Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Bone Miner Res ; 33(8): 1480-1488, 2018 08.
Article in English | MEDLINE | ID: mdl-29693731

ABSTRACT

Pseudohypoparathyroidism type 1A (PHP1A), pseudoPHP (PPHP), and PHP type 1B (PHP1B) are caused by maternal and paternal GNAS mutations and abnormal methylation at maternal GNAS promoter(s), respectively. Adult PHP1A patients are reportedly obese and short, whereas most PPHP patients are born small. In addition to parathyroid hormone (PTH) resistance, PHP1A and PHP1B patients may display early-onset obesity. Because early-onset and severe obesity and short stature are daily burdens for PHP1A patients, we aimed at improving knowledge on the contribution of the GNAS transcripts to fetal and postnatal growth and fat storage. Through an international collaboration, we collected growth and weight data from birth until adulthood for 306 PHP1A/PPHP and 220 PHP1B patients. PHP1A/PPHP patients were smaller at birth than healthy controls, especially PPHP (length Z-score: PHP1A -1.1 ± 1.8; PPHP -3.0 ± 1.5). Short stature is observed in 64% and 59% of adult PHP1A and PPHP patients. PHP1B patients displayed early postnatal overgrowth (height Z-score at 1 year: 2.2 ± 1.3 and 1.3 ± 1.5 in autosomal dominant and sporadic PHP1B) followed by a gradual decrease in growth velocity resulting in normal adult height (Z-score for both: -0.4 ± 1.1). Early-onset obesity characterizes GNAS alterations and is associated with significant overweight and obesity in adults (bodey mass index [BMI] Z-score: 1.4 ± 2.6, 2.1 ± 2.0, and 1.4 ± 1.9 in PPHP, PHP1A, and PHP1B, respectively), indicating that reduced Gsα expression is a contributing factor. The growth impairment in PHP1A/PPHP may be due to Gsα haploinsufficiency in the growth plates; the paternal XLαs transcript likely contributes to prenatal growth; for all disease variants, a reduced pubertal growth spurt may be due to accelerated growth plate closure. Consequently, early diagnosis and close follow-up is needed in patients with GNAS defects to screen and intervene in case of early-onset obesity and decreased growth velocity. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Bone Development/genetics , Chromogranins/genetics , Epigenesis, Genetic , GTP-Binding Protein alpha Subunits, Gs/genetics , Genetic Loci , Genetic Predisposition to Disease , Obesity/genetics , Adult , Age of Onset , Body Mass Index , Female , Humans , Infant, Newborn , Male , Pregnancy , Pseudohypoparathyroidism/genetics , Weight Gain/genetics
2.
Oncotarget ; 9(10): 8849-8869, 2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29507659

ABSTRACT

LRP1 (low-density lipoprotein receptor-related protein 1), a multifunctional endocytic receptor, has recently been identified as a hub within a biomarker network for multi-cancer clinical outcome prediction. As its role in colon cancer has not yet been characterized, we here investigate the relationship between LRP1 and outcome. MATERIALS AND METHODS: LRP1 mRNA expression was determined in colon adenocarcinoma and paired colon mucosa samples, as well as in stromal and tumor cells obtained after laser capture microdissection. Clinical potential was further investigated by immunohistochemistry in a population-based colon cancer series (n = 307). LRP1 methylation, mutation and miR-205 expression were evaluated and compared with LRP1 expression levels. RESULTS: LRP1 mRNA levels were significantly lower in colon adenocarcinoma cells compared with colon mucosa and stromal cells obtained after laser capture microdissection. Low LRP1 immunohistochemical expression in adenocarcinomas was associated with higher age, right-sided tumor, loss of CDX2 expression, Annexin A10 expression, CIMP-H, MSI-H and BRAFV600E mutation. Low LRP1 expression correlated with poor clinical outcome, especially in stage IV patients. While LRP1 expression was downregulated by LRP1 mutation, LRP1 promoter was never methylated. CONCLUSIONS: Loss of LRP1 expression is associated with worse colon cancer outcomes. Mechanistically, LRP1 mutation modulates LRP1 expression.

