Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
1.
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
2.
Front Endocrinol (Lausanne) ; 14: 1268704, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37854194

RESUMEN

Objective: The serum calcium (Ca)-to-phosphorus (P) ratio has been proposed to identify patients with primary hyperparathyroidism and chronic hypoparathyroidism (HPT), but it has never been tested in pseudohypoparathyroidism (PHP). The aim of this study was to test the performance of Ca/P ratio in PHP diagnosis compared with that in healthy subjects and patients with HPT for differential diagnosis. Design: A retrospective, cross-sectional, and observational study was carried out. Methods: Serum Ca, P, creatinine, parathyroid hormone (PTH), and albumin were collected. Ca and P were expressed in mmol/L. Ca/P diagnostic performance was evaluated by receiver operating characteristic curve, sensitivity, specificity, and accuracy. Results: A total of 60 patients with PHP, 60 patients with HPT, and 120 controls were enrolled. The Ca/P ratio was lower in patients with PHP and HPT than that in controls (p < 0.0001). The cutoff of 1.78 (2.32 if Ca and P measured in mg/dL) for Ca/P ratio could identify patients with PHP and HPT among the entire cohort (sensitivity and specificity of 76%). No valid cutoff of Ca/P was found to distinguish patients with PHP from patients with HPT; in this case, PTH above 53.0 ng/dL identified patients with PHP (sensitivity and specificity of 100%). The index (Ca/P × PTH) above 116 ng/L recognized patients with PHP from controls (sensitivity of 84.7% and specificity of 87.4%), whereas (Ca/P × PTH) below 34 ng/L recognized patients with HPT from controls (sensitivity of 88.9% and specificity of 90.8%). Conclusions: The Ca/P ratio below 1.78 (2.32 CU) is highly accurate to identify patients with PHP and HPT, although it is not reliable to differentiate these two conditions. The index (Ca/P × PTH) is excellent to specifically recognize PHP or HPT from healthy subjects.


Asunto(s)
Hipoparatiroidismo , Seudohipoparatiroidismo , Humanos , Calcio , Estudios Retrospectivos , Estudios Transversales , Seudohipoparatiroidismo/diagnóstico , Seudohipoparatiroidismo/metabolismo , Hormona Paratiroidea , Fósforo
3.
Curr Osteoporos Rep ; 21(3): 311-321, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37014531

RESUMEN

PURPOSE OF REVIEW: Pseudohypoparathyroidism (PHP) is a disorder caused by mutations and/or epigenetic changes at the complex GNAS locus. It is characterized by hypocalcemia, hyperphosphatemia, and an elevated parathyroid hormone concentration secondary to the resistance of target tissues to the biological actions of parathyroid hormone. PHP is divided into several subtypes with different yet overlapping phenotypes. Research on the bone status in patients with PHP is sparse and has yielded inconsistent results. This review was performed to summarize the current knowledge on the bone phenotypes and possible mechanisms of PHP. RECENT FINDINGS: Patients with PHP exhibit highly variable bone phenotypes and increased concentrations of bone turnover markers. Long-standing elevation of the parathyroid hormone concentration may lead to hyperparathyroid bone diseases, including rickets and osteitis fibrosa. Compared with normal controls, patients with PHP may exhibit similar, increased, or decreased bone mineral density. Higher bone mineral density has been found in patients with PHP type 1A than in normal controls, whereas decreased bone mass, osteosclerosis, and osteitis fibrosa cystica have been reported in patients with PHP type 1B, indicating more variable bone phenotypes in PHP type 1B. Bone tissues show partial sensitivity to parathyroid hormone in patients with PHP, leading to heterogeneous reactions to parathyroid hormone in different individuals and even in different regions of bone tissues in the same individual. Regions rich in cancellous bone are more sensitive and show more obvious improvement after therapy. Active vitamin D and calcium can significantly improve abnormal bone metabolism in patients with PHP.


