1
|
Kato T, Nakatsuka R, Zhang R, Uemura Y, Yamashita H, Matsuoka Y, Shirouzu Y, Fujioka T, Hattori F, Ogata H, Sakashita A, Honda H, Hitomi H. The role of glial cells missing 2 in induced pluripotent stem cell parathyroid differentiation. Tissue Cell 2025; 92:102634. [PMID: 39615229 DOI: 10.1016/j.tice.2024.102634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024]
Abstract
Glial cells missing 2 (GCM2) has been identified as an essential factor for parathyroid differentiation, and GCM2 silencing in parathyroid cells decreases calcium-sensing receptor (CaSR) expression. However, the role of GCM2 in parathyroid differentiation from induced pluripotent stem cells (iPSCs) is unclear. Here, we investigated the role of GCM2 in parathyroid differentiation from iPSCs using the Tet-On 3 G system. We confirmed that iPS cells transfected with GCM2/TRE3G and pCMV-Tet3G vectors express GCM2 in a doxycycline-dependent manner. Though parathyroid glands derive from the endoderm and differentiate via the third pharyngeal arch (PPE), overexpression of GCM2 in iPSCs significantly abolished the suppression of OCT4 and SOX2, suggesting inhibition of endodermal differentiation. GCM2 overexpression at the stage of differentiation into the third PPE also increased the expression levels of CaSR and parathyroid hormone, and increased the number of CaSR+/EpCAM+ cells. These results suggest that GCM2 regulates parathyroid differentiation after endoderm differentiation rather than at an earlier stage.
Collapse
Affiliation(s)
- Tadashi Kato
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Ryusuke Nakatsuka
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan; Department of Pharmacology, Faculty of Dentistry, Osaka Dental University, Osaka, Japan
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Hiromi Yamashita
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan
| | - Yoshikazu Matsuoka
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan
| | - Yasumasa Shirouzu
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan
| | - Tatsuya Fujioka
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan
| | - Fumiyuki Hattori
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan
| | - Hiroaki Ogata
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Kanagawa, Japan
| | - Akiko Sakashita
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Kanagawa, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hirofumi Hitomi
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan
| |
Collapse
|
2
|
Romanet P, Coppin L, Molin A, Santucci N, Le Bras M, Odou MF. Chapter 5: The roles of genetics in primary hyperparathyroidism. ANNALES D'ENDOCRINOLOGIE 2025; 86:101694. [PMID: 39818301 DOI: 10.1016/j.ando.2025.101694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Around 10% of cases of primary hyperparathyroidism are thought to be genetic in origin, some of which are part of a syndromic form such as multiple endocrine neoplasia types 1, 2A or 4 or hyperparathyroidism-jaw tumor syndrome, while the remainder are cases of isolated familial primary hyperparathyroidism. Recognition of these genetic forms is important to ensure appropriate management according to the gene and type of variant involved, but screening for a genetic cause is not justified in all patients presenting primary hyperparathyroidism. The indications for genetic analysis have made it possible to propose a decision tree that takes into account whether the presentation is familial or sporadic, syndromic or isolated, patient age, and histopathological type of parathyroid lesion. Thus, the first consensus recommendation is to propose genetic screening to any patient with a familial form of primary hyperparathyroidism (≥2 1st or 2nd degree relatives) or in syndromic presentation or a sporadic isolated presentation if the patient is under 50 years of age, or over 50 with a recurrent or multi-glandular form, carcinoma, atypical parathyroid tumor and/or loss of parafibromin expression. The panel of genes currently recommended for first-line treatment comprises MEN1, CDKN1B, CDC73, CASR, GNA11, AP2S1 and GCM2. Other genes may also be involved in familial primary hyperparathyroidism, but in a much more rarely and less consistently. The second recommendation is to propose genetic screening, up to and including whole-genome sequencing in the event of inconclusive panel analysis, to patients with proven familial primary hyperparathyroidism and/or pediatric onset. The role of the genetic practitioner is to interpret the sequencing data by categorizing the variants into 5 classes of pathogenicity. The aim of genetic analysis is to identify the genetic variant involved in the patient's phenotype, in order to make or refute a diagnosis of hereditary primary hyperparathyroidism, and to adapt management and monitoring. Appropriate genetic counseling should then be provided for patient and family.
Collapse
Affiliation(s)
- Pauline Romanet
- Inserm, MMG, Laboratory of Molecular Biology GEnOPé, BIOGENOPOLE, La Timone University Hospital, Aix-Marseille University, AP-HM, Marseille, France.
| | - Lucie Coppin
- Inserm, CNRS, UMR9020-U1277 - CANTHER - Cancer - Heterogeneity Plasticity and Resistance to Therapies, University of Lille, CHU of Lille, Lille, France
| | - Arnaud Molin
- UNICAEN, RU7450 BioTARGen, Department of Genetics, Reference Center for Developmental Disorders and Malformative Syndromes, Anddi-Rares Network, Caen University Hospital, University of Normandy, Caen, France
| | - Nicolas Santucci
- Department of Digestive, Oncological and Endocrine Surgery, Dijon University Hospital Centre, Dijon, France
| | - Maëlle Le Bras
- Department of Endocrinology, Nantes University Hospital, Nantes, France.
| | - Marie-Françoise Odou
- Inserm, U1286 - Infinite, University of Lille, CHU of Lille, 59045 Lille cedex, France; Department of Biochemistry and Molecular Biology, Lille University Hospital, Lille, France.
| |
Collapse
|
3
|
Monticelli S, Giangrande A. Evolutionary Conservation of the Gcm/Glide Cascade: Of Glia and Beyond. BRAIN, BEHAVIOR AND EVOLUTION 2024; 100:58-66. [PMID: 39586239 DOI: 10.1159/000542753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Glia represent a major cell population of the nervous system, and they take part in virtually any process sustaining the development, the functioning, and the pathology of the nervous system. Glial cells diversified significantly during evolution and distinct signals have been adopted to initiate glial development in mammals as compared to flies. In the invertebrate model Drosophila melanogaster, the transcription factor Gcm is necessary and sufficient to generate glial cells. Although Gcm orthologs have been found in protostomes and deuterostomes, they do not act in glial fate commitment as in flies, calling for further investigations of the evolutionarily conserved role of Gcm. SUMMARY Here, we review the impact of the fly Gcm transcription factor in the differentiation of phagocytic competent cells inside and outside the nervous system, glia, and macrophages, respectively. Then, we discuss the evolutionary conservation of Gcm and the neural/nonneural functions of Gcm orthologs. Finally, we present a recent work from Pavlidaki et al. [Cell Rep. 2022;41(3):111506] showing that the Gcm cascade is conserved from fly macrophages to mammalian microglia to counteract acute and chronic inflammation. KEY MESSAGES Gcm has an ancestral role in immunity, and its anti-inflammatory effect is evolutionarily conserved. This opens new avenues to assess Gcm function in other species/animal models, its potential involvement in inflammation-related processes, such as regeneration, and to expand the investigation on glia evolution.
Collapse
Affiliation(s)
- Sara Monticelli
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, Illkirch, France
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, Illkirch, France
| | - Angela Giangrande
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, Illkirch, France
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, Illkirch, France
| |
Collapse
|
4
|
Thambundit A, Martinez-Agosto JA, Kianmahd Shamshoni J, Winer KK, Mittelman SD. Three Siblings With Familial Isolated Hypoparathyroidism: A Diagnostic Journey From CASR to Novel GCM2 Variant. JCEM CASE REPORTS 2024; 2:luae185. [PMID: 39439810 PMCID: PMC11495326 DOI: 10.1210/jcemcr/luae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Indexed: 10/25/2024]
Abstract
We report a patient who initially presented at 4 days old with hypocalcemia, hypoparathyroidism, and elevated phosphorous level. Treatment was initiated with calcitriol, calcium carbonate (CaCO3), vitamin D, and low phosphorous formula. Family history was positive for an activating calcium sensing receptor (CASR) variant (R990G) identified previously in 2 older siblings who were treated with CaCO3 and calcitriol. However, genetic studies were negative for the CASR variant in our patient. She maintained a large calcium requirement and was admitted for multiple episodes of hypocalcemia. Further investigation revealed that the CASR variant identified in the older siblings was now considered a benign, nondisease-causing variant. Whole exome sequencing on our proband revealed a homozygous pathogenic variant in the GCM2 gene (Gln392*) consistent with a molecular diagnosis of familial isolated hypoparathyroidism. Genetic studies revealed the 2 older siblings harbor the same genetic changes and parents are heterozygous carriers for this allele. Due to persistent hypocalcemia, we initiated teriparatide. She weaned off calcitriol and achieved normocalcemia on teriparatide, CaCO3, and vitamin D. Siblings transitioned to the same treatment without complications. These findings demonstrate the importance of adequate diagnostic genetic testing and the role of variant reanalysis over time in promoting accurate diagnoses.
Collapse
Affiliation(s)
- Apisadaporn Thambundit
- Division of Pediatric Endocrinology, UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | | | - Karen K Winer
- Eunice Kennedy Shriver National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven D Mittelman
- Division of Pediatric Endocrinology, UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Şenkal-Turhan S, Bulut-Okumuş E, Aydın M, Başak Türkmen N, Taşlıdere A, Şahin F, Yılmaz Ş, Akkuş Süt P, Doğan A. Induced Pluripotent Stem Cell-Derived Parathyroid Organoids Resemble Parathyroid Morphology and Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407567. [PMID: 39331961 DOI: 10.1002/advs.202407567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/12/2024] [Indexed: 09/29/2024]
Abstract
The primary role of the parathyroid glands is to maintain calcium homeostasis through the secretion of parathyroid hormone (PTH). The limited proliferative capacity and differentiation of parathyroid cells hinder the generation of cell therapy options. In this study, parathyroid organoids are successfully generated from human-induced pluripotent stem cells (hiPSCs). At the end of the 20 days of differentiation, the parathyroid organoids exhibited distinct parathyroid morphology. Stereomicroscope, scanning electron microscopy (SEM), and transmission electron microscopy (TEM) analysis demonstrated the 3D arrangement of the cell layers in which intracellular structures of parathyroid cells resemble human parathyroid cellular morphology. Comprehensive molecular analyses, including RNA sequencing (RNA-Seq) and liquid chromatography/mass spectrometry (LC-MS/MS), confirmed the expression of key parathyroid-related markers. Protein expression of CasR, CxCr4, Gcm2, and PTH are observed in parathyroid organoids. Parathyroid organoids secrete PTH, demonstrate active intercellular calcium signaling, and induce osteogenic differentiation via their secretome. The tissue integration potential of parathyroid organoids is determined by transplantation into parathyroidectomized rats. The organoid transplanted animals showed significant elevations in PTH-related markers (CasR, CxCr4, Foxn1, Gcm2, and PTH). PTH secretion is detected in organoid-transplanted animals. The findings represent a significant advancement in parathyroid organoid culture and may offer a cellular therapy for treating PTH-related diseases, including hypoparathyroidism.
Collapse
Affiliation(s)
- Selinay Şenkal-Turhan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Ezgi Bulut-Okumuş
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Muhterem Aydın
- Department of Veterinary Obstetrics and Gynecology, Faculty of Veterinary Medicine, University of Fırat, Elazığ, 23119, Turkey
| | - Neşe Başak Türkmen
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, University of Inonu, Malatya, 44280, Turkey
| | - Aslı Taşlıdere
- Department of Histology and Embryology, Faculty of Medicine, University of Inonu, Malatya, 44280, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Şahin Yılmaz
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Pınar Akkuş Süt
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, 34755, Turkey
| |
Collapse
|
6
|
Michelson DA, Mathis D. Thymic Mimetic Cells: Ontogeny as Immunology. Annu Rev Cell Dev Biol 2024; 40:283-300. [PMID: 38608315 PMCID: PMC11446667 DOI: 10.1146/annurev-cellbio-112122-023316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Medullary thymic epithelial cells (mTECs) generate immunological self-tolerance by ectopically expressing peripheral-tissue antigens (PTAs) within the thymus to preview the peripheral self to maturing T cells. Recent work, drawing inspiration from old histological observations, has shown that subtypes of mTECs, collectively termed mimetic cells, co-opt developmental programs from throughout the organism to express biologically coherent groups of PTAs. Here, we review key aspects of mimetic cells, especially as they relate to the larger contexts of molecular, cellular, developmental, and evolutionary biology. We highlight lineage-defining transcription factors as key regulators of mimetic cells and speculate as to what other factors, including Aire and the chromatin potential of mTECs, permit mimetic cell differentiation and function. Last, we consider what mimetic cells can teach us about not only the thymus but also other tissues.
Collapse
Affiliation(s)
- Daniel A Michelson
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA;
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
7
|
Kano M. Parathyroid Gland Generation from Pluripotent Stem Cells. Endocrinol Metab (Seoul) 2024; 39:552-558. [PMID: 38853617 PMCID: PMC11375298 DOI: 10.3803/enm.2024.1989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
Patients with permanent hypoparathyroidism require lifelong treatment. Current replacement therapies sometimes have adverse effects (e.g., hypercalciuria and chronic kidney disease). Generating parathyroid glands (PTGs) from the patient's own induced pluripotent stem cells (PSCs), with transplantation of these PTGs, would be an effective treatment option. Multiple methods for generating PTGs from PSCs have been reported. One major trend is in vitro differentiation of PSCs into PTGs. Another is in vivo generation of PSC-derived PTGs by injecting PSCs into PTG-deficient embryos. This review discusses current achievements and challenges in present and future PTG regenerative medicine.
