1
|
Thompson MD, Chidiac P, Jose PA, Hauser AS, Gorvin CM. Genetic variants of accessory proteins and G proteins in human genetic disease. Crit Rev Clin Lab Sci 2025; 62:113-134. [PMID: 39743506 PMCID: PMC11854058 DOI: 10.1080/10408363.2024.2431853] [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: 10/16/2023] [Revised: 01/14/2024] [Accepted: 11/16/2024] [Indexed: 01/04/2025]
Abstract
We present a series of three articles on the genetics and pharmacogenetics of G protein- coupled receptors (GPCR). In the first article, we discuss genetic variants of the G protein subunits and accessory proteins that are associated with human phenotypes; in the second article, we build upon this to discuss "G protein-coupled receptor (GPCR) gene variants and human genetic disease" and in the third article, we survey "G protein-coupled receptor pharmacogenomics". In the present article, we review the processes of ligand binding, GPCR activation, inactivation, and receptor trafficking to the membrane in the context of human genetic disease resulting from pathogenic variants of accessory proteins and G proteins. Pathogenic variants of the genes encoding G protein α and β subunits are examined in diverse phenotypes. Variants in the genes encoding accessory proteins that modify or organize G protein coupling have been associated with disease; these include the contribution of variants of the regulator of G protein signaling (RGS) to hypertension; the role of variants of activator of G protein signaling type III in phenotypes such as hypoxia; the contribution of variation at the RGS10 gene to short stature and immunological compromise; and the involvement of variants of G protein-coupled receptor kinases (GRKs), such as GRK4, in hypertension. Variation in genes that encode proteins involved in GPCR signaling are outlined in the context of the changes in structure and function that may be associated with human phenotypes.
Collapse
Affiliation(s)
- Miles D. Thompson
- Krembil Brain Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Peter Chidiac
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Pedro A. Jose
- Division of Renal Diseases & Hypertension, Departments of Medicine and Pharmacology/Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Alexander S. Hauser
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline M. Gorvin
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, West Midlands, UK
| |
Collapse
|
2
|
Jinnouchi T, Yoshimoto M, Hayashi M. Pseudohypoparathyroidism Type 1b with Digital Clubbing. Intern Med 2025:4760-24. [PMID: 39894498 DOI: 10.2169/internalmedicine.4760-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Pseudohypoparathyroidism (PHP) is a rare disorder characterized by resistance to parathyroid hormone (PTH). Albright's hereditary osteodystrophy (AHO) is a characteristic physical condition associated with PHP. We herein report a 33-year-old Japanese man diagnosed with PHP type 1b who presented with hypocalcemia, preserved PTH secretion, decreased urinary cAMP and phosphate excretion (Ellsworth-Howard test), deletion of exons 5-7 in STX16, hypomethylation of A/B-DMR and AS1-DMR in the guanine nucleotide binding protein alpha stimulating gene. The patient did not have AHO or other comorbidities but showed digital clubbing during childhood. Although digital clubbing is rarely associated with PHP, it may represent a phenotype of this disorder.
Collapse
Affiliation(s)
- Takanobu Jinnouchi
- Department of Endocrinology and Diabetes, NTT Medical Center Tokyo, Japan
| | - Mei Yoshimoto
- Department of Endocrinology and Diabetes, NTT Medical Center Tokyo, Japan
| | - Michio Hayashi
- Department of Endocrinology and Diabetes, NTT Medical Center Tokyo, Japan
| |
Collapse
|
3
|
Li D, Jan de Beur S, Hou C, Ruzhnikov MR, Seeley H, Cutting GR, Sheridan MB, Levine MA. Recurrent small variants in NESP55/NESPAS associated with broad GNAS methylation defects and pseudohypoparathyroidism type 1B. JCI Insight 2024; 9:e185874. [PMID: 39541438 DOI: 10.1172/jci.insight.185874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudohypoparathyroidism type 1B (PHP1B) is associated with epigenetic changes in the maternal allele of the imprinted GNAS gene that inhibit expression of the α subunit of Gs (Gsα), thereby leading to parathyroid hormone resistance in renal proximal tubule cells where expression of Gsα from the paternal GNAS allele is normally silent. Although all patients with PHP1B show loss of methylation for the exon A/B differentially methylated region (DMR), some patients with autosomal dominant PHP1B (AD-PHP1B) and most patients with sporadic PHP1B have additional methylation defects that affect the DMRs corresponding to exons XL, AS1, and NESP. Because the genetic defect is unknown in most of these patients, we sought to identify the underlying genetic basis for AD-PHP1B in 2 multigenerational families with broad GNAS methylation defects and negative clinical exomes. Genome sequencing identified small GNAS variants in each family that were also present in unrelated individuals with PHP1B in a replication cohort. Maternal transmission of one GNAS microdeletion showed reduced penetrance in some unaffected patients. Expression of AS transcripts was increased, and NESP was decreased, in cells from affected patients. These results suggest that the small deletion activated AS transcription, leading to methylation of the NESP DMR with consequent inhibition of NESP transcription, and thereby provide a potential mechanism for PHP1B.
Collapse
Affiliation(s)
- Dong Li
- Center for Applied Genomics, and
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Suzanne Jan de Beur
- Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | - Maura Rz Ruzhnikov
- Neurology and Neurological Sciences, Pediatrics, Division of Medical Genetics, and
| | - Hilary Seeley
- Division of Pediatric Endocrinology, Stanford University and Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Garry R Cutting
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Molly B Sheridan
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael A Levine
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Division of Endocrinology and Diabetes and The Center for Bone Health, The Children's Hospital of Philadelphia, and Department of Pediatrics University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
McMullan P, Maye P, Root SH, Yang Q, Edie S, Rowe D, Kalajzic I, Germain-Lee EL. Hair follicle-resident progenitor cells are a major cellular contributor to heterotopic subcutaneous ossifications in a mouse model of Albright hereditary osteodystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599506. [PMID: 38948860 PMCID: PMC11213030 DOI: 10.1101/2024.06.18.599506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Heterotopic ossifications (HOs) are the pathologic process by which bone inappropriately forms outside of the skeletal system. Despite HOs being a persistent clinical problem in the general population, there are no definitive strategies for their prevention and treatment due to a limited understanding of the cellular and molecular mechanisms contributing to lesion development. One disease in which the development of heterotopic subcutaneous ossifications (SCOs) leads to morbidity is Albright hereditary osteodystrophy (AHO). AHO is caused by heterozygous inactivation of GNAS, the gene that encodes the α-stimulatory subunit (Gαs) of G proteins. Previously, we had shown using our laboratory's AHO mouse model that SCOs develop around hair follicles (HFs). Here we show that SCO formation occurs due to inappropriate expansion and differentiation of HF-resident stem cells into osteoblasts. We also show in AHO patients and mice that Secreted Frizzled Related Protein 2 (SFRP2) expression is upregulated in regions of SCO formation and that elimination of Sfrp2 in male AHO mice exacerbates SCO development. These studies provide key insights into the cellular and molecular mechanisms contributing to SCO development and have implications for potential therapeutic modalities not only for AHO patients but also for patients suffering from HOs with other etiologies.
Collapse
Affiliation(s)
- Patrick McMullan
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT
| | - Peter Maye
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT
| | - Sierra H. Root
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT
| | - Qingfen Yang
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT
| | | | - David Rowe
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT
| | - Emily L. Germain-Lee
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT
- Albright Center, Division of Endocrinology & Diabetes, Connecticut Children’s, Farmington, CT
| |
Collapse
|
5
|
Expósito Raspeño M, Sánchez Escudero V, Pérez de Nanclares Leal G, Ortiz Santamaría M, Sánchez-Dehesa Sáez R, García Cuartero B, González Vergaz A. Diagnosis and approach of pseudohypoparathyroidism type 1A and related disorders during long term follow-up: a case report. J Pediatr Endocrinol Metab 2024; 37:289-295. [PMID: 38353264 DOI: 10.1515/jpem-2023-0454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/03/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVES Pseudohypoparathyroidism type 1A (PHP1A) encompasses the association of resistance to multiple hormones, features of Albright hereditary osteodystrophy and decreased Gsα activity. Little is known about the early signs of PHP1A, with a delay in diagnosis. We report two PHP1A cases and their clinical and biochemical findings during a 20-year follow-up. CASE PRESENTATION Clinical suspicion was based on obesity, TSH resistance and ectopic ossifications which appeared several months before PTH resistance, at almost 3 years of age. Treatment with levothyroxine, calcitriol and calcium was required in both patients. DNA sequencing of GNAS gene detected a heterozygous pathogenic variant within exon 7 (c.569_570delAT) in patient one and a deletion from XLAS to GNAS-exon 5 on the maternal allele in patient 2. In patient 1, ectopic ossifications that required surgical excision were found. Noticeably, patient 2 displayed adult short stature, intracranial calcifications and psychomotor delay. In terms of weight, despite early diagnosis of obesity, dietary measures were established successfully in both cases. CONCLUSIONS GNAS mutations should be considered in patients with obesity, ectopic ossifications and TSH resistance presented in early infancy. These cases emphasize the highly heterogeneous clinical picture PHP1A patients may present, especially in terms of final height and cognitive impairment.
Collapse
|
6
|
Sakran WA, Al-Qahtani M, Alkhalifa M, Alqahtani A. Infant With Pseudohypoparathyroidism Type 1a, Misdiagnosed as Congenital Hypothyroidism. Med Arch 2023; 77:70-73. [PMID: 36923733 PMCID: PMC10010685 DOI: 10.5455/medarh.2023.77.70-73] [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: 12/10/2022] [Accepted: 02/12/2023] [Indexed: 03/09/2023] Open
Abstract
Background Hypothyroidism is a manifestation of multi-hormonal resistance in pseudohypoparathyroidism type Ia (PHP Ia). Objective The aim of this article was to present 9 months old male patient as case of congenital hypothyroidism. Case report We describe a 9 months old male diagnosed with congenital hypothyroidism at age 1.5 month, who developed later (at age 5 months) cyanotic attack associated with hypocalcaemia, hyperphosphatemia, and hyperparathyroidism, patient had typical characters of AHO, so the diagnosis of Pseudohypoparathyroidism 1a associated with resistance (TSH) was established. Conclusion Children diagnosed with PHP 1a should be further evaluated for associated resistance endocrinopathies. The literature on pseudohypoparathyroidism is reviewed with special emphasis on the misdiagnosis with congenital hypothyroidism.
Collapse
Affiliation(s)
- Wessal Al Sakran
- Pediatric Department, Security Forces Hospital, Dammam, Saudi Arabia
| | - Mohammad Al-Qahtani
- Pediatric Department, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammed Alkhalifa
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ali Alqahtani
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
7
|
Krishnan N, McMullan P, Yang Q, Buscarello AN, Germain-Lee EL. Prevalence of Chiari malformation type 1 is increased in pseudohypoparathyroidism type 1A and associated with aberrant bone development. PLoS One 2023; 18:e0280463. [PMID: 36662765 PMCID: PMC9858345 DOI: 10.1371/journal.pone.0280463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Albright hereditary osteodystrophy (AHO) is caused by heterozygous inactivating mutations in GNAS. Patients with maternally-inherited mutations develop pseudohypoparathyroidism type 1A (PHP1A) with multi-hormone resistance and aberrant craniofacial and skeletal development among other abnormalities. Chiari malformation type 1 (CM1), a condition in which brain tissue extends into the spinal canal when the skull is too small, has been reported in isolated cases of PHP1A. It has been hypothesized to be associated with growth hormone (GH) deficiency. Given the adverse clinical sequelae that can occur if CM1 goes unrecognized, we investigated the previously undetermined prevalence of CM1, as well as any potential correlations with GH status, given the known increased prevalence of GH deficiency in PHP1A. We also investigated these metrics for low lying cerebellar tonsils (LLCT), defined as tonsillar descent less than 5 mm below the foramen magnum. In addition, we investigated possible correlations of CM1/LLCT with advanced hand/wrist bone ages and craniofacial abnormalities known to occur in PHP1A to determine whether premature chondrocyte differentiation and/or aberrant craniofacial development could be potential etiologies of CM1/LLCT through both human studies and investigations of our AHO mouse model. METHODS We examined patients with PHP1A in our clinic and noticed CM1 more frequently than expected. Therefore, we set out to determine the true prevalence of CM1 and LLCT in a cohort of 54 mutation-confirmed PHP1A participants who had clinically-indicated brain imaging. We examined potential correlations with GH status, clinical features, biological sex, genotype, and hand/wrist bone age determinations. In addition, we investigated the craniofacial development in our mouse model of AHO (Gnas E1+/-m) by histologic analyses, dynamic histomorphometry, and micro-computerized tomographic imaging (MCT) in order to determine potential etiologies of CM1/LLCT in PHP1A. RESULTS In our cohort of PHP1A, the prevalence of CM1 is 10.8%, which is at least 10-fold higher than in the general population. If LLCT is included, the prevalence increases to 21.7%. We found no correlation with GH status, biological sex, genotype, or hand/wrist bone age. Through investigations of our Gnas E1+/-m mice, the correlate to PHP1A, we identified a smaller cranial vault and increased cranial dome angle with evidence of hyperostosis due to increased osteogenesis. We also demonstrated that there was premature closure of the spheno-occipital synchondrosis (SOS), a cartilaginous structure essential to the development of the cranial base. These findings lead to craniofacial abnormalities and could contribute to CM1 and LLCT development in PHP1A. CONCLUSION The prevalence of CM1 is at least 10-fold higher in PHP1A compared to the general population and 20-fold higher when including LLCT. This is independent of the GH deficiency that is found in approximately two-thirds of patients with PHP1A. In light of potential serious consequences of CM1, clinicians should have a low threshold for brain imaging. Investigations of our AHO mouse model revealed aberrant cranial formation including a smaller cranium, increased cranial dome angle, hyperostosis, and premature SOS closure rates, providing a potential etiology for the increased prevalence of CM1 and LLCT in PHP1A.
Collapse
Affiliation(s)
- Neetu Krishnan
- Department of Pediatrics, Division of Pediatric Endocrinology & Diabetes, University of Connecticut School of Medicine, Farmington, Connecticut, United States of America
- Albright Center, Division of Pediatric Endocrinology & Diabetes, Connecticut Children’s, Farmington, Connecticut, United States of America
| | - Patrick McMullan
- Department of Pediatrics, Division of Pediatric Endocrinology & Diabetes, University of Connecticut School of Medicine, Farmington, Connecticut, United States of America
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, Connecticut, United States of America
| | - Qingfen Yang
- Department of Pediatrics, Division of Pediatric Endocrinology & Diabetes, University of Connecticut School of Medicine, Farmington, Connecticut, United States of America
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, Connecticut, United States of America
| | - Alexzandrea N. Buscarello
- Albright Center, Division of Pediatric Endocrinology & Diabetes, Connecticut Children’s, Farmington, Connecticut, United States of America
| | - Emily L. Germain-Lee
- Department of Pediatrics, Division of Pediatric Endocrinology & Diabetes, University of Connecticut School of Medicine, Farmington, Connecticut, United States of America
- Albright Center, Division of Pediatric Endocrinology & Diabetes, Connecticut Children’s, Farmington, Connecticut, United States of America
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, Connecticut, United States of America
| |
Collapse
|
8
|
Hwang SK, Shim YJ, Oh SH, Jang KM. Early Diagnosis of Pseudohypoparathyroidism before the Development of Hypocalcemia in a Young Infant. CHILDREN 2022; 9:children9050723. [PMID: 35626900 PMCID: PMC9139394 DOI: 10.3390/children9050723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/26/2022]
Abstract
Pseudohypoparathyroidism (PHP) is a rare, heterogeneous disorder characterized by end-organ resistance to parathyroid hormone (PTH). PTH resistance causes elevated PTH levels, hypocalcemia, and hyperphosphatemia. Since hypocalcemia causes life-threatening events, early diagnosis is crucial. However, the diagnosis of PHP is elusive during infancy because PHP is usually diagnosed with hypocalcemia-induced symptoms, which develop later in childhood when calcium requirements increase. A 1-month-old girl was referred to our clinic for elevated thyroid-stimulating hormone (TSH) levels on newborn screening. When measured 1 month after levothyroxine treatment, her TSH level normalized. At 4-months-old, multiple hard nodules were noted on her trunk. A punch skin biopsy revealed osteoma cutis associated with Albright’s hereditary osteodystrophy, a major characteristic of PHP. We performed targeted sanger sequencing of the GNAS gene and detected a heterozygous variant c.150dupA (p.Ser51Ilefs*3) in both the proband and her mother, causing frameshift and premature termination mutations. The patient was diagnosed with PHP Ia when she had normal calcium, phosphorous, and PTH levels. We report the early diagnosis of PHP Ia without hypocalcemia. It emphasizes the importance of meticulous physical examination in patients with congenital hypothyroidism.
