1
|
Undeutsch HJ, Posabella A, Alber AB, Bawa PS, Villacorta-Martin C, Wang F, Ikonomou L, Kotton DN, Hollenberg AN. Derivation of transplantable human thyroid follicular epithelial cells from induced pluripotent stem cells. Stem Cell Reports 2024; 19:1690-1705. [PMID: 39515316 DOI: 10.1016/j.stemcr.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
The production of mature functioning thyroid follicular cells (TFCs) from human induced pluripotent stem cells (iPSCs) is critical for potential novel therapeutic approaches to post-surgical and congenital hypothyroidism. To accomplish this, we developed a novel human iPSC line that expresses fluorophores targeted to the NKX2-1 and PAX8 loci, allowing for the identification and purification of cells destined to become TFCs. Optimizing a sequence of defined, serum-free media to promote stepwise developmental directed differentiation, we found that bone morphogenic protein 4 (BMP4) and fibroblast growth factor 2 (FGF2) stimulated lineage specification into TFCs from multiple iPSC lines. Single-cell RNA sequencing demonstrated that BMP4 withdrawal after lineage specification promoted TFC maturation, with mature TFCs representing the majority of cells present within 1 month. After xenotransplantation into athyreotic immunodeficient mice, engrafted cells exhibited thyroid follicular organization with thyroglobulin protein detected in the lumens of NKX2-1-positive follicles. While our iPSC-derived TFCs presented durable expression of thyroid-specific proteins, they were unable to rescue hypothyroidism in vivo.
Collapse
Affiliation(s)
- Hendrik J Undeutsch
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - Alberto Posabella
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA; University Center of Gastrointestinal and Liver Diseases - Clarunis, University of Basel Faculty of Medicine, Basel, Switzerland
| | - Andrea B Alber
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - Pushpinder S Bawa
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - Carlos Villacorta-Martin
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - Feiya Wang
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
| | - Laertis Ikonomou
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA; Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
| | - Darrell N Kotton
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA; The Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA.
| |
Collapse
|
2
|
Titov SE, Kozorezova ES, Lukyanov SA, Sergiyko SV, Demenkov PS, Veryaskina YA, Vorobyev SL, Sleptsov IV, Chernikov RA, Timofeeva NI, Barashkova SV, Lushnikova EL, Uspenskaya AA, Zolotoukho AV, Romanova OV, Zhimulev IF. Could SLC26A7 Be a Promising Marker for Preoperative Diagnosis of High-Grade Papillary Thyroid Carcinoma? Diagnostics (Basel) 2024; 14:2652. [PMID: 39682560 DOI: 10.3390/diagnostics14232652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES A modern classification distinguishes between two nosological entities posing an intermediate risk between differentiated and anaplastic carcinoma: poorly differentiated thyroid carcinoma and differentiated high-grade thyroid carcinoma. There are currently few studies searching for the preoperative molecular genetic markers of high-grade papillary thyroid carcinoma (PTC HG), primarily because of a recent WHO reclassification and singling out of a separate entity: high-grade follicular cell-derived nonanaplastic thyroid carcinoma. Therefore, this work was aimed at identifying PTC HG-specific microRNAs and mRNAs that reliably distinguish them from differentiated papillary thyroid carcinoma in preoperative cytology specimens (fine-needle aspiration biopsies). METHODS A molecular genetic profile (expression levels of 14 genes and eight microRNAs) was studied in 110 cytology specimens from patients with PTC: 13 PTCs HG and 97 PTCs without features of HG. RESULTS Of the examined eight microRNAs and 14 genes, significant differences in the expression levels between the PTC and PTC HG groups were revealed for genes SLC26A7, TFF3, and TPO. Only one gene (SLC26A7) proved to be crucial for detecting PTC HG. It showed the largest area under the ROC curve (0.816) in differentiation between the PTC and PTC HG groups and was the key element of the decision tree by ensuring 54% sensitivity and 87.6% specificity. CONCLUSIONS Early preoperative diagnosis of PTC HG in patients with early stages of this cancer type will allow clinicians to modify a treatment strategy toward a larger surgery volume and lymph node dissection and may provide indications for subsequent radioactive iodine therapy.
Collapse
Affiliation(s)
- Sergei E Titov
- Department of the Structure and Function of Chromosomes, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- PCR Laboratory, AO Vector-Best, Novosibirsk 630117, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Evgeniya S Kozorezova
- National Center of Clinical Morphological Diagnostics, Saint Petersburg 192283, Russia
| | - Sergei A Lukyanov
- Department of General and Pediatric Surgery, South Ural State Medical University, Chelyabinsk 454092, Russia
| | - Sergei V Sergiyko
- Department of General and Pediatric Surgery, South Ural State Medical University, Chelyabinsk 454092, Russia
| | - Pavel S Demenkov
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Yulia A Veryaskina
- Department of the Structure and Function of Chromosomes, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey L Vorobyev
- National Center of Clinical Morphological Diagnostics, Saint Petersburg 192283, Russia
| | - Ilya V Sleptsov
- Department of Endocrine Surgery, Saint Petersburg State University Hospital, Saint Petersburg 199034, Russia
| | - Roman A Chernikov
- Department of Endocrine Surgery, Saint Petersburg State University Hospital, Saint Petersburg 199034, Russia
| | - Natalia I Timofeeva
- Department of Endocrine Surgery, Saint Petersburg State University Hospital, Saint Petersburg 199034, Russia
| | - Svetlana V Barashkova
- National Center of Clinical Morphological Diagnostics, Saint Petersburg 192283, Russia
| | - Elena L Lushnikova
- Department of Molecular Pathology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
| | - Anna A Uspenskaya
- Department of Endocrine Surgery, Saint Petersburg State University Hospital, Saint Petersburg 199034, Russia
| | - Anna V Zolotoukho
- Department of Endocrine Surgery, Saint Petersburg State University Hospital, Saint Petersburg 199034, Russia
| | - Olga V Romanova
- N.N. Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia
| | - Igor F Zhimulev
- Department of the Structure and Function of Chromosomes, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
3
|
Hiron TK, Aguiar J, Williams JM, Falcone S, Norman PA, Elliott J, Fowkes RC, Syme HM, Davison LJ. Transcriptomic analysis reveals a critical role for activating G sα mutations in spontaneous feline hyperthyroidism. Sci Rep 2024; 14:28749. [PMID: 39567583 PMCID: PMC11579033 DOI: 10.1038/s41598-024-79564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
Feline hyperthyroidism (FHT) is a debilitating disease affecting > 10% of elderly cats. It is generally characterised by chronic elevation of thyroid hormone in the absence of circulating TSH. Understanding of the molecular pathogenesis of FHT is currently limited. However, FHT shares clinical and histopathological similarities with human toxic multinodular goitre, which has been associated with activating mutations in TSH receptor (TSHR) and Gsα encoding genes. Using RNA-seq transcriptomic analysis of thyroid tissue from hyperthyroid and euthyroid cats, we identified differentially expressed genes and dysregulated pathways in FHT, many of which are downstream of TSHR. In addition, we detected missense variants in thyroid RNA-seq reads that alter the structure of both TSHR and Gsα. All FHT-associated mutations were absent in germline sequence from paired blood samples. Only a small number of hyperthyroid cats demonstrated TSHR variation, however all thyroids from advanced cases of FHT carried at least one missense variant affecting Gsα. The activating nature of the acquired Gsα mutations was demonstrated by increased cAMP production in vitro. These data indicate that constitutive activation of signalling downstream of TSHR is central to the TSH-independent production of thyroid hormone in FHT, offering a novel therapeutic target pathway in this common disease.