Asunto(s)
Enfermedades Óseas , Seudohipoparatiroidismo , Humanos , Huesos/metabolismo , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/metabolismo , Hormona Paratiroidea/metabolismo , Enfermedades Óseas/complicaciones , Fenotipo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Cromograninas/genética , Cromograninas/metabolismo
4.
Mol Cell Endocrinol ; 531: 111311, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33989717

RESUMEN

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).


Asunto(s)
Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/metabolismo , Seudohipoparatiroidismo/metabolismo , Calcio/metabolismo , Humanos , Transducción de Señal
5.
Horm Metab Res ; 53(4): 225-235, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33513624

RESUMEN

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.


Asunto(s)
Trastornos de los Cromosomas/genética , Epigenómica , Seudohipoparatiroidismo/genética , Adolescente , Adulto , Cromograninas/genética , Cromograninas/metabolismo , Trastornos de los Cromosomas/metabolismo , Metilación de ADN , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Seudohipoparatiroidismo/metabolismo , Sintaxina 16/genética , Sintaxina 16/metabolismo , Transducina/genética , Transducina/metabolismo , Seudohipoparatiroidismo
6.
Nucleic Acids Res ; 48(20): 11394-11407, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33053156

RESUMEN

Genomic imprinting is an epigenetic process regulated by germline-derived DNA methylation that is resistant to embryonic reprogramming, resulting in parental origin-specific monoallelic gene expression. A subset of individuals affected by imprinting disorders (IDs) displays multi-locus imprinting disturbances (MLID), which may result from aberrant establishment of imprinted differentially methylated regions (DMRs) in gametes or their maintenance in early embryogenesis. Here we investigated the extent of MLID in a family harbouring a ZFP57 truncating variant and characterize the interactions between human ZFP57 and the KAP1 co-repressor complex. By ectopically targeting ZFP57 to reprogrammed loci in mouse embryos using a dCas9 approach, we confirm that ZFP57 recruitment is sufficient to protect oocyte-derived methylation from reprogramming. Expression profiling in human pre-implantation embryos and oocytes reveals that unlike in mice, ZFP57 is only expressed following embryonic-genome activation, implying that other KRAB-zinc finger proteins (KZNFs) recruit KAP1 prior to blastocyst formation. Furthermore, we uncover ZNF202 and ZNF445 as additional KZNFs likely to recruit KAP1 to imprinted loci during reprogramming in the absence of ZFP57. Together, these data confirm the perplexing link between KZFPs and imprint maintenance and highlight the differences between mouse and humans in this respect.


Asunto(s)
Metilación de ADN , Embrión de Mamíferos/metabolismo , Impresión Genómica , Células Germinativas/metabolismo , Oocitos/metabolismo , Proteínas Represoras/metabolismo , Síndrome de Beckwith-Wiedemann/metabolismo , Estudios de Cohortes , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Humanos , Análisis por Micromatrices , Mutación , Linaje , Seudohipoparatiroidismo/metabolismo , RNA-Seq , Proteínas Represoras/genética , Hermanos , Transcriptoma , Proteína 28 que Contiene Motivos Tripartito
7.
BMC Endocr Disord ; 20(1): 61, 2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32393234

