Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Clin Endocrinol Metab ; 106(8): e3005-e3020, 2021 07 13.
Article in English | MEDLINE | ID: mdl-33780542

ABSTRACT

CONTEXT: Pseudohypoparathyroidism (PHP) is a group of disorders characterized by hypocalcemia, hyperphosphatemia, and elevated parathyroid hormone (PTH) levels as a result of end-organ resistance to PTH. OBJECTIVE: To describe a cohort of 26 patients with PHP followed in a single tertiary center. METHODS: Clinical, biochemical, radiological, and genetic analysis of the GNAS gene in 26 patients recruited since 2002. RESULTS: Ten patients harbored a GNAS mutation, 15 epigenetic abnormalities at the GNAS locus, and 1 did not show genetic or epigenetic abnormalities. According to clinical, biochemical, and genetic features, patients were classified as PHP1A, PHP1B, and pseudopseudohypoparathyroidism. Patients with PHP1A had an earlier diagnosis and more cases with family history, Albright hereditary osteodystrophy (AHO) features, hormonal resistance, and hypertension. Obesity was a common feature. No difference in biochemical values was present among PHP1A and PHP1B. Intracerebral calcification occurred in 72% of patients with no difference among PHP1A and PHP1B subgroups. No significant difference was observed between patients with and without intracerebral calcification for the time-weighted average values of total serum calcium, phosphate, calcium-phosphate product, and PTH fold increase. A borderline association between cerebral calcification and age at the time of diagnosis (P = .04) was found in the whole cohort of patients. No renal calcifications were found in the overall cohort. CONCLUSION: Patients with PHP1A more frequently have AHO features as well as hypertension than patients with PHP1B. Patients with PHP presented a high rate of intracerebral calcification with no significant difference between subgroups. No increased risk of renal calcifications was also found in the entire cohort.


Subject(s)
Brain Diseases/genetics , Calcinosis/genetics , Chromogranins/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Kidney Diseases/genetics , Mutation , Pseudohypoparathyroidism/genetics , Adolescent , Adult , Brain Diseases/diagnostic imaging , Brain Diseases/pathology , Calcinosis/diagnostic imaging , Calcinosis/pathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Child , Child, Preschool , Humans , Kidney/diagnostic imaging , Kidney/pathology , Kidney Diseases/diagnostic imaging , Kidney Diseases/pathology , Middle Aged , Pseudohypoparathyroidism/diagnostic imaging , Pseudohypoparathyroidism/pathology , Ultrasonography , Young Adult
2.
Eur J Endocrinol ; 184(2): 311-320, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33270042

ABSTRACT

OBJECTIVE: Pseudohypoparathyroidism and related disorders belong to a group of heterogeneous rare diseases that share an impaired signaling downstream of Gsα-protein-coupled receptors. Affected patients may present with various combination of symptoms including resistance to PTH and/or to other hormones, ectopic ossifications, brachydactyly type E, early onset obesity, short stature and cognitive difficulties. Several years ago we proposed a novel nomenclature under the term of inactivating PTH/PTHrP signaling disorders (iPPSD). It is now of utmost importance to validate these criteria and/or improve the basis of this new classification. DESIGN: Retrospective study of a large international series of 459 probands and 85 relatives molecularly characterized. METHODS: Information on major and minor criteria associated with iPPSD and genetic results were retrieved from patient files. We compared the presence of each criteria according to the iPPSD subtype, age and gender of the patients. RESULTS: More than 98% of the probands met the proposed criteria for iPPSD classification. Noteworthy, most patients (85%) presented a combination of symptoms rather than a single sign suggestive of iPPSD and the overlap among the different genetic forms of iPPSD was confirmed. The clinical and molecular characterization of relatives identified familial history as an additional important criterion predictive of the disease. CONCLUSIONS: The phenotypic analysis of this large cohort confirmed the utility of the major and minor criteria and their combination to diagnose iPPSD. This report shows the importance of having simple and easily recognizable signs to diagnose with confidence these rare disorders and supports a better management of patients.