Collapse
Affiliation(s)
- Thomas K Hiron
- Department of Clinical Science and Services, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Joana Aguiar
- Department of Clinical Science and Services, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
- Dick White Referrals, Station Farm, London Road, Six Mile Bottom, Cambridgeshire, CB8 0UH, UK
| | - Jonathan M Williams
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
| | - Sara Falcone
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Paul A Norman
- Department of Clinical Science and Services, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
| | - Jonathan Elliott
- Department of Comparative Biomedical Science, The Royal Veterinary College, London, NW1 0TU, UK
| | - Robert C Fowkes
- Department of Comparative Biomedical Science, The Royal Veterinary College, London, NW1 0TU, UK
- Department of Small Animal Clinical Sciences, Michigan State University College of Veterinary Medicine, East Lansing, MI, 48824, USA
| | - Harriet M Syme
- Department of Clinical Science and Services, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
| | - Lucy J Davison
- Department of Clinical Science and Services, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.
| |
Collapse
|
4
|
Narumi S. Deciphering the mystery of CHNG3. Ann Pediatr Endocrinol Metab 2024; 29:279-283. [PMID: 39506342 PMCID: PMC11541093 DOI: 10.6065/apem.2448186.093] [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: 08/07/2024] [Revised: 08/28/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
Congenital hypothyroidism (CH), characterized by insufficient thyroid hormone production due to abnormalities in the hypothalamic-pituitary-thyroid axis, is the most common congenital endocrine disorder. We previously conducted comprehensive genetic screening of 102 patients with permanent CH born in Kanagawa Prefecture, Japan and identified mutations in several genes in 19 CH patients, including defects in genes encoding dual oxidase 2, thyroglobulin, thyrotropin receptor, thyroid peroxidase, and paired-box 8. Despite these findings, approximately 80% of cases remain unexplained. CH pedigrees unexplained by known genetic forms of CH have been reported in the literature and registered as congenital hypothyroidism, nongoitrous, 3 (CHNG3; %609893) in Online Mendelian Inheritance in Man. We also identified a Japanese pedigree of CH that was compatible with CHNG3. However, the exact genetic cause of CHNG3 was not revealed by standard analysis methods such as exome sequencing and array comparative genomic hybridization. We therefore took a combined approach and analyzed a total of 11 undiagnosed CH pedigrees by whole genome sequencing to analyze a 3-Mb linkage region, and found a disease-causing variant affecting a TTTG microsatellite in a noncoding region on chromosome 15. Further analysis revealed that 13.9% of 989 Japanese CH patients had abnormalities involving the TTTG microsatellite, with a substantial proportion (41.5%) of familial CH cases carrying these mutations. Identification of the genetic cause of CHNG3 provides new insights into the pathogenesis of CH, and highlights the need for continued exploration of noncoding genomic regions in Mendelian disorders of unknown etiology.
Collapse
Affiliation(s)
- Satoshi Narumi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Sciarroni E, Montanelli L, Di Cosmo C, Bagattini B, Comi S, Pignata L, Brancatella A, De Marco G, Ferrarini E, Nencetti C, Sessa MR, Latrofa F, Santini F, Tonacchera M, Agretti P. Late-onset dyshormonogenic goitrous hypothyroidism due to a homozygous mutation of the SLC26A7 gene: a case report. Ital J Pediatr 2024; 50:106. [PMID: 38812002 PMCID: PMC11137895 DOI: 10.1186/s13052-024-01672-3] [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: 08/08/2023] [Accepted: 05/06/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND In this study, we used targeted next-generation sequencing (NGS) to investigate the genetic basis of congenital hypothyroidism (CH) in a 19-year-old Tunisian man who presented with severe hypothyroidism and goiter. CASE PRESENTATION The propositus reported the appearance of goiter when he was 18. Importantly, he did not show signs of mental retardation, and his growth was proportionate. A partial organification defect was detected through the perchlorate-induced iodide discharge test. NGS identified a novel homozygous mutation in exon 18 of the SLC26A7 gene (P628Qfs*11), which encodes for a new iodide transporter. This variant is predicted to result in a truncated protein. Notably, the patient's euthyroid brother was heterozygous for the same mutation. No renal acid-base abnormalities were found and the administration of 1 mg of iodine failed to correct hypothyroidism. CONCLUSIONS We described the first case of goitrous CH due to a homozygous mutation of the SLC26A7 gene diagnosed during late adolescence.
Collapse
Affiliation(s)
- Elisabetta Sciarroni
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy.
| | - Lucia Montanelli
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Caterina Di Cosmo
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Brunella Bagattini
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Simone Comi
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Luisa Pignata
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Alessandro Brancatella
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Giuseppina De Marco
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Eleonora Ferrarini
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Chiara Nencetti
- Laboratory of Chemistry and Endocrinology, University Hospital of Pisa, 56124, Pisa, Italy
| | - Maria Rita Sessa
- Laboratory of Chemistry and Endocrinology, University Hospital of Pisa, 56124, Pisa, Italy
| | - Francesco Latrofa
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Ferruccio Santini
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Massimo Tonacchera
- Department of Clinical and Experimental Medicine, Endocrine Unit, Research Center of Excellence AmbiSEN, University of Pisa, 56124, Pisa, Italy
| | - Patrizia Agretti
- Laboratory of Chemistry and Endocrinology, University Hospital of Pisa, 56124, Pisa, Italy
| |
Collapse
|
6
|
Lee D, Hong JH. Chloride/Multiple Anion Exchanger SLC26A Family: Systemic Roles of SLC26A4 in Various Organs. Int J Mol Sci 2024; 25:4190. [PMID: 38673775 PMCID: PMC11050216 DOI: 10.3390/ijms25084190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Solute carrier family 26 member 4 (SLC26A4) is a member of the SLC26A transporter family and is expressed in various tissues, including the airway epithelium, kidney, thyroid, and tumors. It transports various ions, including bicarbonate, chloride, iodine, and oxalate. As a multiple-ion transporter, SLC26A4 is involved in the maintenance of hearing function, renal function, blood pressure, and hormone and pH regulation. In this review, we have summarized the various functions of SLC26A4 in multiple tissues and organs. Moreover, the relationships between SLC26A4 and other channels, such as cystic fibrosis transmembrane conductance regulator, epithelial sodium channel, and sodium chloride cotransporter, are highlighted. Although the modulation of SLC26A4 is critical for recovery from malfunctions of various organs, development of specific inducers or agonists of SLC26A4 remains challenging. This review contributes to providing a better understanding of the role of SLC26A4 and development of therapeutic approaches for the SLC26A4-associated hearing loss and SLC26A4-related dysfunction of various organs.
Collapse
Affiliation(s)
| | - Jeong Hee Hong
- Department of Health Sciences and Technology, GAIHST (Gachon Advanced Institute for Health Sciences and Technology), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| |
Collapse
|
7
|
Geertsma ER, Oliver D. SLC26 Anion Transporters. Handb Exp Pharmacol 2024; 283:319-360. [PMID: 37947907 DOI: 10.1007/164_2023_698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Solute carrier family 26 (SLC26) is a family of functionally diverse anion transporters found in all kingdoms of life. Anions transported by SLC26 proteins include chloride, bicarbonate, and sulfate, but also small organic dicarboxylates such as fumarate and oxalate. The human genome encodes ten functional homologs, several of which are causally associated with severe human diseases, highlighting their physiological importance. Here, we review novel insights into the structure and function of SLC26 proteins and summarize the physiological relevance of human members.
Collapse
Affiliation(s)
- Eric R Geertsma
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, Marburg, Giessen, Germany.
| |
Collapse
|
8
|
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
|
9
|
Li J, Huang S, Liu S, Liao X, Yan S, Liu Q. SLC26 family: a new insight for kidney stone disease. Front Physiol 2023; 14:1118342. [PMID: 37304821 PMCID: PMC10247987 DOI: 10.3389/fphys.2023.1118342] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
The solute-linked carrier 26 (SLC26) protein family is comprised of multifunctional transporters of substrates that include oxalate, sulphate, and chloride. Disorders of oxalate homeostasis cause hyperoxalemia and hyperoxaluria, leading to urinary calcium oxalate precipitation and urolithogenesis. SLC26 proteins are aberrantly expressed during kidney stone formation, and consequently may present therapeutic targets. SLC26 protein inhibitors are in preclinical development. In this review, we integrate the findings of recent reports with clinical data to highlight the role of SLC26 proteins in oxalate metabolism during urolithogenesis, and discuss limitations of current studies and potential directions for future research.