Collapse
Affiliation(s)
- Su Kyeong Hwang
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu 37224, Korea;
| | - Ye Jee Shim
- Department of Pediatrics, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, Daegu 37224, Korea;
| | - Seung Hwan Oh
- Department of Laboratory Medicine, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Korea;
| | - Kyung Mi Jang
- Department of Pediatrics, Yeungnam University School of Medicine, Yeungnam University Hospital, Daegu 42415, Korea
- Correspondence: ; Tel.: +82-53-620-3532
| |
Collapse
|
9
|
Paolo C, Andrea G, Elli FM, Rossella G, Erika P, Milena B, Thiele S, Francesca G, Giovanna M, Franco A. A complex pheotype in a girl with a novel heterozygous missense variant (p.Ile56Phe) of the GNAS gene. Orphanet J Rare Dis 2022; 17:83. [PMID: 35197096 PMCID: PMC8867619 DOI: 10.1186/s13023-022-02252-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 02/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND GNAS is a complex gene that encodes Gsα, a signaling protein that triggers a complex network of pathways. Heterozygous inactivating mutations in Gsα-coding GNAS exons cause hormonal resistance; on the contrary, activating mutations in Gsα result in constitutive cAMP stimulation. Recent research has described a clinical condition characterized by both gain and loss of Gsα function, due to a heterozygous de novo variant of the maternal GNAS allele. PATIENTS AND METHODS We describe a girl with a complex combination of clinical signs and a new heterozygous GNAS variant. For the molecular analysis of GNAS gene, DNA samples of the proband and her parents were extracted from their peripheral blood samples. In silico analysis was performed to predict the possible in vivo effect of the detected novel genetic variant. The activity of Gsα protein was in vitro analyzed from samples of erythrocyte membranes, recovered from heparinized blood samples. RESULTS We found a new heterozygous missense c.166A > T-(p.Ile56Phe) GNAS variant in exon 2, inherited from the mother that determined a reduced activity of 50% of Gsα protein function. The analysis of her parents showed a 20-25% reduction in Gsα protein activity in the mother and a normal function in the father. Clinically our patient presented a multisystemic disorder characterized by hyponatremia compatible with a nephrogenic syndrome of inappropriate antidiuresis, subclinical hyperthyroidism, subclinical hypercortisolism, precocious thelarche and pubarche and congenital bone abnormalities. CONCLUSIONS This is the first time that the new variant c.166A > T (p.Ile56Phe) on exon 2 of GNAS gene, originated on maternal allele, has been described as probable cause of a multisystemic disorder. Although the mutation is associated with a reduced activity of the function of Gsα protein, this unusual phenotype on the contrary suggests a mild functional gain.
Collapse
Affiliation(s)
- Cavarzere Paolo
- Pediatric Division, Department of Pediatrics, University Hospital of Verona, Piazzale Stefani 1, 37126, Verona, Italy.
| | - Gastaldi Andrea
- Pediatric Division, Department of Pediatrics, University Hospital of Verona, Piazzale Stefani 1, 37126, Verona, Italy
| | - Francesca Marta Elli
- Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gaudino Rossella
- Pediatric Division, Department of Pediatrics, University Hospital of Verona, Piazzale Stefani 1, 37126, Verona, Italy.,Pediatric Clinic, Department Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Peverelli Erika
- Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Brugnara Milena
- Pediatric Division, Department of Pediatrics, University Hospital of Verona, Piazzale Stefani 1, 37126, Verona, Italy
| | - Susanne Thiele
- Division of Paediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Luebeck, Germany
| | - Granata Francesca
- General Medicine Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mantovani Giovanna
- Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antoniazzi Franco
- Pediatric Division, Department of Pediatrics, University Hospital of Verona, Piazzale Stefani 1, 37126, Verona, Italy.,Pediatric Clinic, Department Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy.,Regional Center for the Diagnosis and Treatment of Children and Adolescents Rare Skeletal Disorders. Pediatric Clinic, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| |
Collapse
|
10
|
McMullan P, Germain-Lee EL. Aberrant Bone Regulation in Albright Hereditary Osteodystrophy dueto Gnas Inactivation: Mechanisms and Translational Implications. Curr Osteoporos Rep 2022; 20:78-89. [PMID: 35226254 DOI: 10.1007/s11914-022-00719-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW This review highlights the impact of Gnas inactivation on both bone remodeling and the development of heterotopic subcutaneous ossifications in Albright hereditary osteodystrophy (AHO). Here we discuss recent advancements in understanding the pathophysiologic mechanisms of the aberrant bone development in AHO as well as potential translational implications. RECENT FINDINGS Gnas inactivation can regulate the differentiation and function of not only osteoblasts but also osteoclasts and osteocytes. Investigations utilizing a mouse model of AHO generated by targeted disruption of Gnas have revealed that bone formation and resorption are differentially affected based upon the parental origin of the Gnas mutation. Data suggest that Gnas inactivation leads to heterotopic bone formation within subcutaneous tissue by changing the connective tissue microenvironment, thereby promoting osteogenic differentiation of tissue-resident mesenchymal progenitors. Observed variations in bone formation and resorption based upon the parental origin of the Gnas mutation warrant future investigations and may have implications in the management and treatment of AHO and related conditions. Additionally, studies of heterotopic bone formation due to Gnas inactivation have identified an essential role of sonic hedgehog signaling, which could have therapeutic implications not only for AHO and related conditions but also for heterotopic bone formation in a wide variety of settings in which aberrant bone formation is a cause of significant morbidity.
Collapse
Affiliation(s)
- Patrick McMullan
- Department of Pediatrics, Division of Pediatric Endocrinology & Diabetes, University of Connecticut School of Medicine, 505 Farmington Ave, 2nd floor, Farmington, CT, 06032, USA
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| | - Emily L Germain-Lee
- Department of Pediatrics, Division of Pediatric Endocrinology & Diabetes, University of Connecticut School of Medicine, 505 Farmington Ave, 2nd floor, Farmington, CT, 06032, USA.
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT, USA.
- Albright Center, Connecticut Children's, Farmington, CT, USA.
| |
Collapse
|
11
|
Januś D, Roztoczyńska D, Janeczko M, Starzyk JB. New insights into thyroid dysfunction in patients with inactivating parathyroid hormone/parathyroid hormone-related protein signalling disorder (the hormonal and ultrasound aspects): One-centre preliminary results. Front Endocrinol (Lausanne) 2022; 13:1012658. [PMID: 36213284 PMCID: PMC9539917 DOI: 10.3389/fendo.2022.1012658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE This study aimed to present the spectrum of thyroid dysfunction, including hormonal and ultrasound aspects, in a cohort of paediatric and adult patients diagnosed with inactivating parathyroid hormone (PTH)/PTH-related protein signalling disorders 2 and 3 (iPPSD). METHODS The medical records of 31 patients from 14 families diagnosed with iPPSD between 1980 and 2021 in a single tertiary unit were retrospectively analysed. Biochemical, hormonal, molecular, and ultrasonographic parameters were assessed. RESULTS In total, 28 patients from 13 families were diagnosed with iPPSD2 (previously pseudohypoparathyroidism [PHP], PHP1A, and pseudo-PHP) at a mean age of 12.2 years (ranging from infancy to 48 years), and three patients from one family were diagnosed with iPPSD3 (PHP1B). Thyroid dysfunction was diagnosed in 21 of the 28 (75%) patients with iPPSD2. Neonatal screening detected congenital hypothyroidism (CH) in 4 of the 20 (20%) newborns. The spectrum of thyroid dysfunction included: CH, 3/21 (14.2%); CH and autoimmune thyroiditis with nodular goitre, 1/21 (4.8%); subclinical hypothyroidism, 10/21 (47.6%); subclinical hypothyroidism and nodular goitre, 1/21 (4.8%); primary hypothyroidism, 4/21 (19%); and autoimmune thyroiditis (Hashimoto and Graves' disease), 2/21 (9.6%). Thyroid function was normal in 7 of the 28 (25%) patients with iPPSD2 and in all patients with iPPSD3. Ultrasound evaluation of the thyroid gland revealed markedly inhomogeneous echogenicity and structure in all patients with thyroid dysfunction. Goitre was found in three patients. CONCLUSION The spectrum of thyroid dysfunction in iPPSD ranges from CH to autoimmune thyroiditis and nodular goitre. Ultrasonography of the thyroid gland may reveal an abnormal thyroid parenchyma.
Collapse
Affiliation(s)
- Dominika Januś
- Department of Paediatric and Adolescent Endocrinology, Chair of Paediatrics, Institute of Paediatrics, Jagiellonian University Medical College, Krakow, Poland
- Department of Paediatric and Adolescent Endocrinology, University Children’s Hospital, Krakow, Poland
- *Correspondence: Dominika Januś,
| | - Dorota Roztoczyńska
- Department of Paediatric and Adolescent Endocrinology, University Children’s Hospital, Krakow, Poland
| | - Magdalena Janeczko
- Department of Genetics, Chair of Paediatrics, University Children’s Hospital, Krakow, Poland
| | - Jerzy B. Starzyk
- Department of Paediatric and Adolescent Endocrinology, Chair of Paediatrics, Institute of Paediatrics, Jagiellonian University Medical College, Krakow, Poland
- Department of Paediatric and Adolescent Endocrinology, University Children’s Hospital, Krakow, Poland
| |
Collapse
|
12
|
McMullan P, Maye P, Yang Q, Rowe DW, Germain‐Lee EL. Parental Origin of
Gsα
Inactivation Differentially Affects Bone Remodeling in a Mouse Model of Albright Hereditary Osteodystrophy. JBMR Plus 2021; 6:e10570. [PMID: 35079678 PMCID: PMC8771002 DOI: 10.1002/jbm4.10570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/25/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023] Open
Abstract
Albright hereditary osteodystrophy (AHO) is caused by heterozygous inactivation of GNAS, a complex locus that encodes the alpha‐stimulatory subunit of heterotrimeric G proteins (Gsα) in addition to NESP55 and XLαs due to alternative first exons. AHO skeletal manifestations include brachydactyly, brachymetacarpia, compromised adult stature, and subcutaneous ossifications. AHO patients with maternally‐inherited GNAS mutations develop pseudohypoparathyroidism type 1A (PHP1A) with resistance to multiple hormones that mediate their actions through G protein‐coupled receptors (GPCRs) requiring Gsα (eg, parathyroid hormone [PTH], thyroid‐stimulating hormone [TSH], growth hormone–releasing hormone [GHRH], calcitonin) and severe obesity. Paternally‐inherited GNAS mutations cause pseudopseudohypoparathyroidism (PPHP), in which patients have AHO skeletal features but do not develop hormonal resistance or marked obesity. These differences between PHP1A and PPHP are caused by tissue‐specific reduction of paternal Gsα expression. Previous reports in mice have shown loss of Gsα causes osteopenia due to impaired osteoblast number and function and suggest that AHO patients could display evidence of reduced bone mineral density (BMD). However, we previously demonstrated PHP1A patients display normal‐increased BMD measurements without any correlation to body mass index or serum PTH. Due to these observed differences between PHP1A and PPHP, we utilized our laboratory's AHO mouse model to address whether Gsα heterozygous inactivation differentially affects bone remodeling based on the parental inheritance of the mutation. We identified fundamental distinctions in bone remodeling between mice with paternally‐inherited (GnasE1+/−p) versus maternally‐inherited (GnasE1+/−m) mutations, and these findings were observed predominantly in female mice. Specifically, GnasE1+/−p mice exhibited reduced bone parameters due to impaired bone formation and enhanced bone resorption. GnasE1+/−m mice, however, displayed enhanced bone parameters due to both increased osteoblast activity and normal bone resorption. These in vivo distinctions in bone remodeling between GnasE1+/−p and GnasE1+/−m mice could potentially be related to changes in the bone microenvironment driven by calcitonin‐resistance within GnasE1+/−m osteoclasts. Further studies are warranted to assess how Gsα influences osteoblast–osteoclast coupling. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Patrick McMullan
- Department of Pediatrics University of Connecticut School of Medicine Farmington CT USA
- Department of Reconstructive Sciences University of Connecticut School of Dental Medicine Farmington CT USA
- Center for Regenerative Medicine and Skeletal Development University of Connecticut School of Dental Medicine Farmington CT USA
| | - Peter Maye
- Department of Reconstructive Sciences University of Connecticut School of Dental Medicine Farmington CT USA
- Center for Regenerative Medicine and Skeletal Development University of Connecticut School of Dental Medicine Farmington CT USA
| | - Qingfen Yang
- Department of Pediatrics University of Connecticut School of Medicine Farmington CT USA
- Department of Reconstructive Sciences University of Connecticut School of Dental Medicine Farmington CT USA
- Center for Regenerative Medicine and Skeletal Development University of Connecticut School of Dental Medicine Farmington CT USA
| | - David W. Rowe
- Department of Reconstructive Sciences University of Connecticut School of Dental Medicine Farmington CT USA
- Center for Regenerative Medicine and Skeletal Development University of Connecticut School of Dental Medicine Farmington CT USA
| | - Emily L. Germain‐Lee
- Department of Pediatrics University of Connecticut School of Medicine Farmington CT USA
- Department of Reconstructive Sciences University of Connecticut School of Dental Medicine Farmington CT USA
- Center for Regenerative Medicine and Skeletal Development University of Connecticut School of Dental Medicine Farmington CT USA
- Albright Center, Division of Pediatric Endocrinology Connecticut Children's Farmington CT USA
| |
Collapse
|
13
|
Danzig J, Li D, Jan de Beur S, Levine MA. High-throughput Molecular Analysis of Pseudohypoparathyroidism 1b Patients Reveals Novel Genetic and Epigenetic Defects. J Clin Endocrinol Metab 2021; 106:e4603-e4620. [PMID: 34157100 PMCID: PMC8677598 DOI: 10.1210/clinem/dgab460] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Patients with pseudohypoparathyroidism type 1b (PHP1b) show disordered imprinting of the maternal GNAS allele or paternal uniparental disomy (UPD). Genetic deletions in STX16 or in upstream exons of GNAS are present in many familial but not sporadic cases. OBJECTIVE Characterization of epigenetic and genetic defects in patients with PHP1b. DESIGN AND PATIENTS DNA from 84 subjects, including 26 subjects with sporadic PHP1b, 27 affected subjects and 17 unaffected and/or obligate gene carriers from 12 PHP1b families, 11 healthy individuals, and 3 subjects with PHP1a was subjected to quantitative pyrosequencing of GNAS differentially methylated regions (DMRs), microarray analysis, and microsatellite haplotype analysis. SETTING Academic medical center. MAIN OUTCOME MEASUREMENTS Molecular pathology of PHP1b. RESULTS Healthy subjects, unaffected family members and obligate carriers of paternal PHP1b alleles, and subjects with PHP1a showed normal methylation of all DMRs. All PHP1b subjects showed loss of methylation (LOM) at the exon A/B DMR. Affected members of 9 PHP1b kindreds showed LOM only at the exon A/B DMR, which was associated with a 3-kb deletion of STX16 exons 4 through 6 in 7 families and a novel deletion of STX16 and adjacent NEPEPL1 in 1 family. A novel NESP deletion was found in 1 of 2 other families with more extensive methylation defects. One sporadic PHP1b had UPD of 20q, 2 had 3-kb STX16 deletions, and 5 had apparent epigenetic mosaicism. CONCLUSIONS We found diverse patterns of defective methylation and identified novel or previously known mutations in 9 of 12 PHP1b families.