Collapse
Affiliation(s)
- Mayuko Kano
- Department of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
8
|
Wang G, Du Y, Cui X, Xu T, Li H, Dong M, Li W, Li Y, Cai W, Xu J, Li S, Yang X, Wu Y, Chen H, Li X. Directed differentiation of human embryonic stem cells into parathyroid cells and establishment of parathyroid organoids. Cell Prolif 2024; 57:e13634. [PMID: 38494923 PMCID: PMC11294423 DOI: 10.1111/cpr.13634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/25/2024] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Differentiation of human embryonic stem cells (hESCs) into human embryonic stem cells-derived parathyroid-like cells (hESC-PT) has clinical significance in providing new therapies for congenital and acquired parathyroid insufficiency conditions. However, a highly reproducible, well-documented method for parathyroid differentiation remains unavailable. By imitating the natural process of parathyroid embryonic development, we proposed a new hypothesis about the in vitro differentiation of parathyroid-like cells. Transcriptome, differentiation marker protein detection and parathyroid hormone (PTH) secretion assays were performed after the completion of differentiation. To optimize the differentiation protocol and further improve the differentiation rate, we designed glial cells missing transcription factor 2 (GCM2) overexpression lentivirus transfection assays and constructed hESCs-derived parathyroid organoids. The new protocol enabled hESCs to differentiate into hESC-PT. HESC-PT cells expressed PTH, GCM2 and CaSR proteins, low extracellular calcium culture could stimulate hESC-PT cells to secrete PTH. hESC-PT cells overexpressing GCM2 protein secreted PTH earlier than their counterpart hESC-PT cells. Compared with the two-dimensional cell culture environment, hESCs-derived parathyroid organoids secreted more PTH. Both GCM2 lentiviral transfection and three-dimensional cultures could make hESC-PT cells functionally close to human parathyroid cells. Our study demonstrated that hESCs could differentiate into hESC-PT in vitro, which paves the road for applying the technology to treat hypoparathyroidism and introduces new approaches in the field of regenerative medicine.
Collapse
Affiliation(s)
- Ge Wang
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yaying Du
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoqing Cui
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tao Xu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hanning Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Menglu Dong
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Li
- Department of Clinical and Diagnostic SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Yajie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wenjun Cai
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xue Yang
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yonglin Wu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
9
|
Höppner J, Jüppner H. Rare genetic disorders that impair parathyroid hormone synthesis, secretion, or bioactivity provide insights into the diagnostic utility of different parathyroid hormone assays. Curr Opin Nephrol Hypertens 2024; 33:375-382. [PMID: 38701324 PMCID: PMC12038852 DOI: 10.1097/mnh.0000000000000999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
PURPOSE OF REVIEW Parathyroid hormone (PTH) is the major peptide hormone regulator of blood calcium homeostasis. Abnormal PTH levels can be observed in patients with various congenital and acquired disorders, including chronic kidney disease (CKD). This review will focus on rare human diseases caused by PTH mutations that have provided insights into the regulation of PTH synthesis and secretion as well as the diagnostic utility of different PTH assays. RECENT FINDINGS Over the past years, numerous diseases affecting calcium and phosphate homeostasis have been defined at the molecular level that are responsible for reduced or increased serum PTH levels. The underlying genetic mutations impair parathyroid gland development, involve the PTH gene itself, or alter function of the calcium-sensing receptor (CaSR) or its downstream signaling partners that contribute to regulation of PTH synthesis or secretion. Mutations in the pre sequence of the mature PTH peptide can, for instance, impair hormone synthesis or intracellular processing, while amino acid substitutions affecting the secreted PTH(1-84) impair PTH receptor (PTH1R) activation, or cause defective cleavage of the pro-sequence and thus secretion of a pro- PTH with much reduced biological activity. Mutations affecting the secreted hormone can alter detection by different PTH assays, thus requiring detailed knowledge of the utilized diagnostic test. SUMMARY Rare diseases affecting PTH synthesis and secretion have offered helpful insights into parathyroid biology and the diagnostic utility of commonly used PTH assays, which may have implications for the interpretation of PTH measurements in more common disorders such as CKD.
Collapse
Affiliation(s)
- Jakob Höppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Hassan A, Khalaily N, Kilav-Levin R, Del Castello B, Manley NR, Ben-Dov IZ, Naveh-Many T. Dicer-Mediated mTORC1 Signaling and Parathyroid Gland Integrity and Function. J Am Soc Nephrol 2024; 35:00001751-990000000-00339. [PMID: 38819931 PMCID: PMC11387037 DOI: 10.1681/asn.0000000000000394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/20/2024] [Indexed: 06/02/2024] Open
Abstract
Key Points
Maintaining parathyroid gland integrity is a dynamic process regulated by the parathyroid microRNA–mechanistic target of rapamycin complex 1 axis.This axis is essential for preserving intact parathyroid glands throughout life, with relevance to CKD-induced secondary hyperparathyroidism.
Background
Secondary hyperparathyroidism of CKD contributes significantly to patient morbidity and mortality. The underlining mechanisms of CKD-induced secondary hyperparathyroidism remain elusive. We previously demonstrated that PT-Dicer
−/−
mice, with parathyroid-specific deletion of the microRNA (miRNA)-processing enzyme Dicer and consequently miRNA, maintain normal basal serum parathyroid hormone (PTH) levels but do not develop secondary hyperparathyroidism induced by CKD. In addition, we showed that the parathyroid mechanistic target of rapamycin complex 1 (mTORC1) pathway is activated in CKD. We now explored the roles of Dicer/miRNA and mTORC1 in parathyroid development and function.
Methods
We generated mice with parathyroid-specific Dicer (PT-Dicer
−/−
), mechanistic target of rapamycin (PT-mTOR
−/−
), or tuberous sclerosis complex 1 (PT-Tsc1
−/−
) deficiency combined with yellow fluorescent protein (YFP) or tdTomato expression to identify the parathyroids by fluorescence microscopy. CKD was induced by an adenine-rich high-phosphate diet.
Results
Despite normal basal serum PTH levels, PT-Dicer
−/−
mice displayed apoptotic loss of intact parathyroid glands postnatally and reduced mechanistic target of rapamycin activity. PT-mTOR
−/−
mice lacked intact parathyroid glands yet maintained normal serum PTH levels, mirroring the phenotype of PT-Dicer
−/−
mice. Conversely, PT-Tsc1
−/−
mice with hyperactivated mTORC1 exhibited enlarged glands along with elevated basal serum PTH and calcium levels. Significantly, PT-Dicer
−/−
;Tsc1
−/−
double knockout mice preserved intact parathyroid glands and reinstated CKD-induced secondary hyperparathyroidism.
Conclusions
mTORC1 operates downstream of Dicer and miRNA in the parathyroid and is essential for maintaining postnatal parathyroid gland integrity throughout life and for the pathogenesis of CKD-induced secondary hyperparathyroidism.
Collapse
Affiliation(s)
- Alia Hassan
- Minerva Center for Bone and Mineral Research, Nephrology Services, Hadassah Hebrew University Medical Center and Faculty of Medicine, Jerusalem, Israel
| | - Nareman Khalaily
- Minerva Center for Bone and Mineral Research, Nephrology Services, Hadassah Hebrew University Medical Center and Faculty of Medicine, Jerusalem, Israel
| | - Rachel Kilav-Levin
- Minerva Center for Bone and Mineral Research, Nephrology Services, Hadassah Hebrew University Medical Center and Faculty of Medicine, Jerusalem, Israel
- School of Nursing, Jerusalem College of Technology, Faculty of Life and Health Sciences, Jerusalem, Israel
| | - Barbara Del Castello
- Department of Genetics, University of Georgia, Athens, Georgia
- CRDF Global, Arlington, Virginia
| | - Nancy Ruth Manley
- Department of Genetics, University of Georgia, Athens, Georgia
- Current address: School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Iddo Z Ben-Dov
- Laboratory of Medical Transcriptomics, Nephrology and Internal Medicine B, Hadassah Hebrew University Medical Center and Faculty of Medicine, Jerusalem, Israel
| | - Tally Naveh-Many
- Minerva Center for Bone and Mineral Research, Nephrology Services, Hadassah Hebrew University Medical Center and Faculty of Medicine, Jerusalem, Israel
- Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
11
|
He(何璇) XA, Berenson A, Bernard M, Weber C, Cook LE, Visel A, Fuxman Bass JI, Fisher S. Identification of conserved skeletal enhancers associated with craniosynostosis risk genes. Hum Mol Genet 2024; 33:837-849. [PMID: 37883470 PMCID: PMC11070136 DOI: 10.1093/hmg/ddad182] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/12/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023] Open
Abstract
Craniosynostosis, defined by premature fusion of one or multiple cranial sutures, is a common congenital defect affecting more than 1/2000 infants and results in restricted brain expansion. Single gene mutations account for 15%-20% of cases, largely as part of a syndrome, but the majority are nonsyndromic with complex underlying genetics. We hypothesized that the two noncoding genomic regions identified by a GWAS for craniosynostosis contain distal regulatory elements for the risk genes BMPER and BMP2. To identify such regulatory elements, we surveyed conserved noncoding sequences from both risk loci for enhancer activity in transgenic Danio rerio. We identified enhancers from both regions that direct expression to skeletal tissues, consistent with the endogenous expression of bmper and bmp2. For each locus, we also found a skeletal enhancer that also contains a sequence variant associated with craniosynostosis risk. We examined the activity of each enhancer during craniofacial development and found that the BMPER-associated enhancer is active in the restricted region of cartilage closely associated with frontal bone initiation. The same enhancer is active in mouse skeletal tissues, demonstrating evolutionarily conserved activity. Using enhanced yeast one-hybrid assays, we identified transcription factors that bind each enhancer and observed differential binding between alleles, implicating multiple signaling pathways. Our findings help unveil the genetic mechanism of the two craniosynostosis risk loci. More broadly, our combined in vivo approach is applicable to many complex genetic diseases to build a link between association studies and specific genetic mechanisms.
Collapse
Affiliation(s)
- Xuan Anita He(何璇)
- Department of Pharmacology, Physiology & Biophysics, Boston University, 700 Albany St, W607, Boston, MA 02118, United States
- Graduate Program in Biomolecular Medicine, Boston University, 72 East Concord St, Boston, MA 02118, United States
| | - Anna Berenson
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States
- Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, 5 Cummington Mall, Boston, MA 02215, United States
| | - Michelle Bernard
- Department of Pharmacology, Physiology & Biophysics, Boston University, 700 Albany St, W607, Boston, MA 02118, United States
- College of Arts and Sciences, Boston University, 5 Cummington Mall, Boston, MA 02215, United States
| | - Chris Weber
- Department of Cell and Developmental Biology, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104-6058, United States
| | - Laura E Cook
- Environmental Genomics & System Biology Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, United States
| | - Axel Visel
- Environmental Genomics & System Biology Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, United States
- U.S. Department of Energy Joint Genome Institute, 1 Cyclotron Road, Berkeley, CA 94720, United States
- School of Natural Sciences, 5200 Lake Road, University of California Merced, Merced, CA 95343, United States
| | - Juan I Fuxman Bass
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States
| | - Shannon Fisher
- Department of Pharmacology, Physiology & Biophysics, Boston University, 700 Albany St, W607, Boston, MA 02118, United States
| |
Collapse
|
12
|
English KA, Lines KE, Thakker RV. Genetics of hereditary forms of primary hyperparathyroidism. Hormones (Athens) 2024; 23:3-14. [PMID: 38038882 PMCID: PMC10847196 DOI: 10.1007/s42000-023-00508-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023]
Abstract
Primary hyperparathyroidism (PHPT), a relatively common disorder characterized by hypercalcemia with raised or inappropriately normal serum parathyroid hormone (PTH) concentrations, may occur as part of a hereditary syndromic disorder or as a non-syndromic disease. The associated syndromic disorders include multiple endocrine neoplasia types 1-5 (MEN1-5) and hyperparathyroidism with jaw tumor (HPT-JT) syndromes, and the non-syndromic forms include familial hypocalciuric hypercalcemia types 1-3 (FHH1-3), familial isolated hyperparathyroidism (FIHP), and neonatal severe hyperparathyroidism (NS-HPT). Such hereditary forms may occur in > 10% of patients with PHPT, and their recognition is important for implementation of gene-specific screening protocols and investigations for other associated tumors. Syndromic PHPT tends to be multifocal and multiglandular with most patients requiring parathyroidectomy with the aim of limiting end-organ damage associated with hypercalcemia, particularly osteoporosis, nephrolithiasis, and renal failure. Some patients with non-syndromic PHPT may have mutations of the MEN1 gene or the calcium-sensing receptor (CASR), whose loss of function mutations usually cause FHH1, a disorder associated with mild hypercalcemia and may follow a benign clinical course. Measurement of the urinary calcium-to-creatinine ratio clearance (UCCR) may help to distinguish patients with FHH from those with PHPT, as the majority of FHH patients have low urinary calcium excretion (UCCR < 0.01). Once genetic testing confirms a hereditary cause of PHPT, further genetic testing can be offered to the patients' relatives and subsequent screening can be carried out in these affected family members, which prevents inappropriate testing in normal individuals.
