Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.586
Filtrar
2.
Mol Biol Rep ; 51(1): 790, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38990390

RESUMEN

INTRODUCTION: Creating induced pluripotent stem cells (iPSCs) from somatic cells of patients with genetic diseases offers a pathway to generate disease-specific iPSCs carrying genetic markers. Differentiating these iPSCs into renal tubular cells can aid in understanding the pathophysiology of rare inherited renal tubular diseases through cellular experiments. MATERIALS AND METHODS: Two Japanese patients with Pseudohypoparathyroidism (PHP), a 49-year-old woman and a 71-year-old man, were studied. iPSC-derived tubular cells were established from their peripheral blood mononuclear cells (PBMCs). We examined changes in intracellular and extracellular cyclic adenosine monophosphate (cAMP) levels in these cells in response to parathyroid hormone (PTH) stimulation. RESULTS: Renal tubular cells, differentiated from iPSCs of a healthy control (648A1), showed a PTH-dependent increase in both intracellular and extracellular cAMP levels. However, the renal tubular cells derived from the PHP patients' iPSCs showed inconsistent changes in cAMP levels upon PTH exposure. CONCLUSION: We successfully created disease-specific iPSCs from PHP patients' PBMCs, differentiated them into tubular cells, and replicated the distinctive response of the disease to PTH in vitro. This approach could enhance our understanding of the pathophysiology of inherited renal tubular diseases and contribute to developing effective treatments.


Asunto(s)
Diferenciación Celular , AMP Cíclico , Células Madre Pluripotentes Inducidas , Túbulos Renales , Leucocitos Mononucleares , Hormona Paratiroidea , Seudohipoparatiroidismo , Humanos , Hormona Paratiroidea/farmacología , Hormona Paratiroidea/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/metabolismo , Femenino , Diferenciación Celular/efectos de los fármacos , Masculino , AMP Cíclico/metabolismo , Túbulos Renales/metabolismo , Túbulos Renales/patología , Persona de Mediana Edad , Anciano , Leucocitos Mononucleares/metabolismo , Células Cultivadas
3.
BMC Pediatr ; 24(1): 271, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664677

RESUMEN

BACKGROUND: Pseudohypoparathyroidism (PHP) is caused by loss-of-function mutations at the GNAS gene (as in the PHP type 1A; PHP1A), de novo or inherited at heterozygous state, or by epigenetic alterations at the GNAS locus (as in the PHP1B). The condition of PHP refers to a heterogeneous group of disorders that share common clinical and biological features of PTH resistance. Manifestations related to resistance to other hormones are also reported in many patients with PHP, in association with the phenotypic picture of Albright hereditary osteodystrophy characterized by short stature, round facies, subcutaneous ossifications, brachydactyly, mental retardation and, in some subtypes, obesity. The purpose of our study is to report a new mutation in the GNAS gene and to describe the significant phenotypic variability of three sisters with PHP1A bearing the same mutation. CASE PRESENTATION: We describe the cases of three sisters with PHP1A bearing the same mutation but characterized by a significantly different phenotypic picture at onset and during follow-up in terms of clinical features, auxological pattern and biochemical changes. Clinical exome sequencing revealed a never before described heterozygote mutation in the GNAS gene (NM_000516.5 c.118_139 + 51del) of autosomal dominant maternal transmission in the three siblings, confirming the diagnosis of PHP1A. CONCLUSIONS: This study reported on a novel mutation of GNAS gene and highlighted the clinical heterogeneity of PHP1A characterized by wide genotype-phenotype variability. The appropriate diagnosis has crucial implications for patient care and long-term multidisciplinary follow-up.