Collapse
Affiliation(s)
- Jennifer Danzig
- Division of Endocrinology and Diabetes, and The Children’s Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Dong Li
- Center for Applied Genomics, The Children’s Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Suzanne Jan de Beur
- Division of Endocrinology and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael A Levine
- Division of Endocrinology and Diabetes, and The Children’s Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Mendes de Oliveira E, Keogh JM, Talbot F, Henning E, Ahmed R, Perdikari A, Bounds R, Wasiluk N, Ayinampudi V, Barroso I, Mokrosiński J, Jyothish D, Lim S, Gupta S, Kershaw M, Matei C, Partha P, Randell T, McAulay A, Wilson LC, Cheetham T, Crowne EC, Clayton P, Farooqi IS. Obesity-Associated GNAS Mutations and the Melanocortin Pathway. N Engl J Med 2021; 385:1581-1592. [PMID: 34614324 DOI: 10.1056/nejmoa2103329] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND GNAS encodes the Gαs (stimulatory G-protein alpha subunit) protein, which mediates G protein-coupled receptor (GPCR) signaling. GNAS mutations cause developmental delay, short stature, and skeletal abnormalities in a syndrome called Albright's hereditary osteodystrophy. Because of imprinting, mutations on the maternal allele also cause obesity and hormone resistance (pseudohypoparathyroidism). METHODS We performed exome sequencing and targeted resequencing in 2548 children who presented with severe obesity, and we unexpectedly identified 22 GNAS mutation carriers. We investigated whether the effect of GNAS mutations on melanocortin 4 receptor (MC4R) signaling explains the obesity and whether the variable clinical spectrum in patients might be explained by the results of molecular assays. RESULTS Almost all GNAS mutations impaired MC4R signaling. A total of 6 of 11 patients who were 12 to 18 years of age had reduced growth. In these patients, mutations disrupted growth hormone-releasing hormone receptor signaling, but growth was unaffected in carriers of mutations that did not affect this signaling pathway (mean standard-deviation score for height, -0.90 vs. 0.75, respectively; P = 0.02). Only 1 of 10 patients who reached final height before or during the study had short stature. GNAS mutations that impaired thyrotropin receptor signaling were associated with developmental delay and with higher thyrotropin levels (mean [±SD], 8.4±4.7 mIU per liter) than those in 340 severely obese children who did not have GNAS mutations (3.9±2.6 mIU per liter; P = 0.004). CONCLUSIONS Because pathogenic mutations may manifest with obesity alone, screening of children with severe obesity for GNAS deficiency may allow early diagnosis, improving clinical outcomes, and melanocortin agonists may aid in weight loss. GNAS mutations that are identified by means of unbiased genetic testing differentially affect GPCR signaling pathways that contribute to clinical heterogeneity. Monogenic diseases are clinically more variable than their classic descriptions suggest. (Funded by Wellcome and others.).
Collapse
Affiliation(s)
- Edson Mendes de Oliveira
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Julia M Keogh
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Fleur Talbot
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Elana Henning
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Rachel Ahmed
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Aliki Perdikari
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Rebecca Bounds
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Natalia Wasiluk
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Vikram Ayinampudi
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Inês Barroso
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Jacek Mokrosiński
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Deepthi Jyothish
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Sharon Lim
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Sanjay Gupta
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Melanie Kershaw
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Cristina Matei
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Praveen Partha
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Tabitha Randell
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Antoinette McAulay
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Louise C Wilson
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Tim Cheetham
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Elizabeth C Crowne
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - Peter Clayton
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| | - I Sadaf Farooqi
- From the University of Cambridge Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge (E.M.O., J.M.K., F.T., E.H., R.A., A.P., R.B., N.W., V.A., J.M., I.S.F.), the Exeter Centre of Excellence for Diabetes Research, University of Exeter Medical School, Exeter (I.B.), Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham (D.J., M.K.), Broomfield Hospital, Chelmsford (S.L.), Hull University Teaching Hospitals NHS Trust, Hull (S.G.), East and North Hertfordshire NHS Trust Lister Hospital, Stevenage (C.M.), County Durham and Darlington NHS Foundation Trust, Darlington (P.P.), Nottingham Children's Hospital, Nottingham (T.R.), University Hospitals Dorset NHS Foundation Trust, Poole (A.M.), Great Ormond Street Hospital for Children NHS Foundation Trust, London (L.C.W.), the Translational and Clinical Research Institute, Newcastle University, and Great North Children's Hospital, Royal Victoria Infirmary, Newcastle upon Tyne (T.C.), University Hospitals Bristol and Weston NHS Foundation Trust, Bristol (E.C.C.), and the Division of Developmental Biology and Medicine, University of Manchester, Manchester (P.C.) - all in the United Kingdom
| |
Collapse
|
15
|
Romanet P, Galluso J, Kamenicky P, Hage M, Theodoropoulou M, Roche C, Graillon T, Etchevers HC, De Murat D, Mougel G, Figarella-Branger D, Dufour H, Cuny T, Assié G, Barlier A. Somatotroph Tumors and the Epigenetic Status of the GNAS Locus. Int J Mol Sci 2021; 22:ijms22147570. [PMID: 34299200 PMCID: PMC8306130 DOI: 10.3390/ijms22147570] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/16/2022] Open
Abstract
Forty percent of somatotroph tumors harbor recurrent activating GNAS mutations, historically called the gsp oncogene. In gsp-negative somatotroph tumors, GNAS expression itself is highly variable; those with GNAS overexpression most resemble phenotypically those carrying the gsp oncogene. GNAS is monoallelically expressed in the normal pituitary due to methylation-based imprinting. We hypothesize that changes in GNAS imprinting of gsp-negative tumors affect GNAS expression levels and tumorigenesis. We characterized the GNAS locus in two independent somatotroph tumor cohorts: one of 23 tumors previously published (PMID: 31883967) and classified by pan-genomic analysis, and a second with 82 tumors. Multi-omics analysis of the first cohort identified a significant difference between gsp-negative and gsp-positive tumors in the methylation index at the known differentially methylated region (DMR) of the GNAS A/B transcript promoter, which was confirmed in the larger series of 82 tumors. GNAS allelic expression was analyzed using a polymorphic Fok1 cleavage site in 32 heterozygous gsp-negative tumors. GNAS expression was significantly reduced in the 14 tumors with relaxed GNAS imprinting and biallelic expression, compared to 18 tumors with monoallelic expression. Tumors with relaxed GNAS imprinting showed significantly lower SSTR2 and AIP expression levels. Altered A/B DMR methylation was found exclusively in gsp-negative somatotroph tumors. 43% of gsp-negative tumors showed GNAS imprinting relaxation, which correlated with lower GNAS, SSTR2 and AIP expression, indicating lower sensitivity to somatostatin analogues and potentially aggressive behavior.
Collapse
Affiliation(s)
- Pauline Romanet
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
| | - Justine Galluso
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
| | - Peter Kamenicky
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, 94270 Le Kremlin-Bicêtre, Île-de-France, France; (P.K.); (M.H.)
| | - Mirella Hage
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse, 94270 Le Kremlin-Bicêtre, Île-de-France, France; (P.K.); (M.H.)
| | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, Ludwig Maximilian University Munich, 80336 Munich, Germany;
| | - Catherine Roche
- APHM, La Conception Hospital, Laboratory of Molecular Biology, 13385 Marseille, France;
| | - Thomas Graillon
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Timone Hospital Department of Neurosurgery, 13385 Marseille, France; (T.G.); (H.D.)
| | - Heather C. Etchevers
- Aix Marseille Univ, INSERM, MMG, UMR1251, Marmara Institute, 13385 Marseille, France;
| | - Daniel De Murat
- Université de Paris, Institut Cochin, Inserm U1016, CNRS UMR8104, F-75014 Paris, France; (D.D.M.); (G.A.)
| | - Grégory Mougel
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
| | - Dominique Figarella-Branger
- Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, CHU Timone, Service d’Anatomie Pathologique et de Neuropathologie, 13385 Marseille, France;
| | - Henry Dufour
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Timone Hospital Department of Neurosurgery, 13385 Marseille, France; (T.G.); (H.D.)
| | - Thomas Cuny
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, Department of Endocrinology, Hospital La Conception, 13385 Marseille, France;
| | - Guillaume Assié
- Université de Paris, Institut Cochin, Inserm U1016, CNRS UMR8104, F-75014 Paris, France; (D.D.M.); (G.A.)
- Department of Endocrinology, Center for Rare Adrenal Diseases, Assistance Publique—Hôpitaux de Paris, Hôpital Cochin, 75014 Paris, France
| | - Anne Barlier
- Aix Marseille Univ, INSERM, APHM, MMG, UMR1251, Marmara Institute, La Conception, Hospital Laboratory of Molecular Biology, 13385 Marseille, France; (P.R.); (J.G.); (G.M.)
- Correspondence:
| |
Collapse
|
16
|
Abstract
Parathyroid hormone (PTH), which is primarily regulated by extracellular calcium changes, controls calcium and phosphate homeostasis. Different diseases are derived from PTH deficiency (hypoparathyroidism), excess (hyperparathyroidism) and resistance (pseudohypoparathyroidism, PHP). Pseudohypoparathyroidism was historically classified into subtypes according to the presence or not of inherited PTH resistance associated or not with features of Albright's hereditary osteodystrophy and deep and progressive ectopic ossifications. The growing knowledge on the PTH/PTHrP signaling pathway showed that molecular defects affecting different members of this pathway determined distinct, yet clinically related disorders, leading to the proposal of a new nomenclature and classification encompassing all disorders, collectively termed inactivating PTH/PTHrP signaling disorders (iPPSD).
Collapse
Affiliation(s)
- Giovanna Mantovani
- University of Milan, Dept. Clinical Sciences and Commmunity Health, Via Lamarmora 5, Milan, Italy; Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Via Lamarmora 5, 20122, Milan, Italy.
| | - Francesca Marta Elli
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Via Lamarmora 5, 20122, Milan, Italy.
| |
Collapse
|
17
|
Kaulfers AMD, Lim WY, Bhowmick SK. Unusual Endocrinopathies in 18q Deletion Syndrome: Pseudoparathyroidism and Hyper-/Hypo-Thyroidism. AACE Clin Case Rep 2021; 7:192-194. [PMID: 34095486 PMCID: PMC8165197 DOI: 10.1016/j.aace.2020.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Objective To describe new and unusual endocrinopathies in children with de novo 18q deletion (18q-) syndrome. Methods We describe 2 patients who have atypical thyroid conditions and 1 who also developed symptomatic hypocalcemia. Results The first patient developed hyperthyroidism at the age of 3 years, with a free thyroxine level of 3.9 (range, 0.8-1.8) ng/dL. Thyroid peroxidase antibodies were 262 (range, 0-32) IU/mL, and thyroid-stimulating immunoglobulin antibodies were 384% (range, 0-139%). On low-dose methimazole treatment, she developed hypothyroidism. Thyroid-stimulating hormone (TSH) level was 163 (range, 0.4-4.5) mIU/mL. Moreover, she later developed growth hormone deficiency. The second patient developed hypothyroidism at the age of 4 years, with a TSH level of 46 mIU/mL. However, TSH remained elevated at levels of 10 to 24 mIU/mL for 3 years, despite appropriate treatment, suggesting TSH resistance. She then developed hypocalcemic seizures and was diagnosed with pseudohypoparathyroidism. Her total calcium level was 6.6 (range, 8.5-10.5) mg/dL and parathyroid hormone level was 432 (range, 15-65) pg/dL. Conclusion The first patient had a mixed picture of autoimmune hypothyroidism and hyperthyroidism, requiring a combination of methimazole and levothyroxine to achieve a euthyroid state. For the second patient, the mild TSH resistance was possibly the early suggestion of a parathyroid hormone resistant state. Although growth hormone deficiency and hypothyroidism are common in patients with 18q- syndrome, the occurrence of hyperthyroidism due to Graves’ disease with the coexistence of Hashimoto’s hypothyroidism is rare. Pseudohypoparathyroidism has not yet been reported in patients with 18q- syndrome.
Collapse
Affiliation(s)
| | - Whei Ying Lim
- Department of Pediatric Endocrinology, University of South Alabama, Mobile, Alabama
| | - Samar K Bhowmick
- Department of Pediatric Endocrinology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
18
|
Elli FM, Mantovani G. Pseudohypoparathyroidism, acrodysostosis, progressive osseous heteroplasia: different names for the same spectrum of diseases? Endocrine 2021; 72:611-618. [PMID: 33179219 PMCID: PMC8159830 DOI: 10.1007/s12020-020-02533-9] [Citation(s) in RCA: 8] [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: 06/09/2020] [Accepted: 10/24/2020] [Indexed: 12/27/2022]
Abstract
Pseudohypoparathyroidism (PHP), the first known post-receptorial hormone resistance, derives from a partial deficiency of the α subunit of the stimulatory G protein (Gsα), a key component of the PTH/PTHrP signaling pathway. Since its first description, different studies unveiled, beside the molecular basis for PHP, the existence of different subtypes and of diseases in differential diagnosis associated with genetic alterations in other genes of the PTH/PTHrP pathway. The clinical and molecular overlap among PHP subtypes and with different but related disorders make both differential diagnosis and genetic counseling challenging. Recently, a proposal to group all these conditions under the novel term "inactivating PTH/PTHrP signaling disorders (iPPSD)" was promoted and, soon afterwards, the first international consensus statement on the diagnosis and management of these disorders has been published. This review will focus on the major and minor features characterizing PHP/iPPSDs as a group and on the specificities as well as the overlap associated with the most frequent subtypes.