RESUMEN

BACKGROUND: Cardiac damage triggered by severe hypocalcemia is well known. However, the role of chronic hypoparathyroidism (HP) and pseudohypoparathyroidism (PHP) in cardiac health is still unclear. We investigated the effect of chronic HP and PHP on cardiac structure and conductive function in patients compiling with treatment. METHODS: The study included 18 patients with HP and eight with PHP aged 45.4 ± 15.4 and 22.1 ± 6.4 years, respectively with a previously regular follow-up. In addition, 26 age- and sex-matched healthy controls were included. General characteristics and biochemical indices were recorded. Cardiac function and structure were assessed by estimation of myocardial enzymes, B-type natriuretic peptide (BNP), and echocardiography. The 12-lead electrocardiogram and 24-h Holter electrocardiography were performed to evaluate the conductive function. RESULTS: Levels of serum calcium in HP and PHP were 2.05 ± 0.16 mmol/L and 2.25 ± 0.19 mmol/L, respectively. The levels of myocardial enzyme and BNP were within the normal range. Adjusting for age at evaluation and body mass index, all M-mode measurements, left ventricular mass (LVM), LVM index (LVMI) and relative wall thickness (RWT) were comparable between patients and controls. Prolongation of corrected QT (QTc) intervals occurred in 52.6% (10/19) of patients, and 6.7% (1/15) of patients manifested more than 100 episodes of supraventricular and ventricular extrasystoles, as well as supraventricular tachycardia. None of the above arrhythmias was related to a severe clinical event. CONCLUSIONS: From this pilot study, patients diagnosed with HP and PHP and well-controlled serum calcium levels manifested normal cardiac morphology and ventricular function, except for prolonged QTc intervals, and a small percentage of mild arrhythmias needing further investigation.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Hipoparatiroidismo/fisiopatología , Seudohipoparatiroidismo/fisiopatología , Disfunción Ventricular Izquierda/fisiopatología , Adolescente , Adulto , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Complejos Atriales Prematuros/etiología , Complejos Atriales Prematuros/metabolismo , Complejos Atriales Prematuros/fisiopatología , Calcio/metabolismo , Estudios de Casos y Controles , Enfermedad Crónica , Ecocardiografía , Electrocardiografía , Electrocardiografía Ambulatoria , Femenino , Humanos , Hipoparatiroidismo/complicaciones , Hipoparatiroidismo/metabolismo , Síndrome de QT Prolongado/etiología , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/fisiopatología , Masculino , Persona de Mediana Edad , Péptido Natriurético Encefálico/metabolismo , Proyectos Piloto , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/metabolismo , Taquicardia Supraventricular/etiología , Taquicardia Supraventricular/metabolismo , Taquicardia Supraventricular/fisiopatología , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/metabolismo , Complejos Prematuros Ventriculares/etiología , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología , Adulto Joven
8.
Am J Med Genet A ; 179(7): 1330-1337, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31041856

RESUMEN

Albright hereditary osteodystrophy (AHO) is a complex disorder defined by the presence of a short adult stature relative to the height of an unaffected parent and brachydactyly type E, as well as a stocky build, round face, and ectopic calcifications. AHO and pseudohypoparathyroidism (PHP) have been used interchangeably in the past. The term PHP describes end-organ resistance to parathyroid hormone (PTH), occurring with or without the physical features of AHO. Conversely, pseudopseudohypoparathyroidism (PPHP) describes individuals with AHO features in the absence of PTH resistance. PHP and PPHP are etiologically linked and caused by genetic and/or epigenetic alterations in the guanine nucleotide-binding protein alpha-stimulating (Gs α) locus (GNAS) in chromosome 20q13. Another less-recognized group of skeletal dysplasias, termed acrodysostosis, partially overlap with skeletal, endocrine, and neurodevelopmental features of AHO/PHP and can be overlooked in clinical practice, causing confusion in the literature. Acrodysostosis is caused by defects in two genes, PRKAR1A and PDE4D, both encoding important components of the Gs α-cyclic adenosine monophosphate-protein kinase A signaling pathway. We describe the clinical course and genotype of two adult patients with overlapping AHO features who harbored novel pathogenic variants in GNAS (c.2273C > G, p.Pro758Arg, NM_080425.2) and PRKAR1A (c.803C > T, p.Ala268Val, NM_002734.4), respectively. We highlight the value of expert radiological opinion and molecular testing in establishing correct diagnoses and discuss phenotypic features of our patients, including the first description of subcutaneous ossification and spina bifida occulta in PRKAR1A-related acrodysostosis, in the context of the novel inactivating PTH/PTH related peptide signaling disorder classification system.


Asunto(s)
Disostosis/genética , Discapacidad Intelectual/genética , Osteocondrodisplasias/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/metabolismo , Seudohipoparatiroidismo/genética , Transducción de Señal , Disostosis/metabolismo , Humanos , Discapacidad Intelectual/metabolismo , Masculino , Persona de Mediana Edad , Osteocondrodisplasias/metabolismo , Seudohipoparatiroidismo/metabolismo
9.
Front Horm Res ; 51: 147-159, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30641531

RESUMEN

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.