Asunto(s)
Cromograninas , Subunidades alfa de la Proteína de Unión al GTP Gs , Seudohipoparatiroidismo , Humanos , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/diagnóstico , Cromograninas/genética , Femenino , Niño , Fenotipo , Linaje , Mutación , Adolescente , Preescolar
4.
Medicina (Kaunas) ; 60(4)2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38674241

RESUMEN

Soft tissue calcifications frequently appear on imaging studies, representing a prevalent but non-specific discovery, varying from a local reaction without clear cause to suggesting an underlying systemic condition. Because calcifications like these can arise from various causes, an accurate differential diagnosis is crucial. Differential diagnosis entails a methodical assessment of the patient, encompassing clinical presentation, medical history, radiological and pathological findings, and other pertinent factors. Through scrutiny of the patient's medical and trauma history, we can refine potential causes of calcification to vascular, metabolic, autoimmune, neoplastic, or traumatic origins. Furthermore, routine laboratory assessments, including serum levels of calcium, phosphorus, ionized calcium, vitamin D metabolites, and parathyroid hormone (PTH), aid in identifying metabolic etiologies. We describe a rare occurrence of osteoma cutis in a 15-year-old female patient with a history of pseudohypoparathyroidism (PHP) and Albright's hereditary osteodystrophy (AHO). The patient presented with a painful mass on the lateral side of her left foot. The diagnosis was based on medical history, laboratory tests, and imaging, leading to an excisional biopsy and complete pain relief post-surgery. Understanding such rare occurrences and related conditions is crucial for accurate diagnosis and management.


Asunto(s)
Calcinosis , Seudohipoparatiroidismo , Humanos , Femenino , Calcinosis/complicaciones , Calcinosis/diagnóstico por imagen , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/diagnóstico , Adolescente , Diagnóstico Diferencial , Pie , Enfermedades Óseas Metabólicas/complicaciones , Enfermedades Óseas Metabólicas/diagnóstico
5.
J Pediatr Endocrinol Metab ; 37(3): 289-295, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38353264

RESUMEN

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.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs , Seudohipoparatiroidismo , Adulto , Humanos , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Seudohipoparatiroidismo/diagnóstico , Seudohipoparatiroidismo/genética , Mutación , Obesidad , Tirotropina , Cromograninas/genética
6.
BMJ Case Rep ; 17(2)2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38423572

RESUMEN

Pseudohypoparathyroidism (PHP) is a rare genetic disorder characterised by a non-functioning PTH. Usually, the diagnosis is made following (symptomatic) hypocalcaemia. We describe a case in which epileptic seizures and abnormalities in dental development were the main clinical manifestation of PHP type 1B. This case demonstrates the importance of screening for hypocalcaemia in patients with de novo epileptic seizures. In addition, antiepileptic medications themselves may interfere with calcium-phosphate metabolism, causing or aggravating a hypocalcaemia as well. By correcting the calcium level, a resolution of these symptoms could be obtained.


Asunto(s)
Epilepsia , Hipocalcemia , Seudohipoparatiroidismo , Humanos , Hipocalcemia/diagnóstico , Hipocalcemia/tratamiento farmacológico , Hipocalcemia/etiología , Calcio/uso terapéutico , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/diagnóstico , Epilepsia/complicaciones , Convulsiones/complicaciones
7.
Endocrine ; 85(1): 331-340, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38393510

RESUMEN

BACKGROUND: 2q37 microdeletion syndrome is a rare clinical condition characterized by a series of physical abnormalities. Its Albright hereditary osteodystrophy (AHO)-like manifestations and possible complication of biochemical abnormalities indicating PTH resistance greatly increased the likelihood of misdiagnosis with classic pseudohypoparathyroidism (PHP) caused by GNAS mutation or methylation alteration, even though there have only been six reports of such clinical occasions. PURPOSE: to investigate the underlying genetic defect in a male patient presenting hypocalcemia, elevated PTH and with a history of kyphosis. METHOD: clinical information was collected, while the DNA was extracted from peripheral blood and subjected to methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) and exome sequencing. RESULT: Physical characteristics featuring short stature, obesity, round face, short neck, and shortened 4th metacarpal and laboratory examination of the patient suggested the presence of PTH resistance, which is indicative of PHP. MS-MLPA did not reveal methylation alterations or deletions of GNAS, STX16 or other monogenetic alterations responsible for iPPSDs, but WES revealed a long-range deletion of approximately 4.18 Mb of the 2q37 region that spanned AGAP1 to NDUFA10, indicating that the patient had 2q37 microdeletion syndrome with PTH resistance. CONCLUSION: After undergoing MS-MLPA and exome sequencing, a novel deletion spanning 4.18 Mb on the 2q37 region was identified in one male patient, clarifying the diagnosis of 2q37 microdeletion syndrome with PTH resistance. The new genetic discovery added to our understanding of the molecular defects that cause inactivating PTH/PTH-related protein signaling disorders (iPPSDs).