Collapse
Affiliation(s)
- Francesca Marta Elli
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanna Mantovani
- Endocrinology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| |
Collapse
|
19
|
Abstract
Pseudohypoparathyroidism (PHP) and pseudopseudohypoparathyroidism (PPHP) are caused by mutations and/or epigenetic changes at the complex GNAS locus on chromosome 20q13.3 that undergoes parent-specific methylation changes at several differentially methylated regions (DMRs). GNAS encodes the alpha-subunit of the stimulatory G protein (Gsα) and several splice variants thereof. PHP type Ia (PHP1A) is caused by heterozygous inactivating mutations involving the maternal exons 1-13. Heterozygosity of these maternal GNAS mutations cause PTH-resistant hypocalcemia and hyperphosphatemia because paternal Gsα expression is suppressed in certain organs thus leading to little or no Gsα protein in the proximal renal tubules and other tissues. Besides biochemical abnormalities, PHP1A patients show developmental abnormalities, referred to as Albright's hereditary osteodystrophy (AHO). Some, but not all of these AHO features are encountered also in patients affected by PPHP, who carry paternal Gsα-specific mutations and typically show no laboratory abnormalities. Autosomal dominant PHP type Ib (AD-PHP1B) is caused by heterozygous maternal deletions within GNAS or STX16, which are associated with loss of methylation at the A/B DMR alone or at all maternally methylated GNAS exons. Loss of methylation of exon A/B and the resulting biallelic expression of A/B transcript reduces Gsα expression thus leading to hormonal resistance. Epigenetic changes at all differentially methylated GNAS regions are also observed in sporadic PHP1B, which is the most frequent PHP1B variant. However, this disease variant remains unresolved at the molecular level, except for rare cases with paternal uniparental isodisomy or heterodisomy of chromosome 20q (patUPD20q).
Collapse
Affiliation(s)
- Harald Jüppner
- Endocrine Unit, Department of Medicine and Pediatric Nephrology Unit, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Correspondence: Harald Jüppner, MD, Endocrine Unit, Thier 10, 50 Blossom Street, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
20
|
Zhadina M, Roszko KL, Geels RES, de Castro LF, Collins MT, Boyce AM. Genotype-Phenotype Correlation in Fibrous Dysplasia/McCune-Albright Syndrome. J Clin Endocrinol Metab 2021; 106:1482-1490. [PMID: 33512531 PMCID: PMC8522305 DOI: 10.1210/clinem/dgab053] [Citation(s) in RCA: 10] [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: 09/23/2020] [Indexed: 02/07/2023]
Abstract
CONTEXT Fibrous dysplasia/McCune-Albright syndrome (FD/MAS) is a rare bone and endocrine disorder resulting in fractures, pain, and disability. There are no targeted or effective therapies to alter the disease course. Disease arises from somatic gain-of-function variants at the R201 codon in GNAS, replacing arginine by either cysteine or histidine. The relative pathogenicity of these variants is not fully understood. OBJECTIVE This work aimed 1) to determine whether the most common GNAS variants (R201C and R201H) are associated with a specific clinical phenotype, and 2) to determine the prevalence of the most common GNAS variants in a large patient cohort. METHODS This retrospective cross-sectional analysis measured the correlation between genotype and phenotype characterized by clinical, biochemical, and radiographic data. RESULTS Sixty-one individuals were genotyped using DNA extracted from tissue or circulating cell-free DNA. Twenty-two patients (36.1%) had the R201C variant, and 39 (63.9%) had the R201H variant. FD skeletal disease burden, hypophosphatemia prevalence, fracture incidence, and ambulation status were similar between the 2 groups. There was no difference in the prevalence of endocrinopathies, ultrasonographic gonadal or thyroid abnormalities, or pancreatic involvement. There was a nonsignificant association of cancer with the R201H variant. CONCLUSION There is no clear genotype-phenotype correlation in patients with the most common FD/MAS pathogenic variants. The predominance of the R201H variant observed in our cohort and reported in the literature indicates it is likely responsible for a larger burden of disease in the overall population of patients with FD/MAS, which may have important implications for the future development of targeted therapies.
Collapse
Affiliation(s)
- Maria Zhadina
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kelly L Roszko
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Raya E S Geels
- Department of Medicine, Division of Endocrinology, Centre for Bone Quality, Leiden University Medical Centre, ZA Leiden, the Netherlands
| | - Luis F de Castro
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Michael T Collins
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Alison M Boyce
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
- Metabolic Bone Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland , USA
- Correspondence: Alison Boyce, MD, Metabolic Bone Disorders Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health, 30 Convent Dr, Bldg 30, Rm 228, MSC 4320, Bethesda, MD 20892, USA.
| |
Collapse
|
21
|
Luo D, Qi X, Liu L, Su Y, Fang L, Guan Q. Genetic and Epigenetic Characteristics of Autosomal Dominant Pseudohypoparathyroidism Type 1B: Case Reports and Literature Review. Horm Metab Res 2021; 53:225-235. [PMID: 33513624 DOI: 10.1055/a-1341-9891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Autosomal dominant pseudohypoparathyroidism 1B (AD-PHP1B) is a rare endocrine and imprinted disorder. The objective of this study is to clarify the imprinted regulation of the guanine nucleotide binding-protein α-stimulating activity polypeptide 1 (GNAS) cluster in the occurrence and development of AD-PHP1B based on animal and clinical patient studies. The methylation-specific multiples ligation-dependent probe amplification (MS-MLPA) was conducted to detect the copy number variation in syntaxin-16 (STX16) gene and methylation status of the GNAS differentially methylated regions (DMRs). Long-range PCR was used to confirm deletion at STX16 gene. In the first family, DNA analysis of the proband and proband's mother revealed an isolated loss of methylation (LOM) at exon A/B and a 3.0 kb STX16 deletion. The patient's healthy grandmother had the 3.0 kb STX16 deletion but no epigenetic abnormality. The patient's healthy maternal aunt showed no genetic or epigenetic abnormality. In the second family, the analysis of long-range PCR revealed the 3.0 kb STX16 deletion for the proband but not her children. In this study, 3.0 kb STX16 deletion causes isolated LOM at exon A/B in two families, which is the most common genetic mutation of AD-PHP1B. The deletion involving NESP55 or AS or genomic rearrangements of GNAS can also result in AD-PHP1B, but it's rare. LOM at exon A/B DMR is prerequisite methylation defect of AD-PHP1B. STX16 and NESP55 directly control the imprinting at exon A/B, while AS controls the imprinting at exon A/B by regulating the transcriptional level of NESP55.
Collapse
Affiliation(s)
- Dandan Luo
- Department of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Xiangyu Qi
- Department of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Luna Liu
- Department of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Yu Su
- Department of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Li Fang
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
| | - Qingbo Guan
- Department of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
22
|
Tessaris D, Bonino E, Weber G, Wasniewska M, Corica D, Pitea M, Scirè G, Caruso-Nicoletti M, Fintini D, de Sanctis L. Pseudohypoparathyroidism: application of the Italian common healthcare-pathway for a homogeneous clinical approach and a shared follow up. Ital J Pediatr 2021; 47:48. [PMID: 33663571 PMCID: PMC7934261 DOI: 10.1186/s13052-021-01000-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pseudohypoparathyroidism (PHP) represents a heterogeneous group of rare endocrine disorders caused by (epi) genetic abnormalities affecting the GNAS locus. It is mainly characterized by resistance to PTH and TSH, and by peculiar clinical features such as short stature, obesity, cognitive impairment, subcutaneous ossifications and brachydactyly. Delayed puberty, GHRH and calcitonin resistances have also been described. The healthcare-pathway recently proposed by the Italian Society of Pediatric Endocrinology and Diabetology (ISPED) has provided a standardized clinical approach to these conditions. The purpose of the present study was to evaluate its application in clinical practice, and to collect data for setting future specific studies. METHODS Through a semi-structured survey, based on the indications of the care-pathway, data on PHP clinical management were collected. The compilation of each data in the survey was read as an index of the adoption of the healthcare-pathway in clinical practice. RESULTS In addition to the proposing Center, 4 Centers joined the study, thus obtaining a large collection of data on 48 PHP patients. Highest rates in the completion of data were obtained for diagnostic history, auxological measurements and subcutaneous ossifications evaluation. As expected, the availability of data for the other investigated fields was lower, coming from recent research studies. More information has been obtained on hormonal resistance classically involved in PHP (PTH, TSH, GHRH and GnRH) and on cognitive impairment, while a few data has been collected on bone mineral status, calcitonin levels and glucolipid metabolism. CONCLUSIONS The presented data show that the ISPED healthcare-pathway could represent a valid tool both to confirm the clinical approach to PHP patients and to allow homogeneous data collection; however, it has not yet been fully adopted. The strengthening of the network among the major Italian Endocrine Centers will contribute to improve its application in clinical practice, optimizing the follow-up of these patients and increasing knowledge on PHP.
Collapse
Affiliation(s)
- Daniele Tessaris
- Department of Pediatric Endocrinology, Regina Margherita Children's Hospital - Health and Science City, Turin, Italy. .,Department of Public Health and Pediatrics, University of Turin, Piazza Polonia 94, 10126, Torino (TO), Italy.
| | - Elisa Bonino
- Department of Pediatric Endocrinology, Regina Margherita Children's Hospital - Health and Science City, Turin, Italy
| | - Giovanna Weber
- Department of Pediatrics, San Raffaele Hospital, University of Milan, Milan, Italy
| | - Malgorzata Wasniewska
- Department of Pediatrics, Gynecological, Microbiological and Biomedical Sciences, University of Messina, Messina, Italy
| | - Domenico Corica
- Department of Pediatrics, Gynecological, Microbiological and Biomedical Sciences, University of Messina, Messina, Italy
| | - Marco Pitea
- Department of Pediatrics, San Raffaele Hospital, University of Milan, Milan, Italy
| | - Giuseppe Scirè
- Endocrinology Unit, Pediatric University Department, "Bambino Gesù" Children's Hospital-IRCCS, Rome, Italy
| | | | - Danilo Fintini
- Endocrinology Unit, Pediatric University Department, "Bambino Gesù" Children's Hospital-IRCCS, Rome, Italy
| | - Luisa de Sanctis
- Department of Pediatric Endocrinology, Regina Margherita Children's Hospital - Health and Science City, Turin, Italy
| |
Collapse
|
23
|
Reyes M, Kagami M, Kawashima S, Pallotta J, Schnabel D, Fukami M, Jüppner H. A Novel GNAS Duplication Associated With Loss-of-Methylation Restricted to Exon A/B Causes Pseudohypoparathyroidism Type Ib (PHP1B). J Bone Miner Res 2021; 36:546-552. [PMID: 33180333 PMCID: PMC8048081 DOI: 10.1002/jbmr.4209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/03/2020] [Accepted: 11/01/2020] [Indexed: 11/09/2022]
Abstract
Pseudohypoparathyroidism type Ib (PHP1B) is characterized by resistance to parathyroid hormone (PTH) leading to hypocalcemia and hyperphosphatemia, and in some cases resistance toward additional hormones. Patients affected by this disorder all share a loss-of-methylation (LOM) at the differentially methylated GNAS exon A/B, which reduces expression of the stimulatory G protein α-subunit (Gsα) from the maternal allele. This leads in the proximal renal tubules, where the paternal GNAS allele does not contribute much to expression of this signaling protein, to little or no Gsα expression thereby causing PTH resistance. We now describe a PHP1B patient with a de novo genomic GNAS duplication of approximately 88 kb, which is associated with LOM restricted to exon A/B alone. Multiplex ligation-dependent probe amplification (MLPA), comparative genomic hybridization (CGH), and whole-genome sequencing (WGS) established that the duplicated DNA fragment extends from GNAS exon AS1 (telomeric breakpoint) to a small region between two imperfect repeats just upstream of LOC105372695 (centromeric breakpoint). Our novel duplication is considerably shorter than previously described duplications/triplications in that portion of chromosome 20q13 and it does not affect methylation at exons AS and XL. Based on these and previous findings, it appears plausible that the identified genomic abnormality disrupts in cis the actions of a transcript that is required for establishing or maintaining exon A/B methylation. Our findings extend the molecular causes of PHP1B and provide additional insights into structural GNAS features that are required for maintaining maternal Gsα expression and for preventing PTH-resistance. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Monica Reyes
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Sayaka Kawashima
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Johanna Pallotta
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dirk Schnabel
- Center for Chronically Sick Children, Pediatric Endocrinology, Charité University Medicine, Berlin, Germany
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Li D, Bupp C, March ME, Hakonarson H, Levine MA. Intragenic Deletions of GNAS in Pseudohypoparathyroidism Type 1A Identify a New Region Affecting Methylation of Exon A/B. J Clin Endocrinol Metab 2020; 105:5841615. [PMID: 32436958 PMCID: PMC7947960 DOI: 10.1210/clinem/dgaa286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Pseudohypoparathyroidism type 1A (PHP1A) and pseudopseudohypoparathyroidism (PPHP) are caused by inactivating mutations in the exons of GNAS that encode the alpha-subunit of the stimulatory G protein (Gsα). In some cases abnormal methylation of exon A/B of GNAS, a hallmark of PHP1B, has been reported. OBJECTIVE To identify the underlying genetic basis for PHP1A/PPHP in patients in whom molecular defects were not detected by GNAS sequencing and microarray-based analysis of copy number variations. METHODS Whole genome sequencing (WGS) and pyrosequencing of differentially methylated regions (DMRs) of GNAS using genomic deoxyribonucleic acid from affected patients. RESULTS We identified 2 novel heterozygous GNAS deletions: a 6.4 kb deletion that includes exon 2 of GNAS in the first proband that was associated with normal methylation (57%) of exon A/B DMR, and a 1438 bp deletion in a second PHP1A patient that encompasses the promoter region and 5' untranslated region of Gsα transcripts, which was inherited from his mother with PPHP. This deletion was associated with reduced methylation (32%) of exon A/B DMR. CONCLUSIONS WGS can detect exonic and intronic mutations, including deletions that are too small to be identified by microarray analysis, and therefore is more sensitive than other techniques for molecular analysis of PHP1A/PPHP. One of the deletions we identified led to reduced methylation of exon A/B DMR, further refining a region needed for normal imprinting of this DMR. We propose that deletion of this region can explain why some PHP1A patients have reduced of methylation of the exon A/B DMR.
Collapse
Affiliation(s)
- Dong Li
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | | | - Michael E March
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Michael A Levine
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Correspondence and Reprint Requests: Michael A. Levine, MD, Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Abramson Research Center, Room 510A, 3615 Civic Center Boulevard, Philadelphia, PA 19104. E-mail:
| |
Collapse
|
25
|
Wang Y, Tian H, Chen X. The Distinct Role of the Extra-Large G Protein ɑ-Subunit XLɑs. Calcif Tissue Int 2020; 107:212-219. [PMID: 32596800 DOI: 10.1007/s00223-020-00714-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/17/2020] [Indexed: 02/05/2023]
Abstract
GNAS is one of the most complex gene loci in the human genome and encodes multiple gene products including Gsα, XLαs, NESP55, A/B, and AS transcripts. XLαs, the extra-large G protein ɑ-subunit, is paternally expressed. XLɑs and Gsɑ share the common 2-13 exons with different promoters and first exons. Therefore, XLɑs contains most of the functional domains of Gsα including receptor and effector binding sites. In vitro studies suggest a "Gsɑ"-like function of XLɑs regarding the stimulation of cAMP generation in response to receptor activation with different cellular actions. However, it is unclear whether XLαs has an important physiological function in humans. Pseudopseudohypoparathyroidism (PPHP) and progressive osseous heteroplasia (POH) are caused by paternally inherited mutations of GNAS. Maternal uniparental disomy of chromosome 20 [UPD(20)mat] lacks paternal chromosome 20. Therefore, the phenotypes of these diseases may be secondary to the abnormal functions of XLɑs, at least partly. From the phenotypes of human diseases like PPHP, POH, and UPD(20)mat, as well as some animal models with deficient XLɑs functions, it could be seen that XLɑs is involved in the growth and development of the mammalian fetus, plays a different role in glucose, lipid, and energy metabolism when compared with Gsɑ, and could prevent heterotopic ossification in humans and mice. More in vivo and in vitro studies, especially the development of conditional XLɑs knockout mice, are needed to clarify the physiopathologic roles and related signal pathways of XLɑs.