Asunto(s)
Enfermedades Óseas Metabólicas , Disostosis , Discapacidad Intelectual , Osificación Heterotópica , Osteocondrodisplasias , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/metabolismo , Seudohipoparatiroidismo , Transducción de Señal/fisiología , Enfermedades Cutáneas Genéticas , Enfermedades Óseas Metabólicas/clasificación , Enfermedades Óseas Metabólicas/diagnóstico , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/terapia , Disostosis/clasificación , Disostosis/diagnóstico , Disostosis/metabolismo , Disostosis/terapia , Humanos , Discapacidad Intelectual/clasificación , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/terapia , Osificación Heterotópica/clasificación , Osificación Heterotópica/diagnóstico , Osificación Heterotópica/metabolismo , Osificación Heterotópica/terapia , Osteocondrodisplasias/clasificación , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/terapia , Seudohipoparatiroidismo/clasificación , Seudohipoparatiroidismo/diagnóstico , Seudohipoparatiroidismo/metabolismo , Seudohipoparatiroidismo/terapia , Enfermedades Cutáneas Genéticas/clasificación , Enfermedades Cutáneas Genéticas/diagnóstico , Enfermedades Cutáneas Genéticas/metabolismo , Enfermedades Cutáneas Genéticas/terapia
10.
J Clin Endocrinol Metab ; 103(11): 4265-4274, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30085125

RESUMEN

Context: Pseudohypoparathyroidism (PHP) is a rare genetic disorder characterized by early-onset obesity and multihormone resistance. To treat abnormal weight gain and prevent complications such as diabetes, we must understand energy balance and glucose homeostasis in PHP types 1A and 1B. Objective: The aim of this study was to evaluate food intake, energy expenditure, and glucose homeostasis in children with PHP. Design: Assessments included resting energy expenditure (REE), physical activity, food intake, sucrose preference, questionnaires, endocrine status, and auxological status. All patients underwent an oral glucose tolerance test (OGTT). Setting: Vanderbilt University Medical Center. Patients: We assessed 16 children with PHP1A, three with PHP1B, and 15 healthy controls. Main Outcome Measures: Food intake during an ad lib buffet meal and glucose at five time points during OGTT. Results: PHP1A and control groups were well matched. Participants with PHP1A had significantly lower REE without concomitant change in food intake or physical activity. At baseline, participants with PHP1A had significantly lower fasting glucose and insulin resistance. During OGTT, participants with PHP1A had significantly delayed peak glucose and a slower rate of glucose decline despite similar oral glucose insulin sensitivity. Participants with PHP1A had 0.46% lower HbA1c levels than controls from a clinic database after adjustment for OGTT 2-hour glucose. The PHP1B group was similar to the PHP1A group. Conclusions: In contrast to other monogenic obesity syndromes, our results support reduced energy expenditure, not severe hyperphagia, as the primary cause of abnormal weight gain in PHP. Patients with PHP are at high risk for dysglycemia without reduced insulin sensitivity.


Asunto(s)
Glucemia/metabolismo , Cromograninas/genética , Metabolismo Energético/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Homeostasis/genética , Hiperfagia/diagnóstico , Seudohipoparatiroidismo/metabolismo , Adolescente , Glucemia/análisis , Niño , Cromograninas/metabolismo , Estudios Transversales , Conducta Alimentaria/fisiología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Pruebas Genéticas , Prueba de Tolerancia a la Glucosa , Humanos , Hiperfagia/etiología , Hiperfagia/metabolismo , Resistencia a la Insulina/genética , Masculino , Estudios Prospectivos , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/diagnóstico , Seudohipoparatiroidismo/genética , Índice de Severidad de la Enfermedad , Aumento de Peso/genética
11.
Best Pract Res Clin Endocrinol Metab ; 32(2): 141-154, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29678282

RESUMEN

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.


