Pediatric Endocrine and Diabetes Unit, Ramat-Gan, Israel,Tel Aviv University, Sackler School of Medicine, Tel Aviv, Israel
| | - Yonatan Yeshayahu
- Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Pediatric Endocrine and Diabetes Unit, Ramat-Gan, Israel,Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Assuta Medical Center, Pediatric Endocrine and Diabetes Unit, Ashdod, Israel
| |
Collapse
|
38
|
Abstract
Hypothyroidism is the common clinical condition of thyroid hormone deficiency and, if left untreated, can lead to serious adverse health effects on multiple organ systems, with the cardiovascular system as the most robustly studied target. Overt primary hypothyroidism is defined as elevated thyroid-stimulating hormone (TSH) concentration in combination with free thyroxine (fT4) concentration below the reference range. Subclinical hypothyroidism, commonly considered an early sign of thyroid failure, is defined by elevated TSH concentrations but fT4 concentrations within the reference range. Hypothyroidism is classified as primary, central or peripheral based on pathology in the thyroid, the pituitary or hypothalamus, or peripheral tissue, respectively. Acquired primary hypothyroidism is the most prevalent form and can be caused by severe iodine deficiency but is more frequently caused by chronic autoimmune thyroiditis in iodine-replete areas. The onset of hypothyroidism is insidious in most cases and symptoms may present relatively late in the disease process. There is a large variation in clinical presentation and the presence of hypothyroid symptoms, especially in pregnancy and in children. Levothyroxine (LT4) is the mainstay of treatment and is one of the most commonly prescribed drugs worldwide. After normalization of TSH and fT4 concentrations, a considerable proportion of patients treated with LT4 continue to have persistent complaints, compromising quality of life. Further research is needed regarding the appropriateness of currently applied reference ranges and treatment thresholds, particularly in pregnancy, and the potential benefit of LT4/liothyronine combination therapy for thyroid-related symptom relief, patient satisfaction and long-term adverse effects.
Collapse
|
39
|
Uthayaseelan K, Kadari M, Subhan M, Saji Parel N, Krishna PV, Gupta A, Uthayaseelan K. Congenital Anomalies in Infant With Congenital Hypothyroidism: A Review of Pathogenesis, Diagnostic Options, and Management Protocols. Cureus 2022; 14:e24669. [PMID: 35663669 PMCID: PMC9162097 DOI: 10.7759/cureus.24669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
Thyroid hormones (TH) regulate growth, nervous system myelination, metabolism, and physiologic functions in nearly every organ system. Congenital hypothyroidism (CH) is one of the most common endocrinopathies in children and has potentially devastating neurologic and developmental consequences. The etiology and clinical manifestations of hypothyroidism in children differ from adults. And hence, pediatric medical care requires a detailed understanding of thyroid function and dysfunction in children. The perinatal risk factors include female sex, preterm birth, low birth weight, postmature birth, additional birth abnormalities, and being delivered in multiple births. In countries where newborn screening is practiced, CH is detected after birth through screening tests. It aids in determining the underlying cause, though some patients may be able to start treatment without these tests. Early detection and treatment prevent irreversible and permanent nervous system damage. Thus, in addition to exploring the development of CH, this article has also covered the epidemiological data, clinical aspects, and management stemming from pediatric hypothyroidism.
Collapse
|
40
|
Wei X, Tan J, Gao H. Role of sirtuin 1 in the brain development in congenital hypothyroidism rats via the regulation of p53 signaling pathway. Bioengineered 2022; 13:9455-9466. [PMID: 35383535 PMCID: PMC9161855 DOI: 10.1080/21655979.2022.2060626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The present study aimed to investigate the expression, role, and underlying mechanism of action of sirtuin 1 (SIRT1) in congenital hypothyroidism (CH). A CH model was established in rats, and neuronal cells were isolated from the hippocampal tissues of normal rats. Free thyroxine (fT4) and thyroid-Stimulating hormone (TSH) concentrations were determined to confirm CH model conduction. The cognitive behavior of rats with CH was examined using open field and forced swimming tests. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to detect the expression levels of SIRT1, p53, B-cell lymphoma-extra-large (Bcl-xl), Bcl-2-associated X (Bax), and cytochrome c in the hippocampal tissues and neuronal cells. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and flow cytometry were performed to evaluate cell viability and apoptosis, respectively. The results revealed that SIRT1 was expressed at low levels in the hippocampal tissues of rats with CH. Moreover, overexpression of SIRT1 in the hippocampal tissues of rats with CH and improved rat behavior, while reducing the CH-induced nerve cell apoptosis. In addition, this overexpression increased the viability, inhibited apoptosis, and reduced the expression of p53, Bax, and cytochrome c, while increasing the expression of Bcl-xl in cultured neurons. In contrast, SIRT1-small interfering RNA exhibited the opposite effects in cultured neurons. In conclusion, SIRT1 plays a role in the occurrence and development of CH by regulating nerve cell apoptosis.