3.
J Clin Endocrinol Metab ; 101(10): 3657-3668, 2016 10.
Article in English | MEDLINE | ID: mdl-27428667

ABSTRACT

CONTEXT: The term pseudohypoparathyroidism (PHP) was coined to describe the clinical condition resulting from end-organ resistance to parathormone (rPTH), caused by genetic and/or epigenetic alterations within or upstream of GNAS. Although knowledge about PHP is growing, there are few data on the prevalence of underlying molecular defects. OBJECTIVE: The purpose of our study was to ascertain the relative prevalence of PHP-associated molecular defects. DESIGN: With a specially designed questionnaire, we collected data from all patients (n = 407) clinically and molecularly characterized to date by expert referral centers in France, Italy, and Spain. RESULTS: Isolated rPTH (126/407, 31%) was caused only by epigenetic defects, 70% of patients showing loss of imprinting affecting all four GNAS differentially methylated regions and 30% loss of methylation restricted to the GNAS A/B:TSS-DMR. Multihormone resistance with no Albright's hereditary osteodystrophy (AHO) signs (61/407, 15%) was essentially due to epigenetic defects, although 10% of patients had point mutations. In patients with rPTH and AHO (40/407, 10%), the rate of point mutations was higher (28%) and methylation defects lower (about 70%). In patients with multihormone resistance and AHO (155/407, 38%), all types of molecular defects appeared with different frequencies. Finally, isolated AHO (18/407, 4%) and progressive osseous heteroplasia (7/407, 2%) were exclusively caused by point mutations. CONCLUSION: With European data, we have established the prevalence of various genetic and epigenetic lesions in PHP-affected patients. Using these findings, we will develop objective criteria to guide cost-effective strategies for genetic testing and explore the implications for management and prognosis.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/deficiency , Parathyroid Hormone , Pseudohypoparathyroidism/genetics , Adolescent , Adult , Child , Chromogranins , Epigenesis, Genetic , Female , France/epidemiology , Humans , Italy/epidemiology , Male , Mutation , Prevalence , Pseudohypoparathyroidism/epidemiology , Spain/epidemiology , Young Adult
4.
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
6.
J Clin Endocrinol Metab ; 100(4): E623-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25603460

ABSTRACT

CONTEXT: GNAS is one of few genetic loci that undergo allelic-specific methylation resulting in the parent-specific expression of at least four different transcripts. Due to monoallelic expression, heterozygous GNAS mutations affecting either paternally or maternally derived transcripts cause different forms of pseudohypoparathyroidism (PHP), including autosomal-dominant PHP type Ib (AD-PHP1B) associated with loss of methylation (LOM) at exon A/B alone or sporadic PHP1B (sporPHP1B) associated with broad GNAS methylation changes. Similar to effects other imprinted genes have on early development, we recently observed severe intrauterine growth retardation in newborns, later diagnosed with pseudopseudohypoparathyroidism (PPHP) because of paternal GNAS loss-of-function mutations. OBJECTIVES: This study aimed to determine whether GNAS methylation abnormalities affect intrauterine growth. PATIENTS AND METHODS: Birth parameters were collected of patients who later developed sporPHP1B or AD-PHP1B, and of their healthy siblings. Comparisons were made to newborns affected by PPHP or PHP1A. RESULTS: As newborns, AD-PHP1B patients were bigger than their healthy siblings and well above the reference average; increased sizes were particularly evident if the mothers were unaffected carriers of STX16 deletions. SporPHP1B newborns were slightly above average for weight and length, but their overgrowth was less pronounced than that of AD-PHP1B newborns from unaffected mothers. CONCLUSION: LOM at GNAS exon A/B due to maternal STX16 deletions and the resulting biallelic A/B expression are associated with enhanced fetal growth. These findings are distinctly different from those of PPHP patients with paternal GNAS exons 2-13 mutations, whose birth parameters are almost 4.5 z-scores below those of AD-PHP1B patients born to healthy mothers.


Subject(s)
DNA Methylation , Fetal Development/genetics , Fetal Macrosomia/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Birth Weight/genetics , Chromogranins , Exons/genetics , Female , Gene Deletion , Humans , Infant, Newborn , Male , Pregnancy , Pseudohypoparathyroidism/genetics , Retrospective Studies , Syntaxin 16/genetics , Pseudohypoparathyroidism
7.
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
8.
Stem Cell Reports ; 3(3): 432-43, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25241742