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 2 , Seudohipoparatiroidismo , Humanos , Masculino , Cromosomas Humanos Par 2/genética , Seudohipoparatiroidismo/genética , Trastornos de los Cromosomas/genética , Hormona Paratiroidea/sangre , China , Pueblos del Este de Asia
8.
Nephrology (Carlton) ; 29(5): 300-304, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38233937

RESUMEN

We describe a unique case of 27-year-old male with Gitelman syndrome (GS) co-exist with pseudohypoparathyroidism type 1B (PHP1B). The patient presented with a 5-year history of seizures, tetany, and numbness of the extremities. Further examinations showed recurrent hypokalemia, inappropriate kaliuresis, hypocalcemia, hyperphosphatemia, and elevated PTH levels. A novel variant of autosomal recessive GS (p.Val287Met SLC12A3) and a novel 492.3Kb deletion containing the whole of STX16, were discovered by a whole-exome sequencing. Following the diagnosis, calcitriol, calcium, and potassium supplements were started. Hematuria calcium and phosphorus levels, as well as blood potassium levels, have recovered and remained within normal ranges after 3 years of follow-up. Our findings have important consequences for supporting the idea that heterozygosity for variants have effects on the patients' clinical performance with autosomal recessive inheritance disorders. Further study is need for the putative effects of the variant. Likewise, further investigation with regards to the gene-gene interaction relations between GS and other electrolyte imbalance disorders is warranted.


Asunto(s)
Síndrome de Gitelman , Hipopotasemia , Seudohipoparatiroidismo , Desequilibrio Hidroelectrolítico , Masculino , Humanos , Adulto , Síndrome de Gitelman/complicaciones , Síndrome de Gitelman/diagnóstico , Síndrome de Gitelman/genética , Hipopotasemia/complicaciones , Calcio , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/diagnóstico , Seudohipoparatiroidismo/genética , Convulsiones/etiología , Convulsiones/genética , Desequilibrio Hidroelectrolítico/complicaciones , Calcio de la Dieta , Epigénesis Genética , Potasio
9.
JCI Insight ; 9(5)2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38290008

RESUMEN

Pseudohypoparathyroidism type 1B (PHP1B) results from aberrant genomic imprinting at the GNAS gene. Defining the underlying genetic cause in new patients is challenging because various genetic alterations (e.g., deletions, insertions) within the GNAS genomic region, including the neighboring STX16 gene, can cause PHP1B, and the genotype-epigenotype correlation has not been clearly established. Here, by analyzing patients with PHP1B with a wide variety of genotypes and epigenotypes, we identified a GNAS differentially methylated region (DMR) of distinct diagnostic value. This region, GNAS AS2, was hypomethylated in patients with genetic alterations located centromeric but not telomeric of this DMR. The AS2 methylation status was captured by a single probe of the methylation-sensitive multiplex ligation-dependent probe amplification (MS-MLPA) assay utilized to diagnose PHP1B. In human embryonic stem cells, where NESP55 transcription regulates GNAS methylation status on the maternal allele, AS2 methylation depended on 2 imprinting control regions (STX16-ICR and NESP-ICR) essential for NESP55 transcription. These results suggest that the AS2 methylation status in patients with PHP1B reflects the position at which the genetic alteration affects NESP55 transcription during an early embryonic period. Therefore, AS2 methylation levels can enable mechanistic PHP1B categorization based on genotype-epigenotype correlation and, thus, help identify the underlying molecular defect in patients.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs , Seudohipoparatiroidismo , Humanos , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Metilación de ADN , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/diagnóstico , Impresión Genómica , Alelos , Cromograninas/genética
12.
J Clin Endocrinol Metab ; 109(2): 424-438, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-37669316