Collapse
Affiliation(s)
- Katherine A English
- OCDEM, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, OX3 7LJ, UK
| | - Kate E Lines
- OCDEM, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK
| | - Rajesh V Thakker
- OCDEM, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, OX3 7LJ, UK.
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK.
| |
Collapse
|
13
|
Alcantara EH, Kwon JH, Kang MK, Cho YE, Kwun IS. Zinc Deficiency Promotes Calcification in Vascular Smooth Muscle Cells Independent of Alkaline Phosphatase Action and Partly Impacted by Pit1 Upregulation. Nutrients 2024; 16:291. [PMID: 38257184 PMCID: PMC10819640 DOI: 10.3390/nu16020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Inorganic phosphate (Pi) is a critical determinant of calcification, and its concentration is regulated by alkaline phosphatase (ALP) and Pit1. ALP is a key regulator of osteogenic calcification and acts by modulating local inorganic phosphate (Pi) concentrations through hydrolyzing pyrophosphate in the extracellular matrix (ECM). Pit1, a sodium-dependent phosphate transporter, regulates calcification via facilitating phosphate uptake within the cells. To investigate whether zinc differentially regulates osteoblastic and vascular calcifications, we examined ALP activity and Pit1 in osteoblastic and vascular smooth muscle cells (VSMCs). Our findings demonstrate that calcification in osteoblastic MC3T3-E1 cells is decreased via diminished ALP action under zinc deficiency. In contrast, zinc-deficiency-induced calcification in VSMCs is independent of ALP action, as demonstrated by very weak ALP activity and expression in calcified VSMCs. In zinc-deficient A7r5 VSMC, P accumulation increased with increasing Na phosphate concentration (3-7 mM) but not with β-GP treatment, which requires ALP activity to generate Pi. Ca deposition also increased with Na phosphate in a dose-dependent manner; in contrast, β-GP did not affect Ca deposition. In osteoblastic cells, Pit1 expression was not affected by zinc treatments. In contrast, Pit1 expression is highly upregulated in A7r5 VSMC under zinc deficiency. Using phosphonoformic acid, a competitive inhibitor of Pit1, we showed that calcification is inhibited in both A7r5 and MC3T3-E1 cells, indicating a requirement for Pit1 in both calcifications. Moreover, the downregulation of VSMC markers under zinc deficiency was restored by blocking Pit1. Taken together, our results imply that zinc-deficiency-induced calcification in VSMC is independent of ALP action in contrast to osteoblastic calcification. Moreover, Pit1 expression in VSMCs is a target for zinc deficiency and may mediate the inhibition of VSMC marker expression under zinc deficiency.
Collapse
Affiliation(s)
| | | | | | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong 36729, Republic of Korea; (E.H.A.); (J.-H.K.); (M.-K.K.)
| | - In-Sook Kwun
- Department of Food and Nutrition, Andong National University, Andong 36729, Republic of Korea; (E.H.A.); (J.-H.K.); (M.-K.K.)
| |
Collapse
|
14
|
Szalat A, Shpitzen S, Pollack R, Mazeh H, Durst R, Meiner V. GCM2 p.Tyr394Ser variant in Ashkenazi Israeli patients with suspected familial isolated hyperparathyroidism. Front Endocrinol (Lausanne) 2023; 14:1254156. [PMID: 38130397 PMCID: PMC10733520 DOI: 10.3389/fendo.2023.1254156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Context A germline mutation can be identified in up to 10% of patients with primary hyperparathyroidism (PHPT). In 2017, a high frequency of the GCM2 [(NM_ 004752.4) c.1181A> C; p.Tyr394Ser; rs142287570] variant was reported in PHPT Ashkenazi Jews (AJ). Objective To evaluate the presence of the GCM2 p.Tyr394Ser variant in Israeli patients addressed for genetic evaluation to characterize their phenotype and clinical management. Method Patients with PHPT who underwent addressed for genetic screening for suspected familial hypocalciuric hypercalcemia (FHH), a family history of isolated hyperparathyroidism (FIHP), or failed parathyroidectomy with persistent PHPT were recruited. Those with normal initial selected gene sequencing or hyperparathyroid genetic panel completed the GCM2 p.Tyr394Ser variant sequencing. The prevalence of this variant was evaluated using our local genomic database. Results A total of 42 single individuals from unrelated kindreds were evaluated. A disease-causing mutation was found in 11 (26.1%) patients: 10 were diagnosed with FHH (eight CASR and two AP2S1 mutations), and one patient had a CKN2B mutation. In 28 of the remaining patients, the GCM2 p.Tyr394Ser variant was positive in three (10.7%), and all were AJ. Within AJ (15/28, 53.5%), the rate of the p.Tyr394Ser variant was 3/15 (20%), and of those, two had a history of familial isolated hyperparathyroidism. Multi-glandular parathyroid adenoma/hyperplasia was also observed in two of these patients. No clinical or laboratory findings could discriminate patients with the GCM2 p.Tyr394Ser variant from those with FHH. Cinacalcet normalized the calcium levels in one patient. The prevalence of the GCM2 p.Tyr394Ser variant in 15,407 tests in our local genomic database was 0.98%. Conclusion In contrast to previous observations, the GCM2 p.Tyr394Ser variant-associated phenotype may be mild in AJ with FIHP, sometimes mimicking FHH. Because surgery may be curative, surgeons should be aware of the possibility of multiple gland diseases in these patients. The clinical spectrum and clinical utility of screening for this variant warrant further investigation.
Collapse
Affiliation(s)
- Auryan Szalat
- Endocrinology and Metabolism Service, Department of Internal Medicine, Osteoporosis Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Shoshana Shpitzen
- Center for Research, Prevention and Treatment of Atherosclerosis, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rena Pollack
- Endocrinology and Metabolism Service, Department of Internal Medicine, Osteoporosis Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Haggi Mazeh
- Department of Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ronen Durst
- Center for Research, Prevention and Treatment of Atherosclerosis, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Vardiella Meiner
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
15
|
Parekh VI, Brinster LR, Guan B, Simonds WF, Weinstein LS, Agarwal SK. A Knock-In Mouse Model of the Gcm2 Variant p.Y392S Develops Normal Parathyroid Glands. J Endocr Soc 2023; 7:bvad126. [PMID: 37885910 PMCID: PMC10599131 DOI: 10.1210/jendso/bvad126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Indexed: 10/28/2023] Open
Abstract
Context The glial cells missing 2 (GCM2) gene functions as a transcription factor that is essential for parathyroid gland development, and variants in this gene have been associated with 2 parathyroid diseases: isolated hypoparathyroidism in patients with homozygous germline inactivating variants and primary hyperparathyroidism in patients with heterozygous germline activating variants. A recurrent germline activating missense variant of GCM2, p.Y394S, has been reported in patients with familial primary hyperparathyroidism. Objective To determine whether the GCM2 p.Y394S missense variant causes overactive and enlarged parathyroid glands in a mouse model. Methods CRISPR/Cas9 gene editing technology was used to generate a mouse model with the germline heterozygous Gcm2 variant p.Y392S that corresponds to the human GCM2 p.Y394S variant. Wild-type (Gcm2+/+) and germline heterozygous (Gcm2+/Y392S) mice were evaluated for serum biochemistry and parathyroid gland morphology. Results Gcm2 +/Y392S mice did not show any change compared to Gcm2+/+ mice in serum calcium and parathyroid hormone levels, parathyroid gland histology, cell proliferation, or parathyroid gland size. Conclusion The mouse model of the p.Y392S variant of Gcm2 shows that this variant is tolerated in mice, as it does not increase parathyroid gland cell proliferation and circulating calcium or PTH levels. Further investigation of Gcm2+/Y392S mice to study the effect of this variant of Gcm2 on early events in parathyroid gland development will be of interest.
Collapse
Affiliation(s)
- Vaishali I Parekh
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lauren R Brinster
- Office of Research Services, Division of Veterinary Resources, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bin Guan
- Opthalmic Genomics Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - William F Simonds
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sunita K Agarwal
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Jha S, Simonds WF. Molecular and Clinical Spectrum of Primary Hyperparathyroidism. Endocr Rev 2023; 44:779-818. [PMID: 36961765 PMCID: PMC10502601 DOI: 10.1210/endrev/bnad009] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/09/2023] [Accepted: 03/17/2023] [Indexed: 03/25/2023]
Abstract
Recent data suggest an increase in the overall incidence of parathyroid disorders, with primary hyperparathyroidism (PHPT) being the most prevalent parathyroid disorder. PHPT is associated with morbidities (fractures, kidney stones, chronic kidney disease) and increased risk of death. The symptoms of PHPT can be nonspecific, potentially delaying the diagnosis. Approximately 15% of patients with PHPT have an underlying heritable form of PHPT that may be associated with extraparathyroidal manifestations, requiring active surveillance for these manifestations as seen in multiple endocrine neoplasia type 1 and 2A. Genetic testing for heritable forms should be offered to patients with multiglandular disease, recurrent PHPT, young onset PHPT (age ≤40 years), and those with a family history of parathyroid tumors. However, the underlying genetic cause for the majority of patients with heritable forms of PHPT remains unknown. Distinction between sporadic and heritable forms of PHPT is useful in surgical planning for parathyroidectomy and has implications for the family. The genes currently known to be associated with heritable forms of PHPT account for approximately half of sporadic parathyroid tumors. But the genetic cause in approximately half of the sporadic parathyroid tumors remains unknown. Furthermore, there is no systemic therapy for parathyroid carcinoma, a rare but potentially fatal cause of PHPT. Improved understanding of the molecular characteristics of parathyroid tumors will allow us to identify biomarkers for diagnosis and novel targets for therapy.
Collapse
Affiliation(s)
- Smita Jha
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1752, USA
| | - William F Simonds
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1752, USA
| |
Collapse
|
17
|
Kameda Y. Cellular and molecular mechanisms of the organogenesis and development, and function of the mammalian parathyroid gland. Cell Tissue Res 2023; 393:425-442. [PMID: 37410127 DOI: 10.1007/s00441-023-03785-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/12/2023] [Indexed: 07/07/2023]
Abstract
Serum calcium homeostasis is mainly regulated by parathormone (PTH) secreted by the parathyroid gland. Besides PTH and Gcm2, a master gene for parathyroid differentiation, many genes are expressed in the gland. Especially, calcium-sensing receptor (CaSR), vitamin D receptor (VDR), and Klotho function to prevent increased secretion of PTH and hyperplasia of the parathyroid gland under chronic hypocalcemia. Parathyroid-specific dual deletion of Klotho and CaSR induces a marked enlargement of the glandular size. The parathyroid develops from the third and fourth pharyngeal pouches except murine species in which the gland is derived from the third pouch only. The development of the murine parathyroid gland is categorized as follows: (1) formation and differentiation of the pharyngeal pouches, (2) appearance of parathyroid domain in the third pharyngeal pouch together with thymus domain, (3) migration of parathyroid primordium attached to the top of thymus, and (4) contact with the thyroid lobe and separation from the thymus. The transcription factors and signaling molecules involved in each of these developmental stages are elaborated. In addition, mesenchymal neural crest cells surrounding the pharyngeal pouches and parathyroid primordium and invading the parathyroid parenchyma participate in the development of the gland.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
18
|
Kano M, Mizuno N, Sato H, Kimura T, Hirochika R, Iwasaki Y, Inoshita N, Nagano H, Kasai M, Yamamoto H, Yamaguchi T, Suga H, Masaki H, Mizutani E, Nakauchi H. Functional calcium-responsive parathyroid glands generated using single-step blastocyst complementation. Proc Natl Acad Sci U S A 2023; 120:e2216564120. [PMID: 37379351 PMCID: PMC10334775 DOI: 10.1073/pnas.2216564120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/24/2023] [Indexed: 06/30/2023] Open
Abstract
Patients with permanent hypoparathyroidism require lifelong replacement therapy to avoid life-threatening complications, The benefits of conventional treatment are limited, however. Transplanting a functional parathyroid gland (PTG) would yield better results. Parathyroid gland cells generated from pluripotent stem cells in vitro to date cannot mimic the physiological responses to extracellular calcium that are essential for calcium homeostasis. We thus hypothesized that blastocyst complementation (BC) could be a better strategy for generating functional PTG cells and compensating loss of parathyroid function. We here describe generation of fully functional PTGs from mouse embryonic stem cells (mESCs) with single-step BC. Using CRISPR-Cas9 knockout of Glial cells missing2 (Gcm2), we efficiently produced aparathyroid embryos for BC. In these embryos, mESCs differentiated into endocrinologically mature PTGs that rescued Gcm2-/- mice from neonatal death. The mESC-derived PTGs responded to extracellular calcium, restoring calcium homeostasis on transplantation into mice surgically rendered hypoparathyroid. We also successfully generated functional interspecies PTGs in Gcm2-/- rat neonates, an accomplishment with potential for future human PTG therapy using xenogeneic animal BC. Our results demonstrate that BC can produce functional endocrine organs and constitute a concept in treatment of hypoparathyroidism.