Asunto(s)
Resistencia a Medicamentos , Enfermedades del Sistema Endocrino , Hormonas/metabolismo , Resistencia a Medicamentos/genética , Enfermedades del Sistema Endocrino/genética , Enfermedades del Sistema Endocrino/metabolismo , Humanos , Hormona Paratiroidea/metabolismo , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/genética
12.
Bone ; 110: 230-237, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29471062

RESUMEN

GNAS mutations leading to constitutively active stimulatory G protein alpha-subunit (Gsα) cause different tumors, fibrous dysplasia of bone, and McCune-Albright syndrome, which are typically not associated with short stature. Enhanced signaling of the parathyroid hormone/parathyroid hormone-related peptide receptor, which couples to multiple G proteins including Gsα, leads to short bones with delayed endochondral ossification. It has remained unknown whether constitutive Gsα activity also impairs bone growth. Here we generated mice expressing a constitutively active Gsα mutant (Gsα-R201H) conditionally upon Cre recombinase (cGsαR201H mice). Gsα-R201H was expressed in cultured bone marrow stromal cells from cGsαR201H mice upon adenoviral-Cre transduction. When crossed with mice in which Cre is expressed in a tamoxifen-regulatable fashion (CAGGCre-ER™), tamoxifen injection resulted in mosaic expression of the transgene in double mutant offspring. We then crossed the cGsαR201H mice with Prx1-Cre mice, in which Cre is expressed in early limb-bud mesenchyme. The double mutant offspring displayed short limbs at birth, with narrow hypertrophic chondrocyte zones in growth plates and delayed formation of secondary ossification center. Consistent with enhanced Gsα signaling, bone marrow stromal cells from these mice demonstrated increased levels of c-fos mRNA. Our findings indicate that constitutive Gsα activity during limb development disrupts endochondral ossification and bone growth. Given that Gsα haploinsufficiency also leads to short bones, as in patients with Albright's hereditary osteodystrophy, these results suggest that a tight control of Gsα activity is essential for normal growth plate physiology.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Animales , Desarrollo Óseo/genética , Desarrollo Óseo/fisiología , Células Cultivadas , Cromograninas/genética , Cromograninas/metabolismo , AMP Cíclico/metabolismo , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Integrasas/genética , Integrasas/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Osteogénesis/genética , Osteogénesis/fisiología , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/metabolismo , Tamoxifeno/farmacología
13.
Best Pract Res Clin Endocrinol Metab ; 32(6): 941-954, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30665554

RESUMEN

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.


Asunto(s)
Resistencia a Medicamentos , Enfermedades del Sistema Endocrino , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/fisiología , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Resistencia a Medicamentos/genética , Disostosis/genética , Disostosis/metabolismo , Enfermedades del Sistema Endocrino/genética , Enfermedades del Sistema Endocrino/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Osificación Heterotópica/genética , Osificación Heterotópica/metabolismo , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/genética , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/metabolismo , Transducción de Señal/genética , Enfermedades Cutáneas Genéticas/genética , Enfermedades Cutáneas Genéticas/metabolismo , Síndrome
14.
J Clin Endocrinol Metab ; 102(6): 1844-1850, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28323910

RESUMEN

Context: Parathormone (PTH) resistance is characterized by hypocalcaemia, hyperphosphatemia, and elevated PTH in the absence of vitamin D deficiency. Pseudohypoparathyroidism type 1A [PHP1A, or inactivating parathormone (PTH)/PTHrp signaling disorder 2, according to the new classification (iPPSD2)], is caused by mutations in the maternal GNAS allele. Objective: To assess PTH resistance over time in 20 patients affected by iPPSD2 (PHP1A), diagnosed because of family history, ectopic ossification, or short stature, and carrying a GNAS mutation. Methods: We gathered retrospective data for calcium, phosphate, thyrotropin (TSH), and PTH levels at regular intervals. PTH infusion testing (teriparatide) was performed in 1 patient. Results: Patients were diagnosed at a mean age of 3.9 years and had a mean follow-up of 2 years. TSH resistance was already present at diagnosis in all patients (TSH, 13.3 ± 9.0 mIU/L). Over time, PTH levels increased (179 to 306 pg/mL; P < 0.05), and calcium levels decreased (2.31 to 2.21 mmol/L; P < 0.05), but phosphate levels did not decrease with age as expected for healthy individuals. One patient born with ectopic ossifications showed an increase in cyclic adenosine monophosphate upon PTH infusion, similar to that of controls, at 7 months of age, but an impaired response at 4 years of age. Conclusions: In patients with iPPSD2 (PHP1A), PTH resistance and hypocalcemia develop over time. These findings highlight the importance of screening for maternal GNAS mutations in the presence of ectopic ossifications or family history, even in the absence of PTH resistance and hypocalcemia. The follow-up of these patients should include regular assessments of calcium, phosphate, and PTH levels.