RESUMEN

CONTEXT: Pseudohypoparathyroidism type IA (PHPIA) is a rare genetic disorder characterized by hormone resistance and a typical phenotype named Albright hereditary osteodystrophy. Unawareness of this rare disease leads to delays in diagnosis. OBJECTIVE: The aims of this study were to describe the clinical and molecular characteristics of patients with genetically confirmed GNAS mutations and to evaluate their long-term outcomes. METHODS: A retrospective search for all patients diagnosed with PHPIA in 2 referral centers in Israel was conducted. RESULTS: Nine children (8 females) belonging to 6 families were included in the study. Five patients had GNAS missense mutations, 2 had deletions, and 2 had frameshift mutations. Four mutations were novel. Patients were referred at a mean age of 2.4 years due to congenital hypothyroidism (5 patients), short stature (2 patients), or obesity (2 patients), with a follow-up duration of up to 20 years. Early obesity was observed in the majority of patients. Elevated parathyroid hormone was documented at a mean age of 3 years; however, hypocalcemia became evident at a mean age of 5.9 years, about 3 years later. All subjects were diagnosed with mild to moderate mental retardation. Female adult height was very short (mean -2.5 SD) and 5 females had primary or secondary amenorrhea. CONCLUSION: Long-term follow-up of newborns with a combination of congenital hypothyroidism, early-onset obesity, and minor dysmorphic features associated with PHPIA is warranted and molecular analysis is recommended since the complete clinical phenotype may develop a long time after initial presentation.


Asunto(s)
Hipotiroidismo Congénito , Seudohipoparatiroidismo , Recién Nacido , Niño , Adulto , Humanos , Femenino , Preescolar , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Estudios de Seguimiento , Estudios Retrospectivos , Cromograninas/genética , Seudohipoparatiroidismo/diagnóstico , Seudohipoparatiroidismo/genética , Obesidad
13.
J Pediatr Endocrinol Metab ; 37(1): 84-89, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38095637

RESUMEN

OBJECTIVES: Pseudohypoparathyroidism (PHP1B) is most commonly caused by epigenetic defects resulting in loss of methylation at the GNAS locus, although deletions of STX16 leading to GNAS methylation abnormalities have been previously reported. The phenotype of this disorder is variable and can include hormonal resistances and severe infantile obesity with hyperphagia. A possible time relationship between the onset of obesity and endocrinopathies has been previously reported but remains unclear. Understanding of the condition's natural history is limited, partly due to a scarcity of literature, especially in children. CASE PRESENTATION: We report three siblings with autosomal dominant PHP1B caused by a deletion in STX16 who presented with early childhood onset PTH-resistance with normocalcemia with a progressive nature, accompanied by TSH-resistance and severe infantile obesity with hyperphagia in some, not all of the affected individuals. CONCLUSIONS: PHP1B from a STX16 deletion displays intrafamilial phenotypic variation. It is a novel cause of severe infantile obesity, which is not typically included in commercially available gene panels but must be considered in the genetic work-up. Finally, it does not seem to have a clear time relationship between the onset of obesity and hormonal resistance.


Asunto(s)
Obesidad Mórbida , Obesidad Infantil , Seudohipoparatiroidismo , Niño , Humanos , Preescolar , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Hermanos , Obesidad Infantil/genética , Cromograninas/genética , Seudohipoparatiroidismo/genética , Metilación de ADN , Obesidad Mórbida/genética , Fenotipo , Hiperfagia , Sintaxina 16/genética
14.
Osteoporos Int ; 35(1): 195-199, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37644196