Collapse
Affiliation(s)
- Mayuko Kano
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Metabolism and Endocrinology, Department of Medicine, St. Marianna University School of Medicine, Miyamae-ku, Kawasaki, Kanagawa216-8511, Japan
| | - Naoaki Mizuno
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Hideyuki Sato
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Takaharu Kimura
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Rei Hirochika
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Yasumasa Iwasaki
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Mie510-0293, Japan
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi783-8505, Japan
| | - Naoko Inoshita
- Department of Pathology, Moriyama Memorial Hospital, Edogawa-ku, Tokyo134-0081, Japan
| | - Hisato Nagano
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, Tokorozawa, Saitama359-8513, Japan
| | - Mariko Kasai
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Hiromi Yamamoto
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Laboratory of Regenerative Medicine, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo192-0392, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya466-8550, Japan
| | - Hideki Masaki
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Eiji Mizutani
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Hiromitsu Nakauchi
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
19
|
Tao X, Xu T, Lin X, Xu S, Fan Y, Guo B, Deng X, Jiao Q, Chen L, Wei Z, Chen C, Yang W, Zhang Z, Yu X, Yue H. Genomic Profiling Reveals the Variant Landscape of Sporadic Parathyroid Adenomas in Chinese Population. J Clin Endocrinol Metab 2023; 108:1768-1775. [PMID: 36611251 PMCID: PMC10271222 DOI: 10.1210/clinem/dgad002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/09/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To define somatic variants of parathyroid adenoma (PA) and to provide novel insights into the underlying molecular mechanism of sporadic PA. METHODS Basic clinical characteristics and biochemical indices of 73 patients with PA were collected. Whole-exome sequencing was performed on matched tumor-constitutional DNA pairs to detect somatic alterations. Functional annotation was carried out by ingenuity pathway analysis afterward. The protein expression of the variant gene was confirmed by immunohistochemistry, and the relationship between genotype and phenotype was analyzed. RESULTS Somatic variants were identified in 1549 genes, with an average of 69 variants per tumor (range, 13-2109; total, 9083). Several novel recurrent somatic variants were detected, such as KMT2D (15/73), MUC4 (14/73), POTEH (13/73), CD22 (12/73), HSPA2 (12/73), HCFC1 (11/73), MAGEA1 (11/73), and SLC4A3 (11/73), besides the previously reported PA-related genes, including MEN1 (11/73), CASR (6/73), MTOR (4/73), ASXL3 (3/73), FAT1 (3/73), ZFX (5/73), EZH1 (2/73), POT1 (2/73), and EZH2 (1/73). Among them, KMT2D might be the candidate driver gene of PA. Crucially, 5 patients carried somatic mutations in CDC73, showed an aggressive phenotype similar to that of parathyroid carcinoma (PC), and had a decreased expression of parafibromin. Pathway analysis of recurrent potential PA-associated driver variant genes revealed functional enrichments in the signaling pathway of Notch. CONCLUSION Our study expanded the pathogenic variant spectrum of PA and indicated that KMT2D might be a novel candidate driver gene and be considered as a diagnostic biomarker for PA. Meanwhile, CDC73 mutations might be an early developmental event from PA to PC. The results provided insights into elucidating the pathogenesis of parathyroid tumorigenesis and a certain basis for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaohui Tao
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Tian Xu
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaoyun Lin
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Shuqin Xu
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Youben Fan
- Center of Thyroid and Parathyroid, Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Bomin Guo
- Center of Thyroid and Parathyroid, Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xianzhao Deng
- Center of Thyroid and Parathyroid, Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lihui Chen
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhe Wei
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Chengkun Chen
- Center of Thyroid and Parathyroid, Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Wendi Yang
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhenlin Zhang
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiangtian Yu
- Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hua Yue
- Shanghai Clinical Research Center of Bone Diseases, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
20
|
Tolkin L, Klein V, Frankel M, Altarescu G, Beeri R, Munter G. Variant Tyr 394Ser in the GCM2 Gene Is Rare in a Cohort of Ashkenazi Jews With Primary Hyperparathyroidism. J Endocr Soc 2023; 7:bvad086. [PMID: 37362385 PMCID: PMC10289514 DOI: 10.1210/jendso/bvad086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Indexed: 06/28/2023] Open
Abstract
Context Various genes have been associated with familial and sporadic primary hyperparathyroidism (PHPT), including activating mutations of the glial cells missing transcription factor 2 (GCM2) gene. Objective The aim of this study was to assess the prevalence of the GCM2 p.Tyr394Ser variant in the Jerusalem Ashkenazi Jewish (AJ) population with PHPT, and to conclude whether routine genetic testing is justified. Methods The blood of 40 self-reported AJ patients with PHPT and 200 AJ controls was tested for the GCM2 p.Tyr394Ser variant. Demographic and medical information was extracted from the patients' charts and evaluated accordingly. Results Two (5%) PHPT patients and 3 (1.5%) controls were heterozygotes for the tested variant. Our patients were mostly (87.5%) sporadic cases. One of the heterozygote patients had familial PHPT; the other had 2 parathyroid adenomas, and the levels of his blood and urinary calcium were extremely high. Conclusion Our results suggest that in AJ patients with sporadic, single-gland PHPT, the likelihood of the tested variant is low and genetic testing should be limited to those with familial PHPT or multiglandular disease.
Collapse
Affiliation(s)
- Lior Tolkin
- Correspondence: Lior Tolkin, MD, Department of Internal Medicine Endocrine Unit, Shaare Zedek Medical Center Jerusalem affiliated with the Faculty of Medicine, Hebrew University, 12 Shmuel Bait St, PO Box 3235, Jerusalem 3235, Israel.
| | - Vanessa Klein
- Department of Internal Medicine Endocrine Unit, Shaare Zedek Medical Center Jerusalem affiliated with the Faculty of Medicine, Hebrew University, Jerusalem 3235, Israel
| | - Meir Frankel
- Department of Internal Medicine Endocrine Unit, Shaare Zedek Medical Center Jerusalem affiliated with the Faculty of Medicine, Hebrew University, Jerusalem 3235, Israel
| | - Gheona Altarescu
- Genetic Department, Shaare Zedek Medical Center Jerusalem affiliated with the Faculty of Medicine, Hebrew University, Jerusalem 3235, Israel
| | - Rachel Beeri
- Genetic Department, Shaare Zedek Medical Center Jerusalem affiliated with the Faculty of Medicine, Hebrew University, Jerusalem 3235, Israel
| | - Gabriel Munter
- Department of Internal Medicine Endocrine Unit, Shaare Zedek Medical Center Jerusalem affiliated with the Faculty of Medicine, Hebrew University, Jerusalem 3235, Israel
| |
Collapse
|
21
|
Butylina M, Föger-Samwald U, Gelles K, Pietschmann P, Sipos W. Challenges in establishing animal models for studying osteoimmunology of hypoparathyroidism. Front Vet Sci 2023; 10:1163903. [PMID: 37180074 PMCID: PMC10169642 DOI: 10.3389/fvets.2023.1163903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 03/30/2023] [Indexed: 05/15/2023] Open
Abstract
Hypoparathyroidism is a relatively rare human and veterinary disease characterized by deficient or absent production of parathyroid hormone (PTH). PTH is known as a classical regulator of calcium and phosphorus homeostasis. Nevertheless, the hormone also appears to modulate immune functions. For example, increased CD4:CD8 T-cell ratios and elevated interleukin (IL)-6 and IL-17A levels were observed in patients with hyperparathyroidism, whereas gene expression of tumor necrosis factor-α (TNF-α) and granulocyte macrophage-colony stimulating factor (GM-CSF) was decreased in patients with chronic postsurgical hypoparathyroidism. Various immune cell populations are affected differently. So, there is a need for validated animal models for the further characterization of this disease for identifying targeted immune-modulatory therapies. In addition to genetically modified mouse models of hypoparathyroidism, there are surgical rodent models. Parathyroidectomy (PTX) can be well performed in rats-for pharmacological and associated osteoimmunological research and bone mechanical studies, a large animal model could be preferable, however. A major drawback for successfully performing total PTX in large animal species (pigs and sheep) is the presence of accessory glands, thus demanding to develop new approaches for real-time detection of all parathyroid tissues.
Collapse
Affiliation(s)
- Maria Butylina
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ursula Föger-Samwald
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Katharina Gelles
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Pietschmann
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Sipos
- Clinical Department for Farm Animals, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
22
|
Mannstadt M, Cianferotti L, Gafni RI, Giusti F, Kemp EH, Koch CA, Roszko KL, Yao L, Guyatt GH, Thakker RV, Xia W, Brandi ML. Hypoparathyroidism: Genetics and Diagnosis. J Bone Miner Res 2022; 37:2615-2629. [PMID: 36375809 DOI: 10.1002/jbmr.4667] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 01/05/2023]
Abstract
This narrative report summarizes diagnostic criteria for hypoparathyroidism and describes the clinical presentation and underlying genetic causes of the nonsurgical forms. We conducted a comprehensive literature search from January 2000 to January 2021 and included landmark articles before 2000, presenting a comprehensive update of these topics and suggesting a research agenda to improve diagnosis and, eventually, the prognosis of the disease. Hypoparathyroidism, which is characterized by insufficient secretion of parathyroid hormone (PTH) leading to hypocalcemia, is diagnosed on biochemical grounds. Low albumin-adjusted calcium or ionized calcium with concurrent inappropriately low serum PTH concentration are the hallmarks of the disease. In this review, we discuss the characteristics and pitfalls in measuring calcium and PTH. We also undertook a systematic review addressing the utility of measuring calcium and PTH within 24 hours after total thyroidectomy to predict long-term hypoparathyroidism. A summary of the findings is presented here; results of the detailed systematic review are published separately in this issue of JBMR. Several genetic disorders can present with hypoparathyroidism, either as an isolated disease or as part of a syndrome. A positive family history and, in the case of complex diseases, characteristic comorbidities raise the clinical suspicion of a genetic disorder. In addition to these disorders' phenotypic characteristics, which include autoimmune diseases, we discuss approaches for the genetic diagnosis. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Michael Mannstadt
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Luisella Cianferotti
- Bone Metabolic Diseases Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Rachel I Gafni
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Christian A Koch
- Department of Medicine/Endocrinology, Fox Chase Cancer Center, Philadelphia, PA, USA.,Department of Medicine/Endocrinology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kelly L Roszko
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Liam Yao
- Department of Health Research Methods, Evidence, and Impact, and Department of Medicine, McMaster University, Hamilton, Canada
| | - Gordon H Guyatt
- Department of Health Research Methods, Evidence, and Impact, and Department of Medicine, McMaster University, Hamilton, Canada
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, UK.,Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Weibo Xia
- Department of Endocrinology, Peking Union Medical Collage Hospital, Beijing, China
| | - Maria-Luisa Brandi
- Fondazione Italiana sulla Ricerca sulle Malattie dell'Osso (F.I.R.M.O. Foundation), Florence, Italy
| |
Collapse
|
23
|
An anti-inflammatory transcriptional cascade conserved from flies to humans. Cell Rep 2022; 41:111506. [DOI: 10.1016/j.celrep.2022.111506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
|
24
|
Michelson DA, Mathis D. Thymic mimetic cells: tolerogenic masqueraders. Trends Immunol 2022; 43:782-791. [PMID: 36008259 PMCID: PMC9509455 DOI: 10.1016/j.it.2022.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
Medullary thymic epithelial cells (mTECs) clonally delete or divert autoreactive T cells by ectopically expressing a diverse array of peripheral-tissue antigens (PTAs) within the thymus. Although thymic stromal cells with histological features of extra-thymic cell types, like myocytes or neurons, have been observed by light microscopy since the mid-1800s, most modern work on PTA expression has focused on the transcription factor Aire. Here, we highlight recent work that has refocused attention on such 'misplaced' thymic cells, referred to collectively as thymic mimetic cells. We review the molecular underpinnings of mimetic cells and their roles in establishing T cell tolerance, and we propose that mimetic cells play important roles in autoimmunity. Finally, we suggest future directions for this emerging area.
Collapse
Affiliation(s)
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Nagakubo D, Hirakawa M, Iwanami N, Boehm T. Limits to in vivo fate changes of epithelia in thymus and parathyroid by ectopic expression of transcription factors Gcm2 and Foxn1. Sci Rep 2022; 12:13554. [PMID: 35941210 PMCID: PMC9360016 DOI: 10.1038/s41598-022-17844-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/02/2022] [Indexed: 11/09/2022] Open
Abstract
The development of the parathyroid and the thymus from the third pharyngeal pouch depends on the activities of the Gcm2 and Foxn1 transcription factors, respectively, whose expression domains sharply demarcate two regions in the developing third pharyngeal pouch. Here, we have generated novel mouse models to examine whether ectopic co-expression of Gcm2 in the thymic epithelium and of Foxn1 in the parathyroid perturbs the establishment of organ fates in vivo. Expression of Gcm2 in the thymic rudiment does not activate a parathyroid-specific expression programme, even in the absence of Foxn1 activity. Co-expression of Foxn1 in the parathyroid fails to impose thymopoietic capacity. We conclude that the actions of Foxn1 and Gcm2 transcription factors are cell context-dependent and that they each require permissive transcription factor landscapes in order to successfully interfere with organ-specific cell fate.
Collapse
Affiliation(s)
- Daisuke Nagakubo
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108, Freiburg, Germany.,Division of Health and Hygienic Sciences, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji, Hyogo, 670-8524, Japan
| | - Mayumi Hirakawa
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108, Freiburg, Germany.,Division of Immunology and Allergy, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda-City, Chiba, 278-0022, Japan
| | - Norimasa Iwanami
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108, Freiburg, Germany.,Center for Bioscience Research and Education, Utsunomiya University, Utsunomiya, Tochigi, 321-8505, Japan
| | - Thomas Boehm
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108, Freiburg, Germany.
| |
Collapse
|
26
|
Parathyroid Cell Differentiation from Progenitor Cells and Stem Cells: Development, Molecular Mechanism, Function, and Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1387:13-24. [PMID: 34981451 DOI: 10.1007/5584_2021_694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Parathyroid glands are endocrine organs which are located posterior to thyroid glands and control secretion of parathyroid hormone (PTH) in order to regulate blood calcium level. PTH maintains calcium homeostasis by acting on the bone, kidney, and small intestine. PTH deficiency leads to chronic hypocalcemia, organ calcinosis, kidney and heart failure, painful muscle spasms, neuromuscular problems, and memory problems. Since parathyroid cells have inadequate proliferation potential in culture conditions, their utilization as a cellular therapy option is very limited. Although studies conducted so far include parathyroid cell differentiation from various cell types, problems related to successful cellular differentiation and transplantation still remain. Recently, parathyroid tissue engineering has attracted attention as a potential treatment for the parathyroid-related diseases caused by hypoparathyroidism. Although major progression is made in the construction of tissue engineering protocols using parathyroid cells and biomaterials, PTH secretion to mimic its spontaneous harmony in the body is a challenge. This chapter comprehensively defines the derivation of parathyroid cells from various cell sources including pluripotent stem cells, molecular mechanisms, and tissue engineering applications.
Collapse
|
27
|
Kim SW. Gene expression profiles in parathyroid adenoma and normal parathyroid tissue. VITAMINS AND HORMONES 2022; 120:289-304. [PMID: 35953114 DOI: 10.1016/bs.vh.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A parathyroid adenoma comprises 80-85% as a cause of primary hyperparathyroidism. The clonal origin of most parathyroid adenomas suggests a defect at the level of the gene controlling growth of the parathyroid cell or the expression of parathyroid hormone (PTH). Two genes, MEN1 and CCND1, a tumor suppressor and a proto-oncogene respectively, have been solidly established as primary tumorigenic drivers in parathyroid adenomas. As well, germline and somatic mutation of other genes involved in cell cycle regulation or PTH regulation have been discovered in parathyroid adenomas. Moreover, comparative genomic studies between parathyroid adenomas and normal parathyroid tissues have suggested more complex genetic landscape. Microarray analysis have revealed differential expression profiles of genes involved in cell cycle regulation, growth factors, apoptotic pathway, or PTH synthesis or regulation pathway such as CASR, GCM2 and KL (Klotho). Furthermore, recent next-generation sequencing analysis reconfirmed previous finding or revealed novel finding, suggesting signal peptidase complex subunit (SPCS2), ribosomal proteins (RPL23, RPL26, RPN1, RPS25), the endoplasmic reticulum membrane (SEC11C, SEC11A, SEC61G), Klotho, cyclin D1, β-catenin, VDR, CaSR and GCM2 may be important factors contributing to the parathyroid adenoma.
Collapse
Affiliation(s)
- Sang Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Hirschberger C, Gillis JA. The pseudobranch of jawed vertebrates is a mandibular arch-derived gill. Development 2022; 149:275947. [PMID: 35762641 PMCID: PMC9340550 DOI: 10.1242/dev.200184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 06/14/2022] [Indexed: 12/16/2022]
Abstract
The pseudobranch is a gill-like epithelial elaboration that sits behind the jaw of most fishes. This structure was classically regarded as a vestige of the ancestral gill arch-like condition of the gnathostome jaw. However, more recently, hypotheses of jaw evolution by transformation of a gill arch have been challenged, and the pseudobranch has alternatively been considered a specialised derivative of the second (hyoid) pharyngeal arch. Here, we demonstrate in the skate (Leucoraja erinacea) that the pseudobranch does, in fact, derive from the mandibular arch, and that it shares gene expression features and cell types with gills. We also show that the skate mandibular arch pseudobranch is supported by a spiracular cartilage that is patterned by a shh-expressing epithelial signalling centre. This closely parallels the condition seen in the gill arches, where cartilaginous appendages called branchial rays, which support the respiratory lamellae of the gills, are patterned by a shh-expressing gill arch epithelial ridge. Together with similar discoveries in zebrafish, our findings support serial homology of the pseudobranch and gills, and an ancestral origin of gill arch-like anatomical features from the gnathostome mandibular arch. Summary: The skate pseudobranch is a gill serial homologue and reveals the ancestral gill arch-like nature of the jawed vertebrate mandibular arch.
Collapse
Affiliation(s)
- Christine Hirschberger
- University of Cambridge 1 Department of Zoology , , Downing Street, Cambridge CB2 3EJ , UK
| | - J. Andrew Gillis
- University of Cambridge 1 Department of Zoology , , Downing Street, Cambridge CB2 3EJ , UK
- Marine Biological Laboratory 2 , 7 MBL Street, Woods Hole, MA 02543 , USA
| |
Collapse
|
29
|
Ilenwabor BP, Schigt H, Kompatscher A, Bos C, Zuidscherwoude M, van der Eerden BCJ, Hoenderop JGJ, de Baaij JHF. FAM111A is dispensable for electrolyte homeostasis in mice. Sci Rep 2022; 12:10211. [PMID: 35715480 PMCID: PMC9205974 DOI: 10.1038/s41598-022-14054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Autosomal dominant mutations in FAM111A are causative for Kenny-Caffey syndrome type 2. Patients with Kenny-Caffey syndrome suffer from severe growth retardation, skeletal dysplasia, hypoparathyroidism, hypocalcaemia, hyperphosphataemia and hypomagnesaemia. While recent studies have reported FAM111A to function in antiviral response and DNA replication, its role in regulating electrolyte homeostasis remains unknown. In this study, we assessed the role of FAM111A in the regulation of serum electrolyte balance using a Fam111a knockout (Fam111a-/-) C57BL/6 N mouse model. Fam111a-/- mice displayed normal weight and serum parathyroid hormone (PTH) concentration and exhibited unaltered magnesium, calcium and phosphate levels in serum and 24-hour urine. Expression of calciotropic (including Cabp28k, Trpv5, Klotho and Cyp24a1), magnesiotropic (including Trpm6, Trpm7, Cnnm2 and Cnnm4) and phosphotropic (Slc20a1, Slc20a2, Slc34a1 and Slc34a3) genes in the kidneys, duodenum and colon were not affected by Fam111a depletion. Only Slc34a2 expression was significantly upregulated in the duodenum, but not in the colon. Analysis of femurs showed unaffected bone morphology and density in Fam111a-/- mice. Kidney and parathyroid histology were also normal in Fam111a-/- mice. In conclusion, our study is the first to characterise the function of FAM111A in vivo and we report that mice lacking FAM111A exhibit normal electrolyte homeostasis on a standard diet.
Collapse
Affiliation(s)
- Barnabas P Ilenwabor
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Heidi Schigt
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Andreas Kompatscher
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Caro Bos
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Malou Zuidscherwoude
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
30
|
Vincze S, Peters NV, Kuo CL, Brown TC, Korah R, Murtha TD, Bellizzi J, Riccardi A, Parham K, Carling T, Costa-Guda J, Arnold A. GCM2 Variants in Familial and Multiglandular Primary Hyperparathyroidism. J Clin Endocrinol Metab 2022; 107:e2021-e2026. [PMID: 34967908 DOI: 10.1210/clinem/dgab929] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Multiglandular and familial parathyroid disease constitute important fractions of primary hyperparathyroidism (PHPT). Germline missense variants of GCM2, a regulator of parathyroid development, were observed in familial isolated hyperparathyroidism and sporadic PHPT. However, as these previously reported GCM2 variants occur at relatively high frequencies in the population, understanding their potential clinical utility will require both additional penetrance data and functional evidence relevant to tumorigenicity. OBJECTIVE Determine the frequency of GCM2 variants of interest among patients with sporadic multigland or familial parathyroid disease and assess their penetrance. DESIGN AND PATIENTS DNA-encoding PHPT-associated GCM2 germline variants were polymerase chain reaction-amplified and sequenced from 107 patients with either sporadic multigland or suspected/confirmed familial parathyroid tumors. RESULTS GCM2 variants were observed in 9 of 107 cases (8.4%): Y282D in 4 patients (6.3%) with sporadic multigland disease; Y394S in 2 patients (11.1%) with familial PHPT and 3 (4.8%) with sporadic multigland disease. Compared with the general population, Y282D was enriched 5.9-fold in multigland disease, but its penetrance was very low (0.02%). Y394S was enriched 79-fold in sporadic multigland disease and 93-fold in familial PHPT, but its penetrance was low (1.33% and 1.04%, respectively). CONCLUSIONS Observed in vitro-activating GCM2 variant alleles are significantly overrepresented in PHPT patients with multiglandular or familial disease compared to the general population, yet penetrance values are very low; that is, most individuals with these variants in the population have a very low risk of developing PHPT. The potential clinical utility of detecting these GCM2 variants requires further investigation, including assessing their possible role as pathogenic/low-penetrance alleles.
Collapse
Affiliation(s)
- Sarah Vincze
- Center for Molecular Oncology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Nicholas V Peters
- Yale Endocrine Neoplasia Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Chia-Ling Kuo
- Biostatistics Center, Connecticut Institute for Clinical and Translational Science, University of Connecticut, Farmington, CT, USA
| | - Taylor C Brown
- Yale Endocrine Neoplasia Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Washington University School of Medicine, St. Louis, MO,USA
| | - Reju Korah
- Yale Endocrine Neoplasia Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Timothy D Murtha
- Yale Endocrine Neoplasia Laboratory, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Justin Bellizzi
- Center for Molecular Oncology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Aaliyah Riccardi
- Center for Molecular Oncology, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Kourosh Parham
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Tobias Carling
- Biostatistics Center, Connecticut Institute for Clinical and Translational Science, University of Connecticut, Farmington, CT, USA
- Carling Adrenal Center, Hospital for Endocrine Surgery, Tampa, FL, USA
| | - Jessica Costa-Guda
- Center for Molecular Oncology, University of Connecticut School of Medicine, Farmington, CT, USA
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| | - Andrew Arnold
- Center for Molecular Oncology, University of Connecticut School of Medicine, Farmington, CT, USA
- Division of Endocrinology and Metabolism, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
31
|
Egstrand S, Mace ML, Morevati M, Nordholm A, Engelholm LH, Thomsen JS, Brüel A, Naveh-Many T, Guo Y, Olgaard K, Lewin E. Hypomorphic expression of parathyroid Bmal1 disrupts the internal parathyroid circadian clock and increases parathyroid cell proliferation in response to uremia. Kidney Int 2022; 101:1232-1250. [PMID: 35276205 DOI: 10.1016/j.kint.2022.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 01/22/2023]
Abstract
The molecular circadian clock is an evolutionary adaptation to anticipate recurring changes in the environment and to coordinate variations in activity, metabolism and hormone secretion. Parathyroid hyperplasia in uremia is a significant clinical challenge. Here, we examined changes in the transcriptome of the murine parathyroid gland over 24 hours and found a rhythmic expression of parathyroid signature genes, such as Casr, Vdr, Fgfr1 and Gcm2. Overall, 1455 genes corresponding to 6.9% of all expressed genes had significant circadian rhythmicity. Biological pathway analysis indicated that the circadian clock system is essential for the regulation of parathyroid cell function. To study this, a novel mouse strain with parathyroid gland-specific knockdown of the core clock gene Bmal1 (PTHcre;Bmal1flox/flox) was created. Dampening of the parathyroid circadian clock rhythmicity was found in these knockdown mice, resulting in abrogated rhythmicity of regulators of parathyroid cell proliferation such as Sp1, Mafb, Gcm2 and Gata3, indicating circadian clock regulation of these genes. Furthermore, the knockdown resulted in downregulation of genes involved in mitochondrial function and synthesis of ATP. When superimposed by uremia, these PTHcre;Bmal1flox/flox mice had an increased parathyroid cell proliferative response, compared to wild type mice. Thus, our findings indicate a role of the internal parathyroid circadian clock in the development of parathyroid gland hyperplasia in uremia.
Collapse
Affiliation(s)
- Søren Egstrand
- Nephrological Department B, Herlev Hospital, University of Copenhagen, Denmark; Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Maria Lerche Mace
- Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Marya Morevati
- Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Anders Nordholm
- Nephrological Department B, Herlev Hospital, University of Copenhagen, Denmark; Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Lars Henning Engelholm
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark; Finsen Laboratory, University of Copenhagen, Denmark
| | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Tally Naveh-Many
- Minerva Center for Calcium and Bone Metabolism, Nephrology Services, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yuliu Guo
- Department of Genomic Medicine, Rigshospitalet, Centre of Diagnostics, Copenhagen, Denmark
| | - Klaus Olgaard
- Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark
| | - Ewa Lewin
- Nephrological Department B, Herlev Hospital, University of Copenhagen, Denmark; Nephrological Department P, Rigshospitalet, University of Copenhagen, Denmark.
| |
Collapse
|
32
|
Rapti G. Open Frontiers in Neural Cell Type Investigations; Lessons From Caenorhabditis elegans and Beyond, Toward a Multimodal Integration. Front Neurosci 2022; 15:787753. [PMID: 35321480 PMCID: PMC8934944 DOI: 10.3389/fnins.2021.787753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Nervous system cells, the building blocks of circuits, have been studied with ever-progressing resolution, yet neural circuits appear still resistant to schemes of reductionist classification. Due to their sheer numbers, complexity and diversity, their systematic study requires concrete classifications that can serve reduced dimensionality, reproducibility, and information integration. Conventional hierarchical schemes transformed through the history of neuroscience by prioritizing criteria of morphology, (electro)physiological activity, molecular content, and circuit function, influenced by prevailing methodologies of the time. Since the molecular biology revolution and the recent advents in transcriptomics, molecular profiling gains ground toward the classification of neurons and glial cell types. Yet, transcriptomics entails technical challenges and more importantly uncovers unforeseen spatiotemporal heterogeneity, in complex and simpler nervous systems. Cells change states dynamically in space and time, in response to stimuli or throughout their developmental trajectory. Mapping cell type and state heterogeneity uncovers uncharted terrains in neurons and especially in glial cell biology, that remains understudied in many aspects. Examining neurons and glial cells from the perspectives of molecular neuroscience, physiology, development and evolution highlights the advantage of multifaceted classification schemes. Among the amalgam of models contributing to neuroscience research, Caenorhabditis elegans combines nervous system anatomy, lineage, connectivity and molecular content, all mapped at single-cell resolution, and can provide valuable insights for the workflow and challenges of the multimodal integration of cell type features. This review reflects on concepts and practices of neuron and glial cells classification and how research, in C. elegans and beyond, guides nervous system experimentation through integrated multidimensional schemes. It highlights underlying principles, emerging themes, and open frontiers in the study of nervous system development, regulatory logic and evolution. It proposes unified platforms to allow integrated annotation of large-scale datasets, gene-function studies, published or unpublished findings and community feedback. Neuroscience is moving fast toward interdisciplinary, high-throughput approaches for combined mapping of the morphology, physiology, connectivity, molecular function, and the integration of information in multifaceted schemes. A closer look in mapped neural circuits and understudied terrains offers insights for the best implementation of these approaches.
Collapse
|
33
|
Liang J, Qian J, Yang L, Chen X, Wang X, Lin X, Wang X, Zhao B. Modeling Human Thyroid Development by Fetal Tissue-Derived Organoid Culture. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105568. [PMID: 35064652 PMCID: PMC8948548 DOI: 10.1002/advs.202105568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/06/2022] [Indexed: 05/29/2023]
Abstract
Euthyroidism is of profound importance for lifetime health. However, the early diagnosis or therapeutics of thyroid developmental defects has not been established, mainly due to limited understanding of human thyroid development and a lack of recapitulating research model. Herein, the authors elaborate the cell atlas and potential regulatory signaling of the evolution of heterogeneous thyrocyte population from 12 to 16 gestational weeks. Moreover, they establish a long-term culture of human fetal thyroid organoids (hFTOs) system, which retains the fetal thyroid lineages and molecular signatures, as well as the ability to generate functional human thyroid follicles post mice renal transplantation. Notably, cAMP signaling activation in hFTOs by forskolin boosts the maturation of follicle and thus thyroid hormone T4 secretion, which recapitulates the key developmental events of fetal thyroid. Employing this ex vivo system, it is found that enhanced chromatin accessibility at thyroid maturation genes (such as TPO and TG) loci permits the transcription for hormone production. This study provides the cell atlas of and an organoid model for human thyroid development, which will facilitate thyroid research and prospective medicine.
Collapse
Affiliation(s)
- Jianqing Liang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| | - Jun Qian
- State Key Laboratory of Medical Molecular BiologyDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijing100730China
| | - Li Yang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| | - Xiaojun Chen
- Obstetrics and Gynecology Hospital of Fudan UniversityShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai200011China
| | - Xiaoning Wang
- School of Laboratory Medicine and BiotechnologySouthern Medical UniversitySchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510000China
| | - Xinhua Lin
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| | - Xiaoyue Wang
- State Key Laboratory of Medical Molecular BiologyDepartment of Biochemistry and Molecular BiologyInstitute of Basic Medical Sciences Chinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijing100730China
| | - Bing Zhao
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteZhongshan HospitalFudan UniversityShanghai200438China
| |
Collapse
|
34
|
Development of Surgically Transplantable Parathyroid Hormone-Releasing Microbeads. Biomedicines 2022; 10:biomedicines10020440. [PMID: 35203648 PMCID: PMC8962264 DOI: 10.3390/biomedicines10020440] [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: 12/16/2021] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 01/27/2023] Open
Abstract
Hypoparathyroidism is an endocrine disorder that occurs because of the inability to produce parathyroid hormone (PTH) effectively. Previously, we reported the efficacy of tonsil-derived mesenchymal stem cells (TMSCs) differentiated into parathyroid-like cells for the treatment of hypoparathyroidism. Here, we investigated the feasibility of three-dimensional structural microbeads fabricated with TMSCs and alginate, a natural biodegradable polymer, to treat hypoparathyroidism. Alginate microbeads were fabricated by dropping a 2% (w/v) alginate solution containing TMSCs into a 5% CaCl2 solution and then differentiated into parathyroid-like cells using activin A and sonic hedgehog for 7 days. The protein expression of PTH, a specific marker of the parathyroid gland, was significantly higher in differentiated alginate microbeads with TMSCs (Al-dT) compared with in undifferentiated alginate microbeads with TMSCs. For in vivo experiments, we created the hypoparathyroidism animal model by parathyroidectomy (PTX) and implanted alginate microbeads in the dorsal interscapular region. The PTX rats with Al-dT (PTX+Al-dT) showed the highest survival rate and weight change and a gradual increase in serum intact PTH levels. We also detected a higher expression of PTH in retrieved tissues of PTX+Al-dT using immunofluorescence analysis. This study demonstrates that alginate microbeads are potential a new tool as a surgically scalable therapy for treating hypoparathyroidism.
Collapse
|
35
|
Canaff L, Guarnieri V, Kim Y, Wong BYL, Nolin-Lapalme A, Cole DEC, Minisola S, Eller-Vainicher C, Cetani F, Repaci A, Turchetti D, Corbetta S, Scillitani A, Goltzman D. Novel Glial Cells Missing-2 (GCM2) variants in parathyroid disorders. Eur J Endocrinol 2022; 186:351-366. [PMID: 35038313 DOI: 10.1016/10.1530/eje-21-0433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 01/13/2022] [Indexed: 05/25/2023]
Abstract
OBJECTIVE The aim of this study was to analyze variants of the gene glial cells missing-2 (GCM2), encoding a parathyroid cell-specific transcription factor, in familial hypoparathyroidism and in familial isolated hyperparathyroidism (FIHP) without and with parathyroid carcinoma. DESIGN We characterized 2 families with hypoparathyroidism and 19 with FIHP in which we examined the mechanism of action of GCM2 variants. METHODS Leukocyte DNA of hypoparathyroid individuals was Sanger sequenced for CASR, PTH, GNA11 and GCM2 mutations. DNA of hyperparathyroid individuals underwent MEN1, CDKN1B, CDC73, CASR, RET and GCM2 sequencing. The actions of identified GCM2 variants were evaluated by in vitro functional analyses. RESULTS A novel homozygous p.R67C GCM2 mutation which failed to stimulate transcriptional activity in a luciferase assay was identified in affected members of two hypoparathyroid families. Oligonucleotide pull-down assay and in silico structural modeling indicated that this mutant had lost the ability to bind the consensus GCM recognition sequence of DNA. Two novel (p.I383M and p.T386S) and one previously reported (p.Y394S) heterozygous GCM2 variants that lie within a C-terminal conserved inhibitory domain were identified in three affected individuals of the hyperparathyroid families. One family member, heterozygous for p.I138M, had parathyroid carcinoma (PC), and a heterozygous p.V382M variant was found in another patient affected by sporadic PC. These variants exerted significantly enhanced in vitrotranscriptional activity, including increased stimulation of the PTH promoter. CONCLUSIONS We provide evidence that two novel GCM2 R67C inactivating mutations with an inability to bind DNA are causative of hypoparathyroidism. Additionally, we provide evidence that two novel GCM2 variants increased transactivation of the PTH promoter in vitro and are associated with FIHP. Furthermore, our studies suggest that activating GCM2 variants may contribute to facilitating more aggressive parathyroid disease.
Collapse
Affiliation(s)
- Lucie Canaff
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | - Vito Guarnieri
- Division of Medical Genetics and Unit of Endocrinology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Yoojung Kim
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | - Betty Y L Wong
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Alexis Nolin-Lapalme
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | - David E C Cole
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Salvatore Minisola
- Department of Internal Medicine and Medical Disciplines, 'Sapienza' Rome University, Rome, Italy
| | - Cristina Eller-Vainicher
- Department of Medical Sciences and Community, Fondazione Ca'Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Andrea Repaci
- Unit of Endocrinology, S. Orsola Malpighi Hospital, Bologna, Italy
| | - Daniela Turchetti
- Center for the Studies of Hereditary Cancers, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Sabrina Corbetta
- Endocrinology and Diabetology Service, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Alfredo Scillitani
- Division of Medical Genetics and Unit of Endocrinology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - David Goltzman
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
36
|
Canaff L, Guarnieri V, Kim Y, Wong BYL, Nolin-Lapalme A, Cole DEC, Minisola S, Eller-Vainicher C, Cetani F, Repaci A, Turchetti D, Corbetta S, Scillitani A, Goltzman D. Novel Glial Cells Missing-2 (GCM2) variants in parathyroid disorders. Eur J Endocrinol 2022; 186:351-366. [PMID: 35038313 PMCID: PMC8859918 DOI: 10.1530/eje-21-0433] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 01/13/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The aim of this study was to analyze variants of the gene glial cells missing-2 (GCM2), encoding a parathyroid cell-specific transcription factor, in familial hypoparathyroidism and in familial isolated hyperparathyroidism (FIHP) without and with parathyroid carcinoma. DESIGN We characterized 2 families with hypoparathyroidism and 19 with FIHP in which we examined the mechanism of action of GCM2 variants. METHODS Leukocyte DNA of hypoparathyroid individuals was Sanger sequenced for CASR, PTH, GNA11 and GCM2 mutations. DNA of hyperparathyroid individuals underwent MEN1, CDKN1B, CDC73, CASR, RET and GCM2 sequencing. The actions of identified GCM2 variants were evaluated by in vitro functional analyses. RESULTS A novel homozygous p.R67C GCM2 mutation which failed to stimulate transcriptional activity in a luciferase assay was identified in affected members of two hypoparathyroid families. Oligonucleotide pull-down assay and in silico structural modeling indicated that this mutant had lost the ability to bind the consensus GCM recognition sequence of DNA. Two novel (p.I383M and p.T386S) and one previously reported (p.Y394S) heterozygous GCM2 variants that lie within a C-terminal conserved inhibitory domain were identified in three affected individuals of the hyperparathyroid families. One family member, heterozygous for p.I138M, had parathyroid carcinoma (PC), and a heterozygous p.V382M variant was found in another patient affected by sporadic PC. These variants exerted significantly enhanced in vitrotranscriptional activity, including increased stimulation of the PTH promoter. CONCLUSIONS We provide evidence that two novel GCM2 R67C inactivating mutations with an inability to bind DNA are causative of hypoparathyroidism. Additionally, we provide evidence that two novel GCM2 variants increased transactivation of the PTH promoter in vitro and are associated with FIHP. Furthermore, our studies suggest that activating GCM2 variants may contribute to facilitating more aggressive parathyroid disease.