Collapse
Affiliation(s)
- Yan Wang
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Haoming Tian
- Department of Endocrinology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Chen
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, China.
| |
Collapse
|
26
|
Mantovani G, Bastepe M, Monk D, de Sanctis L, Thiele S, Ahmed SF, Bufo R, Choplin T, De Filippo G, Devernois G, Eggermann T, Elli FM, Ramirez AG, Germain-Lee EL, Groussin L, Hamdy NA, Hanna P, Hiort O, Jüppner H, Kamenický P, Knight N, Le Norcy E, Lecumberri B, Levine MA, Mäkitie O, Martin R, Martos-Moreno GÁ, Minagawa M, Murray P, Pereda A, Pignolo R, Rejnmark L, Rodado R, Rothenbuhler A, Saraff V, Shoemaker AH, Shore EM, Silve C, Turan S, Woods P, Zillikens MC, de Nanclares GP, Linglart A. Recommendations for Diagnosis and Treatment of Pseudohypoparathyroidism and Related Disorders: An Updated Practical Tool for Physicians and Patients. Horm Res Paediatr 2020; 93:182-196. [PMID: 32756064 PMCID: PMC8140671 DOI: 10.1159/000508985] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Patients affected by pseudohypoparathyroidism (PHP) or related disorders are characterized by physical findings that may include brachydactyly, a short stature, a stocky build, early-onset obesity, ectopic ossifications, and neurodevelopmental deficits, as well as hormonal resistance most prominently to parathyroid hormone (PTH). In addition to these alterations, patients may develop other hormonal resistances, leading to overt or subclinical hypothyroidism, hypogonadism and growth hormone (GH) deficiency, impaired growth without measurable evidence for hormonal abnormalities, type 2 diabetes, and skeletal issues with potentially severe limitation of mobility. PHP and related disorders are primarily clinical diagnoses. Given the variability of the clinical, radiological, and biochemical presentation, establishment of the molecular diagnosis is of critical importance for patients. It facilitates management, including prevention of complications, screening and treatment of endocrine deficits, supportive measures, and appropriate genetic counselling. Based on the first international consensus statement for these disorders, this article provides an updated and ready-to-use tool to help physicians and patients outlining relevant interventions and their timing. A life-long coordinated and multidisciplinary approach is recommended, starting as far as possible in early infancy and continuing throughout adulthood with an appropriate and timely transition from pediatric to adult care.
Collapse
Affiliation(s)
- Giovanna Mantovani
- Endocrinology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Murat Bastepe
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Monk
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Luisa de Sanctis
- Pediatric Endocrinology Unit, Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Susanne Thiele
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - S. Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Roberto Bufo
- Italian Progressive Osseous Heteroplasia Association (IPOHA), Foggia, Italy
| | - Timothée Choplin
- K20, French PHP and Related Disorders Patient Association, Jouars-Pontchartrain, France
| | - Gianpaolo De Filippo
- APHP, Department of Medicine for Adolescents, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicetre, France
| | - Guillemette Devernois
- K20, French PHP and Related Disorders Patient Association, Jouars-Pontchartrain, France
| | - Thomas Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Francesca M. Elli
- Endocrinology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Emily L. Germain-Lee
- Albright Center and Center for Rare Bone Disorders, Division of Pediatric Endocrinology and Diabetes, Connecticut Children’s Medical Center, Farmington, CT, USA,Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lionel Groussin
- APHP, Department of Endocrinology, Cochin Hospital (HUPC), Paris, France,University of Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Neveen A.T. Hamdy
- Division of Endocrinology and Centre for Bone Quality, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick Hanna
- INSERM U1185, Bicêtre Paris Sud – Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Olaf Hiort
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Harald Jüppner
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Kamenický
- INSERM U1185, Bicêtre Paris Sud – Paris Saclay University, Le Kremlin-Bicêtre, France,APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Saclay for Rare Diseases and Filière OSCAR, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France,APHP, Department of Endocrinology and Reproductive Diseases, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Nina Knight
- Acrodysostosis Support and Research patients’ group, London, UK
| | - Elvire Le Norcy
- University of Paris Descartes, Sorbonne Paris Cité, Paris, France,APHP, Department of Odontology, Bretonneau Hospital (PNVS), Paris, France
| | - Beatriz Lecumberri
- Department of Endocrinology and Nutrition, La Paz University Hospital, Madrid, Spain,Department of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Michael A. Levine
- Division of Endocrinology and Diabetes and Center for Bone Health, Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Outi Mäkitie
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Regina Martin
- Osteometabolic Disorders Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Hospital das Clínicas HCFMUSP, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Gabriel Ángel Martos-Moreno
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Hospital La Princesa Institute for Health Research (IIS La Princesa), Madrid, Spain,Department of Pediatrics, Autonomous University of Madrid (UAM), Madrid, Spain,CIBERobn, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Philip Murray
- Department of Paediatric Endocrinology, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Arrate Pereda
- Molecular (Epi)Genetics Laboratory, BioAraba Research Health Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Spain
| | | | - Lars Rejnmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Rebeca Rodado
- AEPHP, Spanish PHP and Related Disorders Patient Association, Almeria, Spain
| | - Anya Rothenbuhler
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Saclay for Rare Diseases and Filière OSCAR, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France,APHP, Endocrinology and Diabetes for Children, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Vrinda Saraff
- Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham, UK
| | - Ashley H. Shoemaker
- Pediatric Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eileen M. Shore
- Departments of Orthopedic Surgery and Genetics, Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Silve
- APHP, Service de Biochimie et Génétique Moléculaires, Hôpital Cochin, Paris, France
| | - Serap Turan
- Department of Pediatrics, Division of Endocrinology and Diabetes, Marmara University, Istanbul, Turkey
| | - Philip Woods
- Acrodysostosis Support and Research patients’ group, London, UK
| | - M. Carola Zillikens
- Department of Internal Medicine, Bone Center Erasmus MC – University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guiomar Perez de Nanclares
- Molecular (Epi)Genetics Laboratory, BioAraba Research Health Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Spain
| | - Agnès Linglart
- INSERM U1185, Bicêtre Paris Sud – Paris Saclay University, Le Kremlin-Bicêtre, France,APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Saclay for Rare Diseases and Filière OSCAR, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France,APHP, Endocrinology and Diabetes for Children, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW This review is timely given the 2018 publication of the first international Consensus Statement for the diagnosis and management of pseudohypoparathyroidism (PHP) and related disorders. The purpose of this review is to provide the knowledge needed to recognize and manage PHP1A, pseudopseudohypoparathyroidism (PPHP) and PHP1B - the most common of the subtypes - with an overview of the entire spectrum and to provide a concise summary of management for clinical use. This review will draw from recent literature as well as personal experience in evaluating hundreds of children and adults with PHP. RECENT FINDINGS Progress is continually being made in understanding the mechanisms underlying the PHP spectrum. Every year, through clinical and laboratory studies, the phenotypes are elucidated in more detail, as are clinical issues such as short stature, brachydactyly, subcutaneous ossifications, cognitive/behavioural impairments, obesity and metabolic disturbances. Headed by a European PHP consortium, experts worldwide published the first international Consensus that provides detailed guidance in a systematic manner and will lead to exponential progress in understanding and managing these disorders. SUMMARY As more knowledge is gained from clinical and laboratory investigations, the mechanisms underlying the abnormalities associated with PHP are being uncovered as are improvements in management.
Collapse
|
28
|
Labbadia R, Bizzarri C, Mucciolo M, Di Zazzo G, Guzzo I, Cappa M, Emma F, Dello Strologo L. Renal Tubular Dysfunction Fully Accounts for Plasma Biochemical Abnormalities in Type 1A Pseudohypoparathyroidism. J Clin Endocrinol Metab 2019; 104:823-826. [PMID: 30418563 DOI: 10.1210/jc.2018-01193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/05/2018] [Indexed: 11/19/2022]
Abstract
CONTEXT Type 1A pseudohypoparathyroidism (PHP-1A) is characterized by target organ resistance to PTH. Patients can present with various dysmorphic features; however, renal failure has not been classically described. CASE DESCRIPTION A female patient came to our attention at the age of 7 years with characteristic signs of PTH resistance (i.e., hypocalcemia, hyperphosphatemia, and high serum PTH levels). She also presented with hypothyroidism, early-onset obesity, short metacarpal bones, and multiple subcutaneous ossifications, leading to a clinical diagnosis of pseudohypoparathyroidism. In addition to her genetic condition, she had bilateral renal hypodysplasia that was slowly progressing to end-stage kidney disease. She received a kidney transplant at the age of 16 years and, after transplantation, experienced rapidly normalized calcium, phosphate, and PTH levels, allowing f withdrawal of vitamin D supplementation. CONCLUSIONS To the best of our knowledge, ours is the first report of a patient with PHP-1A undergoing kidney transplantation. Normalization of biochemical parameters after the procedure demonstrated that renal tubular resistance to PTH is sufficient to explain the calcium/phosphate abnormalities observed in PHP-1A.
Collapse
Affiliation(s)
- Raffaella Labbadia
- Nephrology Unit, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Carla Bizzarri
- Endocrinology Unit, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Mafalda Mucciolo
- Medical Genetics Laboratory, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Giacomo Di Zazzo
- Nephrology Unit, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Isabella Guzzo
- Nephrology Unit, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Marco Cappa
- Endocrinology Unit, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Francesco Emma
- Nephrology Unit, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Luca Dello Strologo
- Nephrology Unit, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| |
Collapse
|
29
|
Abstract
A central dogma of mammalian reproductive biology is that the size of the primordial follicle pool represents reproductive capacity in females. The assembly of the primordial follicle starts after the primordial germ cells (PGCs)-derived oocyte releases from the synchronously dividing germline cysts. PGCs initiate meiosis during fetal development. However, after synapsis and recombination of homologous chromosomes, they arrest at the diplotene stage of the first meiotic prophase (MI). The diplotene-arrested oocyte, together with the surrounding of a single layer of flattened granulosa cells, forms a basic unit of the ovary, the primordial follicle. At the start of each estrous (animal) or menstrual cycle (human), in response to a surge of luteinizing hormone (LH) from the pituitary gland, a limited number of primordial follicles are triggered to develop into primary follicles, preantral follicles, antral follicles and reach to preovulatory follicle stage. During the transition from the preantral to antral stages, the enclosed oocyte gradually acquires the capacity to resume meiosis. Meiotic resumption from the prophase of MI is morphologically characterized by the dissolution of the oocyte nuclear envelope, which is generally termed the "germinal vesicle breakdown" (GVBD). Following GVBD and completion of MI, the oocyte enters meiosis II without an obvious S-phase and arrests at metaphase phase II (MII) until fertilization. The underlying mechanism of meiotic arrest has been widely explored in numerous studies. Many studies indicated that two cellular second messengers, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) play an essential role in maintaining oocyte meiotic arrest. This review will discuss how these two cyclic nucleotides regulate oocyte maturation by blocking or initiating meiotic processes, and to provide an insight in future research.
Collapse
Affiliation(s)
- Bo Pan
- Department of Animal Biosciences, University of Guelph, 50 Stone Road E, Building #70, Guelph, ON, N1G 2W1, Canada
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, 50 Stone Road E, Building #70, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
30
|
Elli FM, Pereda A, Linglart A, Perez de Nanclares G, Mantovani G. Parathyroid hormone resistance syndromes - Inactivating PTH/PTHrP signaling disorders (iPPSDs). Best Pract Res Clin Endocrinol Metab 2018; 32:941-954. [PMID: 30665554 DOI: 10.1016/j.beem.2018.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metabolic disorders caused by impairments of the Gsα/cAMP/PKA pathway affecting the signaling of PTH/PTHrP lead to features caused by non-responsiveness of target organs, in turn leading to manifestations similar to the deficiency of the hormone itself. Pseudohypoparathyroidism (PHP) and related disorders derive from a defect of the α subunit of the stimulatory G protein (Gsα) or of downstream effectors of the same pathway, such as the PKA regulatory subunit 1A and the phosphodiesterase type 4D. The increasing knowledge on these diseases made the actual classification of PHP outdated as it does not include related conditions such as acrodysostosis (ACRDYS) or progressive osseous heteroplasia (POH), so that a new nomenclature and classification has been recently proposed grouping these disorders under the term "inactivating PTH/PTHrP signaling disorder" (iPPSD). This review will focus on the pathophysiology, clinical and molecular aspects of these rare, heterogeneous but closely related diseases.
Collapse
Affiliation(s)
- Francesca Marta Elli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Arrate Pereda
- Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, OSI Araba University Hospital, Vitoria-Gasteiz, Spain.
| | - Agnès Linglart
- APHP, Department of Paediatric Endocrinology and Diabetes for Children, Bicêtre Paris-Sud Hospital, Le Kremlin-Bicêtre, France; APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Filière OSCAR and Plateforme d'Expertise Maladies Rares Paris-Sud, Bicêtre Paris-Sud Hospital, Le Kremlin Bicêtre, France.
| | - Guiomar Perez de Nanclares
- Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, OSI Araba University Hospital, Vitoria-Gasteiz, Spain.
| | - Giovanna Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| |
Collapse
|
31
|
Abstract
Pseudohypoparathyroidism (PHP) is a rare group of disorders characterised by end-organ resistance to the parathyroid hormone (PTH). A 16-year-old boy presented with a 2-year history of involuntary dystonic movements involving mainly the left hand, initially after writing and later during physical exercise. Serum calcium was 1.37 mmol/L (2.20-2.69), phosphate 2.1 mmol/L (0.8-1.45) and PTH 302 ng/L (12-88). CT scan of the head demonstrated multiple subcortical and diffuse basal ganglia calcifications. Genetic analysis confirmed a methylation defect in the GNAS cluster on chromosome 20q13.32 which established the diagnosis. Treatment with calcitriol and calcium carbonate led to complete remission of symptoms. Causes of hypocalcaemia should be considered in evaluating patients with movement disorders. The diagnosis of PHP-1B is challenging but the overall prognosis is excellent.