RESUMEN

Bone lytic lesions are a possible complication of pseudohypoparathyroidism type 1B, in undertreated adult patients. Whole body [18F] F-fluorocholine PET/CT is a useful imaging tool to assess brown tumor progression in this context. We describe the case of a 33-year-old woman, referred for the diagnostic evaluation of lytic bone lesions of the lower limbs, in the context of asymptomatic pseudohypoparathyroidism. She had been treated with alfacalcidol and calcium during her childhood. Treatment was discontinued at the age of 18 years old because of the lack of symptoms. A femur biopsy revealed a lesion rich in giant cells, without malignancy, consistent with a brown tumor. Laboratory tests showed a parathyroid level at 1387 pg/ml (14-50). Whole-body Fluorocholine PET/CT revealed hypermetabolism of bone lesions. The final diagnosis was brown tumors related to hyperparathyroidism complicating an untreated pseudohypoparathyroidism. Genetic testing confirmed PHP type 1B. Pseudohypoparathyroidism with radiographic evidence of hyperparathyroid bone disease, is a very rare condition due to parathyroid hormone resistance in target organs, i.e., kidney resistance, but with conserved bone cell sensitivity. It has been reported in only a few cases of pseudohypoparathyroidism type Ib. Long-term vitamin D treatment was required to correct bone hyperparathyroidism. With this rationale, the patient was treated with calcium, alfacalcidol, and cholecalciferol. One-year follow-up showed complete resolution of pain, improvement in serum calcium, and regression of bone lesions on [18F]F-fluorocholine PET/CT. This case illustrates the usefulness of [18F]F-fluorocholine PET/CT for the imaging of brown tumors in pseudohypoparathyroidism type 1B, and emphasizes the importance of calcium and vitamin D treatment in adult patients, to avoid the deleterious effects of high parathyroid hormone on skeletal integrity.


Asunto(s)
Enfermedades Óseas , Colina/análogos & derivados , Hiperparatiroidismo , Neoplasias , Osteítis Fibrosa Quística , Seudohipoparatiroidismo , Humanos , Adulto , Femenino , Niño , Adolescente , Calcio/uso terapéutico , Tomografía Computarizada por Tomografía de Emisión de Positrones , Osteítis Fibrosa Quística/complicaciones , Seudohipoparatiroidismo/complicaciones , Hormona Paratiroidea , Hiperparatiroidismo/complicaciones , Vitaminas , Vitamina D/uso terapéutico
16.
J Mol Endocrinol ; 72(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37965945

RESUMEN

Several human disorders are caused by genetic or epigenetic changes involving the GNAS locus on chromosome 20q13.3 that encodes the alpha-subunit of the stimulatory G protein (Gsα) and several splice variants thereof. Thus, pseudohypoparathyroidism type Ia (PHP1A) is caused by heterozygous inactivating mutations involving the maternal GNAS exons 1-13 resulting in characteristic abnormalities referred to as Albright's hereditary osteodystrophy (AHO) that are associated with resistance to several agonist ligands, particularly to parathyroid hormone (PTH), thereby leading to hypocalcemia and hyperphosphatemia. GNAS mutations involving the paternal Gsα exons also cause most of these AHO features, but without evidence for hormonal resistance, hence the term pseudopseudohypoparathyroidism (PPHP). Autosomal dominant pseudohypoparathyroidism type Ib (PHP1B) due to maternal GNAS or STX16 mutations (deletions, duplications, insertions, and inversions) is associated with epigenetic changes at one or several differentially methylated regions (DMRs) within GNAS. Unlike the inactivating Gsα mutations that cause PHP1A and PPHP, hormonal resistance is caused in all PHP1B variants by impaired Gsα expression due to loss of methylation at GNAS exon A/B, which can be associated in some familial cases with epigenetic changes at the other maternal GNAS DMRs. The genetic defect(s) responsible for sporadic PHP1B, the most frequent variant of this disorder, remain(s) unknown for the majority of patients. However, characteristic epigenetic GNAS changes can be readily detected that include a gain of methylation at the neuroendocrine secretory protein (NESP) DMR. Multiple genetic or epigenetic GNAS abnormalities can thus impair Gsα function or expression, consequently leading to inadequate cAMP-dependent signaling events downstream of various Gsα-coupled receptors.