Collapse
Affiliation(s)
- Jialin Li
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Sigen Huang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Shengyin Liu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xinzhi Liao
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Sheng Yan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Quanliang Liu
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
10
|
Nagasaki K, Minamitani K, Nakamura A, Kobayashi H, Numakura C, Itoh M, Mushimoto Y, Fujikura K, Fukushi M, Tajima T. Guidelines for Newborn Screening of Congenital Hypothyroidism (2021 Revision). Clin Pediatr Endocrinol 2022; 32:26-51. [PMID: 36761493 PMCID: PMC9887297 DOI: 10.1297/cpe.2022-0063] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/06/2022] [Indexed: 12/04/2022] Open
Abstract
Purpose of developing the guidelines: Newborn screening (NBS) for congenital hypothyroidism (CH) was started in 1979 in Japan, and early diagnosis and treatment improved the intelligence prognosis of CH patients. The incidence of CH was once about one in 5,000-8,000 births, but has been increased with diagnosis of subclinical CH. The disease requires continuous treatment and specialized medical facilities should conduct differential diagnosis and treatment in patients who are positive by NBS to avoid unnecessary treatment. The Guidelines for Mass Screening of Congenital Hypothyroidism (1998 version) were developed by the Mass Screening Committee of the Japanese Society for Pediatric Endocrinology in 1998. Subsequently, the guidelines were revised in 2014. Here, we have added minor revisions to the 2014 version to include the most recent findings. Target disease/conditions: Primary congenital hypothyroidism. Users of the Guidelines: Physician specialists in pediatric endocrinology, pediatric specialists, physicians referring pediatric practitioners, general physicians, laboratory technicians in charge of mass screening, and patients.
Collapse
Affiliation(s)
- Keisuke Nagasaki
- Mass Screening Committee, Japanese Society for Pediatric Endocrinology
- Thyroid Committee, Japanese Society for Pediatric Endocrinology
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kanshi Minamitani
- Thyroid Committee, Japanese Society for Pediatric Endocrinology
- Department of Pediatrics, Teikyo University Chiba Medical Center, Chiba, Japan
| | - Akie Nakamura
- Mass Screening Committee, Japanese Society for Pediatric Endocrinology
- Department of Pediatrics, Hokkaido University School of Medicine, Sapporo, Japan
| | - Hironori Kobayashi
- Mass Screening Committee, Japanese Society for Pediatric Endocrinology
- Laboratories Division, Shimane University Hospital, Izumo, Japan
| | - Chikahiko Numakura
- Mass Screening Committee, Japanese Society for Pediatric Endocrinology
- Department of Pediatrics, Yamagata University School of Medicine, Yamagata, Japan
| | - Masatsune Itoh
- Thyroid Committee, Japanese Society for Pediatric Endocrinology
- Department of Pediatrics, Kanazawa Medical University, Kanazawa, Japan
| | - Yuichi Mushimoto
- Thyroid Committee, Japanese Society for Pediatric Endocrinology
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kaori Fujikura
- Japanese Society for Neonatal Screening
- Sapporo City Institute of Public Health, Sapporo, Japan
| | - Masaru Fukushi
- Japanese Society for Neonatal Screening
- Sapporo Immuno Diagnostic Laboratory (IDL), Sapporo, Japan
| | - Toshihiro Tajima
- Mass Screening Committee, Japanese Society for Pediatric Endocrinology
- Department of Pediatrics, Jichi Medical University Tochigi Children's Medical Center, Tochigi, Japan
| |
Collapse
|
11
|
Kiriya M, Kawashima A, Fujiwara Y, Tanimura Y, Yoshihara A, Nakamura Y, Tanigawa K, Kondo T, Suzuki K. Thyroglobulin regulates the expression and localization of the novel iodide transporter solute carrier family 26 member 7 (SLC26A7) in thyrocytes. Endocr J 2022; 69:1217-1225. [PMID: 35644541 DOI: 10.1507/endocrj.ej22-0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Solute carrier family 26 member 7 (SLC26A7), identified as a causative gene for congenital hypothyroidism, was found to be a novel iodide transporter expressed on the apical side of the follicular epithelium of the thyroid. We recently showed that TSH suppressed the expression of SLC26A7 and induces its localization to the plasma membrane, where it functions. We also showed that the ability of TSH to induce thyroid hormone synthesis is completely reversed by an autocrine negative-feedback action of thyroglobulin (Tg) stored in the follicular lumen. In the present study, we investigated the potential effect of follicular Tg on SLC26A7 expression and found that follicular Tg significantly suppressed the promoter activity, mRNA level, and protein level of SLC26A7 in rat thyroid FRTL-5 cells. In addition, follicular Tg inhibited the ability of TSH to induce the membrane localization of SLC26A7. In rat thyroid sections, the expression of SLC26A7 was weaker in follicles with a higher concentration of Tg, as evidenced by immunofluorescence staining. These results indicate that Tg stored in the follicular lumen is a feedback suppressor of the expression and membrane localization of SLC26A7, thereby downregulating the transport of iodide into the follicular lumen.
Collapse
Affiliation(s)
- Mitsuo Kiriya
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo 173-8605, Japan
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Akira Kawashima
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo 173-8605, Japan
| | - Yoko Fujiwara
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo 173-8605, Japan
| | - Yuta Tanimura
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo 173-8605, Japan
| | - Aya Yoshihara
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo 173-8605, Japan
- Center for Medical Education, Faculty of Medicine, Toho University, Tokyo 143-8540, Japan
| | - Yasuhiro Nakamura
- Center for Promotion of Pharmaceutical Education & Research, Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
| | - Kazunari Tanigawa
- Department of Molecular Pharmaceutics, Faculty of Pharma-Science, Teikyo University, Tokyo 173-8605, Japan
| | - Tetsuo Kondo
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Koichi Suzuki
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo 173-8605, Japan
| |
Collapse
|
12
|
Jing L, Zhang Q. Intrathyroidal feedforward and feedback network regulating thyroid hormone synthesis and secretion. Front Endocrinol (Lausanne) 2022; 13:992883. [PMID: 36187113 PMCID: PMC9519864 DOI: 10.3389/fendo.2022.992883] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Thyroid hormones (THs), including T4 and T3, are produced and released by the thyroid gland under the stimulation of thyroid-stimulating hormone (TSH). The homeostasis of THs is regulated via the coordination of the hypothalamic-pituitary-thyroid axis, plasma binding proteins, and local metabolism in tissues. TH synthesis and secretion in the thyrocytes-containing thyroid follicles are exquisitely regulated by an elaborate molecular network comprising enzymes, transporters, signal transduction machineries, and transcription factors. In this article, we synthesized the relevant literature, organized and dissected the complex intrathyroidal regulatory network into structures amenable to functional interpretation and systems-level modeling. Multiple intertwined feedforward and feedback motifs were identified and described, centering around the transcriptional and posttranslational regulations involved in TH synthesis and secretion, including those underpinning the Wolff-Chaikoff and Plummer effects and thyroglobulin-mediated feedback regulation. A more thorough characterization of the intrathyroidal network from a systems biology perspective, including its topology, constituent network motifs, and nonlinear quantitative properties, can help us to better understand and predict the thyroidal dynamics in response to physiological signals, therapeutic interventions, and environmental disruptions.