Collapse
Affiliation(s)
- Catarina Garcia
- a Department of Pediatrics , Hospital Professor Doutor Fernando Fonseca, EPE , Lisbon , Portugal
| | - Cátia R Correia
- b Department of Pediatrics , Centro Hospitalar Lisboa Ocidental, EPE , Lisbon , Portugal
| | - Lurdes Lopes
- c Endocrinology Unit, Department of Pediatrics , Centro Hospitalar Lisboa Central, EPE , Lisbon , Portugal
| |
Collapse
|
32
|
Parish AJ, Nguyen V, Goodman AM, Murugesan K, Frampton GM, Kurzrock R. GNAS, GNAQ, and GNA11 alterations in patients with diverse cancers. Cancer 2018; 124:4080-4089. [PMID: 30204251 DOI: 10.1002/cncr.31724] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/23/2018] [Accepted: 06/26/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Advances in deep sequencing technology have uncovered a widespread, protumorigenic role of guanine nucleotide-binding (G protein) α (GNA) subunits, particularly GNA subunits Gs (GNAS), Gq (GNAQ), and G11 (GNA11) (GNA*), in a diverse collection of malignancies. The objectives of the current study were: 1) to determine GNA* aberration status in a cohort of 1348 patients with cancer and 2) to examine tumor mutational burden, overall survival rates, and treatment outcomes in patients with GNA*-positive tumors versus those with tumors that had wild-type GNA*. METHODS For each patient, clinical and genomic data were collected from medical records. Next-generation sequencing was performed for each patient (range, 182-236 genes). RESULTS Aberrations of GNA* genes were identified in a subset of patients who had 8 of the 12 cancer types examined, and a significant association was observed for appendiceal cancer and ocular melanoma (P < .0001 for both; multivariate analysis). Overall, 4.1% of the cancer population was affected. GNA* abnormalities were associated with higher numbers of co-alterations in univariate (but not multivariate) analysis and were most commonly accompanied by Aurora kinase A (AURKA), Cbl proto-oncogene (CBL), and LYN proto-oncogene (LYN) co-alterations (all P < .0001; multivariate analysis). GNA* alterations were correlated with a trend toward lower median overall survival (P = .085). The median tumor mutational burden was 4 mutations per megabase in both GNA*-altered and GNA* wild-type tumors. For this limited sample of GNA*-positive patients, longer survival was not correlated with any specific treatment regimens. CONCLUSIONS In the current sample, the genes GNAS, GNAQ, and GNA11 were widely altered across cancer types, and these alterations often were accompanied by specific genomic abnormalities in AURKA, CBL, and LYN. Therefore, targeting GNA* alterations may require drugs that address the GNA* signal and important co-alterations. Cancer 2018;00:000-000. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Austin J Parish
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, Louisiana Jolla, California
| | - Vi Nguyen
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, Louisiana Jolla, California
| | - Aaron M Goodman
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, Louisiana Jolla, California.,Division of Blood and Marrow Transplantation, Department of Medicine, University of California San Diego, Moores Cancer Center, Louisiana Jolla, California.,Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, Louisiana Jolla, California
| | | | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, Louisiana Jolla, California.,Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, Louisiana Jolla, California
| |
Collapse
|
33
|
Mantovani G, Bastepe M, Monk D, de Sanctis L, Thiele S, Usardi A, Ahmed SF, Bufo R, Choplin T, De Filippo G, Devernois G, Eggermann T, Elli FM, Freson K, García Ramirez A, Germain-Lee EL, Groussin L, Hamdy N, Hanna P, Hiort O, Jüppner H, Kamenický P, Knight N, Kottler ML, Le Norcy E, Lecumberri B, Levine MA, Mäkitie O, Martin R, Martos-Moreno GÁ, Minagawa M, Murray P, Pereda A, Pignolo R, Rejnmark L, Rodado R, Rothenbuhler A, Saraff V, Shoemaker AH, Shore EM, Silve C, Turan S, Woods P, Zillikens MC, Perez de Nanclares G, Linglart A. Diagnosis and management of pseudohypoparathyroidism and related disorders: first international Consensus Statement. Nat Rev Endocrinol 2018; 14:476-500. [PMID: 29959430 PMCID: PMC6541219 DOI: 10.1038/s41574-018-0042-0] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This Consensus Statement covers recommendations for the diagnosis and management of patients with pseudohypoparathyroidism (PHP) and related disorders, which comprise metabolic disorders characterized by physical findings that variably include short bones, short stature, a stocky build, early-onset obesity and ectopic ossifications, as well as endocrine defects that often include resistance to parathyroid hormone (PTH) and TSH. The presentation and severity of PHP and its related disorders vary between affected individuals with considerable clinical and molecular overlap between the different types. A specific diagnosis is often delayed owing to lack of recognition of the syndrome and associated features. The participants in this Consensus Statement agreed that the diagnosis of PHP should be based on major criteria, including resistance to PTH, ectopic ossifications, brachydactyly and early-onset obesity. The clinical and laboratory diagnosis should be confirmed by a molecular genetic analysis. Patients should be screened at diagnosis and during follow-up for specific features, such as PTH resistance, TSH resistance, growth hormone deficiency, hypogonadism, skeletal deformities, oral health, weight gain, glucose intolerance or type 2 diabetes mellitus, and hypertension, as well as subcutaneous and/or deeper ectopic ossifications and neurocognitive impairment. Overall, a coordinated and multidisciplinary approach from infancy through adulthood, including a transition programme, should help us to improve the care of patients affected by these disorders.
Collapse
Affiliation(s)
- Giovanna Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Murat Bastepe
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Monk
- Imprinting and Cancer Group, Cancer Epigenetic and Biology Program (PEBC), Institut d'Investigació Biomedica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Luisa de Sanctis
- Pediatric Endocrinology Unit, Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Susanne Thiele
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Alessia Usardi
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- APHP, Endocrinology and diabetes for children, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Roberto Bufo
- IPOHA, Italian Progressive Osseous Heteroplasia Association, Cerignola, Foggia, Italy
| | - Timothée Choplin
- K20, French PHP and related disorders patient association, Jouars Pontchartrain, France
| | - Gianpaolo De Filippo
- APHP, Department of medicine for adolescents, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Guillemette Devernois
- K20, French PHP and related disorders patient association, Jouars Pontchartrain, France
| | - Thomas Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Francesca M Elli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Aurora García Ramirez
- AEPHP, Spanish PHP and related disorders patient association, Huércal-Overa, Almería, Spain
| | - Emily L Germain-Lee
- Albright Center & Center for Rare Bone Disorders, Division of Pediatric Endocrinology & Diabetes, Connecticut Children's Medical Center, Farmington, CT, USA
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lionel Groussin
- APHP, Department of Endocrinology, Cochin Hospital (HUPC), Paris, France
- University of Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Neveen Hamdy
- Department of Medicine, Division of Endocrinology and Centre for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
| | - Patrick Hanna
- INSERM U1169, Bicêtre Paris Sud, Paris Sud - Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Olaf Hiort
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Harald Jüppner
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Kamenický
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- APHP, Department of Endocrinology and Reproductive Diseases, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- INSERM U1185, Paris Sud - Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Nina Knight
- UK acrodysostosis patients' group, London, UK
| | - Marie-Laure Kottler
- Department of Genetics, Reference Centre for Rare Disorders of Calcium and Phosphate Metabolism, Caen University Hospital, Caen, France
- BIOTARGEN, UNICAEN, Normandie University, Caen, France
| | - Elvire Le Norcy
- University of Paris Descartes, Sorbonne Paris Cité, Paris, France
- APHP, Department of Odontology, Bretonneau Hospital (PNVS), Paris, France
| | - Beatriz Lecumberri
- Department of Endocrinology and Nutrition, La Paz University Hospital, Madrid, Spain
- Department of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain
- Endocrine Diseases Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Michael A Levine
- Division of Endocrinology and Diabetes and Center for Bone Health, Children's Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Outi Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Regina Martin
- Osteometabolic Disorders Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Hospital das Clínicas HCFMUSP, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Gabriel Ángel Martos-Moreno
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, CIBERobn, ISCIII, Madrid, Spain
- Department of Pediatrics, Autonomous University of Madrid (UAM), Madrid, Spain
- Endocrine Diseases Research Group, Hospital La Princesa Institute for Health Research (IIS La Princesa), Madrid, Spain
| | | | - Philip Murray
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Arrate Pereda
- Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, Hospital Universitario Araba-Txagorritxu, Vitoria-Gasteiz, Alava, Spain
| | | | - Lars Rejnmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Rebecca Rodado
- AEPHP, Spanish PHP and related disorders patient association, Huércal-Overa, Almería, Spain
| | - Anya Rothenbuhler
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- APHP, Endocrinology and diabetes for children, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Vrinda Saraff
- Department of Endocrinology and Diabetes, Birmingham Children's Hospital, Birmingham, UK
| | - Ashley H Shoemaker
- Pediatric Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eileen M Shore
- Departments of Orthopaedic Surgery and Genetics, Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Silve
- APHP, Service de Biochimie et Génétique Moléculaires, Hôpital Cochin, Paris, France
| | - Serap Turan
- Department of Pediatrics, Division of Endocrinology and Diabetes, Marmara University, Istanbul, Turkey
| | | | - M Carola Zillikens
- Department of Internal Medicine, Bone Center Erasmus MC - University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Guiomar Perez de Nanclares
- Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, Hospital Universitario Araba-Txagorritxu, Vitoria-Gasteiz, Alava, Spain.
| | - Agnès Linglart
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France.
- APHP, Endocrinology and diabetes for children, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France.
- INSERM U1169, Bicêtre Paris Sud, Paris Sud - Paris Saclay University, Le Kremlin-Bicêtre, France.
| |
Collapse
|
34
|
Mantovani G, Elli FM. Multiple hormone resistance and alterations of G-protein-coupled receptors signaling. Best Pract Res Clin Endocrinol Metab 2018; 32:141-154. [PMID: 29678282 DOI: 10.1016/j.beem.2018.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Metabolic disorders deriving from the non-responsiveness of target organs to hormones, which manifest clinically similar to the deficiency of a given hormone itself, derive from molecular alterations affecting specific hormone receptors. Pseudohypoparathyroidism (PHP) and related disorders exemplify an unusual form of hormone resistance as the underlying molecular defect is a partial deficiency of the α subunit of the stimulatory G protein (Gsα), a key regulator of cAMP signaling pathway, or, as more recently described, of downstream effector proteins of the same pathway, such as PKA regulatory subunit 1A (R1A) and phosphodyestarase type 4D (PDE4D). In this group of diseases, resistance to hormones such as PTH, TSH, gonadotropins and GHRH may be variably present, so that the clinical and molecular overlap among these different but related disorders represents a challenge for endocrinologists as to differential diagnosis and genetic counseling. This review will describe the presenting features of multiple resistance in PHP and related disorders, focusing on both our current understanding and future challenges.
Collapse
Affiliation(s)
- Giovanna Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Francesca Marta Elli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| |
Collapse
|
35
|
Abstract
GNAS is a complex imprinted gene encoding the alpha-subunit of the stimulatory heterotrimeric G protein (Gsα). GNAS gives rise to additional gene products that exhibit exclusively maternal or paternal expression, such as XLαs, a large variant of Gsα that shows exclusively paternal expression and is partly identical to the latter. Gsα itself is expressed biallelically in most tissues, although the expression occurs predominantly from the maternal allele in a small set of tissues, such as renal proximal tubules. Inactivating mutations in Gsα-coding GNAS exons are responsible for Albright's hereditary osteodystrophy (AHO), which refers to a constellation of physical and developmental disorders including obesity, short stature, brachydactyly, cognitive impairment, and heterotopic ossification. Patients with Gsα mutations can present with AHO in the presence or absence of end-organ resistance to multiple hormones including parathyroid hormone. Maternal Gsα mutations lead to AHO with hormone resistance (i.e. pseudohypoparathyroidism type-Ia), whereas paternal mutations cause AHO alone (i.e. pseudo-pseudohypoparathyroidism). Heterotopic ossification associated with AHO develops through intramembranous bone formation and is limited to dermis and subcutis. In rare cases carrying Gsα mutations, however, ossifications progress into deep connective tissue and skeletal muscle, a disorder termed progressive osseous heteroplasia (POH). Here I briefly review the genetic, clinical, and molecular aspects of these disorders caused by inactivating GNAS mutations, with particular emphasis on heterotopic ossification.
Collapse
Affiliation(s)
- Murat Bastepe
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, United States.
| |
Collapse
|
36
|
Chu X, Zhu Y, Wang O, Nie M, Quan T, Xue Y, Wang W, Jiang Y, Li M, Xia W, Xing X. Clinical and genetic characteristics of Pseudohypoparathyroidism in the Chinese population. Clin Endocrinol (Oxf) 2018; 88:285-294. [PMID: 29136292 DOI: 10.1111/cen.13516] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Pseudohypoparathyroidism (PHP) is caused by mutations and epimutations in the GNAS locus, and characterized by the possibility of resistance to multiple hormones and Albright's hereditary osteodystrophy. PHP can be classified into the forms 1A/C, sporadic 1B and familial 1B. OBJECTIVES To obtain an overall view of the clinical and genetic characteristics of the Chinese PHP patient population. METHODS From 2000 to 2016, 120 patients were recruited and studied using Sanger sequencing, methylation-specific multiple ligation-dependent probe amplification (MS-MLPA) and combined bisulfite restriction analysis (COBRA). Of these patients, 104 had positive molecular alterations indicative of certain forms of PHP and were included in data analysis. Clinical and laboratory features were compared between PHP1A/C and PHP1B patients. RESULTS Ten PHP1A/C, 21 familial PHP1B and 73 sporadic PHP1B patients were identified. Four novel GNAS mutations were discovered in these patients, including c.1038+1G>T, c.530+2T>C, c.880_883delCAAG and c.311_312delAAG, insT. The most common symptoms in this series were recurrent tetany (89.4%) and epilepsy (47.1%). The prevalence of weight excess increased with age for PHP1B (10%-35%) and PHP1A/C (50%-75%). Intracranial calcification had a prevalence of 94.6% and correlated with seizures (r = .227, P = .029). Cataracts occurred in 56.2% PHP patients, and there was a trend towards longer disease duration in patients with cataracts (P = .051). Statistically significant differences (P < .05) were observed when comparing certain clinical characteristics between PHP1B and PHP1A/C patients, including age of onset (10 vs 7 year), short stature (21.3% vs 70%), rounded face (60.6% vs 100%), brachydactyly (25.5% vs 100%), ectopic ossification (1.1% vs 40%) and TSH resistance (44.6% vs 90%), respectively. CONCLUSIONS This study is the largest single-centre series of PHP patients and summarizes the clinical and genetic features of the Chinese PHP population. While there was substantial clinical overlap between PHP1A/C and PHP1B, differences in disease progression were observed.
Collapse
Affiliation(s)
- Xueying Chu
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yan Zhu
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Ou Wang
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Min Nie
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Tingting Quan
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yu Xue
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Wenbo Wang
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yan Jiang
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Mei Li
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Weibo Xia
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Xiaoping Xing
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences (CAMS), Beijing, China
| |
Collapse
|
37
|
Ray A, Macwan I, Singh S, Silwal S, Patra P. A Computational Approach for Understanding the Interactions between Graphene Oxide and Nucleoside Diphosphate Kinase with Implications for Heart Failure. NANOMATERIALS 2018; 8:nano8020057. [PMID: 29360759 PMCID: PMC5853690 DOI: 10.3390/nano8020057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/13/2018] [Accepted: 01/20/2018] [Indexed: 01/05/2023]
Abstract
During a heart failure, an increased content and activity of nucleoside diphosphate kinase (NDPK) in the sarcolemmal membrane is responsible for suppressing the formation of the second messenger cyclic adenosine monophosphate (cAMP)-a key component required for calcium ion homeostasis for the proper systolic and diastolic functions. Typically, this increased NDPK content lets the surplus NDPK react with a mutated G protein in the beta-adrenergic signal transduction pathway, thereby inhibiting cAMP synthesis. Thus, it is thus that inhibition of NDPK may cause a substantial increase in adenylate cyclase activity, which in turn may be a potential therapy for end-stage heart failure patients. However, there is little information available about the molecular events at the interface of NDPK and any prospective molecule that may potentially influence its reactive site (His118). Here we report a novel computational approach for understanding the interactions between graphene oxide (GO) and NDPK. Using molecular dynamics, it is found that GO interacts favorably with the His118 residue of NDPK to potentially prevent its binding with adenosine triphosphate (ATP), which otherwise would trigger the phosphorylation of the mutated G protein. Therefore, this will result in an increase in cAMP levels during heart failure.