Asunto(s)
Cromograninas , Seudohipoparatiroidismo , Humanos , Cromograninas/genética , Cromograninas/metabolismo , Seudohipoparatiroidismo/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Epigénesis Genética , Metilación de ADN
17.
Mutat Res Rev Mutat Res ; 793: 108487, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38103632

RESUMEN

BACKGROUND: GNAS (guanine nucleotide-binding protein, alpha stimulating) is an imprinted gene that encodes Gsα, the α subunit of the heterotrimeric stimulatory G protein. This subunit mediates the signalling of a diverse array of G protein-coupled receptors (GPCRs), including the melanocortin 4 receptor (MC4R) that serves a pivotal role in regulating food intake, energy homoeostasis, and body weight. Genetic or epigenetic alterations in GNAS are known to cause pseudohypoparathyroidism in its different subtypes and have been recently associated with isolated, early-onset, severe obesity. Given the diverse biological functions that Gsα serves, multiple molecular mechanisms involving various GPCRs, such as MC4R, ß2- and ß3-adrenoceptors, and corticotropin-releasing hormone receptor, have been implicated in the pathophysiology of severe, early-onset obesity that results from genetic or epigenetic GNAS changes. SCOPE OF REVIEW: This review examines the structure and function of GNAS and provides an overview of the disorders that are caused by defects in this gene and may feature early-onset obesity. Moreover, it elucidates the potential molecular mechanisms underlying Gsα deficiency-induced early-onset obesity, highlighting some of their implications for the diagnosis, management, and treatment of this complex condition. MAJOR CONCLUSIONS: Gsα deficiency is an underappreciated cause of early-onset, severe obesity. Therefore, screening children with unexplained, severe obesity for GNAS defects is recommended, to enhance the molecular diagnosis and management of this condition.


Asunto(s)
Cromograninas , Epigénesis Genética , Subunidades alfa de la Proteína de Unión al GTP Gs , Obesidad , Humanos , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Cromograninas/genética , Epigénesis Genética/genética , Obesidad/genética , Animales , Seudohipoparatiroidismo/genética , Mutación/genética , Receptor de Melanocortina Tipo 4/genética , Edad de Inicio
18.
Eur J Endocrinol ; 189(6): 590-600, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38039118

RESUMEN

OBJECTIVE: Pseudohypoparathyroidism type 1B (PHP1B) caused by methylation defects of differentially methylated regions (DMRs) on the GNAS locus can be categorized into groups according to etiologies and methylation defect patterns of the DMRs. The aim of this study was to clarify the clinical characteristics of each group. DESIGN: Comprehensive molecular analyses consisting of methylation, copy number, and microsatellite analyses. METHODS: Eighty-four patients with PHP1B were included in this study. We classified them into 5 groups, namely, autosomal dominant inheritance-PHP1B (Group 1, G1), sporadic-PHP1B (G2), and atypical-PHP1B (G3-G5), based on the methylation defect patterns in 4 DMRs on the GNAS locus and etiologies and evaluated the clinical findings in each group and compared them among the groups. RESULTS: G2 had the youngest age and the highest serum intact parathyroid hormone levels among the 5 groups at the time of diagnosis. The most common symptoms at the time of diagnosis were tetany in G1, and seizures or loss of consciousness in G2. Albright's hereditary osteodystrophy and PHP-suggestive features were most frequently observed in the G2 proband. Nine patients had neurodevelopmental disorders (NDs) consisting of mild to borderline intellectual disability and/or developmental delay. There were no significant correlations between the average methylation ratios of 7 CpG sites in the GNAS-A/B:TSS-DMR and hormonal and biochemical findings. CONCLUSION: This study revealed the differences in some clinical characteristics, particularly clinical features, and ages at the time of diagnosis between G2 and other groups and detailed NDs observed in some patients with PHP1B.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs , Seudohipoparatiroidismo , Humanos , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Cromograninas/genética , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/diagnóstico , Familia , Metilación de ADN/genética
19.
Med. clín (Ed. impr.) ; 161(11): 493-497, dic. 2023.
Artículo en Español | IBECS | ID: ibc-228154

RESUMEN

Antecedentes y objetivo El solapamiento clínico y bioquímico de diversas enfermedades del metabolismo fosfocálcico puede conllevar un erróneo diagnóstico y su consecuente abordaje clínico. Un ejemplo es el seudohipoparatiroidismo, que puede confundirse con el raquitismo dependiente de vitamina D (VDDR1) si no se hacen las determinaciones bioquímicas adecuadas. Pacientes y métodos Dos parejas de hermanos, de familias independientes, fueron diagnosticados clínicamente en la adolescencia de seudohipoparatiroidismo al presentar hipocalcemia, niveles elevados de hormona paratiroidea y valores normales o elevados de fósforo. Tras descartar alteraciones en GNAS, se realizó un estudio, mediante secuenciación masiva, de genes asociados a otros diagnósticos diferenciales. Resultados Se identificaron 2variantes genéticas en el gen CYP27B1 potencialmente asociadas con el fenotipo. Variantes patogénicas en este gen se asocian con VDDR1A. La reevaluación clínica-bioquímica de los pacientes confirmó dicho diagnóstico y se adecuó el tratamiento. Conclusiones Si bien la VDDR1A es un trastorno del metabolismo de diagnóstico infrecuente en la edad adulta, en casos de hipocalcemia con valores elevados de PTH es relevante la determinación de las formas 1,25(OH)2D3 y 25(OH)D3 de la vitamina D para alcanzar un diagnóstico correcto (AU)


Background and objective The clinical and biochemical overlap of various pathologies of phosphocalcic metabolism can lead to misdiagnosis and consequent clinical management. One example is pseudohypoparathyroidism, which can be confused with vitamin D-dependent rickets (VDDR1) if appropriate biochemical determinations are not performed. Patients and methods Two pairs of siblings, from independent families, were clinically diagnosed in adolescence with pseudohypoparathyroidism due to hypocalcaemia, elevated parathyroid hormone levels and normal or elevated phosphorus values. After ruling out alterations in GNAS, a massive sequencing study of genes associated with other differential diagnoses was carried out. Results Two genetic variants in the CYP27B1 gene potentially associated with the phenotype were identified. Pathogenic variants in this gene are associated with VDDR1A. Clinical-biochemical re-evaluation of the patients confirmed this diagnosis and treatment was adapted. Conclusions Although VDDR1A is an infrequently diagnosed pathology in adulthood, in cases of hypocalcaemia with elevated PTH values, determination of the 1,25(OH)2D3 and 25(OH)D3 forms of vitamin D is relevant to reach a correct diagnosis (AU)


Asunto(s)
Humanos , Femenino , Persona de Mediana Edad , Deficiencia de Vitamina D/diagnóstico , Deficiencia de Vitamina D/genética , Seudohipoparatiroidismo/diagnóstico , Seudohipoparatiroidismo/genética , Raquitismo Hipofosfatémico/diagnóstico , Raquitismo Hipofosfatémico/genética
20.
Front Endocrinol (Lausanne) ; 14: 1255864, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37920253

RESUMEN

GNASis a complex locus characterized by multiple transcripts and an imprinting effect. It orchestrates a variety of physiological processes via numerous signaling pathways. Human diseases associated with the GNAS gene encompass fibrous dysplasia (FD), Albright's Hereditary Osteodystrophy (AHO), parathyroid hormone(PTH) resistance, and Progressive Osseous Heteroplasia (POH), among others. To facilitate the study of the GNAS locus and its associated diseases, researchers have developed a range of mouse models. In this review, we will systematically explore the GNAS locus, its related signaling pathways, the bone diseases associated with it, and the mouse models pertinent to these bone diseases.


Asunto(s)
Enfermedades Óseas Metabólicas , Osificación Heterotópica , Seudohipoparatiroidismo , Animales , Ratones , Humanos , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Cromograninas/genética , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/genética , Osificación Heterotópica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...