Collapse
Affiliation(s)
- Li Jing
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| |
Collapse
|
13
|
Bernal Barquero CE, Geysels RC, Jacques V, Carro GH, Martín M, Peyret V, Abregú MC, Papendieck P, Masini-Repiso AM, Savagner F, Chiesa AE, Citterio CE, Nicola JP. Targeted Next-Generation Sequencing of Congenital Hypothyroidism-Causative Genes Reveals Unexpected Thyroglobulin Gene Variants in Patients with Iodide Transport Defect. Int J Mol Sci 2022; 23:ijms23169251. [PMID: 36012511 PMCID: PMC9409291 DOI: 10.3390/ijms23169251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 01/12/2023] Open
Abstract
Congenital iodide transport defect is an uncommon autosomal recessive disorder caused by loss-of-function variants in the sodium iodide symporter (NIS)-coding SLC5A5 gene and leading to dyshormonogenic congenital hypothyroidism. Here, we conducted a targeted next-generation sequencing assessment of congenital hypothyroidism-causative genes in a cohort of nine unrelated pediatric patients suspected of having a congenital iodide transport defect based on the absence of 99mTc-pertechnetate accumulation in a eutopic thyroid gland. Although, unexpectedly, we could not detect pathogenic SLC5A5 gene variants, we identified two novel compound heterozygous TG gene variants (p.Q29* and c.177-2A>C), three novel heterozygous TG gene variants (p.F1542Vfs*20, p.Y2563C, and p.S523P), and a novel heterozygous DUOX2 gene variant (p.E1496Dfs*51). Splicing minigene reporter-based in vitro assays revealed that the variant c.177-2A>C affected normal TG pre-mRNA splicing, leading to the frameshift variant p.T59Sfs*17. The frameshift TG variants p.T59Sfs*17 and p.F1542Vfs*20, but not the DUOX2 variant p.E1496Dfs*51, were predicted to undergo nonsense-mediated decay. Moreover, functional in vitro expression assays revealed that the variant p.Y2563C reduced the secretion of the TG protein. Our investigation revealed unexpected findings regarding the genetics of congenital iodide transport defects, supporting the existence of yet to be discovered mechanisms involved in thyroid hormonogenesis.
Collapse
Affiliation(s)
- Carlos Eduardo Bernal Barquero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Romina Celeste Geysels
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Virginie Jacques
- Laboratoire de Biochimie, Institut Fédératif de Biologie, Le Centre Hospitalier Universitaire de Toulouse, 31300 Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1297, 31432 Toulouse, France
| | - Gerardo Hernán Carro
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Victoria Peyret
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - María Celeste Abregú
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Patricia Papendieck
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1006, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires 1120, Argentina
| | - Ana María Masini-Repiso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Frédérique Savagner
- Laboratoire de Biochimie, Institut Fédératif de Biologie, Le Centre Hospitalier Universitaire de Toulouse, 31300 Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1297, 31432 Toulouse, France
| | - Ana Elena Chiesa
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1006, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires 1120, Argentina
| | - Cintia E. Citterio
- Instituto de Inmunología, Genética y Metabolismo, Consejo Nacional de Investigaciones Científicas y Técnicas (INIGEM-CONIET), Buenos Aires 1120, Argentina
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
- Correspondence: ; Tel.: +54-0351-535-3850 (ext. 55423)
| |
Collapse
|
14
|
Yamaguchi N, Suzuki A, Yoshida A, Tanaka T, Aoyama K, Oishi H, Hara Y, Ogi T, Amano I, Kameo S, Koibuchi N, Shibata Y, Ugawa S, Mizuno H, Saitoh S. The iodide transporter Slc26a7 impacts thyroid function more strongly than Slc26a4 in mice. Sci Rep 2022; 12:11259. [PMID: 35788623 PMCID: PMC9253019 DOI: 10.1038/s41598-022-15151-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 06/20/2022] [Indexed: 11/09/2022] Open
Abstract
SLC26A4 is a known iodide transporter, and is localized at the apical membrane of thyrocytes. Previously, we reported that SLC26A7 is also involved in iodide transport and that Slc26a7 is a novel causative gene for congenital hypothyroidism. However, its detailed role in vivo remains to be elucidated. We generated mice that were deficient in Slc26a7 and Slc26a4 to delineate differences and associations in their roles in iodide transport. Slc26a7-/- mice showed goitrous congenital hypothyroidism and mild growth failure on a normal diet. Slc26a7-/- mice with a low iodine environment showed marked growth failure. In contrast, Slc26a4-/- mice showed no growth failure and hypothyroidism in the same low iodine environment. Double-deficient mice showed more severe growth failure than Slc26a7-/- mice. RNA-seq analysis revealed that the number of differentially expressed genes (DEGs) in Slc26a7-/- mice was significantly higher than that in Slc26a4-/- mice. These indicate that SLC26A7 is more strongly involved in iodide transport and the maintenance of thyroid function than SLC26A4.
Collapse
Affiliation(s)
- Naoya Yamaguchi
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Atsushi Suzuki
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Aya Yoshida
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Tatsushi Tanaka
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Kohei Aoyama
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Hisashi Oishi
- Department of Comparative and Experimental Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuichiro Hara
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Satomi Kameo
- Department of Nutrition, Koshien University, Takarazuka, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuhiro Shibata
- Department of Anatomy and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shinya Ugawa
- Department of Anatomy and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Haruo Mizuno
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| |
Collapse
|
15
|
Li M, Li X, Wang F, Ren Y, Zhang X, Wang J, Shen L, Zhao D, ShiguoLiu. Genetic analysis of iodide transporter and recycling (NIS, PDS, SLC26A7, IYD) in patients with congenital hypothyroidism. Gene X 2022; 824:146402. [PMID: 35276235 DOI: 10.1016/j.gene.2022.146402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 12/18/2022] Open
Affiliation(s)
- Miaomiao Li
- Department of Medical Genetic, the Affiliated Hospital of Qingdao University, Qingdao, China; Prenatal Diagnosis Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaole Li
- Neonatal Screening Center, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Wang
- Endocrinology Department, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yubao Ren
- Neonatal Screening Center, Shengli Hospital of Shengli Oilfield, Dongying, China
| | - Xiao Zhang
- Department of Medical Genetic, the Affiliated Hospital of Qingdao University, Qingdao, China; Prenatal Diagnosis Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingli Wang
- Department of Medical Genetic, the Affiliated Hospital of Qingdao University, Qingdao, China; Prenatal Diagnosis Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Shen
- Department of Medical Genetic, the Affiliated Hospital of Qingdao University, Qingdao, China; Prenatal Diagnosis Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dehua Zhao
- Neonatal Screening Center, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - ShiguoLiu
- Department of Medical Genetic, the Affiliated Hospital of Qingdao University, Qingdao, China; Prenatal Diagnosis Center, the Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
16
|
Acar S, Gürsoy S, Arslan G, Nalbantoğlu Ö, Hazan F, Köprülü Ö, Özkaya B, Özkan B. Screening of 23 candidate genes by next-generation sequencing of patients with permanent congenital hypothyroidism: novel variants in TG, TSHR, DUOX2, FOXE1, and SLC26A7. J Endocrinol Invest 2022; 45:773-786. [PMID: 34780050 DOI: 10.1007/s40618-021-01706-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/07/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To date, many genes have been associated with congenital hypothyroidism (CH). Our aim was to identify the mutational spectrum of 23 causative genes in Turkish patients with permanent CH, including thyroid dysgenesis (TD) and dyshormonogenesis (TDH) cases. METHODS A total of 134 patients with permanent CH (130 primary, 4 central) were included. To identify the genetic etiology, we screened 23 candidate genes associated with CH by next-generation sequencing. For confirmation and to detect the status of the specific familial variant in relatives, Sanger sequencing was also performed. RESULTS Possible pathogenic variants were found in 5.2% of patients with TD and in 64.0% of the patients with normal-sized thyroid or goiter. In all patients, variants were most frequently found in TSHR, followed by TPO and TG. The same homozygous TSHB variant (c.162 + 5G > A) was identified in four patients with central CH. In addition, we detected novel variants in the TSHR, TG, SLC26A7, FOXE1, and DUOX2. CONCLUSION Genetic causes were determined in the majority of CH patients with TDH, however, despite advances in genetics, we were unable to identify the genetic etiology of most CH patients with TD, suggesting the effect of unknown genes or environmental factors. The previous studies and our findings suggest that TSHR and TPO mutations is the main genetic defect of CH in the Turkish population.