Collapse
Affiliation(s)
- Xiaofang Wei
- Department of Obstetrics, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Juan Tan
- Department of Genetics and Prenatal Diagnosis, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu, China
| | - Hui Gao
- Department of Pediatrics, Lianyungang Affiliated Hospital Nanjing University of Traditional Chinese Medicine, Lianyungang, Jiangsu, China
| |
Collapse
|
41
|
Walsh JP. Thyroid Function across the Lifespan: Do Age-Related Changes Matter? Endocrinol Metab (Seoul) 2022; 37:208-219. [PMID: 35417936 PMCID: PMC9081302 DOI: 10.3803/enm.2022.1463] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/15/2022] Open
Abstract
Circulating concentrations of thyrotropin (TSH) and thyroxine (T4) are tightly regulated. Each individual has setpoints for TSH and free T4 which are genetically determined, and subject to environmental and epigenetic influence. Pituitary-thyroid axis setpoints are probably established in utero, with maturation of thyroid function continuing until late gestation. From neonatal life (characterized by a surge of TSH and T4 secretion) through childhood and adolescence (when free triiodothyronine levels are higher than in adults), thyroid function tests display complex, dynamic patterns which are sexually dimorphic. In later life, TSH increases with age in healthy older adults without an accompanying fall in free T4, indicating alteration in TSH setpoint. In view of this, and evidence that mild subclinical hypothyroidism in older people has no health impact, a strong case can be made for implementation of age-related TSH reference ranges in adults, as is routine in children.
Collapse
Affiliation(s)
- John P. Walsh
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, Australia
- Medical School, University of Western Australia, Crawley, Australia
- Corresponding author: John P. Walsh Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Hospital Avenue, Nedlands, Western Australia 6009, Australia Tel: +61-864572466, Fax: +61-864573221, E-mail:
| |
Collapse
|
42
|
Hammersen J, Bettendorf M, Bonfig W, Schönau E, Warncke K, Eckert AJ, Fricke-Otto S, Palm K, Holl RW, Woelfle J. Twenty years of newborn screening for congenital adrenal hyperplasia and congenital primary hypothyroidism - experiences from the DGKED/AQUAPE study group for quality improvement in Germany. MED GENET-BERLIN 2022; 34:29-40. [PMID: 38836017 PMCID: PMC11007626 DOI: 10.1515/medgen-2022-2114] [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: 11/01/2021] [Accepted: 03/01/2022] [Indexed: 06/06/2024]
Abstract
Congenital primary hypothyroidism (CH) and congenital adrenal hyperplasia (CAH) are targeted by the German and Austrian newborn screening. For both diseases, there are registries for quality improvement, based on standardized observational data from long-term patient follow-up, under the auspices of the DGKED study group. By September 2021, the CH registry HypoDOK includes datasets from 23,348 visits of 1,840 patients, and the CAH registry contains datasets from 36,237 visits of 1,976 patients. Here, we report on the recruitment process, patient characteristics, and research contributions from the registries, and underline that the registries are an important tool to improve patient care and outcomes. Registries for rare conditions should thus be considered as an important public health measure and they should be adequately institutionalized and funded.
Collapse
Affiliation(s)
- Johanna Hammersen
- Department of Paediatrics, University Hospital Erlangen, Erlangen, Germany
| | - Markus Bettendorf
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | - Walter Bonfig
- Department of Paediatrics, Klinikum Wels-Grieskirchen, Wels, Austria
| | - Eckhard Schönau
- Children's Hospital, University Hospital of Cologne and UniReha, University Hospital of Cologne, Cologne, Germany
| | - Katharina Warncke
- Department of Paediatrics, Kinderklinik München Schwabing, Technical University of Munich School of Medicine, Munich, Germany
| | - Alexander J Eckert
- Institute of Epidemiology and Medical Biometry, ZIBMT, Ulm University, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | | | - Katja Palm
- Otto-von-Guericke-University Magdeburg, Department of Paediatrics, Magdeburg, Germany
| | - Reinhard W Holl
- Institute of Epidemiology and Medical Biometry, ZIBMT, Ulm University, Ulm, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - Joachim Woelfle
- Department of Paediatrics, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
43
|
Shakerian N, Mard-Soltani M, Nasri S, Rasaee MJ, Khalili S. Different combinations of monoclonal antibodies and polyclonal antibodies in the design of neonatal hypothyroidism diagnostic kit. Appl Biochem Biotechnol 2022; 194:3167-3181. [PMID: 35349083 DOI: 10.1007/s12010-022-03888-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 03/14/2022] [Indexed: 11/25/2022]
Abstract
Neonatal hypothyroidism is a deficiency of thyroid hormones at birth that can cause lifelong mental and physical disorders in humans. Lack of timely detection could lead to irreversible damage by neonatal hypothyroidism. However, it could be managed quickly and efficiently via timely diagnosis. The screening programs rely on immunoassays to diagnose neonatal hypothyroidism in most countries. This method is time-consuming, needs laboratory equipment, and should be performed by trained and skilled technicians. Given these circumstances, the ELISA method is not a preferable method for the diagnosing of neonatal hypothyroidism. However, it can be used as a confirmatory method in infants with suspected and unknown neonatal hypothyroidism. In the present study, the homemade SR95-1, SR95-2, and SR95-3 anti-β-TSH polyclonal and the commercially available monoclonal antibodies were used to detect β-TSH in a rapid assay kit design hypothyroidism screening. To design the kit, the different combinations of the antibodies were used to establish a sandwich immune-chromatography method. The designed rapid neonatal hypothyroidism tests were used to measure neonatal β-TSH in 100 dry blood samples. This study showed that the best antibody pair in terms of sensitivity is the SR95-1 antibody as capture antibody and the SR95-2 as a conjugated antibody. Using 100 clinical samples, the designed assay was shown to have 94% sensitivity, 83% specificity, and 94% accuracy. The results showed that polyclonal antibodies (SR95-1 as capture) and SR95-2 (as detector) antibodies can detect the reference range of β-TSH in dried blood samples and can be used in the screening of neonatal hypothyroidism.
Collapse
Affiliation(s)
- Neda Shakerian
- Department of Biology, Payame Noor University, Tehran, Iran
| | - Maysam Mard-Soltani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran.
| | - Sima Nasri
- Department of Biology, Payame Noor University, Tehran, Iran
| | - Mohammad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
44
|
NKX2-5 Variant in Two Siblings with Thyroid Hemiagenesis. Int J Mol Sci 2022; 23:ijms23063414. [PMID: 35328834 PMCID: PMC8950672 DOI: 10.3390/ijms23063414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/04/2022] Open
Abstract
Thyroid hemiagenesis (THA) is an inborn absence of one thyroid lobe of largely unknown etiopathogenesis. The aim of the study was to reveal genetic factors responsible for thyroid maldevelopment in two siblings with THA. None of the family members presented with congenital heart defect. The samples were subjected to whole-exome sequencing (WES) (Illumina, TruSeq Exome Enrichment Kit, San Diego, CA 92121, USA). An ultra-rare variant c.839C>T (p.Pro280Leu) in NKX2-5 gene (NM_004387.4) was identified in both affected children and an unaffected father. In the mother, the variant was not present. This variant is reported in population databases with 0.0000655 MAF (GnomAD v3, dbSNP rs761596254). The affected amino acid position is moderately conserved (positive scores in PhyloP: 1.364 and phastCons: 0.398). Functional prediction algorithms showed deleterious impact (dbNSFP v4.1, FATHMM, SIFT) or benign (CADD, PolyPhen-2, Mutation Assessor). According to ACMG criteria, variant is classified as having uncertain clinical significance. For the first time, NKX2-5 gene variants were found in two siblings with THA, providing evidence for its potential contribution to the pathogenesis of this type of thyroid dysgenesis. The presence of the variant in an unaffected parent, carrier of p.Pro280Leu variant, suggests potential contribution of yet unidentified additional factors determining the final penetrance and expression.
Collapse
|
45
|
Levaillant L, Huet F, Bretones P, Corne C, Dupuis C, Reynaud R, Somma C, Barat P, Corcuff J, Bouhours-Nouet N, Gauthereau V, Polak M, Leger J, Cheillan D, Coutant R. Neonatal screening for congenital hypothyroidism: Time to lower the TSH threshold in France. Arch Pediatr 2022; 29:253-257. [DOI: 10.1016/j.arcped.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 12/20/2021] [Accepted: 02/04/2022] [Indexed: 10/18/2022]
|
46
|
Liang J, Qian J, Yang L, Chen X, Wang X, Lin X, Wang X, Zhao B. Modeling Human Thyroid Development by Fetal Tissue-Derived Organoid Culture. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105568. [PMID: 35064652 PMCID: PMC8948548 DOI: 10.1002/advs.202105568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/06/2022] [Indexed: 05/29/2023]
Abstract
Euthyroidism is of profound importance for lifetime health. However, the early diagnosis or therapeutics of thyroid developmental defects has not been established, mainly due to limited understanding of human thyroid development and a lack of recapitulating research model. Herein, the authors elaborate the cell atlas and potential regulatory signaling of the evolution of heterogeneous thyrocyte population from 12 to 16 gestational weeks. Moreover, they establish a long-term culture of human fetal thyroid organoids (hFTOs) system, which retains the fetal thyroid lineages and molecular signatures, as well as the ability to generate functional human thyroid follicles post mice renal transplantation. Notably, cAMP signaling activation in hFTOs by forskolin boosts the maturation of follicle and thus thyroid hormone T4 secretion, which recapitulates the key developmental events of fetal thyroid. Employing this ex vivo system, it is found that enhanced chromatin accessibility at thyroid maturation genes (such as TPO and TG) loci permits the transcription for hormone production. This study provides the cell atlas of and an organoid model for human thyroid development, which will facilitate thyroid research and prospective medicine.
Collapse
Affiliation(s)
- Jianqing Liang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| | - Jun Qian
- State Key Laboratory of Medical Molecular BiologyDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijing100730China
| | - Li Yang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| | - Xiaojun Chen
- Obstetrics and Gynecology Hospital of Fudan UniversityShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai200011China
| | - Xiaoning Wang
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversitySchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510000China
| | - Xinhua Lin
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| | - Xiaoyue Wang
- State Key Laboratory of Medical Molecular BiologyDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijing100730China
| | - Bing Zhao
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| |
Collapse
|
47
|
Петеркова ВА, Безлепкина ОБ, Ширяева ТЮ, Вадина ТА, Нагаева ЕВ, Чикулаева ОА, Шредер ЕВ, Конюхова МБ, Макрецкая НА, Шестопалова ЕА, Митькина ВБ. [Clinical guideline of «congenital hypothyroidism»]. PROBLEMY ENDOKRINOLOGII 2022; 68:90-103. [PMID: 35488760 PMCID: PMC9764271 DOI: 10.14341/probl12880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/09/2023]
Abstract
Congenital hypothyroidism is an important issue of pediatric endocrinology at which timely diagnosis and treatment can prevent the development of severe cases of the disease. The developed clinical guidelines are a working tool for a practicing physician. The target audience is pediatric endocrinologists and pediatricians. They briefly and logically set out the main definition of the disease, epidemiology, classification, methods of diagnosis and treatment, based on the principles of -evidence-based medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - М. Б. Конюхова
- Московский центр неонатального скрининга Морозовской детской городской клинической больницы
| | | | | | - В. Б. Митькина
- Московский центр неонатального скрининга Морозовской детской городской клинической больницы
| |
Collapse
|
48
|
Abstract
Fetal and neonatal dysfunctions include rare serious disorders involving abnormal thyroid function during the second half of gestation, which may persist throughout life, as for most congenital thyroid disorders, or be transient, resolving in the first few weeks of life, as in autoimmune hyperthyroidism or hypothyroidism and some cases of congenital hypothyroidism (CH) with the thyroid gland in situ. Primary CH is diagnosed by neonatal screening, which has been implemented for 40 years in developed countries and should be introduced worldwide, as early treatment prevents irreversible neurodevelopmental delay. Central CH is a rarer entity occurring mostly in association with multiple pituitary hormone deficiencies. Other rare disorders impair the action of thyroid hormones. Neonatal Graves' disease (GD) results from the passage of thyrotropin receptor antibodies (TRAbs) across the placenta, from mother to fetus. It may affect the fetuses and neonates of mothers with a history of current or past GD, but hyperthyroidism develops only in those with high levels of stimulatory TRAb activity. The presence of antibodies predominantly blocking thyroid-stimulating hormone receptors may result in transient hypothyroidism, possibly followed by neonatal hyperthyroidism, depending on the balance between the antibodies present. Antithyroid drugs taken by the mother cross the placenta, treating potential fetal hyperthyroidism, but they may also cause transient fetal and neonatal hypothyroidism. Early diagnosis and treatment are key to optimizing the child's prognosis. This review focuses on the diagnosis and management of these patients during the fetal and neonatal periods. It includes the description of a case of fetal and neonatal autoimmune hyperthyroidism.
Collapse
Affiliation(s)
- Juliane Léger
- Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Pediatric Endocrinology-Diabetology Department, Reference Center for Growth and Development Endocrine Diseases, Paris, France
- Université de Paris; NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Clemence Delcour
- Université de Paris; NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Gynecology and Obstetric Department, Paris, France
| | - Jean-Claude Carel
- Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Pediatric Endocrinology-Diabetology Department, Reference Center for Growth and Development Endocrine Diseases, Paris, France
- Université de Paris; NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| |
Collapse
|
49
|
Castets S, Villanueva C, Vergier J, Brue T, Saveanu A, Reynaud R. Clinical, radiological, and molecular diagnosis of congenital pituitary diseases causing short stature. Arch Pediatr 2022; 28:8S33-8S38. [PMID: 37870532 DOI: 10.1016/s0929-693x(22)00041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Short stature in children can be caused by congenital pituitary disorders involving at least one form of growth hormone deficiency. Clinical and radiological evaluations of the index case and family history assessments are essential to guide genetic diagnostic testing and interpret results. The first-line approach is panel testing of genes involved in pituitary development with variants known to be pathogenic in this context. It identifies a genetic cause in less than 10% of cases, however. Whole-exome and whole-genome sequencing techniques may provide original information but also raise new questions regarding the pathophysiological role of identified variants. These new tools can make genetic counselling more complex. The role of clinicians in these interpretations is therefore important. © 2022 French Society of Pediatrics. Published by Elsevier Masson SAS. All rights reserved.
Collapse
Affiliation(s)
- S Castets
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital Timone Enfants, Service de Pédiatrie Multidisciplinaire, Marseille, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France.
| | - C Villanueva
- Hospices Civils de Lyon (HCL), Hôpital Femme Mère Enfant (HFME), Service d'Endocrinologie pédiatrique, Bron, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France
| | - J Vergier
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital Timone Enfants, Service de Pédiatrie Multidisciplinaire, Marseille, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France
| | - T Brue
- Hospices Civils de Lyon (HCL), Hôpital Femme Mère Enfant (HFME), Service d'Endocrinologie pédiatrique, Bron, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France; Assistance Publique-Hôpitaux de Marseille (AP-HM), Service d'Endocrinologie, Hôpital de la Conception, Marseille, France; Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| | - A Saveanu
- Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France; Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Marseille Medical Genetics (MMG), U 1251, Marseille, France; Institut Marseille Maladies Rares (MarMaRa), Marseille, France; Assistance Publique-Hôpitaux de Marseille (AP-HM), Laboratoire de Biologie Moléculaire, Hôpital de la Conception, Marseille, France
| | - R Reynaud
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital Timone Enfants, Service de Pédiatrie Multidisciplinaire, Marseille, France; Centre de Référence des Maladies Rares de l'Hypophyse HYPO, Marseille, France; Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Marseille Medical Genetics (MMG), U 1251, Marseille, France; Institut Marseille Maladies Rares (MarMaRa), Marseille, France
| |
Collapse
|
50
|
Lipska E, Lecka-Ambroziak A, Witkowski D, Szamotulska K, Mierzejewska E, Ołtarzewski M. Primary Congenital Hypothyroidism in Children Below 3 Years Old - Etiology and Treatment With Overtreatment and Undertreatment Risks, a 5-Year Single Centre Experience. Front Endocrinol (Lausanne) 2022; 13:895507. [PMID: 35832434 PMCID: PMC9271568 DOI: 10.3389/fendo.2022.895507] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/18/2022] [Indexed: 12/02/2022] Open
Abstract
Worldwide neonatal screening for congenital hypothyroidism (CH) is a gold standard of active surveillance in newborns. Prompt diagnosis, subsequent timely treatment implementation, and proper dosage of levothyroxine (L-T4) are crucial for normal growth and development, especially of the central nervous system. However, overtreatment may have a potential negative impact on further neurodevelopment. We retrospectively analysed data of 99 newborns with CH diagnosis, referred to the Endocrinology Outpatient Clinic of the Institute of Mother and Child in Warsaw, Poland from the CH screening program from 2017 to 2021. We evaluated the diagnostic process and treatment up to the age of 3 years. We compared groups of children from the first and the second screening groups (FSG, SSG) in the neonatal screening with an evaluation of ultrasound examination (thyroid dysgenesis vs. gland in situ, GIS). The overtreatment and undertreatment risks were assessed and an analysis of the new TSH thresholds was performed. Treatment was implemented at a median of 9 days of life (3 - 27); 8 days (3 - 17) in FSG and 19 (6 - 27) in SSG. The dose of L-T4 differed between FSG and SSG at all three analysed time points (start of the therapy, 12 months, and 3 years) with significantly higher doses in FSG. The same was observed for the patients with thyroid dysgenesis vs. GIS. Screening TSH level was ≥ 28mIU/l in 91.7% of patients with thyroid dysgenesis in comparison to 74.0% of patients with GIS (p= 0.038). The optimally treated group (fT4 in the upper half of the reference range, according to the guidelines) was up to 58.0% of the children during the follow-up. The risk for overtreatment was present in 1/5 of the study group after 12 months and 1/4 after 3 years of L-T4 therapy. Analysis of new TSH thresholds showed an increased prevalence of mild hypothyroidism, GIS, and either euthyroid state or overtreatment while treating with lower L-T4 doses in comparison to the rest of the cohort. The study confirmed the general efficacy of the CH diagnostic pathway and the timely implemented L-T4 therapy. The suspected overtreatment after the first 12 months of L-T4 therapy requires consideration of the earlier diagnosis re-evaluation.
Collapse
Affiliation(s)
- Elżbieta Lipska
- Endocrinology Outpatient Clinic, Institute of Mother and Child, Warsaw, Poland
- *Correspondence: Elżbieta Lipska,
| | - Agnieszka Lecka-Ambroziak
- Endocrinology Outpatient Clinic, Institute of Mother and Child, Warsaw, Poland
- Department of Endocrinology and Diabetology, Children’s Memorial Health Institute, Warsaw, Poland
| | - Daniel Witkowski
- Endocrinology Outpatient Clinic, Institute of Mother and Child, Warsaw, Poland
| | - Katarzyna Szamotulska
- Department of Epidemiology and Biostatistics, Institute of Mother and Child, Warsaw, Poland
| | - Ewa Mierzejewska
- Department of Epidemiology and Biostatistics, Institute of Mother and Child, Warsaw, Poland
| | - Mariusz Ołtarzewski
- Department of Screening and Metabolic Diagnostics, Institute of Mother and Child, Warsaw, Poland
| |
Collapse
|