Collapse
Affiliation(s)
- Lucie Canaff
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | - Vito Guarnieri
- Division of Medical Genetics and Unit of Endocrinology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Yoojung Kim
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | - Betty Y L Wong
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Alexis Nolin-Lapalme
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
| | - David E C Cole
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Salvatore Minisola
- Department of Internal Medicine and Medical Disciplines, ‘Sapienza’ Rome University, Rome, Italy
| | - Cristina Eller-Vainicher
- Department of Medical Sciences and Community, Fondazione Ca’Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Andrea Repaci
- Unit of Endocrinology, S. Orsola Malpighi Hospital, Bologna, Italy
| | - Daniela Turchetti
- Center for the Studies of Hereditary Cancers, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Sabrina Corbetta
- Endocrinology and Diabetology Service, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Alfredo Scillitani
- Division of Medical Genetics and Unit of Endocrinology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - David Goltzman
- Metabolic Complications and Disorders, Research Institute-McGill University Health Centre, Montreal, Quebec, Canada
- Correspondence should be addressed to D Goltzman;
| |
Collapse
|
37
|
Stenhouse C, Suva LJ, Gaddy D, Wu G, Bazer FW. Phosphate, Calcium, and Vitamin D: Key Regulators of Fetal and Placental Development in Mammals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1354:77-107. [PMID: 34807438 DOI: 10.1007/978-3-030-85686-1_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Normal calcium and bone homeostasis in the adult is virtually fully explained by the interactions of several key regulatory hormones, including parathyroid hormone, 1,25 dihydroxy vitamin D3, fibroblast growth factor-23, calcitonin, and sex steroids (estradiol and testosterone). In utero, bone and mineral metabolism is regulated differently from the adult. During development, it is the placenta and not the fetal kidneys, intestines, or skeleton that is the primary source of minerals for the fetus. The placenta is able to meet the almost inexhaustible needs of the fetus for minerals by actively driving the transport of calcium and phosphorus from the maternal circulation to the growing fetus. These fundamentally important minerals are maintained in the fetal circulation at higher concentrations than those in maternal blood. Maintenance of these inordinately higher fetal levels is necessary for the developing skeleton to accrue sufficient minerals by term. Importantly, in livestock species, prenatal mineralization of the skeleton is crucial for the high levels of offspring activity soon after birth. Calcium is required for mineralization, as well as a plethora of other physiological functions. Placental calcium and phosphate transport are regulated by several mechanisms that are discussed in this review. It is clear that phosphate and calcium metabolism is intimately interrelated and, therefore, placental transport of these minerals cannot be considered in isolation.
Collapse
Affiliation(s)
- Claire Stenhouse
- Departments of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Larry J Suva
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Dana Gaddy
- Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Guoyao Wu
- Departments of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Fuller W Bazer
- Department of Animal Science, Kleberg Center, Texas A&M University, College Station, TX, 77843-2471, USA.
| |
Collapse
|
38
|
Berger JM, Karsenty G. Osteocalcin and the Physiology of Danger. FEBS Lett 2021; 596:665-680. [PMID: 34913486 PMCID: PMC9020278 DOI: 10.1002/1873-3468.14259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/02/2022]
Abstract
Bone biology has long been driven by the question as to what molecules affect cell differentiation or the functions of bone. Exploring this issue has been an extraordinarily powerful way to improve our knowledge of bone development and physiology. More recently, a second question has emerged: does bone have other functions besides making bone? Addressing this conundrum revealed that the bone-derived hormone osteocalcin affects a surprisingly large number of organs and physiological processes, including acute stress response. This review will focus on this emerging aspect of bone biology taking osteocalcin as a case study and will show how classical and endocrine functions of bone help to define a new functional identity for this tissue.
Collapse
Affiliation(s)
- Julian Meyer Berger
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, NY, 10032, USA
| | - Gerard Karsenty
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, NY, 10032, USA
| |
Collapse
|
39
|
Singh P, Bhadada SK, Dahiya D, Saikia UN, Arya AK, Sachdeva N, Kaur J, Behera A, Brandi ML, Rao SD. GCM2 Silencing in Parathyroid Adenoma Is Associated With Promoter Hypermethylation and Gain of Methylation on Histone 3. J Clin Endocrinol Metab 2021; 106:e4084-e4096. [PMID: 34077544 PMCID: PMC8475237 DOI: 10.1210/clinem/dgab374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT Glial cells missing 2 (GCM2), a zinc finger-transcription factor, is essentially required for the development of the parathyroid glands. OBJECTIVE We sought to identify whether the epigenetic alterations in GCM2 transcription are involved in the pathogenesis of sporadic parathyroid adenoma. In addition, we examined the association between promoter methylation and histone modifications with disease indices. METHODS Messenger RNA (mRNA) and protein expression of GCM2 were analyzed by reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry in 33 adenomatous and 10 control parathyroid tissues. DNA methylation and histone methylation/acetylation of the GCM2 promoter were measured by bisulfite sequencing and chromatin immunoprecipitation-qPCR. Additionally, we investigated the role of epigenetic modifications on GCM2 and DNA methyltransferase 1 (DNMT1) expression in parathyroid (PTH)-C1 cells by treating with 5-aza-2'-deoxycytidine (DAC) and BRD4770 and assessed for GCM2 mRNA and DNMT1 protein levels. RESULTS mRNA and protein expression of GCM2 were lower in sporadic adenomatous than in control parathyroid tissues. This reduction correlated with hypermethylation (P < .001) and higher H3K9me3 levels in the GCM2 promoter (P < .04) in adenomas. In PTH-C1 cells, DAC treatment resulted in increased GCM2 transcription and decreased DNMT1 protein expression, while cells treated with the BRD4770 showed reduced H3K9me3 levels but a nonsignificant change in GCM2 transcription. CONCLUSION These findings suggest the concurrent association of promoter hypermethylation and higher H3K9me3 with the repression of GCM2 expression in parathyroid adenomas. Treatment with DAC restored GCM2 expression in PTH-C1 cells. Our results showed a possible epigenetic landscape in the tumorigenesis of parathyroid adenoma and also that DAC may be a promising avenue of research for parathyroid adenoma therapeutics.
Collapse
Affiliation(s)
- Priyanka Singh
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Divya Dahiya
- Department of General Surgery, PGIMER, Chandigarh, 160012, India
| | | | - Ashutosh Kumar Arya
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Jyotdeep Kaur
- Department of Biochemistry, PGIMER, Chandigarh, 160012, India
| | - Arunanshu Behera
- Department of General Surgery, PGIMER, Chandigarh, 160012, India
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50121, Italy
| | - Sudhaker Dhanwada Rao
- Bone and Mineral Research Laboratory, Henry Ford Hospital, Detroit, Michigan 48202, USA
| |
Collapse
|
40
|
Arnold A, Dennison E, Kovacs CS, Mannstadt M, Rizzoli R, Brandi ML, Clarke B, Thakker RV. Hormonal regulation of biomineralization. Nat Rev Endocrinol 2021; 17:261-275. [PMID: 33727709 DOI: 10.1038/s41574-021-00477-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2021] [Indexed: 01/31/2023]
Abstract
Biomineralization is the process by which organisms produce mineralized tissues. This crucial process makes possible the rigidity and flexibility that the skeleton needs for ambulation and protection of vital organs, and the hardness that teeth require to tear and grind food. The skeleton also serves as a source of mineral in times of short supply, and the intestines absorb and the kidneys reclaim or excrete minerals as needed. This Review focuses on physiological and pathological aspects of the hormonal regulation of biomineralization. We discuss the roles of calcium and inorganic phosphate, dietary intake of minerals and the delicate balance between activators and inhibitors of mineralization. We also highlight the importance of tight regulation of serum concentrations of calcium and phosphate, and the major regulators of biomineralization: parathyroid hormone (PTH), the vitamin D system, vitamin K, fibroblast growth factor 23 (FGF23) and phosphatase enzymes. Finally, we summarize how developmental stresses in the fetus and neonate, and in the mother during pregnancy and lactation, invoke alternative hormonal regulatory pathways to control mineral delivery, skeletal metabolism and biomineralization.
Collapse
Affiliation(s)
- Andrew Arnold
- Division of Endocrinology & Metabolism and Center for Molecular Oncology, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Elaine Dennison
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Christopher S Kovacs
- Faculty of Medicine - Endocrinology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Michael Mannstadt
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - René Rizzoli
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Maria Luisa Brandi
- Department of Biochemical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Bart Clarke
- Mayo Clinic Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Rochester, MN, USA
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Caprio C, Lania G, Bilio M, Ferrentino R, Chen L, Baldini A. EZH2 is required for parathyroid and thymic development through differentiation of the third pharyngeal pouch endoderm. Dis Model Mech 2021; 14:dmm.046789. [PMID: 33608392 PMCID: PMC7969367 DOI: 10.1242/dmm.046789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
The Ezh2 gene encodes a histone methyltransferase of the polycomb repressive complex 2 that methylates histone H3 lysine 27. In this study, we investigated whether EZH2 has a role in the development of the pharyngeal apparatus and whether it regulates the expression of the Tbx1 gene, which encodes a key transcription factor required in pharyngeal development. To these ends, we performed genetic in vivo experiments with mouse embryos and used mouse embryonic stem cell (ESC)-based protocols to probe endoderm and cardiogenic mesoderm differentiation. Results showed that EZH2 occupies the Tbx1 gene locus in mouse embryos, and that suppression of EZH2 was associated with reduced expression of Tbx1 in differentiated mouse ESCs. Conditional deletion of Ezh2 in the Tbx1 expression domain, which includes the pharyngeal endoderm, did not cause cardiac defects but revealed that the gene has an important role in the morphogenesis of the third pharyngeal pouch (PP). We found that in conditionally deleted embryos the third PP was hypoplastic, had reduced expression of Tbx1, lacked the expression of Gcm2, a gene that marks the parathyroid domain, but expressed FoxN1, a gene marking the thymic domain. Consistently, the parathyroids did not develop, and the thymus was hypoplastic. Thus, Ezh2 is required for parathyroid and thymic development, probably through a function in the pouch endoderm. This discovery also provides a novel interpretational key for the finding of Ezh2 activating mutations in hyperparathyroidism and parathyroid cancer. Summary: EZH2 in the pharyngeal apparatus is required in the endoderm for the development of the parathyroids and thymus, and positively regulates the expression of the Tbx1 gene.
Collapse
Affiliation(s)
- Cinzia Caprio
- Institute of Genetics and Biophysics, CNR, Naples, Italy
| | | | - Marchesa Bilio
- Institute of Genetics and Biophysics, CNR, Naples, Italy
| | | | - Li Chen
- Department of Biology and Biochemistry, University of Houston, TX
| | - Antonio Baldini
- Institute of Genetics and Biophysics, CNR, Naples, Italy .,Department of Molecular Medicine and Medical Biotechnologies, University Federico II, Naples, Italy
| |
Collapse
|
42
|
Five patients with disorders of calcium metabolism presented with GCM2 gene variants. Sci Rep 2021; 11:2968. [PMID: 33536578 PMCID: PMC7859196 DOI: 10.1038/s41598-021-82661-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
The GCM2 gene encodes a transcription factor predominantly expressed in parathyroid cells that is known to be critical for development, proliferation and maintenance of the parathyroid cells. A cohort of 127 Spanish patients with a disorder of calcium metabolism were screened for mutations by Next-Generation Sequencing (NGS). A targeted panel for disorders of calcium and phosphorus metabolism was designed to include 65 genes associated with these disorders. We observed two variants of uncertain significance (p.(Ser487Phe) and p.Asn315Asp), one likely pathogenic (p.Val382Met) and one benign variant (p.Ala393_Gln395dup) in the GCM2 gene in the heterozygous state in five families (two index cases had hypocalcemia and hypoparathyroidism, respectively, and three index cases had primary hyperparathyroidism). Our study shows the utility of NGS in unravelling the genetic origin of some disorders of the calcium and phosphorus metabolism, and confirms the GCM2 gene as an important element for the maintenance of calcium homeostasis. Importantly, a novel variant in the GCM2 gene (p.(Ser487Phe)) has been found in a patient with hypocalcemia.
Collapse
|
43
|
Zhang H, Zhang Y, Qiu C, Zhu W, Wen M, Lao X. Differentiation of human umbilical cord mesenchymal stem cells into parathyroid cells by editing the PTH gene with the CRISPR/Cas9 system. BIOTECHNOL BIOTEC EQ 2021. [DOI: 10.1080/13102818.2021.1961608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Affiliation(s)
- Huiting Zhang
- Department of General Surgery, Zhaoqing First People’s Hospital, Zhaoqing, Guangdong, PR China
| | - Yiting Zhang
- Department of General Surgery, Zhaoqing First People’s Hospital, Zhaoqing, Guangdong, PR China
| | - Changhong Qiu
- Department of General Surgery, Zhaoqing First People’s Hospital, Zhaoqing, Guangdong, PR China
| | - Wentian Zhu
- Department of General Surgery, Zhaoqing First People’s Hospital, Zhaoqing, Guangdong, PR China
| | - Mingbo Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, PR China
| | - Xuejun Lao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, PR China
| |
Collapse
|
44
|
Asmat MW, Ramzan MH, Ramzan F. Exogenous Neurokinin B Administration May Have a Strong Effect on Negative Feedback Loop of Hypothalamic Pituitary Thyroid Axis. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09925-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
45
|
Rivas VN, Magdesian KG, Fagan S, Slovis NM, Luethy D, Javsicas LH, Caserto BG, Miller AD, Dahlgren AR, Peterson J, Hales EN, Peng S, Watson KD, Khokha MK, Finno CJ. A nonsense variant in Rap Guanine Nucleotide Exchange Factor 5 (RAPGEF5) is associated with equine familial isolated hypoparathyroidism in Thoroughbred foals. PLoS Genet 2020; 16:e1009028. [PMID: 32986719 PMCID: PMC7544121 DOI: 10.1371/journal.pgen.1009028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/08/2020] [Accepted: 08/05/2020] [Indexed: 11/18/2022] Open
Abstract
Idiopathic hypocalcemia in Thoroughbred (TB) foals causes tetany and seizures and is invariably fatal. Based upon the similarity of this disease with human familial hypoparathyroidism and occurrence only in the TB breed, we conducted a genetic investigation on two affected TB foals. Familial hypoparathyroidism was identified, and pedigree analysis suggested an autosomal recessive (AR) mode of inheritance. We performed whole-genome sequencing of the two foals, their unaffected dams and four unaffected, unrelated TB horses. Both homozygosity mapping and an association analysis were used to prioritize potential genetic variants. Of the 2,808 variants that significantly associated with the phenotype using an AR mode of inheritance (P<0.02) and located within a region of homozygosity, 1,507 (54%) were located in a 9.7 Mb region on chr4 (44.9-54.6 Mb). Within this region, a nonsense variant (RAPGEF5 c.2624C>A,p.Ser875*) was significantly associated with the hypoparathyroid phenotype (Pallelic = 0.008). Affected foals were homozygous for the variant, with two additional affected foals subsequently confirmed in 2019. Necropsies of all affected foals failed to identify any histologically normal parathyroid glands. Because the nonsense mutation in RAPGEF5 was near the C-terminal end of the protein, the impact on protein function was unclear. Therefore, we tested the variant in our Xenopus overexpression model and demonstrated RAPGEF5 loss-of-function. This RAPGEF5 variant represents the first genetic variant for hypoparathyroidism identified in any domestic animal species.