Collapse
Affiliation(s)
- S Acar
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, İsmet Kaptan Mh, Sezer Doğan Sokağı No:11, 35210, Konak/Izmir, Turkey.
| | - S Gürsoy
- Division of Pediatric Genetics, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - G Arslan
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, İsmet Kaptan Mh, Sezer Doğan Sokağı No:11, 35210, Konak/Izmir, Turkey
| | - Ö Nalbantoğlu
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, İsmet Kaptan Mh, Sezer Doğan Sokağı No:11, 35210, Konak/Izmir, Turkey
| | - F Hazan
- Department of Medical Genetics, Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Ö Köprülü
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, İsmet Kaptan Mh, Sezer Doğan Sokağı No:11, 35210, Konak/Izmir, Turkey
| | - B Özkaya
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, İsmet Kaptan Mh, Sezer Doğan Sokağı No:11, 35210, Konak/Izmir, Turkey
| | - B Özkan
- Division of Pediatric Endocrinology, Dr. Behçet Uz Children's Education and Research Hospital, İsmet Kaptan Mh, Sezer Doğan Sokağı No:11, 35210, Konak/Izmir, Turkey
| |
Collapse
|
17
|
Albader N, Zou M, BinEssa HA, Abdi S, Al-Enezi AF, Meyer BF, Alzahrani AS, Shi Y. Insights of Noncanonical Splice-site Variants on RNA Splicing in Patients With Congenital Hypothyroidism. J Clin Endocrinol Metab 2022; 107:e1263-e1276. [PMID: 34632506 DOI: 10.1210/clinem/dgab737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Congenital hypothyroidism (CH) is caused by mutations in the genes for thyroid hormone synthesis. In our previous investigation of CH patients, approximately 53% of patients had mutations in either coding exons or canonical splice sites of causative genes. Noncanonical splice-site variants in the intron were detected but their pathogenic significance was not known. OBJECTIVE This work aims to evaluate noncanonical splice-site variants on pre-messenger RNA (pre-mRNA) splicing of CH-causing genes. METHODS Next-generation sequencing data of 55 CH cases in 47 families were analyzed to identify rare intron variants. The effects of variants on pre-mRNA splicing were investigated by minigene RNA-splicing assay. RESULTS Four intron variants were found in 3 patients: solute carrier family 26 member 4 (SLC26A4) c.1544+9C>T and c.1707+94C>T in one patient, and solute carrier family 5 member 5 (SLC5A5) c.970-48G>C and c.1652-97A>C in 2 other patients. The c.1707+94C>T and c.970-48G>C caused exons 15 and 16 skipping, and exon 8 skipping, respectively. The remaining variants had no effect on RNA splicing. Furthermore, we analyzed 28 previously reported noncanonical splice-site variants (4 in TG and 24 in SLC26A4). Among them, 15 variants (~ 54%) resulted in aberrant splicing and 13 variants had no effect on RNA splicing. These data were compared with 3 variant-prediction programs (FATHMM-XF, FATHMM-MKL, and CADD). Among 32 variants, FATHMM-XF, FATHMM-MKL, and CADD correctly predicted 20 (63%), 17 (53%), and 26 (81%) variants, respectively. CONCLUSION Two novel deep intron mutations have been identified in SLC26A4 and SLC5A5, bringing the total number of solved families with disease-causing mutations to approximately 45% in our cohort. Approximately 46% (13/28) of reported noncanonical splice-site mutations do not disrupt pre-mRNA splicing. CADD provides highest prediction accuracy of noncanonical splice-site variants.
Collapse
Affiliation(s)
- Najla Albader
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Minjing Zou
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Huda A BinEssa
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Saba Abdi
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Anwar F Al-Enezi
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Brian F Meyer
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Ali S Alzahrani
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Yufei Shi
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
18
|
Relationships between SLC26A7 expressions and extra-thyroid metastasis of papillary thyroid carcinoma. Chin Med J (Engl) 2021; 135:225-227. [PMID: 34593695 PMCID: PMC8769115 DOI: 10.1097/cm9.0000000000001662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
19
|
Tanimura Y, Kiriya M, Kawashima A, Mori H, Luo Y, Kondo T, Suzuki K. Regulation of solute carrier family 26 member 7 (Slc26a7) by thyroid stimulating hormone in thyrocytes. Endocr J 2021; 68:691-699. [PMID: 33583874 DOI: 10.1507/endocrj.ej20-0502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Iodine transportation is an important step in thyroid hormone biosynthesis. Uptake of iodine into the thyroid follicle is mediated mainly by the basolateral sodium-iodide symporter (NIS or solute carrier family 5 member 5: SLC5A5), and iodine efflux across the apical membrane into the follicular lumen is mediated by pendrin (SLC26A4). In addition to these transporters, SLC26A7, which has recently been identified as a causative gene for congenital hypothyroidism, was found to encode a novel apical iodine transporter in the thyroid. Although SLC5A5 and SLC26A4 have been well-characterized, little is known about SLC26A7, including its regulation by TSH, the central hormone regulator of thyroid function. Using rat thyroid FRTL-5 cells, we showed that the mRNA levels of Slc26a7 and Slc26a4, two apical iodine transporters responsible for iodine efflux, were suppressed by TSH, whereas the mRNA level of Slc5a5 was induced. Forskolin and dibutyryl cAMP (dbcAMP) had the same effect as that of TSH on the mRNA levels of these transporters. TSH, forskolin and dbcAMP also had suppressive effects on SLC26A7 promoter activity, as assessed by luciferase reporter gene assays, and protein levels, as determined by Western blot analysis. TSH, forskolin and dbcAMP also induced strong localization of Slc26a7 to the cell membrane according to immunofluorescence staining and confocal laser scanning microscopy. Together, these results suggest that TSH suppresses the expression level of Slc26a7 but induces its accumulation at the cell membrane, where it functions as an iodine transporter.
Collapse
Affiliation(s)
- Yuta Tanimura
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | - Mitsuo Kiriya
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| | - Akira Kawashima
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| | - Hitomi Mori
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| | - Yuqian Luo
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital and Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China
| | - Tetsuo Kondo
- Department of Pathology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | - Koichi Suzuki
- Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Itabashi, Tokyo 173-8605, Japan
| |
Collapse
|
20
|
Zou M, BinEssa HA, Al-Malki YH, Al-Yahya S, Al-Alwan M, Al-Jammaz I, Khabar KSA, Almohanna F, Assiri AM, Meyer BF, Alzahrani AS, Al-Mohanna FA, Shi Y. β-Catenin Attenuation Inhibits Tumor Growth and Promotes Differentiation in a BRAF V600E-Driven Thyroid Cancer Animal Model. Mol Cancer Ther 2021; 20:1603-1613. [PMID: 34224366 DOI: 10.1158/1535-7163.mct-21-0037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/23/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022]
Abstract
BRAFV600E mutation is the most frequent genetic alteration in papillary thyroid cancer (PTC). β-Catenin (Ctnnb1) is a key downstream component of canonical Wnt signaling pathway and is frequently overexpressed in PTC. BRAF V600E-driven tumors have been speculated to rely on Wnt/β-catenin signaling to sustain its growth, although many details remain to be elucidated. In this study, we investigated the role of β-catenin in BrafV600E -driven thyroid cancer in a transgenic mouse model. In Braf V600E mice with wild-type (WT) Ctnnb1 (BVE-Ctnnb1WT or BVE), overexpression of β-catenin was observed in thyroid tumors. In Braf V600E mice with Ctnnb1 knockout (BVE-Ctnnb1null), thyroid tumor growth was slowed with significant reduction in papillary architecture. This was associated with increased expression of genes involved in thyroid hormone synthesis, elevated 124iodine uptake, and serum T4. The survival of BVE-Ctnnb1null mice was increased by more than 50% during 14-month observation. Mechanistically, downregulation of MAPK, PI3K/Akt, and TGFβ pathways and loss of epithelial-mesenchymal transition (EMT) were demonstrated in the BVE-Ctnnb1null tumors. Treatment with dual β-catenin/KDM4A inhibitor PKF118-310 dramatically improved the sensitivity of BVE-Ctnnb1WT tumor cells to BRAFV600E inhibitor PLX4720, resulting in significant growth arrest and apoptosis in vitro, and tumor regression and differentiation in vivo These findings indicate that β-catenin signaling plays an important role in thyroid cancer growth and resistance to BRAFV600E inhibitors. Simultaneously targeting both Wnt/β-catenin and MAPK signaling pathways may achieve better therapeutic outcome in BRAFV600E inhibitor-resistant and/or radioiodine-refractory thyroid cancer.
Collapse
Affiliation(s)
- Minjing Zou
- Department of Genetics and Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Huda A BinEssa
- Department of Genetics and Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Yousif H Al-Malki
- Department of Cyclotron and Radiopharmaceuticals, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Suhad Al-Yahya
- Department of BioMolecular Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Monther Al-Alwan
- Department of Stem Cell & Tissue Re-engineering, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ibrahim Al-Jammaz
- Department of Cyclotron and Radiopharmaceuticals, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khalid S A Khabar
- Department of BioMolecular Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Falah Almohanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdullah M Assiri
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Brian F Meyer
- Department of Genetics and Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali S Alzahrani
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Yufei Shi
- Department of Genetics and Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| |
Collapse
|
21
|
Kostopoulou E, Miliordos K, Spiliotis B. Genetics of primary congenital hypothyroidism-a review. Hormones (Athens) 2021; 20:225-236. [PMID: 33400193 DOI: 10.1007/s42000-020-00267-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Congenital primary hypothyroidism (CH) is a state of inadequate thyroid hormone production detected at birth, caused either by absent, underdeveloped or ectopic thyroid gland (dysgenesis), or by defected thyroid hormone biosynthesis (dyshormonogenesis). A genetic component has been identified in many cases of CH. This review summarizes the clinical and biochemical features of the genetic causes of primary CH. METHODS A literature review was conducted of gene defects causing congenital hypothyroidism. RESULTS Mutations in five genes have predominantly been implicated in thyroid dysgenesis (TSHR, FOXE1, NKX2-1, PAX8, and NKX2-5), the primary cause of CH (85%), and mutations in seven genes in thyroid dyshormonogenesis (SLC5A5, TPO, DUOX2, DUOXA2, SLC6A4, Tg, and DEHAL1). These genes encode for proteins that regulate genes expressed during the differentiation of the thyroid, such as TPO and Tg genes, or genes that regulate iodide organification, thyroglobulin synthesis, iodide transport, and iodotyrosine deiodination. Besides thyroid dysgenesis and dyshormonogenesis, additional causes of congenital hypothyroidism, such as iodothyronine transporter defects and resistance to thyroid hormones, have also been associated with genetic mutations. CONCLUSION The identification of the underlying genetic defects of CH is important for genetic counseling of families with an affected member, for identifying additional clinical characteristics or the risk for thyroid neoplasia and for diagnostic and management purposes.
Collapse
Affiliation(s)
- Eirini Kostopoulou
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics|, University of Patras School of Medicine, Patras, Greece.
| | - Konstantinos Miliordos
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics|, University of Patras School of Medicine, Patras, Greece
| | - Bessie Spiliotis
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics|, University of Patras School of Medicine, Patras, Greece
| |
Collapse
|
22
|
Citterio CE, Rivolta CM, Targovnik HM. Structure and genetic variants of thyroglobulin: Pathophysiological implications. Mol Cell Endocrinol 2021; 528:111227. [PMID: 33689781 DOI: 10.1016/j.mce.2021.111227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/22/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
Thyroglobulin (TG) plays a main role in the biosynthesis of thyroid hormones (TH), and, thus, it is involved in a wide range of vital functions throughout the life cycle of all vertebrates. Deficiency of TH production due to TG genetic variants causes congenital hypothyroidism (CH), with devastating consequences such as intellectual disability and impaired growth if untreated. To this day, 229 variations in the human TG gene have been identified while the 3D structure of TG has recently appeared. Although TG deficiency is thought to be of autosomal recessive inheritance, the introduction of massive sequencing platforms led to the identification of a variety of monoallelic TG variants (combined with mutations in other thyroid gene products) opening new questions regarding the possibility of oligogenic inheritance of the disease. In this review we discuss remarkable advances in the understanding of the TG architecture and the pathophysiology of CH associated with TG defects, providing new insights for the management of congenital disorders as well as counseling benefits for families with a history of TG abnormalities. Moreover, we summarize relevant aspects of TH synthesis within TG and offer an updated analysis of animal and cellular models of TG deficiency for pathophysiological studies of thyroid dyshormonogenesis while highlighting perspectives for new investigations. All in all, even though there has been sustained progress in understanding the role of TG in thyroid pathophysiology during the past 50 years, functional characterization of TG variants remains an important area of study for future advancement in the field.
Collapse
Affiliation(s)
- Cintia E Citterio
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Carina M Rivolta
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Héctor M Targovnik
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina.
| |
Collapse
|
23
|
Pio MG, Molina MF, Siffo S, Chiesa A, Rivolta CM, Targovnik HM. A novel mutation in intron 11 donor splice site, responsible of a rare genotype in thyroglobulin gene by altering the pre-mRNA splincing process. Cell expression and bioinformatic analysis. Mol Cell Endocrinol 2021; 522:111124. [PMID: 33321114 DOI: 10.1016/j.mce.2020.111124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 01/09/2023]
Abstract
Thyroglobulin (TG) is a homodimeric glycoprotein synthesized by the thyroid gland. To date, two hundred twenty-seven variations of the TG gene have been identified in humans. Thyroid dyshormonogenesis due to TG gene mutations have an estimated incidence of approximately 1 in 100,000 newborns. The clinical spectrum ranges from euthyroid to mild or severe hypothyroidism. The purpose of the present study was to identify and characterize new variants in the TG gene. We report an Argentine patient with congenital hypothyroidism, enlarged thyroid gland and low levels of serum TG. Sequencing of DNA, expression of chimeric minigenes as well as bioinformatics analysis were performed. DNA sequencing identified the presence of compound heterozygous mutations in the TG gene: the maternal mutation consists of a c.3001+5G > A, whereas the paternal mutation consists of p.Arg296*. Minigen analysis of the variant c.3001+5A performed in HeLa, CV1 and Hek293T cell lines, showed a total lack of transcript expression. So, in order to validate that the loss of expression was caused by such variation, site-directed mutagenesis was performed on the mutated clone, which previously had a pSPL3 vector change, to give rise to a wild-type clone c.3001+5G, endorsing that the mutation c.3001+5G > A is the cause of the total lack of expression. In conclusion, we demonstrate that the c.3001+5G > A mutation causes a rare genotype, altering the splicing of the pre-mRNA. This work contributes to elucidating the molecular bases of TG defects associated with congenital hypothyroidism and expands our knowledge in relation to the pathologic roles of the position 5 in the donor splice site.
Collapse
Affiliation(s)
- Mauricio Gomes Pio
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Maricel F Molina
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Sofia Siffo
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Ana Chiesa
- Centro de Investigaciones Endocrinológicas, CEDIE-CONICET, División Endocrinología, Hospital de Niños "Ricardo Gutiérrez", Buenos Aires, Argentina
| | - Carina M Rivolta
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Héctor M Targovnik
- Universidad de Buenos Aires. Facultad de Farmacia y Bioquímica. Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires. Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina.
| |
Collapse
|
24
|
Venugopalan V, Al-Hashimi A, Rehders M, Golchert J, Reinecke V, Homuth G, Völker U, Manirajah M, Touzani A, Weber J, Bogyo MS, Verrey F, Wirth EK, Schweizer U, Heuer H, Kirstein J, Brix K. The Thyroid Hormone Transporter Mct8 Restricts Cathepsin-Mediated Thyroglobulin Processing in Male Mice through Thyroid Auto-Regulatory Mechanisms That Encompass Autophagy. Int J Mol Sci 2021; 22:ijms22010462. [PMID: 33466458 PMCID: PMC7796480 DOI: 10.3390/ijms22010462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
The thyroid gland is both a thyroid hormone (TH) generating as well as a TH responsive organ. It is hence crucial that cathepsin-mediated proteolytic cleavage of the precursor thyroglobulin is regulated and integrated with the subsequent export of TH into the blood circulation, which is enabled by TH transporters such as monocarboxylate transporters Mct8 and Mct10. Previously, we showed that cathepsin K-deficient mice exhibit the phenomenon of functional compensation through cathepsin L upregulation, which is independent of the canonical hypothalamus-pituitary-thyroid axis, thus, due to auto-regulation. Since these animals also feature enhanced Mct8 expression, we aimed to understand if TH transporters are part of the thyroid auto-regulatory mechanisms. Therefore, we analyzed phenotypic differences in thyroid function arising from combined cathepsin K and TH transporter deficiencies, i.e., in Ctsk-/-/Mct10-/-, Ctsk-/-/Mct8-/y, and Ctsk-/-/Mct8-/y/Mct10-/-. Despite the impaired TH export, thyroglobulin degradation was enhanced in the mice lacking Mct8, particularly in the triple-deficient genotype, due to increased cathepsin amounts and enhanced cysteine peptidase activities, leading to ongoing thyroglobulin proteolysis for TH liberation, eventually causing self-thyrotoxic thyroid states. The increased cathepsin amounts were a consequence of autophagy-mediated lysosomal biogenesis that is possibly triggered due to the stress accompanying intrathyroidal TH accumulation, in particular in the Ctsk-/-/Mct8-/y/Mct10-/- animals. Collectively, our data points to the notion that the absence of cathepsin K and Mct8 leads to excessive thyroglobulin degradation and TH liberation in a non-classical pathway of thyroid auto-regulation.