ABSTRACT

Data from the literature indicate that genomic imprint marks are disturbed in human pluripotent stem cells (PSCs). GNAS is an imprinted locus that produces one biallelic (Gsα) and four monoallelic (NESP55, GNAS-AS1, XLsα, and A/B) transcripts due to differential methylation of their promoters (DMR). To document imprinting at the GNAS locus in PSCs, we studied GNAS locus DMR methylation and transcript (NESP55, XLsα, and A/B) expression in human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) derived from two human fibroblasts and their progenies. Results showed that (1) methylation at the GNAS locus DMRs is DMR and cell line specific, (2) changes in allelic transcript expression can be independent of a change in allele-specific DNA methylation, and (3) interestingly, methylation at A/B DMR is correlated with A/B transcript expression. These results indicate that these models are valuable to study the mechanisms controlling GNAS methylation, factors involved in transcript expression, and possibly mechanisms involved in the pathophysiology of pseudohypoparathyroidism type 1B.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Induced Pluripotent Stem Cells/metabolism , Alleles , Cell Line , Chromogranins , Embryonic Stem Cells/cytology , Genetic Loci , Genomic Imprinting , Humans , Induced Pluripotent Stem Cells/cytology , Polymorphism, Genetic , Promoter Regions, Genetic , Transcription, Genetic
9.
JAMA Surg ; 149(8): 858-62, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24898823

ABSTRACT

GNAS-activating mutations are reported in intraductal papillary mucinous neoplasms (IPMNs) and in McCune-Albright syndrome, characterized by fibrous dysplasia, precocious puberty, and café au lait spots. Recently, IPMNs have been described as a McCune-Albright syndrome-associated tumor, present in about 15% of patients. The aim of the present work was to assess the prevalence of polyostotic fibrous dysplasia and McCune-Albright syndrome among patients operated on for presumptive sporadic IPMNs. All patients operated on for IPMNs between January 1, 2007, and December 31, 2012, with available imaging were retrospectively screened for polyostotic fibrous dysplasia based on their preoperative abdominal or thoracoabdominal spiral computed tomography images. Systematic screening of 272 patients operated on for IPMNs revealed 1 patient with axial and peripheral polyostotic fibrous dysplasia and café au lait spots on clinical examination suggestive of McCune-Albright syndrome. This patient had been operated on for an unusually large invasive colloid adenocarcinoma (pT3N0M0 R0) derived from an intestinal subtype GNAS-mutated IPMN. The patient underwent adjuvant chemotherapy with gemcitabine for 6 months and was alive without recurrence 6 years later. Besides providing additional evidence of a syndromic IPMN as a feature of McCune-Albright syndrome, this observation is further evidence of the functional oncogenic consequences of GNAS mutations in the pancreas.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Fibrous Dysplasia, Polyostotic/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Mutation/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma, Mucinous/complications , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Papillary/complications , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Chromogranins , Fibrous Dysplasia, Polyostotic/complications , Fibrous Dysplasia, Polyostotic/pathology , Humans , Male , Middle Aged , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/pathology , Retrospective Studies
10.
J Clin Endocrinol Metab ; 99(3): E469-73, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24423332

ABSTRACT

BACKGROUND: Except after neck surgery, hypoparathyroidism is a rare disease caused by defects in genes involved in parathyroid gland development (TBX1/22q11.2 del, GCMB, GATA3, TBCE) or function [calcium sensing receptor (CASR), GNA11, PTH], or the autoimmune polyglandular syndrome type 1 (AIRE). Approximately 90% of sporadic cases and 30% of familial cases of isolated hypoparathyroidism remain unexplained. Recurrent missense mutations in AP2S1, a calcium-sensing receptor regulator, have been recently identified in familial hyperparathyroidism. AIM: The aim of the study was to investigate AP2S1 as a putative hypoparathyroidism-causing gene. METHODS: Sequencing analysis and quantitative genomic PCR of the AP2S1 gene in a large cohort of 10 index cases (from nine families) and 50 sporadic cases affected with isolated hypoparathyroidism were investigated. RESULTS AND CONCLUSIONS: None of the 60 patients presented with nucleotidic changes or copy number variation in the AP2S1 gene, thereby excluding AP2S1 defects as a frequent cause of isolated hypoparathyroidism.


Subject(s)
Adaptor Protein Complex 2/genetics , Adaptor Protein Complex sigma Subunits/genetics , Hypoparathyroidism/genetics , Receptors, Calcium-Sensing/metabolism , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Family , Female , Humans , Hypoparathyroidism/metabolism , Infant , Infant, Newborn , Male , Middle Aged , Pedigree , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...