Collapse
Affiliation(s)
- Anushka Ray
- Nashua High School South, Nashua, NH 03062, USA.
| | - Isaac Macwan
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| | - Shrishti Singh
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| | - Sushila Silwal
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| | - Prabir Patra
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| |
Collapse
|
38
|
Salemi P, Skalamera Olson JM, Dickson LE, Germain-Lee EL. Ossifications in Albright Hereditary Osteodystrophy: Role of Genotype, Inheritance, Sex, Age, Hormonal Status, and BMI. J Clin Endocrinol Metab 2018; 103:158-168. [PMID: 29059381 PMCID: PMC5761497 DOI: 10.1210/jc.2017-00860] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 10/16/2017] [Indexed: 12/14/2022]
Abstract
CONTEXT Albright hereditary osteodystrophy (AHO) is caused by heterozygous inactivating mutations in GNAS. Depending on the parental origin of the mutated allele, patients develop either pseudohypoparathyroidism type 1A (PHP1A), with multihormone resistance and severe obesity, or pseudopseudohypoparathyroidism (PPHP), without hormonal abnormalities or marked obesity. Subcutaneous ossifications (SCOs) are a source of substantial morbidity in both PHP1A and PPHP. OBJECTIVE This study investigated the previously undetermined prevalence of SCO formation in PHP1A vs PPHP as well as possible correlations with genotype, sex, age, hormonal resistance, and body mass index (BMI). DESIGN This study evaluated patients with AHO for SCOs by physical examination performed by one consistent physician over 16 years. SETTING Albright Clinic, Kennedy Krieger Institute; Institute for Clinical and Translational Research, Johns Hopkins Hospital; Albright Center, Connecticut Children's Medical Center. PATIENTS We evaluated 67 patients with AHO (49 with PHP1A, 18 with PPHP) with documented mutations in GNAS. MAIN OUTCOME MEASURES Relationships of SCOs to genotype, sex, age, hormonal resistance, and BMI. RESULTS Forty-seven of 67 participants (70.1%) had SCOs. Patients with PHP1A and PPHP had similar prevalences and degrees of ossification formation. Patients with frameshift and nonsense mutations had much more extensive SCOs than those with missense mutations. Males were affected more than females. There was no correlation with hormonal status or BMI. CONCLUSIONS There is a similar prevalence of SCOs in PHP1A and PPHP, and the extent of SCO formation correlates with the severity of the mutation. Males are affected more extensively than females, and the SCOs tend to worsen with age.
Collapse
Affiliation(s)
- Parissa Salemi
- Department of Pediatrics, Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Lauren E Dickson
- Albright Center and Center for Rare Bone Disorders, Division of Pediatric Endocrinology & Diabetes, Connecticut Children's Medical Center, Farmington, Connecticut
| | - Emily L Germain-Lee
- Department of Pediatrics, Division of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Albright Clinic, Kennedy Krieger Institute, Baltimore, Maryland
- Albright Center and Center for Rare Bone Disorders, Division of Pediatric Endocrinology & Diabetes, Connecticut Children's Medical Center, Farmington, Connecticut
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, Connecticut
| |
Collapse
|
39
|
Turan S. Current Nomenclature of Pseudohypoparathyroidism: Inactivating Parathyroid Hormone/Parathyroid Hormone-Related Protein Signaling Disorder. J Clin Res Pediatr Endocrinol 2017; 9:58-68. [PMID: 29280743 PMCID: PMC5790322 DOI: 10.4274/jcrpe.2017.s006] [Citation(s) in RCA: 5] [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] [Indexed: 01/16/2023] Open
Abstract
Disorders related to parathyroid hormone (PTH) resistance and PTH signaling pathway impairment are historically classified under the term of pseudohypoparathyroidism (PHP). The disease was first described and named by Fuller Albright and colleagues in 1942. Albright hereditary osteodystrophy (AHO) is described as an associated clinical entity with PHP, characterized by brachydactyly, subcutaneous ossifications, round face, short stature and a stocky build. The classification of PHP is further divided into PHP-Ia, pseudo-PHP (pPHP), PHP-Ib, PHP-Ic and PHP-II according to the presence or absence of AHO, together with an in vivo response to exogenous PTH and the measurement of Gsα protein activity in peripheral erythrocyte membranes in vitro. However, PHP classification fails to differentiate all patients with different clinical and molecular findings for PHP subtypes and classification become more complicated with more recent molecular characterization and new forms having been identified. So far, new classifications have been established by the EuroPHP network to cover all disorders of the PTH receptor and its signaling pathway. Inactivating PTH/PTH-related protein signaling disorder (iPPSD) is the new name proposed for a group of these disorders and which can be further divided into subtypes - iPPSD1 to iPPSD6. These are termed, starting from PTH receptor inactivation mutation (Eiken and Blomstrand dysplasia) as iPPSD1, inactivating Gsα mutations (PHP-Ia, PHP-Ic and pPHP) as iPPSD2, loss of methylation of GNAS DMRs (PHP-Ib) as iPPSD3, PRKAR1A mutations (acrodysostosis type 1) as iPPSD4, PDE4D mutations (acrodysostosis type 2) as iPPSD5 and PDE3A mutations (autosomal dominant hypertension with brachydactyly) as iPPSD6. iPPSDx is reserved for unknown molecular defects and iPPSDn+1 for new molecular defects which are yet to be described. With these new classifications, the aim is to clarify the borders of each different subtype of disease and make the classification according to molecular pathology. The iPPSD group is designed to be expandable and new classifications will readily fit into it as necessary.
Collapse
Affiliation(s)
- Serap Turan
- Marmara University Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey
,* Address for Correspondence: Marmara University Faculty of Medicine, Department of Pediatric Endocrinology, İstanbul, Turkey Phone: +90 216 625 45 45 E-mail:
| |
Collapse
|
40
|
Reyes M, Karaca A, Bastepe M, Gulcelik NE, Jüppner H. A novel deletion involving GNAS exon 1 causes PHP1A and further refines the region required for normal methylation at exon A/B. Bone 2017; 103:281-286. [PMID: 28711660 PMCID: PMC5943703 DOI: 10.1016/j.bone.2017.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/20/2017] [Accepted: 07/11/2017] [Indexed: 11/25/2022]
Abstract
GNAS exons 1-13 encode the biallelically expressed alpha-subunit of the stimulatory G protein (Gαs). Additional transcripts derived from this locus use alternative first exons that undergo parent-specific methylation, thus allowing transcription only from the non-modified allele. Pseudohypoparathyroidism type Ia (PHP1A) is characterized by Albright's Hereditary Osteodystrophy (AHO) and resistance to multiple hormones; this disorder is caused by maternal inactivating mutations involving Gαs exons. In contrast, pseudohypoparathyroidism type Ib (PHP1B) is characterized mostly by resistance to PTH and often mild TSH resistance, usually without AHO features. The autosomal dominant variant of PHP1B (AD-PHP1B) is caused by maternal deletions in GNAS or STX16 that reduce Gαs expression through loss-of-methylation at GNAS exon A/B alone or at multiple differentially methylated regions (DMR). Several large maternal deletions involve not only GNAS exons 1-13, but also one or several GNAS DMRs, thus causing PHP1A combined with apparent GNAS epigenetic changes that are indistinguishable from those observed in PHP1B. Some of these deletions include a large CpG island extending from exon A/B to the intron between GNAS exons 1 and 2, but there is no evidence for parent-specific exon 1 methylation. We now describe a family in which the female proband and her daughter presented with hypocalcemia, elevated PTH levels, shortened metacarpals, and obesity, but without obvious neurocognitive abnormalities. A maternally inherited 2015-bp deletion that includes GNAS exon 1 was identified thereby establishing the diagnosis of PHP1A. The centromeric deletion breakpoint is located 178bp upstream of exon 1, yet no methylation changes were observed at exon A/B. This novel deletion therefore refines further the region between exon A/B and exon 1 that is critical for establishing or maintaining normal methylation at GNAS exon A/B.
Collapse
Affiliation(s)
- Monica Reyes
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anara Karaca
- Ankara Training and Research Hospital, Endocrinology and Metabolism, Ankara, Turkey
| | - Murat Bastepe
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nese Ersoz Gulcelik
- Ankara Training and Research Hospital, Endocrinology and Metabolism, Ankara, Turkey
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Bastepe M, Turan S, He Q. Heterotrimeric G proteins in the control of parathyroid hormone actions. J Mol Endocrinol 2017; 58:R203-R224. [PMID: 28363951 PMCID: PMC5650080 DOI: 10.1530/jme-16-0221] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 02/17/2017] [Indexed: 12/17/2022]
Abstract
Parathyroid hormone (PTH) is a key regulator of skeletal physiology and calcium and phosphate homeostasis. It acts on bone and kidney to stimulate bone turnover, increase the circulating levels of 1,25 dihydroxyvitamin D and calcium and inhibit the reabsorption of phosphate from the glomerular filtrate. Dysregulated PTH actions contribute to or are the cause of several endocrine disorders. This calciotropic hormone exerts its actions via binding to the PTH/PTH-related peptide receptor (PTH1R), which couples to multiple heterotrimeric G proteins, including Gs and Gq/11 Genetic mutations affecting the activity or expression of the alpha-subunit of Gs, encoded by the GNAS complex locus, are responsible for several human diseases for which the clinical findings result, at least partly, from aberrant PTH signaling. Here, we review the bone and renal actions of PTH with respect to the different signaling pathways downstream of these G proteins, as well as the disorders caused by GNAS mutations.
Collapse
Affiliation(s)
- Murat Bastepe
- Endocrine UnitDepartment of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Serap Turan
- Department of Pediatric EndocrinologyMarmara University School of Medicine, Istanbul, Turkey
| | - Qing He
- Endocrine UnitDepartment of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Elhamamsy AR. Role of DNA methylation in imprinting disorders: an updated review. J Assist Reprod Genet 2017; 34:549-562. [PMID: 28281142 PMCID: PMC5427654 DOI: 10.1007/s10815-017-0895-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022] Open
Abstract
Genomic imprinting is a complex epigenetic process that contributes substantially to embryogenesis, reproduction, and gametogenesis. Only small fraction of genes within the whole genome undergoes imprinting. Imprinted genes are expressed in a monoallelic parent-of-origin-specific manner, which means that only one of the two inherited alleles is expressed either from the paternal or maternal side. Imprinted genes are typically arranged in clusters controlled by differentially methylated regions or imprinting control regions. Any defect or relaxation in imprinting process can cause loss of imprinting in the key imprinted loci. Loss of imprinting in most cases has a harmful effect on fetal development and can result in neurological, developmental, and metabolic disorders. Since DNA methylation and histone modifications play a key role in the process of imprinting. This review focuses on the role of DNA methylation in imprinting process and describes DNA methylation aberrations in different imprinting disorders.
Collapse
Affiliation(s)
- Amr Rafat Elhamamsy
- Department of Clinical Pharmacy, School of Pharmacy, Tanta University, Tanta, 31512, Gharbia, Egypt.
| |
Collapse
|
43
|
Tafaj O, Jüppner H. Pseudohypoparathyroidism: one gene, several syndromes. J Endocrinol Invest 2017; 40:347-356. [PMID: 27995443 DOI: 10.1007/s40618-016-0588-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 11/25/2016] [Indexed: 01/04/2023]
Abstract
Pseudohypoparathyroidism (PHP) and pseudopseudohypoparathyroidism (PPHP) are caused by mutations and/or epigenetic changes at the complex GNAS locus on chromosome 20q13.3 that undergoes parent-specific methylation changes at several sites. GNAS encodes the alpha-subunit of the stimulatory G protein (Gsα) and several splice variants thereof. Heterozygous inactivating mutations involving the maternal GNAS exons 1-13 cause PHP type Ia (PHP1A). Because of much reduced paternal Gsα expression in certain tissues, such as the proximal renal tubules, thyroid, and pituitary, there is little or no Gsα protein in the presence of maternal GNAS mutations, thus leading to PTH-resistant hypocalcemia and hyperphosphatemia. When located on the paternal allele, the same or similar GNAS mutations are the cause of PPHP. Besides biochemical abnormalities, patients affected by PHP1A show developmental abnormalities, referred to as Albrights hereditary osteodystrophy (AHO). Some, but not all of these AHO features are encountered also in patients affected by PPHP, who typically show no laboratory abnormalities. Autosomal dominant PHP type Ib (AD-PHP1B) is caused by heterozygous maternal deletions within GNAS or STX16, which are associated with loss-of-methylation (LOM) at exon A/B alone or at all maternally methylated GNAS exons. LOM at exon A/B and the resulting biallelic expression of A/B transcripts reduces Gsα expression, thus leading to hormonal resistance. Epigenetic changes at all differentially methylated GNAS regions are also observed in sporadic PHP1B, the most frequent disease variant, which remains unresolved at the molecular level, except for rare cases with paternal uniparental isodisomy or heterodisomy of chromosome 20q (patUPD20q).
Collapse
Affiliation(s)
- O Tafaj
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Thier 10, 50 Blossom Street, Boston, MA, 02114, USA
| | - H Jüppner
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Thier 10, 50 Blossom Street, Boston, MA, 02114, USA.