Collapse
Affiliation(s)
- Victor N. Rivas
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, California, United States of America
| | - K. Gary Magdesian
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, California, United States of America
| | - Sophia Fagan
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale School of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Nathan M. Slovis
- Hagyard Equine Medical Hospital, Lexington, Kentucky, United States of America
| | - Daniela Luethy
- Department of Clinical Studies–New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Laura H. Javsicas
- Rhinebeck Equine L.L.P., Rhinebeck, New York, United States of America
| | | | - Andrew D. Miller
- Department of Biomedical Sciences, Section of Anatomic Pathology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Anna R. Dahlgren
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, California, United States of America
| | - Janel Peterson
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, California, United States of America
| | - Erin N. Hales
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, California, United States of America
| | - Sichong Peng
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, California, United States of America
| | - Katherine D. Watson
- Department of Anatomic Pathology, Veterinary Medical Teaching Hospital, University of California-Davis, Davis, California, United States of America
| | - Mustafa K. Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale School of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Carrie J. Finno
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California-Davis, Davis, California, United States of America
| |
Collapse
|
46
|
Thymus Inception: Molecular Network in the Early Stages of Thymus Organogenesis. Int J Mol Sci 2020; 21:ijms21165765. [PMID: 32796710 PMCID: PMC7460828 DOI: 10.3390/ijms21165765] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 11/17/2022] Open
Abstract
The thymus generates central immune tolerance by producing self-restricted and self-tolerant T-cells as a result of interactions between the developing thymocytes and the stromal microenvironment, mainly formed by the thymic epithelial cells. The thymic epithelium derives from the endoderm of the pharyngeal pouches, embryonic structures that rely on environmental cues from the surrounding mesenchyme for its development. Here, we review the most recent advances in our understanding of the molecular mechanisms involved in early thymic organogenesis at stages preceding the expression of the transcription factor Foxn1, the early marker of thymic epithelial cells identity. Foxn1-independent developmental stages, such as the specification of the pharyngeal endoderm, patterning of the pouches, and thymus fate commitment are discussed, with a special focus on epithelial–mesenchymal interactions.
Collapse
|
47
|
Uchiyama T, Ohkido I, Nakashima A, Saito Y, Okabe M, Yokoo T. Severe chronic kidney disease environment reduced calcium-sensing receptor expression in parathyroid glands of adenine-induced rats even without high phosphorus diet. BMC Nephrol 2020; 21:219. [PMID: 32517664 PMCID: PMC7285719 DOI: 10.1186/s12882-020-01880-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/02/2020] [Indexed: 02/08/2023] Open
Abstract
Background Chronic kidney disease (CKD) disrupts mineral homeostasis and its main underlying cause is secondary hyperparathyroidism (SHPT). We previously reported that calcium-sensing receptor (CaSR) mRNA and protein expression in parathyroid glands (PTGs) significantly decreased in a CKD rat model induced by a 5/6 nephrectomy that were fed a high phosphorus diet. However, there was a significant difference in the severity of CKD between high phosphorus and adequate phosphorus diet groups. Thus, it was unclear whether CKD environment or the high phosphorus diet influenced CaSR expression, and the underlying mechanism remains largely unknown. Methods CKD was induced in rats with 0.75% adenine-containing diet. CKD and control rats were maintained for 5 days and 2 weeks on diets with 0.7% or 1.3% phosphorus. For gene expression analysis, quantitative real-time polymerase chain reaction was performed with TaqMan probes. Protein expression was analyzed by immunohistochemistry. Results PTG CaSR expression significantly decreased in the presence of a severe CKD environment, even without the high phosphate load. Ki67 expressing cells in PTGs were significantly higher only in the CKD rats fed a high phosphorus diet. Furthermore, among the many genes that could affect CaSR expression, only vitamin D receptor (VDR) and glial cells missing 2 (Gcm2) showed significant changes. Moreover, Gcm2 was significantly reduced at an early stage without significant changes in serum calcium, phosphorus and 1,25(OH)2 vitamin D, and there was no significant reduction in CaSR and VDR expressions. Then, significantly elevated Ki67-positive cell numbers were also only observed in the early CKD PTGs with high-phosphorus diets. Conclusions Our data suggest that the cause of the decreased PTG CaSR expression is the reduction in VDR and Gcm2 expression; Gcm2 may play a role in the onset and progression of SHPT.
Collapse
Affiliation(s)
- Taketo Uchiyama
- Division of Nephrology and Hypertension, Department of Internal Medicine, the Jikei University School of Medicine, Tokyo, Japan
| | - Ichiro Ohkido
- Division of Nephrology and Hypertension, Department of Internal Medicine, the Jikei University School of Medicine, Tokyo, Japan.
| | - Akio Nakashima
- Division of Nephrology and Hypertension, Department of Internal Medicine, the Jikei University School of Medicine, Tokyo, Japan
| | - Yatsumu Saito
- Division of Nephrology and Hypertension, Department of Internal Medicine, the Jikei University School of Medicine, Tokyo, Japan
| | - Masataka Okabe
- Department of Anatomy, the Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, the Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
48
|
Verdelli C, Tavanti GS, Corbetta S. Intratumor heterogeneity in human parathyroid tumors. Histol Histopathol 2020; 35:1213-1228. [PMID: 32468569 DOI: 10.14670/hh-18-230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parathyroid tumors are the second most common endocrine neoplasia after thyroid neoplasia. They are mostly associated with impaired parathormone (PTH) synthesis and release determining the metabolic and clinical condition of primary hyperparathyroidism (PHPT). PHPT is the third most prevalent endocrine disorder, mainly affecting postmenopausal women. Parathyroid benign tumors, both adenomas of a single gland or hyperplasia involving all the glands, are the main histotypes, occurring in more than 95% of PHPT cases. The differential diagnosis between benign and malignant parathyroid lesions is a challenge for clinicians. It relies on histologic features, which display significant overlap between the histotypes with different clinical outcomes. Parathyroid adenomas and hyperplasia have been considered so far as a unique monoclonal/polyclonal entity, while accumulating evidence suggest great heterogeneity. Intratumor parathyroid heterogeneity involves tumor cell type, as well as tumor cell function, in terms of PTH synthesis and secretion, and of expression patterns of membrane and nuclear receptors (calcium sensing receptor, vitamin D receptor, α-klotho receptor and others). Intratumor heterogeneity can also interfere with cell molecular biology, in regard to clonality, oncosuppressor gene expression (such as MEN1 and HRPT2/CDC73), transcription factors (GCM2, TBX1) and microRNA expression. Such heterogeneity is likely involved in the phenotypic variability of the parathyroid tumors, and it should be considered in the clinical management, though at present target therapies are not available, with the exception of the calcium sensing receptor agonists.
Collapse
Affiliation(s)
- C Verdelli
- Laboratory of Experimental Endocrinology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - G S Tavanti
- Laboratory of Experimental Endocrinology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - S Corbetta
- Endocrinology and Diabetology Service, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
49
|
Fabbri S, Zonefrati R, Galli G, Gronchi G, Perigli G, Borrelli A, Brandi ML. In Vitro Control of Genes Critical for Parathyroid Embryogenesis by Extracellular Calcium. J Endocr Soc 2020; 4:bvaa058. [PMID: 32666007 PMCID: PMC7326476 DOI: 10.1210/jendso/bvaa058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Background The expression of the parathyroid transcription factors, encoded by the genes GATA3, GCM2, and MAFB, persists after parathyroid morphogenesis. This suggests a role of these genes in the regulatory program that governs parathyroid function in the adult. Indeed, these 3 genes form a transcriptional cascade able to activate PTH gene expression. Materials and Methods Adult adenoma parathyroid tissues were put in primary cell culture to evaluate the messenger ribonucleic acid (mRNA) expression of the PTH gene, of the genes involved in the calcium regulatory signaling pathway (CaSR, GNA11, and AP2S1), and of the 3 genes (GATA3, GCM2, and MAFB) involved in the parathyroid morphogenesis in the presence of different extracellular calcium concentrations from 0.1 mM to 3.0 mM. Aim The aim of the study was to investigate whether different extracellular calcium conditions could control the expression of transcription factors critical for parathyroid embryogenesis. Results The results of the experiments showed that the mRNA expression of GATA3, GCM2, and MAFB genes follows the same response as the PTH gene to extracellular calcium concentrations, with the highest expression at low calcium (0.1 mM) and the lowest at high calcium (3.0 mM). Conversely, the genes involved in the calcium signaling in the parathyroid cells showed a variable response to the extracellular calcium concentrations, with the CaSR and GNA11 genes exhibiting a sensitivity to low calcium concentrations. Conclusions These findings indicate that transcription factors recognized for their role in parathyroid embryogenesis show a response to extracellular calcium later in adulthood that parallels the behavior of the PTH gene.
Collapse
Affiliation(s)
- Sergio Fabbri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Roberto Zonefrati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Gianna Galli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Giorgio Gronchi
- Department of Neuroscience, Psychology, Drug Research & Child Health, University of Florence, Florence, Italy
| | - Giuliano Perigli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andrea Borrelli
- Bariatric, General Surgery and Metabolic Department, Santa Maria Nuova Hospital, Florence, Italy
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
50
|
Cetani F, Pardi E, Aretini P, Saponaro F, Borsari S, Mazoni L, Apicella M, Civita P, La Ferla M, Caligo MA, Lessi F, Mazzanti CM, Torregossa L, Oppo A, Marcocci C. Whole exome sequencing in familial isolated primary hyperparathyroidism. J Endocrinol Invest 2020; 43:231-245. [PMID: 31486992 DOI: 10.1007/s40618-019-01107-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/29/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Familial isolated hyperparathyroidism (FIHP) is a rare inherited disease accounting for 1% of all cases of primary hyperparathyroidism (PHPT). It is genetically heterogeneous being associated with mutations in different genes, including MEN1, CDC73, CASR, and recently GCM2. The aim of the study was to further investigate the molecular pathogenesis in Italian FIHP kindreds. METHODS We used whole exome sequencing (WES) in the probands of seven unrelated FIHP kindreds. We carried out a separate family-based exome analysis in a large family characterized by the co-occurrence of PHPT with multiple tumors apparently unrelated to the disease. Selected variants were also screened in 18 additional FIHP kindreds. The clinical, biochemical, and pathological characteristics of the families were also investigated. RESULTS Three different variants in GCM2 gene were found in two families, but only one (p.Tyr394Ser), already been shown to be pathogenic in vitro, segregated with the disease. Six probands carried seven heterozygous missense mutations segregating with the disease in the FAT3, PARK2, HDAC4, ITPR2 and TBCE genes. A genetic variant in the APC gene co-segregating with PHPT (p.Val530Ala) was detected in a family whose affected relatives had additional tumors, including colonic polyposis. CONCLUSION We confirm the role of GCM2 germline mutations in the pathogenesis of FIHP, although at a lower rate than in the previous WES study. Further studies are needed to establish the prevalence and the role in the predisposition to FIHP of the novel variants in additional genes.
Collapse
Affiliation(s)
- F Cetani
- University Hospital of Pisa, Endocrine Unit 2, Via Paradisa 2, 56124, Pisa, Italy.
| | - E Pardi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - P Aretini
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - F Saponaro
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - S Borsari
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - L Mazoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - M Apicella
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - P Civita
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - M La Ferla
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - M A Caligo
- Molecular Genetics Unit, Department of Laboratory Medicine, University Hospital of Pisa, Pisa, Italy
| | - F Lessi
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - C M Mazzanti
- Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - L Torregossa
- Division of Surgical Pathology, University Hospital of Pisa, Pisa, Italy
| | - A Oppo
- Endocrinology Unit, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - C Marcocci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|