Subject(s)
Parathyroid Hormone-Related Protein/physiology , Parathyroid Hormone/physiology , Pseudohypoparathyroidism/classification , Pseudohypoparathyroidism/diagnosis , Terminology as Topic , Adolescent , Adult , Aged , Child , Child, Preschool , Dysostoses/classification , Dysostoses/genetics , Female , France/epidemiology , Gene Silencing , Humans , Infant , Infant, Newborn , Intellectual Disability/classification , Intellectual Disability/genetics , Italy/epidemiology , Male , Middle Aged , Molecular Diagnostic Techniques , Mutation , Ossification, Heterotopic/classification , Ossification, Heterotopic/genetics , Osteochondrodysplasias/classification , Osteochondrodysplasias/genetics , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein/genetics , Pseudohypoparathyroidism/epidemiology , Pseudohypoparathyroidism/genetics , Rare Diseases , Retrospective Studies , Signal Transduction/genetics , Spain/epidemiology , Young Adult
3.
Horm Res Paediatr ; 93(3): 182-196, 2020.
Article in English | MEDLINE | ID: mdl-32756064

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.


Subject(s)
Diabetes Mellitus, Type 2 , Dwarfism, Pituitary , Hypothyroidism , Pseudohypoparathyroidism , Transition to Adult Care , Adult , Child , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/therapy , Dwarfism, Pituitary/diagnosis , Dwarfism, Pituitary/therapy , Humans , Hypothyroidism/diagnosis , Hypothyroidism/therapy , Practice Guidelines as Topic , Pseudohypoparathyroidism/diagnosis , Pseudohypoparathyroidism/therapy
4.
Endocrine ; 67(2): 466-472, 2020 02.
Article in English | MEDLINE | ID: mdl-31939093

ABSTRACT

PURPOSE: Pseudohypoparathyroidism (PHP), characterized by multihormone resistance and Albright's hereditary osteodystrophy (AHO), is caused by GNAS mutations. Whole or partial gene deletions are rare. All disorders due to inactivating mutations of the GNAS gene are now classified as "inactivating PTH/PTHrP signaling disorder type 2" (iPPSD2). This study reports a family harboring a large GNAS gene deletion in order to improve the knowledge of genotype-phenotype correlation of this disease. METHODS: An 18-year-old man with severe diffuse soft ossifications and multihormone resistance underwent to clinical, biochemical, radiological, and genetic studies. A review of the literature of other cases of iPPSD2 due to GNAS large deletions was performed focusing on clinical and biochemical features. RESULTS: The proband presented signs of hypocalcemia and marked AHO features. Laboratory tests revealed hypocalcemia, high levels of serum phosphate, PTH, TSH, and calcitonin despite therapy with calcium carbonate, calcitriol, and levothyroxine. Diffuse soft tissue ossifications and brain calcifications were shown by radiological exams. Family history was remarkable for hypocalcemia, neurocognitive impairment, and cerebral calcifications in his brother and AHO features in the maternal grandfather. The proband's mother showed short stature, whereas physical examination of the father was unremarkable. Genetic analysis of the GNAS gene revealed an unreported large deletion encompassing exons 1-7 in the proband, brother, and mother. By reviewing the literature, only six other cases were described. CONCLUSIONS: We report a kindred harboring a large GNAS deletion. A genotype-phenotype correlation was observed in term of severity of tissue ossifications in the siblings but not in the mother.


Subject(s)
Hypocalcemia , Pseudohypoparathyroidism , Adolescent , Chromogranins/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Hypocalcemia/genetics , Male , Parathyroid Hormone-Related Protein/metabolism , Pseudohypoparathyroidism/genetics
5.
Int J Mol Sci ; 20(4)2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30791553

ABSTRACT

Recently, we found a strict bone association between Fibroblast growth factor 23 (FGF23) and Fetuin-A, both involved in cardiovascular and mineral bone disorders. In this study, an uninvestigated bone marrow positivity for both was found. Though the role of exogenous FGF23 on mesenchymal cells (MSCs) was reported, no information is as yet available on the possible production of this hormone by MSCs. To further analyze these uninvestigated aspects, we studied human primary cells and mouse and human cell lines by means of immunostaining, qRT-PCR, enzyme linked immunosorbent assays, chromatin immunoprecipitation, transfection, and a streamlined approach for the FGF23⁻Fetuin-A interaction called Duolink proximity ligation assay. Mesenchymal cells produce but do not secrete FGF23 and its expression increases during osteo-differentiation. Fibroblast growth factor 23 is also involved in the regulation of Fetuin-A by binding directly to the Fetuin-A promoter and then activating its transcription. Both FGF23 overexpression and addition induced an upregulation of Fetuin-A in the absence of osteo-inducer factors. Fibroblast growth factor 23 and Fetuin-A promoter were increased by osteo-inducer factors with this effect being abolished after FGF23 silencing. In conclusion, both FGF23 and Fetuin-A are present and strictly linked to each other in MSCs with FGF23 driving Fetuin-A production. This mechanism suggests a role for these two proteins in the osteoblast differentiation.


Subject(s)
Fibroblast Growth Factors/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis , alpha-2-HS-Glycoprotein/metabolism , Animals , Biomarkers , Cell Line , Fibroblast Growth Factor-23 , Gene Expression , Gene Silencing , Humans , Male , Mice , Mice, Transgenic , Osteogenesis/genetics , Protein Binding
6.
Front Horm Res ; 51: 147-159, 2019.
Article in English | MEDLINE | ID: mdl-30641531

ABSTRACT

Pseudohypoparathyroidism (PHP), pseudo-PHP, acrodysostosis, and progressive osseous heteroplasia are heterogeneous disorders characterized by physical findings, differently associated in each subtype, including short bones, short stature, a stocky build, ectopic ossifications (features associated with Albright's hereditary osteodystrophy), as well as laboratory abnormalities consistent with hormone resistance, such as hypocalcemia, hyperphosphatemia, and elevated parathyroid hormone (PTH) and thyroid-stimulating hormone levels. All these disorders are caused by impairments in the cAMP-mediated signal transduction pathway and, in particular, in the PTH/PTHrP signaling pathway: the main subtypes of PHP and related disorders are caused by de novo or autosomal dominantly inherited inactivating genetic mutations, and/or epigenetic, sporadic, or genetic-based alterations within or upstream of GNAS, PRKAR1A, PDE4D, and PDE3A. Here we will review the impressive progress that has been made over the past 30 years on the pathophysiology of these diseases and will describe the recently proposed novel nomenclature and classification. The new term "inactivating PTH/PTHrP signaling disorder," iPPSD: (1) defines the common mechanism responsible for all diseases, (2) does not require a confirmed genetic defect, (3) avoids ambiguous terms like "pseudo," and (4) eliminates the clinical or molecular overlap between diseases.


Subject(s)
Bone Diseases, Metabolic , Dysostoses , Intellectual Disability , Ossification, Heterotopic , Osteochondrodysplasias , Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone/metabolism , Pseudohypoparathyroidism , Signal Transduction/physiology , Skin Diseases, Genetic , Bone Diseases, Metabolic/classification , Bone Diseases, Metabolic/diagnosis , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/therapy , Dysostoses/classification , Dysostoses/diagnosis , Dysostoses/metabolism , Dysostoses/therapy , Humans , Intellectual Disability/classification , Intellectual Disability/diagnosis , Intellectual Disability/metabolism , Intellectual Disability/therapy , Ossification, Heterotopic/classification , Ossification, Heterotopic/diagnosis , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/therapy , Osteochondrodysplasias/classification , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/therapy , Pseudohypoparathyroidism/classification , Pseudohypoparathyroidism/diagnosis , Pseudohypoparathyroidism/metabolism , Pseudohypoparathyroidism/therapy , Skin Diseases, Genetic/classification , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/therapy
7.
Nat Rev Endocrinol ; 14(8): 476-500, 2018 08.
Article in English | MEDLINE | ID: mdl-29959430

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.


Subject(s)
Delayed Diagnosis/adverse effects , Parathyroid Hormone/therapeutic use , Practice Guidelines as Topic , Pseudohypoparathyroidism/diagnosis , Pseudohypoparathyroidism/therapy , Consensus , Drug Resistance , Female , Genetic Predisposition to Disease , Humans , Infant, Newborn , Male , Neonatal Screening/organization & administration , Prognosis , Program Development , Pseudohypoparathyroidism/genetics , Risk Assessment
8.
Histopathology ; 71(1): 134-142, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28239886

ABSTRACT

AIMS: Fibrocartilaginous mesenchymoma is a rare intraosseous lesion, with a total of 26 cases described in the literature. This study describes the clinical, radiological and histological features of eight new cases of fibrocartilaginous mesenchymoma collected at a single institution between 1982 and 2016. The presence of GNAS and IDH1/2 mutations and MDM2 amplification was explored to evaluate possible links between fibrocartilaginous mesenchymoma, fibrous dysplasia, de-differentiated chondrosarcoma and low-grade osteosarcoma. METHODS AND RESULTS: Eight new cases of fibrocartilaginous mesenchymoma of bone identified in our archives, dating from 1982 to 2016, were reviewed. The diagnosis was not performed on the initial biopsy in any of these cases, due mainly to the absence of obvious cartilaginous differentiation. On imaging, the tumour contained cartilaginous calcifications and showed a very strong uptake of contrast medium after injection. Histologically, the tumour was characterized by spindle cell proliferation mimicking a low-grade spindle cell sarcoma, associated with epiphyseal growth-plate-like nodules of cartilage and bone production. Molecularly, no GNAS and IDH1/2 mutations or MDM2 amplification were found in the cases analysed. Only one case recurred 1 year following intralesional resection. None died of disease. CONCLUSIONS: This very rare bone tumour has a typical radiological and histological pattern and a favourable survival outcome after treatment. Local recurrences can be prevented with complete surgery. Fibrocartilaginous mesenchymoma does not seem to be related genetically to fibrous dysplasia, low-grade osteosarcoma and de-differentiated chondrosarcoma.


Subject(s)
Bone Neoplasms/pathology , Mesenchymoma/pathology , Adolescent , Adult , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Child , Child, Preschool , Female , Humans , Infant , Male , Mesenchymoma/diagnosis , Mesenchymoma/genetics , Young Adult
9.
Eur J Endocrinol ; 175(6): P1-P17, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27401862

ABSTRACT

OBJECTIVE: Disorders caused by impairments in the parathyroid hormone (PTH) signalling pathway are historically classified under the term pseudohypoparathyroidism (PHP), which encompasses rare, related and highly heterogeneous diseases with demonstrated (epi)genetic causes. The actual classification is based on the presence or absence of specific clinical and biochemical signs together with an in vivo response to exogenous PTH and the results of an in vitro assay to measure Gsa protein activity. However, this classification disregards other related diseases such as acrodysostosis (ACRDYS) or progressive osseous heteroplasia (POH), as well as recent findings of clinical and genetic/epigenetic background of the different subtypes. Therefore, the EuroPHP network decided to develop a new classification that encompasses all disorders with impairments in PTH and/or PTHrP cAMP-mediated pathway. DESIGN AND METHODS: Extensive review of the literature was performed. Several meetings were organised to discuss about a new, more effective and accurate way to describe disorders caused by abnormalities of the PTH/PTHrP signalling pathway. RESULTS AND CONCLUSIONS: After determining the major and minor criteria to be considered for the diagnosis of these disorders, we proposed to group them under the term 'inactivating PTH/PTHrP signalling disorder' (iPPSD). This terminology: (i) defines the common mechanism responsible for all diseases; (ii) does not require a confirmed genetic defect; (iii) avoids ambiguous terms like 'pseudo' and (iv) eliminates the clinical or molecular overlap between diseases. We believe that the use of this nomenclature and classification will facilitate the development of rationale and comprehensive international guidelines for the diagnosis and treatment of iPPSDs.


Subject(s)
Parathyroid Hormone-Related Protein , Parathyroid Hormone , Pseudohypoparathyroidism/classification , Pseudohypoparathyroidism/diagnosis , Bone Diseases, Metabolic/blood , Bone Diseases, Metabolic/classification , Bone Diseases, Metabolic/diagnosis , Dysostoses/blood , Dysostoses/classification , Dysostoses/diagnosis , Europe , Humans , Intellectual Disability/blood , Intellectual Disability/classification , Intellectual Disability/diagnosis , Ossification, Heterotopic/blood , Ossification, Heterotopic/classification , Ossification, Heterotopic/diagnosis , Osteochondrodysplasias/blood , Osteochondrodysplasias/classification , Osteochondrodysplasias/diagnosis , Parathyroid Hormone/blood , Parathyroid Hormone-Related Protein/blood , Pseudohypoparathyroidism/blood , Skin Diseases, Genetic/blood , Skin Diseases, Genetic/classification , Skin Diseases, Genetic/diagnosis
10.
Ann Endocrinol (Paris) ; 76(2): 101-4, 2015 May.
Article in English | MEDLINE | ID: mdl-25910998

ABSTRACT

The term pseudohypoparathryoidism (PHP) refers to a group of rare genetic and epigenetic disorders characterized by resistance to the action of parathyroid hormone (PTH) that activates cAMP signaling in target cells. Together with pseudohypoparathyroidism, Albright hereditary osteodystrophy (AHO) and progressive osseous heteroplasia (POH) represent rare, related and deeply impairing disorders encompassing heterogeneous features, such as brachydactyly, ectopic ossifications, short stature, mental retardation and endocrine deficiencies due to resistance to the action of different hormones. The two main subtypes, PHP-Ia and PHP-Ib, are caused by mutations in GNAS exons 1-13 and methylation defects in the imprinted GNAS cluster respectively, while mutations in the PRKAR1A and PDE4D genes (also involved in mediating cAMP signalling) have been demonstrated in patients with acrodysostosis, a disease of bone formation with characteristics similar to AHO. The molecular overlap among these disorders indicates the need for different classification models and seriously alters our understanding of the mechanisms through which GNAS defects, together with the new recently described defects involving other components of the cAMP signalling cascade, cause AHO-related disorders.


Subject(s)
Pseudohypoparathyroidism/genetics , Pseudohypoparathyroidism/pathology , Chromogranins , Disease Progression , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Receptor, Parathyroid Hormone, Type 1/genetics , Pseudohypoparathyroidism
12.
J Clin Endocrinol Metab ; 100(4): E681-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25594858

ABSTRACT

CONTEXT: Pseudohypoparathyroidism type Ia (PHP1A) is a rare endocrine disorder characterized by hypocalcemia, hyperphosphatemia, multiple hormonal resistance, and features of Albright hereditary osteodystrophy. When the phenotype is present but not associated with hormonal resistance, it is called psedopseudohypoparathyroidism (PPHP). Both entities have been associated to GNAS haploinsufficiency, and are mostly caused by inherited inactivating mutations at GNAS gene that codes for the stimulatory alpha subunit of G protein, although the cause remains unidentified in approximately 30% of patients. OBJECTIVES: The aims of our work were 1) to identify GNAS locus defects in 112 patients with clinical diagnosis of PHP1A/PPHP and no point mutations at GNAS, to improve molecular diagnostic and genetic counseling; 2) to outline the underlying molecular mechanism(s). METHODS: Methylation-specific-multiplex ligation-dependent probe amplification, qPCR, array comparative genomic hybridization, and long-PCR were used to search for genomic rearrangements at chromosome 20q and to identify their boundaries. We used different bioinformatic approaches to assess the involvement of the genomic architecture in the origin of the deletions. RESULTS: We discovered seven novel genomic deletions, ranging from 106-bp to 2.6-Mb. The characterization of five of seven deletion breakpoints and the definition of the putative molecular mechanisms responsible for these rearrangements revealed that Alu sequences play a major role in determining the genetic instability of the region. CONCLUSION: We observed that deletions at GNAS locus represent a significant cause of PPHP/PHP1A and that such defects are mostly associated with Alu-mediated recombination events. Their investigation revealed to be fundamental as, in some cases, they could be misdiagnosed as imprinting defects.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Deletion , Pseudohypoparathyroidism/genetics , Adolescent , Adult , Alu Elements/genetics , Child , Child, Preschool , Chromogranins , Cohort Studies , Comparative Genomic Hybridization , Female , Genetic Loci , Humans , Infant , Male , Molecular Diagnostic Techniques , Young Adult
13.
Eur J Hum Genet ; 23(4): 438-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25005735

ABSTRACT

Pseudohypoparathyroidism is a rare endocrine disorder that can be caused by genetic (mainly maternally inherited inactivating point mutations, although intragenic and gross deletions have rarely been reported) or epigenetic alterations at GNAS locus. Clinical and molecular characterization of this disease is not that easy because of phenotypic, biochemical and molecular overlapping features between both subtypes of the disease. The European Consortium for the study of PHP (EuroPHP) designed the present work with the intention of generating the standards of diagnostic clinical molecular (epi)genetic testing in PHP patients. With this aim, DNA samples of eight independent PHP patients carrying GNAS genetic and/or epigenetic defects (three patients with GNAS deletions, two with 20q uniparental disomy and three with a methylation defect of unknown origin) without GNAS point mutations were anonymized and sent to the five participant laboratories for their routine genetic analysis (methylation-specific (MS)-MLPA, pyrosequencing and EpiTYPER) and interpretations. All laboratories were able to detect methylation defects and, after the data analysis, the Consortium compared the results to define technical advantages and disadvantages of different techniques. To conclude, we propose as first-level investigation in PHP patients copy number and methylation analysis by MS-MLPA. Then, in patients with partial methylation defect, the result should be confirmed by single CpG bisulphite-based methods (ie pyrosequencing), whereas in case of a complete methylation defect without detectable deletion, microsatellites or SNP genotyping should be performed to exclude uniparental disomy 20.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Genetic Testing/standards , Genetic Variation , Pseudohypoparathyroidism/diagnosis , Pseudohypoparathyroidism/genetics , Chromogranins , DNA Methylation , Epigenesis, Genetic , Europe , Gene Deletion , Genotyping Techniques , Humans , Practice Guidelines as Topic , Sequence Analysis, DNA , Syntaxin 16/genetics , Uniparental Disomy/genetics
14.
J Clin Endocrinol Metab ; 99(4): E724-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24438374

ABSTRACT

CONTEXT: Pseudohypoparathyroidism type Ib (PHP-Ib) is a rare imprinting disorder characterized by end-organ resistance to PTH and, frequently, to thyroid-stimulating hormone. PHP-Ib familial form, with an autosomal dominant pattern of transmission (autosomal dominant pseudohypoparathyroidism type Ib [AD-PHP-Ib]), is typically characterized by an isolated loss of methylation at the guanine nucleotide-binding protein α-stimulating activity polypeptide 1 A/B differentially methylated region (DMR), secondary to genetic deletions disrupting the upstream imprinting control region in the syntaxin-16 (STX16) locus. However, deletions described up to now failed to account some cases of patients with a methylation defect limited to the A/B DMR; thus, it is expected the existence of other still unknown rearrangements, undetectable with conventional molecular diagnostic methods. OBJECTIVE: We investigated a PHP-Ib patient with a methylation defect limited to the A/B DMR and no known STX16 deletions to find the underlying primary genetic defect. PATIENT AND METHODS: A PHP-Ib patient (hypocalcaemia, hyperphosphataemia, raised serum PTH levels, no vitamin D deficiency) and his unaffected relatives were investigated by methylation specific-multiplex ligand-dependent probe amplification to search for novel pathogenetic defects affecting the guanine nucleotide-binding protein α-stimulating activity polypeptide 1 and STX16 loci. RESULTS: We report the clinical, biochemical, and molecular analysis of an AD-PHP-Ib patient with a novel STX16 deletion overlapping with previously identified STX16 deletions but that, unlike these genetic defects associated with AD-PHP-Ib, goes unnoticed with commonly used first-level diagnostic techniques. CONCLUSIONS: Our work highlights the importance of performing accurate investigations in PHP-Ib patients with methylation defects to allow precise genetic counseling because, in case of deletions, the segregation ratio is about 50% and the disease phenotype is transmitted in an autosomal dominant fashion via the mother.


Subject(s)
DNA Methylation/genetics , Gene Deletion , Genomic Imprinting/genetics , Pseudohypoparathyroidism/genetics , Syntaxin 16/genetics , Genes, Dominant , Humans , Male , Pedigree , Young Adult , Pseudohypoparathyroidism
15.
J Clin Endocrinol Metab ; 99(3): E508-17, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24423294

ABSTRACT

CONTEXT: Pseudohypoparathyroidism type I (PHP-I) includes two main subtypes, PHP-Ia and -Ib. About 70% of PHP-Ia patients, who show Albright hereditary osteodystrophy (AHO) associated with resistance toward multiple hormones (PTH/TSH/GHRH/gonadotropins), carry heterozygous mutations in the α-subunit of the stimulatory G protein (Gsα) exons 1-13, encoded by the guanine nucleotide binding-protein α-stimulating activity polypeptide 1 (GNAS), whereas the majority of PHP-Ib patients, who classically display hormone resistance limited to PTH and TSH with no AHO sign, have methylation defects in the imprinted GNAS cluster. Recently methylation defects have been detected also in patients with PHP and different degrees of AHO, indicating a molecular overlap between the two forms. OBJECTIVES: The objectives of the study were to collect patients with the following characteristics: clinical PHP-I (with or without AHO), no mutation in Gsα coding sequence, but the presence of GNAS methylation alterations and to investigate the existence of correlations between the degree of the epigenetic defect and the severity of the disease. PATIENTS AND METHODS: We quantified GNAS methylation alterations by both PCR-pyrosequencing and methylation specific-multiplex ligation-dependent probe amplification assay in genomic DNA from 63 patients with PHP-I and correlated these findings with clinical parameters (age at diagnosis; calcium, phosphorus, PTH, TSH levels; presence or absence of each AHO sign). RESULTS: By both approaches, the degree of the imprinting defect did not correlate with the onset of the disease, the severity of endocrine resistances, or with the presence/absence of specific AHO signs. CONCLUSIONS: Similar molecular alterations may lead to a broad spectrum of diseases, from isolated PTH resistance to complete PHP-Ia, and the degree of methylation alterations does not reflect or anticipate the severity and the type of different PHP/AHO manifestations.


Subject(s)
DNA Methylation , Epigenesis, Genetic , GTP-Binding Protein alpha Subunits, Gs/genetics , Pseudohypoparathyroidism/genetics , Adolescent , Adult , Child , Child, Preschool , Chromogranins , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Pseudohypoparathyroidism/diagnosis , Severity of Illness Index , Young Adult
17.
Gene ; 509(1): 168-72, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22771918

ABSTRACT

Wolfram syndrome (WS) is a rare autosomal recessive disorder characterized by diabetes insipidus (DI), insulin-dependent diabetes mellitus (DM), optic atrophy (OA) and deafness caused by mutations in WFS1 gene (4p16.1), which encodes an endoplasmic reticulum protein, called Wolframin. We describe the case of an infant who presented hypernatremia and severe hypoplasia of the left eyeball with alteration of visual evoked potentials. Persistent hypernatremia, iposmolar polyuria and high plasma osmolality suggested DI, confirmed by a normal urine concentration after vasopressin test. Treatment with vasopressin allowed a normalization of sodium levels and urine output. Brain magnetic resonance imaging showed absence of the neurohypophysis hyperintense signal, normal adenohypophysis and optic tracts hypoplasia. The concomitant presence of DI and OA, even in the absence of DM and deafness, prompted the suspicion of WS and complete genetic analysis was performed. Genomic DNA sequencing of WFS1 showed no inactivating mutations described to date, but suggested a structural mutation as markers genotyping revealed a segmental paternal heterodisomy involving the upstream regulatory region (promoter and 5'UTR). cDNA sequencing revealed the coexistence of the wild-type transcript and two splice variants; one variant, probably benign, is known in literature and the other one causes the loss of exon 2, containing the translation initiation site. Western blot confirmed a marked protein reduction. During the clinical follow-up child's condition remained stable and glucose metabolism is still in the standard. In conclusion, the phenotype associated with this structural rearrangement, which substantially reduces the synthesis of Wolframin, confirms a tissue-specific pattern of expression of WFS1, suggests the presence of a different protein dosage sensitivity in different tissues and could be causative of DI and OA in our patient. The "incomplete" phenotype here described, usually absent in typical WS cases, is explained by the residual Wolframin expression that would preserve other organs, i.e. pancreatic islets. A careful longitudinal clinical follow-up will assess any changes in the phenotypic penetrance in our patient.


Subject(s)
Membrane Proteins/genetics , Mutation , Wolfram Syndrome/genetics , Alternative Splicing , Base Sequence , Child, Preschool , Chromosomes, Human, Pair 4/genetics , DNA Mutational Analysis , Gene Rearrangement , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Membrane Proteins/metabolism , Penetrance , Phenotype , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Distribution , Uniparental Disomy/genetics , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
18.
J Clin Endocrinol Metab ; 97(3): 967-77, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22259062

ABSTRACT

CONTEXT: Dopamine agonists (DA) are the first choice treatment of prolactinomas. However, a subset of patients is resistant to DA, due to undefined dopamine D2 receptor (D2R) alterations. Recently, D2R was found to associate with filamin-A (FLNA), a widely expressed cytoskeleton protein with scaffolding properties, in melanoma and neuronal cells. OBJECTIVE: The aim of the study was to investigate the role of FLNA in D2R expression and signaling in human tumorous lactotrophs and rat MMQ and GH3 cells. DESIGN: We analyzed FLNA expression in a series of prolactinomas by immunohistochemistry and Western blotting. We performed FLNA silencing or transfection experiments in cultured cells from DA-sensitive or -resistant prolactinomas and in MMQ and GH3 cells, followed by analysis of D2R expression and signaling. RESULTS: We demonstrated reduced FLNA and D2R expression in DA-resistant tumors. The crucial role of FLNA on D2R was demonstrated by experiments showing that: 1) FLNA silencing in DA-sensitive prolactinomas resulted in 60% reduction of D2R expression and abrogation of DA-induced inhibition of prolactin release and antiproliferative signals, these results being replicated in MMQ cells that endogenously express FLNA and D2R; and 2) FLNA overexpression in DA-resistant prolactinomas restored D2R expression and prolactin responsiveness to DA, whereas this manipulation was ineffective in GH3 cells that express FLNA but not D2R. No alteration in FLNA promoter methylation was detected, ruling out the occurrence of epigenetic FLNA silencing in DA-resistant prolactinomas. CONCLUSIONS: These data indicate that FLNA is crucial for D2R expression and signaling in lactotrophs, suggesting that the impaired response to DA may be related to the reduction of FLNA expression in DA-resistant prolactinomas.


Subject(s)
Contractile Proteins/metabolism , Lactotrophs/metabolism , Microfilament Proteins/metabolism , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation , Contractile Proteins/genetics , Filamins , Humans , Microfilament Proteins/genetics , Phosphorylation , Pituitary Neoplasms/genetics , Prolactinoma/genetics , Rats , Receptors, Dopamine D2/genetics , Tumor Cells, Cultured
19.
J Clin Endocrinol Metab ; 95(2): 651-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20061437

ABSTRACT

CONTEXT: The two main subtypes of pseudohypoparathyroidism (PHP), PHP-Ia and -Ib, are caused by mutations in GNAS exons 1-13 and methylation defects in the imprinted GNAS cluster, respectively. PHP-Ia patients show Albright hereditary osteodystrophy (AHO) and resistance toward PTH and additional hormones, whereas PHP-Ib patients do not have AHO, and hormone resistance appears to be limited to PTH and TSH. Recently, methylation defects have been detected in few patients with PHP and mild AHO, indicating a molecular overlap between the two forms. OBJECTIVES: The aim of the study was to screen patients with clinically diagnosed PHP-Ia for methylation defects and to investigate the presence of correlations between the molecular findings and AHO severity. PATIENTS AND METHODS: We investigated differential methylation of GNAS regions and STX16 microdeletions in genomic DNA from 40 patients with sporadic AHO and multihormone resistance, with no mutations in Gsalpha-coding GNAS exons. RESULTS: Molecular analysis showed GNAS cluster imprinting defects in 24 of the 40 patients analyzed. No STX16 deletion was detected. The presence of imprinting defects was not associated with the severity of AHO or with specific AHO signs. CONCLUSIONS: We report the largest series of the literature of patients with clinical AHO and multihormone resistance and no mutation in the Gsalpha gene. Our findings of frequent GNAS imprinting defects further confirm the existence of an overlap between molecular and clinical features of PHP-Ia and PHP-Ib and highlight the necessity of a new clinical classification of the disease that takes into account the recent knowledge on the molecular basis underlying these defects.


Subject(s)
Epigenesis, Genetic , Fibrous Dysplasia, Polyostotic/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Pseudohypoparathyroidism/genetics , Adolescent , Adult , Child , Chromogranins , DNA Methylation , Female , Humans , Male , Polymorphism, Single Nucleotide , Pseudohypoparathyroidism/classification , Syntaxin 16/genetics
SELECTION OF CITATIONS
SEARCH DETAIL