Asunto(s)
Hiperfosfatemia/metabolismo , Hipocalcemia/metabolismo , Hormona Paratiroidea/metabolismo , Seudohipoparatiroidismo/metabolismo , Tirotropina/metabolismo , Adolescente , Calcio/metabolismo , Niño , Preescolar , Cromograninas/genética , AMP Cíclico/metabolismo , Progresión de la Enfermedad , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Humanos , Hiperfosfatemia/etiología , Hipocalcemia/etiología , Lactante , Recién Nacido , Masculino , Herencia Materna , Mutación , Fosfatos/metabolismo , Seudohipoparatiroidismo/complicaciones , Seudohipoparatiroidismo/genética , Estudios Retrospectivos
15.
J Clin Invest ; 127(2): 500-510, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27991864

RESUMEN

Gsα, encoded by Gnas, mediates hormone and neurotransmitter receptor-stimulated cAMP generation. Heterozygous Gsα-inactivating mutations lead to obesity in Albright hereditary osteodystrophy (AHO) patients, but only when the mutations occur on the maternal allele. This parent-of-origin effect is due to Gsα imprinting in the CNS, although the relevant CNS regions are unknown. We have now shown that mice with a Gnas gene deletion disrupting Gsα expression on the maternal allele, but not the paternal allele, in the dorsomedial nucleus of the hypothalamus (DMH) developed obesity and reduced energy expenditure without hyperphagia. Although maternal Gnas deletion impaired activation of brown adipose tissue (BAT) in mice, their responses to cold environment remained intact. Similar findings were observed in mice with DMH-specific deficiency of melanocortin MC4R receptors, which are known to activate Gsα. Our results show that Gsα imprinting in the DMH underlies the parent-of-origin metabolic phenotype that results from Gsα mutations and that DMH MC4R/Gsα signaling is important for regulation of energy expenditure and BAT activation, but not the metabolic response to cold.


Asunto(s)
Tejido Adiposo Pardo , Cromograninas , Núcleo Hipotalámico Dorsomedial , Metabolismo Energético/genética , Subunidades alfa de la Proteína de Unión al GTP Gs , Impresión Genómica , Mutación , Obesidad , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/fisiopatología , Alelos , Animales , Cromograninas/genética , Cromograninas/metabolismo , Frío , Núcleo Hipotalámico Dorsomedial/metabolismo , Núcleo Hipotalámico Dorsomedial/fisiopatología , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Obesidad/fisiopatología , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/metabolismo , Seudohipoparatiroidismo/fisiopatología , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal/genética
16.
Genes Brain Behav ; 15(7): 669-77, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27415614

RESUMEN

Pseudohypoparathyroidism type 1b (PHP1b) is characterized by hypocalcemia, hyperphosphatemia, increased levels of circulating parathyroid hormone (PTH), and no skeletal or developmental abnormalities. The goal of this study was to perform a full characterization of a familial case of PHP1b with neurological involvement and to identify the genetic cause of disease. The initial laboratory profile of the proband showed severe hypocalcemia, hyperphosphatemia and normal levels of PTH, which was considered to be compatible with primary hypoparathyroidism. With disease progression the patient developed cognitive disturbance, PTH levels were found to be slightly elevated and a picture of PTH resistance syndrome seemed more probable. The diagnosis of PHP1b was established after the study of family members and blunted urinary cAMP results were obtained in a PTH stimulation test. Integration of whole genome genotyping and exome sequencing data supported this diagnosis by revealing a novel homozygous missense mutation in PTH1R (p.Arg186His) completely segregating with the disease. Here, we demonstrate segregation of a novel mutation in PTH1R with a phenotype of PHP1b presenting with neurological symptoms, but no bone defects. This case represents the extreme end of the spectrum of cognitive impairment in PTH dysfunction and defines a possible novel form of PHP1b resulting from the impaired interaction between PTH and PTH1R.