- Pediatric Nephrology Unit, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
Grigelioniene G, Nevalainen PI, Reyes M, Thiele S, Tafaj O, Molinaro A, Takatani R, Ala-Houhala M, Nilsson D, Eisfeldt J, Lindstrand A, Kottler ML, Mäkitie O, Jüppner H. A Large Inversion Involving GNAS Exon A/B and All Exons Encoding Gsα Is Associated With Autosomal Dominant Pseudohypoparathyroidism Type Ib (PHP1B). J Bone Miner Res 2017; 32:776-783. [PMID: 28084650 PMCID: PMC5395346 DOI: 10.1002/jbmr.3083] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/31/2016] [Accepted: 01/11/2017] [Indexed: 01/08/2023]
Abstract
Pseudohypoparathyroidism type Ib (PHP1B) is characterized primarily by resistance to parathyroid hormone (PTH) and thus hypocalcemia and hyperphosphatemia, in most cases without evidence for Albright hereditary osteodystrophy (AHO). PHP1B is associated with epigenetic changes at one or several differentially-methylated regions (DMRs) within GNAS, which encodes the α-subunit of the stimulatory G protein (Gsα) and splice variants thereof. Heterozygous, maternally inherited STX16 or GNAS deletions leading to isolated loss-of-methylation (LOM) at exon A/B alone or at all maternal DMRs are the cause of autosomal dominant PHP1B (AD-PHP1B). In this study, we analyzed three affected individuals, the female proband and her two sons. All three revealed isolated LOM at GNAS exon A/B, whereas the proband's healthy maternal grandmother and uncle showed normal methylation at this locus. Haplotype analysis was consistent with linkage to the STX16/GNAS region, yet no deletion could be identified. Whole-genome sequencing of one of the patients revealed a large heterozygous inversion (1,882,433 bp). The centromeric breakpoint of the inversion is located 7,225 bp downstream of GNAS exon XL, but its DMR showed no methylation abnormality, raising the possibility that the inversion disrupts a regulatory element required only for establishing or maintaining exon A/B methylation. Because our three patients presented phenotypes consistent with PHP1B, and not with PHP1A, the Gsα promoter is probably unaffected by the inversion. Our findings expand the spectrum of genetic mutations that lead to LOM at exon A/B alone and thus biallelic expression of the transcript derived from this alternative first GNAS exon. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Giedre Grigelioniene
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital Stockholm, Stockholm, Sweden
| | - Pasi I Nevalainen
- Endocrine Unit, Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Monica Reyes
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Susanne Thiele
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Olta Tafaj
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Angelo Molinaro
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rieko Takatani
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marja Ala-Houhala
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Daniel Nilsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital Stockholm, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet Science Park, Solna, Sweden
| | - Jesper Eisfeldt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet Science Park, Solna, Sweden
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital Stockholm, Stockholm, Sweden
| | - Marie-Laure Kottler
- Centre Hospitalier Universitaire de Caen, Department of Genetics, Reference Centre for Rare Disorders of Calcium and Phosphorus Metabolism, Caen, France
| | - Outi Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Abstract
Hypothyroidism may occur in association with congenital parathyroid disorders determining parathyroid hormone insufficiency, which is characterized by hypocalcemia and concomitant inappropriately low secretion of parathormone (PTH). The association is often due to loss of function of genes common to thyroid and parathyroid glands embryonic development. Hypothyroidism associated with hypoparathyroidism is generally mild and not associated with goiter; moreover, it is usually part of a multisystemic involvement not restricted to endocrine function as occurs in patients with 22q11 microdeletion/DiGeorge syndrome, the most frequent disorders. Hypothyroidism and hypoparathyroidism may also follow endocrine glands' damages due to autoimmunity or chronic iron overload in thalassemic disorders, both genetically determined conditions. Finally, besides PTH deficiency, hypocalcemia can be due to PTH resistance in pseudohypoparathyroidism; when hormone resistance is generalized, patients can suffer from hypothyroidism due to TSH resistance. In evaluating patients with hypothyroidism and hypocalcemia, physical examination and clinical history are essential to drive the diagnostic process, while routine genetic screening is not recommended.
Collapse
Affiliation(s)
- Giovanna Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Francesca Marta Elli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Sabrina Corbetta
- Endocrinology Service, Department of Biomedical Sciences, University of Milan, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.
| |
Collapse
|
46
|
de Sanctis L, Giachero F, Mantovani G, Weber G, Salerno M, Baroncelli GI, Elli MF, Matarazzo P, Wasniewska M, Mazzanti L, Scirè G, Tessaris D. Genetic and epigenetic alterations in the GNAS locus and clinical consequences in Pseudohypoparathyroidism: Italian common healthcare pathways adoption. Ital J Pediatr 2016; 42:101. [PMID: 27871293 PMCID: PMC5117549 DOI: 10.1186/s13052-016-0310-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 11/12/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Genetic and epigenetic alterations in the GNAS locus are responsible for the Gsα protein dysfunctions causing Pseudohypoparathyroidism (PHP) type Ia/c and Ib, respectively. For these heterogeneous diseases characterized by multiple hormone resistances and Albright's Hereditary Osteodystrophy (AHO) the current classification results inadequate because of the clinical overlap between molecular subtypes and a standard clinical approach is still missing. In the present paper several members of the Study Group Endocrine diseases due to altered function of Gsα protein of the Italian Society of Pediatric Endocrinology and Diabetology (ISPED) have reviewed and updated the clinical-molecular data of the largest case series of (epi)/genetically characterized AHO/PHP patients; they then produced a common healthcare pathway for patients with these disorders. METHODS The molecular analysis of the GNAS gene and locus identified the causal alteration in 74 subjects (46 genetic and 28 epigenetic mutations). The clinical data at the diagnosis and their evolution during up to 15 years follow-up were collected using two different cards. RESULTS In patients with genetic mutations the growth impairment worsen during the time, while obesity prevalence decreases; subcutaneous ossifications seem specific for this group. Brachydactyly has been detected in half of the subjects with epigenetic alterations, in which the disease overts later in life, often with symptomatic hypocalcaemia, and also early TSH and GHRH resistances have been recorded. CONCLUSIONS A dedicated healthcare pathway addressing all these aspects in a systematic way would improve the clinical management, allowing an earlier recognition of some PHP features, the optimization of their medical treatment and a better clinical-oriented molecular analysis. Furthermore, standardized follow-up data would provide new insight into less known aspects.
Collapse
Affiliation(s)
- L de Sanctis
- Department of Public Health and Pediatric Sciences, University of Turin - Regina Margherita Children's Hospital - Health and Science City, Subintensiva Allargata Prima Infanzia, Piazza Polonia 94, 10126, Torino, Italy.
| | - F Giachero
- Kinderklinik, Evangelisches Krankenhaus Oberhausen, Oberhausen, Germany
| | - G Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - G Weber
- Department of Pediatrics, San Raffaele Hospital, University of Milan, Milan, Italy
| | - M Salerno
- Pediatric Endocrine Unit, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - G I Baroncelli
- Department of Obstetrics, Gynecology and Pediatrics, I Pediatric Division, University Hospital, Pisa, Italy
| | - M F Elli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - P Matarazzo
- Pediatric Endocrinology and Diabetology Unit, Regina Margherita Children's Hospital - Health and Science City, Turin, Italy
| | - M Wasniewska
- Department of Pediatric, Gynecological, Microbiological and Biomedical Sciences, University of Messina, Messina, Italy
| | - L Mazzanti
- Pediatric Endocrinology and Rare Diseases, Department of Pediatrics, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - G Scirè
- Endocrinology Ward, Bambin Gesù Children's Hospital, Rome, Italy
| | - D Tessaris
- Department of Public Health and Pediatric Sciences, University of Turin - Regina Margherita Children's Hospital - Health and Science City, Subintensiva Allargata Prima Infanzia, Piazza Polonia 94, 10126, Torino, Italy
| | | |
Collapse
|
47
|
de Lange IM, Verrijn Stuart AA, van der Luijt RB, Ploos van Amstel HK, van Haelst MM. Macrosomia, obesity, and macrocephaly as first clinical presentation of PHP1b caused by STX16 deletion. Am J Med Genet A 2016; 170:2431-5. [PMID: 27338644 DOI: 10.1002/ajmg.a.37818] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 06/12/2016] [Indexed: 01/21/2023]
Abstract
Pseudohypoparathyroidism (PHP) is a genetic disorder with resistance to parathyroid hormone (PTH) as most important feature. Main subtypes of the disease are pseudohypoparathyroidism 1b (PHP1b) and pseudohypoparathyroidism 1a (PHP1a). PHP1b is characterized by PTH resistance of the renal cortex due to reduced activity of the stimulatory G protein α subunit (Gsα) of the PTH receptor. In addition to resistance to PTH, PHP1a patients also lack sensitivity for other hormones that signal their actions through G protein-coupled receptors and display physical features of Albright hereditary osteodystrophy (AHO), which is not classically seen in PHP1b patients. PHP1a is caused by heterozygous loss-of-function mutations in maternally inherited GNAS exons 1-13, which encode Gsα. PHP1b is often caused by deletion of the STX16 gene, which is thought to have an important role in controlling the methylation and thus imprinting at part of the GNAS locus. Here we present a patient with PHP1b caused by the previously described recurrent 3-kb STX16 deletion. The patient's first symptoms were macrosomia, early onset obesity, and macrocephaly. Since this is an atypical but previously described rare presentation of PHP1b, we reemphasize STX16 deletions and PHP1b as a rare cause for early onset obesity and macrosomia. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Iris M de Lange
- Department of Medical Genetics, University Medical Centre, Utrecht, The Netherlands
| | | | - Rob B van der Luijt
- Department of Medical Genetics, University Medical Centre, Utrecht, The Netherlands
| | | | - Mieke M van Haelst
- Department of Medical Genetics, University Medical Centre, Utrecht, The Netherlands
| |
Collapse
|
48
|
Linglart A, Maupetit-Méhouas S, Silve C. GNAS -Related Loss-of-Function Disorders and the Role of Imprinting. Horm Res Paediatr 2016; 79:119-29. [PMID: 23548772 DOI: 10.1159/000348516] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 01/27/2013] [Indexed: 11/19/2022] Open
Abstract
GNAS (guanine nucleotide-binding protein, α stimulating) is a complex imprinted locus coding, besides the α-stimulatory subunit of the G protein, the paternally (extra-large, antisense and A/B) and maternally (neuroendocrine secretory protein) transcripts. Heterozygous mutations in the coding sequence of GNAS produce dominant phenotypes (combination of resistances to hormones signaling through G-protein-coupled receptors, osteodystrophy and obesity) that depend on the parental origin of the mutated allele. Likewise, alterations in the methylation at promoters of GNAS transcripts, associated or not with deletions of imprinting control regions in the nearby STX16 gene or within GNAS, prompt resistance to parathormone when affecting the maternal allele. Therefore, imprinting of GNAS is the determining factor for the variability of the phenotype. Knowledge of the various phenotypes is necessary for genetic counseling as well as an appropriate therapeutic balance between regular follow-up, prevention of disease complications and iatrogeny.
Collapse
Affiliation(s)
- Agnès Linglart
- Endocrinology and Diabetology for Children and French Center of Reference for Rare Disorders of Calcium and Phosphorus Metabolism, Bicêtre-Paris-Sud Hospital, Le Kremlin Bicêtre, France
| | | | | |
Collapse
|
49
|
Mantovani G, Spada A, Elli FM. Pseudohypoparathyroidism and Gsα-cAMP-linked disorders: current view and open issues. Nat Rev Endocrinol 2016; 12:347-56. [PMID: 27109785 DOI: 10.1038/nrendo.2016.52] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pseudohypoparathyroidism exemplifies an unusual form of hormone resistance as the underlying molecular defect is a partial deficiency of the α subunit of the stimulatory G protein (Gsα), a key regulator of the cAMP signalling pathway, rather than of the parathyroid hormone (PTH) receptor itself. Despite the first description of this disorder dating back to 1942, later findings have unveiled complex epigenetic alterations in addition to classic mutations in GNAS underpining the molecular basis of the main subtypes of pseudohypoparathyroidism. Moreover, mutations in PRKAR1A and PDE4D, which encode proteins crucial for Gsα-cAMP-mediated signalling, have been found in patients with acrodysostosis. As acrodysostosis, a disease characterized by skeletal malformations and endocrine disturbances, shares clinical and molecular characteristics with pseudohypoparathyroidism, making a differential diagnosis and providing genetic counselling to patients and families is a challenge for endocrinologists. Accumulating data on the genetic and clinical aspects of this group of diseases highlight the limitation of the current classification system and prompt the need for a new definition as well as for new diagnostic and/or therapeutic algorithms. This Review discusses both the current understanding and future challenges for the clinical and molecular diagnosis, classification and treatment of pseudohypoparathyroidism.
Collapse
MESH Headings
- Bone Diseases, Metabolic/diagnosis
- Bone Diseases, Metabolic/genetics
- Chromogranins/genetics
- Chromosome Deletion
- Chromosomes, Human, Pair 2/genetics
- Cyclic AMP
- Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 4/genetics
- Diagnosis, Differential
- Dysostoses/diagnosis
- Dysostoses/genetics
- Epigenesis, Genetic/genetics
- GTP-Binding Protein alpha Subunits, Gs/genetics
- Humans
- Intellectual Disability/diagnosis
- Intellectual Disability/genetics
- Ossification, Heterotopic/diagnosis
- Ossification, Heterotopic/genetics
- Osteochondrodysplasias/diagnosis
- Osteochondrodysplasias/genetics
- Pseudohypoparathyroidism/classification
- Pseudohypoparathyroidism/diagnosis
- Pseudohypoparathyroidism/genetics
- Signal Transduction
- Skin Diseases, Genetic/diagnosis
- Skin Diseases, Genetic/genetics
Collapse
Affiliation(s)
- Giovanna Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Via Francesco Sforza 35, Milan 20122, Italy
| | - Anna Spada
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Via Francesco Sforza 35, Milan 20122, Italy
| | - Francesca Marta Elli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Via Francesco Sforza 35, Milan 20122, Italy
| |
Collapse
|
50
|
Takatani R, Molinaro A, Grigelioniene G, Tafaj O, Watanabe T, Reyes M, Sharma A, Singhal V, Raymond FL, Linglart A, Jüppner H. Analysis of Multiple Families With Single Individuals Affected by Pseudohypoparathyroidism Type Ib (PHP1B) Reveals Only One Novel Maternally Inherited GNAS Deletion. J Bone Miner Res 2016; 31:796-805. [PMID: 26479409 PMCID: PMC4826817 DOI: 10.1002/jbmr.2731] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/05/2015] [Accepted: 10/12/2015] [Indexed: 01/22/2023]
Abstract
Proximal tubular resistance to parathyroid hormone (PTH) resulting in hypocalcemia and hyperphosphatemia are preeminent abnormalities in pseudohypoparathyroidism type Ib (PHP1B), but resistance toward other hormones as well as variable features of Albright's Hereditary Osteodystrophy (AHO) can occur also. Genomic DNA from PHP1B patients shows epigenetic changes at one or multiple differentially methylated regions (DMRs) within GNAS, the gene encoding Gαs and splice variants thereof. In the autosomal dominant disease variant, these methylation abnormalities are caused by deletions in STX16 or GNAS on the maternal allele. The molecular defect(s) leading to sporadic PHP1B (sporPHP1B) remains in most cases unknown and we therefore analyzed 60 sporPHP1B patients and available family members by microsatellite markers, single nucleotide polymorphisms (SNPs), multiplex ligation-dependent probe amplification (MLPA), and methylation-specific MLPA (MS-MLPA). All investigated cases revealed broad GNAS methylation changes, but no evidence for inheritance of two paternal chromosome 20q alleles. Some patients with partial epigenetic modifications in DNA from peripheral blood cells showed more complete GNAS methylation changes when testing their immortalized lymphoblastoid cells. Analysis of siblings and children of sporPHP1B patients provided no evidence for an abnormal mineral ion regulation and no changes in GNAS methylation. Only one patient revealed, based on MLPA and microsatellite analyses, evidence for an allelic loss, which resulted in the discovery of two adjacent, maternally inherited deletions (37,597 and 1427 bp, respectively) that remove the area between GNAS antisense exons 3 and 5, including exon NESP. Our findings thus emphasize that the region comprising antisense exons 3 and 4 is required for establishing all maternal GNAS methylation imprints. The genetic defect(s) leading in sporPHP1B to epigenetic GNAS changes and thus PTH-resistance remains unknown, but it seems unlikely that this disease variant is caused by heterozygous inherited or de novo mutations involving GNAS.
Collapse
Affiliation(s)
- Rieko Takatani
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Angelo Molinaro
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Giedre Grigelioniene
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Olta Tafaj
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomoyuki Watanabe
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Monica Reyes
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Amita Sharma
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Vibha Singhal
- Pediatric Endocrinology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - F Lucy Raymond
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Agnès Linglart
- Paediatric Endocrinology and Diabetology, French National Reference Centre for Rare Disorders of Mineral Metabolism, AP-HP Hôpital Bicêtre Paris Sud, le Kremlin-Bicêtre, France
- Faculté de Médecine, Université Paris Sud, le Kremlin-Bicêtre, France
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|