Collapse
Affiliation(s)
- Vaishnavi Venugopalan
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Alaa Al-Hashimi
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Maren Rehders
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Janine Golchert
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Vivien Reinecke
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Mythili Manirajah
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Adam Touzani
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Jonas Weber
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Matthew S. Bogyo
- Department of Pathology, School of Medicine, Stanford University, 300 Pasteur Dr., Stanford, CA 94305-5324, USA;
| | - Francois Verrey
- Physiologisches Institut, Universität Zürich, Winterthurerstr. 190, CH-8057 Zürich, Switzerland;
| | - Eva K. Wirth
- Berlin Institute of Health, Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Hessische Str. 3-4, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, D-10115 Berlin, Germany;
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Medizinische Fakultät, Universität Bonn, Nußallee 11, D-53115 Bonn, Germany;
| | - Heike Heuer
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen (AöR), Universität Duisburg-Essen, Hufelandstr. 55, D-45147 Essen, Germany;
| | - Janine Kirstein
- Fachbereich 2 Biologie/Chemie, Faculty of Cell Biology, Universität Bremen, Leobener Straße 5, D-28359 Bremen, Germany;
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
- Correspondence: ; Tel.: +49-421-200-3246
| |
Collapse
|
25
|
Hermanns P, Claßen C, Pohlenz J. A Novel Homozygous Mutation in the Solute Carrier Family 26 Member 7 Gene Causes Thyroid Dyshormonogenesis in a Girl with Congenital Hypothyroidism. Thyroid 2020; 30:1831-1833. [PMID: 32486989 DOI: 10.1089/thy.2020.0293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We investigated the genetic cause of thyroid dyshormonogenesis in a girl with congenital hypothyroidism. Genetic analysis showed that she was homozygous for a hitherto not described mutation (c.1432_1433delGT, p.V478KfsX11) in the solute carrier family 26 member 7 (SLC26A7) gene. SLC26A7 is proposed to be an anion transporter in the thyroid gland. The mutation leads to a frameshift and a premature stop codon. The predicted protein is truncated and very likely to be nonfunctional if it was expressed at all. In addition, in silico studies predict the mutation to be pathogenic.
Collapse
Affiliation(s)
- Pia Hermanns
- Department of Molecular Pediatric Endocrinology, Johannes Gutenberg University of Mainz, Children's Hospital, Mainz, Germany
| | - Charlotte Claßen
- Department of Molecular Pediatric Endocrinology, Johannes Gutenberg University of Mainz, Children's Hospital, Mainz, Germany
| | - Joachim Pohlenz
- Department of Molecular Pediatric Endocrinology, Johannes Gutenberg University of Mainz, Children's Hospital, Mainz, Germany
| |
Collapse
|
26
|
Scoville DW, Kang HS, Jetten AM. Transcription factor GLIS3: Critical roles in thyroid hormone biosynthesis, hypothyroidism, pancreatic beta cells and diabetes. Pharmacol Ther 2020; 215:107632. [PMID: 32693112 PMCID: PMC7606550 DOI: 10.1016/j.pharmthera.2020.107632] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022]
Abstract
GLI-Similar 3 (GLIS3) is a member of the GLIS subfamily of Krüppel-like zinc finger transcription factors that functions as an activator or repressor of gene expression. Study of GLIS3-deficiency in mice and humans revealed that GLIS3 plays a critical role in the regulation of several biological processes and is implicated in the development of various diseases, including hypothyroidism and diabetes. This was supported by genome-wide association studies that identified significant associations of common variants in GLIS3 with increased risk of these pathologies. To obtain insights into the causal mechanisms underlying these diseases, it is imperative to understand the mechanisms by which this protein regulates the development of these pathologies. Recent studies of genes regulated by GLIS3 led to the identification of a number of target genes and have provided important molecular insights by which GLIS3 controls cellular processes. These studies revealed that GLIS3 is essential for thyroid hormone biosynthesis and identified a critical function for GLIS3 in the generation of pancreatic β cells and insulin gene transcription. These observations raised the possibility that the GLIS3 signaling pathway might provide a potential therapeutic target in the management of diabetes, hypothyroidism, and other diseases. To develop such strategies, it will be critical to understand the upstream signaling pathways that regulate the activity, expression and function of GLIS3. Here, we review the recent progress on the molecular mechanisms by which GLIS3 controls key functions in thyroid follicular and pancreatic β cells and how this causally relates to the development of hypothyroidism and diabetes.
Collapse
Affiliation(s)
- David W Scoville
- Cell Biology Group, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Hong Soon Kang
- Cell Biology Group, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Anton M Jetten
- Cell Biology Group, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
27
|
Cao X, Soleimani M, Hughes BA. SLC26A7 constitutes the thiocyanate-selective anion conductance of the basolateral membrane of the retinal pigment epithelium. Am J Physiol Cell Physiol 2020; 319:C641-C656. [PMID: 32726161 DOI: 10.1152/ajpcell.00027.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anion channels in the retinal pigment epithelium (RPE) play an essential role in the transport of Cl- between the outer retina and the choroidal blood to regulate the ionic composition and volume of the subretinal fluid that surrounds the photoreceptor outer segments. Recently, we reported that the anion conductance of the mouse RPE basolateral membrane is highly selective for the biologically active anion thiocyanate (SCN-), a property that does not correspond with any of the Cl- channels that have been found to be expressed in the RPE to date. The purpose of this study was to determine the extent to which SLC26A7, a SCN- permeable-anion exchanger/channel that was reported to be expressed in human RPE, contributes to the RPE basolateral anion conductance. We show by quantitative RT-PCR that Slc26a7 is highly expressed in mouse RPE compared with other members of the Slc26 gene family and Cl- channel genes known to be expressed in the RPE. By applying immunofluorescence microscopy to mouse retinal sections and isolated cells, we localized SLC26A7 to the RPE basolateral membrane. Finally, we performed whole cell and excised patch recordings from RPE cells acutely isolated from Slc26a7 knockout mice to show that the SCN- conductance and permeability of its basolateral membrane are dramatically smaller relative to wild-type mouse RPE cells. These findings establish SLC26A7 as the SCN--selective conductance of the RPE basolateral membrane and provide new insight into the physiology of an anion channel that may participate in anion transport and pH regulation by the RPE.
Collapse
Affiliation(s)
- Xu Cao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan
| | | | - Bret A Hughes
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
28
|
Perinatal Exposure to Triclosan Results in Abnormal Brain Development and Behavior in Mice. Int J Mol Sci 2020; 21:ijms21114009. [PMID: 32503345 PMCID: PMC7312693 DOI: 10.3390/ijms21114009] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022] Open
Abstract
Triclosan (TCS) is one of the most common endocrine-disrupting chemicals (EDCs) present in household and personal wash products. Recently, concerns have been raised about the association between abnormal behavior in children and exposure to EDC during gestation. We hypothesized that exposure to TCS during gestation could affect brain development. Cortical neurons of mice were exposed in vitro to TCS. In addition, we examined in vivo whether maternal TCS administration can affect neurobehavioral development in the offspring generation. We determined that TCS can impair dendrite and axon growth by reducing average length and numbers of axons and dendrites. Additionally, TCS inhibited the proliferation of and promoted apoptosis in neuronal progenitor cells. Detailed behavioral analyses showed impaired acquisition of spatial learning and reference memory in offspring derived from dams exposed to TCS. The TCS-treated groups also showed cognition dysfunction and impairments in sociability and social novelty preference. Furthermore, TCS-treated groups exhibited increased anxiety-like behavior, but there was no significant change in depression-like behaviors. In addition, TCS-treated groups exhibited deficits in nesting behavior. Taken together, our results indicate that perinatal exposure to TCS induces neurodevelopment disorder, resulting in abnormal social behaviors, cognitive impairment, and deficits in spatial learning and memory in offspring.
Collapse
|
29
|
Bruellman RJ, Watanabe Y, Ebrhim RS, Creech MK, Abdullah MA, Dumitrescu AM, Refetoff S, Weiss RE. Increased Prevalence of TG and TPO Mutations in Sudanese Children With Congenital Hypothyroidism. J Clin Endocrinol Metab 2020; 105:5684913. [PMID: 31867598 PMCID: PMC7093074 DOI: 10.1210/clinem/dgz297] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022]
Abstract
CONTEXT Congenital hypothyroidism (CH) is due to dyshormonogenesis in 10% to 15% of subjects worldwide but accounts for 60% of CH cases in the Sudan. OBJECTIVE To investigate the molecular basis of CH in Sudanese families. DESIGN Clinical phenotype reporting and serum thyroid hormone measurements. Deoxyribonucelic acid extraction for whole-exome sequencing and Sanger sequencing. SETTING University research center. PATIENTS Twenty-six Sudanese families with CH. INTERVENTION Clinical evaluation, thyroid function tests, genetic sequencing, and analysis. Our samples and information regarding samples from the literature were used to compare TG (thyroglobulin) and TPO (thyroid peroxidase) mutation rates in the Sudanese population with all populations. RESULTS Mutations were found in dual-oxidase 1 (DUOX1), dual-oxidase 2 (DUOX2), iodotyrosine deiodinase (IYD), solute-carrier (SLC) 26A4, SLC26A7, SLC5A5, TG, and TPO genes. The molecular basis of the CH in 7 families remains unknown. TG mutations were significantly higher on average in the Sudanese population compared with the average number of TG mutations in other populations (P < 0.05). CONCLUSIONS All described mutations occur in domains important for protein structure and function, predicting the CH phenotype. Genotype prediction based on phenotype includes low or undetectable thyroglobulin levels for TG gene mutations and markedly higher thyroglobulin levels for TPO mutations. The reasons for higher incidence of TG gene mutations include gene length and possible positive genetic selection due to endemic iodine deficiency.
Collapse
Affiliation(s)
- Ryan J Bruellman
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Yui Watanabe
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Reham S Ebrhim
- Department of Paediatrics and Child Health, Faculty of Medicine, University of Almughtaribeen, Khartoum, Sudan
| | - Matthew K Creech
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Mohamed A Abdullah
- Department of Paediatrics and Child Health, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Alexandra M Dumitrescu
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Committee on Molecular Medicine and Nutrition, The University of Chicago, Chicago, Illinois
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
- Committee on Genetics, The University of Chicago, Chicago, Illinois
| | - Roy E Weiss
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Correspondence and Reprint Requests: Roy E. Weiss, MD, PhD, University of Miami Miller School of Medicine, 1120 NW 14th Street, Room 310F, Miami, Florida 33136. E-mail:
| |
Collapse
|
30
|
Targovnik HM, Scheps KG, Rivolta CM. Defects in protein folding in congenital hypothyroidism. Mol Cell Endocrinol 2020; 501:110638. [PMID: 31751626 DOI: 10.1016/j.mce.2019.110638] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/21/2019] [Accepted: 11/01/2019] [Indexed: 12/26/2022]
Abstract
Primary congenital hypothyroidism (CH) is the most common endocrine disease in children and one of the most common preventable causes of both cognitive and motor deficits. CH is a heterogeneous group of thyroid disorders in which inadequate production of thyroid hormone occurs due to defects in proteins involved in the gland organogenesis (dysembryogenesis) or in multiple steps of thyroid hormone biosynthesis (dyshormonogenesis). Dysembryogenesis is associated with genes responsible for the development or growth of thyroid cells: such as NKX2-1, FOXE1, PAX8, NKX2-5, TSHR, TBX1, CDCA8, HOXD3 and HOXB3 resulting in agenesis, hypoplasia or ectopia of thyroid gland. Nevertheless, the etiology of the dysembryogenesis remains unknown for most cases. In contrast, the majority of patients with dyshormonogenesis has been linked to mutations in the SLC5A5, SLC26A4, SLC26A7, TPO, DUOX1, DUOX2, DUOXA1, DUOXA2, IYD or TG genes, which usually originate goiter. About 800 genetic mutations have been reported to cause CH in patients so far, including missense, nonsense, in-frame deletion and splice-site variations. Many of these mutations are implicated in specific domains, cysteine residues or glycosylation sites, affecting the maturation of nascent proteins that go through the secretory pathway. Consequently, misfolded proteins are permanently entrapped in the endoplasmic reticulum (ER) and are translocated to the cytosol for proteasomal degradation by the ER-associated degradation (ERAD) machinery. Despite of all these remarkable advances in the field of the CH pathogenesis, several points on the development of this disease remain to be elucidated. The continuous study of thyroid gene mutations with the application of new technologies will be useful for the understanding of the intrinsic mechanisms related to CH. In this review we summarize the present status of knowledge on the disorders in the protein folding caused by thyroid genes mutations.
Collapse
Affiliation(s)
- Héctor M Targovnik
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina.
| | - Karen G Scheps
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Carina M Rivolta
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| |
Collapse
|
31
|
Touré A. Importance of SLC26 Transmembrane Anion Exchangers in Sperm Post-testicular Maturation and Fertilization Potential. Front Cell Dev Biol 2019; 7:230. [PMID: 31681763 PMCID: PMC6813192 DOI: 10.3389/fcell.2019.00230] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/26/2019] [Indexed: 12/17/2022] Open
Abstract
In mammals, sperm cells produced within the testis are structurally differentiated but remain immotile and are unable to fertilize the oocyte unless they undergo a series of maturation events during their transit in the male and female genital tracts. This post-testicular functional maturation is known to rely on the micro-environment of both male and female genital tracts, and is tightly controlled by the pH of their luminal milieus. In particular, within the epididymis, the establishment of a low bicarbonate (HCO3–) concentration contributes to luminal acidification, which is necessary for sperm maturation and subsequent storage in a quiescent state. Following ejaculation, sperm is exposed to the basic pH of the female genital tract and bicarbonate (HCO3–), calcium (Ca2+), and chloride (Cl–) influxes induce biochemical and electrophysiological changes to the sperm cells (cytoplasmic alkalinization, increased cAMP concentration, and protein phosphorylation cascades), which are indispensable for the acquisition of fertilization potential, a process called capacitation. Solute carrier 26 (SLC26) members are conserved membranous proteins that mediate the transport of various anions across the plasma membrane of epithelial cells and constitute important regulators of pH and HCO3– concentration. Most SLC26 members were shown to physically interact and cooperate with the cystic fibrosis transmembrane conductance regulator channel (CFTR) in various epithelia, mainly by stimulating its Cl– channel activity. Among SLC26 members, the function of SLC26A3, A6, and A8 were particularly investigated in the male genital tract and the sperm cells. In this review, we will focus on SLC26s contributions to ionic- and pH-dependent processes during sperm post-testicular maturation. We will specify the current knowledge regarding their functions, based on data from the literature generated by means of in vitro and in vivo studies in knock-out mouse models together with genetic studies of infertile patients. We will also discuss the limits of those studies, the current research gaps and identify some key points for potential developments in this field.
Collapse
Affiliation(s)
- Aminata Touré
- INSERM U1016, Centre National de la Recherche Scientifique, UMR 8104, Institut Cochin, Université de Paris, Paris, France
| |
Collapse
|