Asunto(s)
Mutación , Seudohipoparatiroidismo/genética , Receptor de Hormona Paratiroídea Tipo 1/genética , Anciano , Anciano de 80 o más Años , Femenino , Genoma , Homocigoto , Humanos , Hiperfosfatemia/genética , Hipocalcemia/genética , Masculino , Persona de Mediana Edad , Hormona Paratiroidea/genética , Hormona Paratiroidea/metabolismo , Linaje , Seudohipoparatiroidismo/sangre , Seudohipoparatiroidismo/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Seudohipoparatiroidismo
17.
J Bone Miner Res ; 31(4): 796-805, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26479409

RESUMEN

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.


Asunto(s)
Alelos , Secuencia de Bases , Cromograninas/genética , Cromograninas/metabolismo , Familia , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/metabolismo , Eliminación de Secuencia , Adolescente , Línea Celular Transformada , Niño , Metilación de ADN , Epigénesis Genética , Exones , Femenino , Humanos , Masculino , Sintaxina 16/genética , Sintaxina 16/metabolismo , Seudohipoparatiroidismo
18.
J Clin Endocrinol Metab ; 101(3): 880-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26709970

RESUMEN

CONTEXT: Pseudohypoparathyroidism type 1A (PHP1A) is caused by loss-of-function mutations on the maternally inherited GNAS allele and is associated with early-onset obesity, neurocognitive defects, and resistance to multiple hormones. The role of energy intake vs central regulation of energy expenditure in the pathophysiology of obesity remains unclear. OBJECTIVE: The aim of this study was to evaluate resting energy expenditure (REE) in participants with PHP1A. DESIGN: We assessed REE, biochemical, endocrine, and auxological status of 12 participants with PHP1A who had normal or elevated body mass index; controls were a cohort of 156 obese participants. SETTING: This study took place at Children's Hospital in Philadelphia and Sick Children's Hospital in Toronto. MAIN OUTCOME MEASURES: REE as a percent of predicted REE was the outcome measure. RESULTS: PHP1A participants had normal endocrine status while receiving appropriate hormone replacement therapy, but had significantly decreased REE as a percent of predicted REE (using the modified Schofield equation). CONCLUSION: Our results are consistent with REE being the principal cause of obesity in PHP1A rather than it being caused by excessive energy intake or endocrine dysfunction.


Asunto(s)
Metabolismo Energético , Seudohipoparatiroidismo/metabolismo , Adolescente , Adulto , Composición Corporal , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad
20.
Am J Med Sci ; 350(6): 458-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26488942

RESUMEN

Pseudohypohyperparathyroidism (PHHP) is a rare type of pseudohypoparathyroidism (PHP), which seems to have a normal skeletal response to parathyroid hormone but shows renal resistance. Almost all patients with PHHP have PHP Ib, a subtype of PHP that is usually caused by GNAS methylation defects, often in exon 1A. Some features of Albright hereditary osteodystrophy can occasionally be found in patients with PHHP, but these features are also common in Turner syndrome. The authors report on an extremely rare case of a patient with PHHP and Turner syndrome, a 47-year-old woman who sought medical attention for hypocalcemia and elevated parathyroid hormone. She had no family history of hypocalcemia and no STX16 gene deletions. She had a mosaic karyotype of 46, X, del(X)(p11.4)/45, XO. Pyrosequencing was performed to determine the GNAS exon 1A methylation. The degree of methylation found in exon 1A of the patient was lower than her unaffected relatives.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Seudohipoparatiroidismo/diagnóstico , Síndrome de Turner/diagnóstico , Cromograninas , Metilación de ADN , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Persona de Mediana Edad , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo/metabolismo , Síndrome de Turner/genética , Síndrome de Turner/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA