Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
J Endocrinol Invest ; 47(2): 469-478, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37561279

RESUMO

PURPOSE: Hypoparathyroidism, deafness, and renal dysplasia (HDR) syndrome, also known as Barakat syndrome, is a rare autosomal dominant disease characterized by the triad of hypoparathyroidism, deafness, and renal abnormalities. The disorder is caused by the haploinsufficiency of the zinc finger transcription factor GATA3 and exhibits a great clinical variability with an age-dependent penetrance of each feature. We report two unrelated kindreds whose probands were referred to our outpatient clinic for further evaluation of hypoparathyroidism. METHODS: The proband of family 1, a 17-year-old boy, was referred for severe hypocalcemia (5.9 mg/dL) incidentally detected at routine blood tests. Abdomen ultrasound showed bilateral renal cysts. The audiometric evaluation revealed the presence of bilateral moderate hearing loss although the patient could communicate without any problem. Conversely, the proband of family 2, a 19-year-old man, had severe symptomatic hypocalcemia complicated by epileptic seizure at the age of 14 years; his past medical history was remarkable for right nephrectomy at the age of 4 months due to multicystic renal disease and bilateral hearing loss diagnosed at the age of 18 years. RESULTS: Based on clinical, biochemical, and radiologic data, HDR syndrome was suspected and genetic analysis of the GATA3 gene revealed the presence of two pathogenetic variants in exon 3, c.404dupC and c.431dupG, in the proband of family 1 and 2, respectively. CONCLUSION: HDR syndrome is a rare cause of hypoparathyroidism and must be excluded in all patients with apparently idiopathic hypoparathyroidism. A correct diagnosis is of great importance for early detection of other HDR-related features and genetic counseling.


Assuntos
Surdez , Perda Auditiva Neurossensorial , Hipocalcemia , Hipoparatireoidismo , Nefrose , Masculino , Humanos , Adolescente , Lactente , Adulto Jovem , Adulto , Hipocalcemia/complicações , Hipocalcemia/diagnóstico , Hipocalcemia/genética , Hipoparatireoidismo/complicações , Hipoparatireoidismo/diagnóstico , Hipoparatireoidismo/genética , Surdez/complicações , Surdez/genética , Itália
2.
CEN Case Rep ; 12(4): 413-418, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-36967423

RESUMO

TRPM6 is predominantly expressed in the kidney and colon and encodes a protein containing an ion channel domain and a protein kinase domain. It is crucial for magnesium homeostasis and plays important roles in epithelial magnesium transport and the active magnesium absorption. In this study, we present a 70-day-old Iranian female patient from consanguineous parents with hypomagnesemia and secondary hypocalcemia. She presented with seizures 19 days after birth and refractory watery non-bloody diarrhea. She consequently had failure to thrive. Other features included hypotonia, wide anterior fontanel, ventriculomegaly, and pseudotumor cerebri following administration of nalidixic acid. She had severe hypomagnesemia and hypocalcemia which were treated with magnesium and calcium supplementation. Despite initial unstable response to supplemental magnesium, she eventually improved and the diarrhea discontinued. The patient was discharged by magnesium and calcium therapy. At the last follow-up at age 2.5 years, the patient remained well without any recurrence or complication. Genetic testing by whole-exome sequencing revealed a novel homozygous frameshift insertion-deletion (indel) variant in exon 26 of the TRPM6 gene, c.3693-3699del GCAAGAG ins CTGCTGTTGACATCTGCT, p.L1231Ffs*36. Segregation analysis revealed the TRPM6 heterozygous variant in both parents. Patients with biallelic TRPM6 pathogenic variants typically exhibit hypomagnesemia with secondary hypocalcemia and present with neurologic manifestations including seizures. In some patients, this is also complicated by chronic diarrhea and failure to thrive. Long-term complications are rare and most of the patients show a good prognosis with supplemental magnesium therapy.


Assuntos
Hipocalcemia , Canais de Cátion TRPM , Feminino , Humanos , Cálcio , Diarreia/etiologia , Diarreia/complicações , Insuficiência de Crescimento/etiologia , Hipocalcemia/diagnóstico , Hipocalcemia/genética , Irã (Geográfico) , Magnésio , Convulsões/complicações , Canais de Cátion TRPM/genética , Idoso
3.
Genes (Basel) ; 13(10)2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36292790

RESUMO

The guidelines for management of children with 22q11.2 deletion syndrome (22q11DS) highlight the risk for developing hypocalcemia after surgery and recommend monitoring calcium perioperatively. Despite this guidance, little has been published on postoperative hypocalcemia and 22q11DS. Our goals were to evaluate the frequency of perioperative calcium monitoring and examine how often postoperative hypocalcemia was identified. This is a retrospective chart review of patients in our 22q Center's repository. Inclusion criteria were a diagnosis of 22q11DS and a history of a non-cardiac surgical procedure. Data collected included all non-cardiac surgeries and perioperative calcium labs. In total, 68 patients were included and underwent 305 on-cardiac surgeries. Patients in only 17% of these surgeries had postoperative calcium testing, but of those tested, 58% showed hypocalcemia. Patients with history of hypocalcemia at the time of chart review undergoing non-cardiac surgeries were tested postoperatively 40% of the time; however, 67% of these had hypocalcemia. Similarly, for patients without history of hypocalcemia, postoperative testing occurred 60% of the time, with 52% of these having hypocalcemia. This study demonstrates that postoperative hypocalcemia in children with 22q11DS following non-cardiac surgeries is common and affects patients both with and without prior history of hypocalcemia. These data support establishing a protocol for perioperative testing/management of hypocalcemia for patients with 22q11DS.


Assuntos
Síndrome de DiGeorge , Hipocalcemia , Criança , Humanos , Hipocalcemia/genética , Hipocalcemia/diagnóstico , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/cirurgia , Cálcio , Estudos Retrospectivos
4.
Sci Rep ; 12(1): 10211, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35715480

RESUMO

Autosomal dominant mutations in FAM111A are causative for Kenny-Caffey syndrome type 2. Patients with Kenny-Caffey syndrome suffer from severe growth retardation, skeletal dysplasia, hypoparathyroidism, hypocalcaemia, hyperphosphataemia and hypomagnesaemia. While recent studies have reported FAM111A to function in antiviral response and DNA replication, its role in regulating electrolyte homeostasis remains unknown. In this study, we assessed the role of FAM111A in the regulation of serum electrolyte balance using a Fam111a knockout (Fam111a-/-) C57BL/6 N mouse model. Fam111a-/- mice displayed normal weight and serum parathyroid hormone (PTH) concentration and exhibited unaltered magnesium, calcium and phosphate levels in serum and 24-hour urine. Expression of calciotropic (including Cabp28k, Trpv5, Klotho and Cyp24a1), magnesiotropic (including Trpm6, Trpm7, Cnnm2 and Cnnm4) and phosphotropic (Slc20a1, Slc20a2, Slc34a1 and Slc34a3) genes in the kidneys, duodenum and colon were not affected by Fam111a depletion. Only Slc34a2 expression was significantly upregulated in the duodenum, but not in the colon. Analysis of femurs showed unaffected bone morphology and density in Fam111a-/- mice. Kidney and parathyroid histology were also normal in Fam111a-/- mice. In conclusion, our study is the first to characterise the function of FAM111A in vivo and we report that mice lacking FAM111A exhibit normal electrolyte homeostasis on a standard diet.


Assuntos
Hiperostose Cortical Congênita , Hipocalcemia , Serina Proteases , Canais de Cátion TRPM , Animais , Humanos , Camundongos , Cálcio/metabolismo , Eletrólitos/metabolismo , Hiperostose Cortical Congênita/genética , Hipocalcemia/genética , Magnésio/metabolismo , Camundongos Endogâmicos C57BL , Hormônio Paratireóideo/metabolismo , Receptores Virais , Serina Proteases/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Canais de Cátion TRPM/metabolismo , Equilíbrio Hidroeletrolítico
6.
Artigo em Inglês | MEDLINE | ID: mdl-34012148

RESUMO

BACKGROUND: Primary hypomagnesemia with secondary hypocalcemia (HSH) is a rare genetic disorder. Dysfunctional transient receptor potential melastatin 6 causes impaired intestinal absorption of magnesium, leading to low serum levels accompanied by hypocalcemia. Typical signs at initial manifestation are generalized seizures, tetany, and/or muscle spasms. CASE REPORT: We present a 5 w/o female manifesting tonic-clonic seizures. Laboratory tests detected severe hypomagnesemia and hypocalcemia. The molecular genetic analysis revealed two novel mutations within the TRPM6 gene c.3308dupC (p.Pro1104Thrfs*28) (p.P1104Tfs*28) and c.3958C>T (p.Gln1302*) (p.Q1302*) and the patient was successfully treated with Mg supplementation. CONCLUSION: Ion disbalance should be taken into account in the differential diagnosis of infantile seizures. Accurate diagnosis of HSH together with appropriate treatment are crucial to prevent irreversible neurological outcomes.


Assuntos
Hipocalcemia , Deficiência de Magnésio , Canais de Cátion TRPM , Feminino , Humanos , Hipercalciúria , Hipocalcemia/genética , Magnésio , Deficiência de Magnésio/congênito , Deficiência de Magnésio/genética , Mutação , Nefrocalcinose , Erros Inatos do Transporte Tubular Renal , Convulsões/genética , Canais de Cátion TRPM/genética
7.
Am J Med Genet A ; 185(6): 1903-1907, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33750016

RESUMO

Kenny-Caffey syndrome type 2 (KCS2) and osteocraniostenosis (OCS) are allelic disorders caused by heterozygous pathogenic variants in the FAM111A gene. Both conditions are characterized by gracile bones, characteristic facial features, hypomineralized skull with delayed closure of fontanelles and hypoparathyroidism. OCS and KCS2 are often referred to as FAM111A-related syndromes as a group; although OCS presents with a more severe, perinatal lethal phenotype. We report a novel FAM111A mutation in a fetus with poorly ossified skull, proportionate long extremities with thin diaphysis, and hypoplastic spleen consistent with FAM111A-related syndromes. Trio whole exome sequencing identified a p.Y562S de novo missense variant in the FAM111A gene. The variant shows significant similarity to other reported pathogenic mutations fitting proposed pathophysiologic mechanism which provide sufficient evidence for classification as likely pathogenic. Our report contributed a novel variant to the handful of OCS and KCS2 cases reported with pathogenic variants.


Assuntos
Anormalidades Múltiplas/genética , Doenças do Desenvolvimento Ósseo/genética , Anormalidades Craniofaciais/genética , Nanismo/genética , Hiperostose Cortical Congênita/genética , Hipocalcemia/genética , Receptores Virais/genética , Anormalidades Múltiplas/diagnóstico por imagem , Anormalidades Múltiplas/patologia , Doenças do Desenvolvimento Ósseo/diagnóstico , Doenças do Desenvolvimento Ósseo/diagnóstico por imagem , Doenças do Desenvolvimento Ósseo/patologia , Anormalidades Cardiovasculares/diagnóstico , Anormalidades Cardiovasculares/genética , Anormalidades Cardiovasculares/patologia , Anormalidades Craniofaciais/diagnóstico , Anormalidades Craniofaciais/diagnóstico por imagem , Anormalidades Craniofaciais/patologia , Nanismo/diagnóstico , Nanismo/diagnóstico por imagem , Nanismo/patologia , Ossos Faciais/anormalidades , Ossos Faciais/patologia , Feminino , Feto , Predisposição Genética para Doença , Heterozigoto , Humanos , Hiperostose Cortical Congênita/diagnóstico , Hiperostose Cortical Congênita/diagnóstico por imagem , Hiperostose Cortical Congênita/patologia , Hipocalcemia/diagnóstico , Hipocalcemia/diagnóstico por imagem , Hipocalcemia/patologia , Masculino , Mutação/genética , Gravidez , Crânio/anormalidades , Crânio/patologia , Baço/anormalidades , Baço/diagnóstico por imagem , Sequenciamento do Exoma
8.
Endocrine ; 71(3): 611-617, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33528764

RESUMO

The calcium-sensing receptor (CaSR) provides the major mechanism for the detection of extracellular calcium concentration in several cell types, via the induction of G-protein-coupled signalling. Accordingly, CaSR plays a pivotal role in calcium homeostasis, and the CaSR gene defects are related to diseases characterized by serum calcium level changes. Activating mutations of the CaSR gene cause enhanced sensitivity to extracellular calcium concentration resulting in autosomal dominant hypocalcemia or Bartter-syndrome type V. Inactivating CaSR gene mutations lead to resistance to extracellular calcium. In these cases, familial hypocalciuric hypercalcaemia (FHH1) or neonatal severe hyperparathyroidism (NSHPT) can develop. FHH2 and FHH3 are associated with mutations of genes of partner proteins of calcium signal transduction. The common polymorphisms of the CaSR gene have been reported not to affect the calcium homeostasis itself; however, they may be associated with the increased risk of malignancies.


Assuntos
Hipercalcemia , Hipocalcemia , Cálcio , Humanos , Hipercalcemia/genética , Hipocalcemia/genética , Recém-Nascido , Mutação , Doenças Raras , Receptores de Detecção de Cálcio/genética
9.
Arch. endocrinol. metab. (Online) ; 65(1): 112-116, Jan.-Feb. 2021. tab
Artigo em Inglês | LILACS | ID: biblio-1152878

RESUMO

SUMMARY Pseudohypoparathyroidism comprehends an assorted group of genetically rare disorders that share end-organ resistance to parathyroid hormone. Genetic and epigenetic modifications on guanine nucleotide-binding protein alpha-stimulating gene locus are the most common underlying mechanisms associated with pseudohypoparathyroidism. Biochemical and molecular analysis stratify pseudohypoparathyroidism into types 1A, 1B, 1C, and 2. We describe an unusual case of sporadic pseudohypoparathyroidism type 1B. A 34-year-old Caucasian woman was admitted to the emergency department, with persistent asthenia, limb paresthesias, and tactile hyposensitivity. Her physical examination, previous personal and family histories were unsuspicious, except for mild, intermittent and self-limited complaints of paresthesia during her two pregnancies, but no detailed workup was done. No typical features of Albright hereditary osteodystrophy were observed. The initial laboratory investigation showed elevated parathyroid hormone level (311.2 pg/mL), hypocalcemia (albumin-corrected serum calcium 4.3 mg/dL), hypocalciuria, hyperphosphatemia, hypophosphaturia, and vitamin D deficiency. Combined calcium, vitamin D, and magnesium supplementation was commenced, with symptomatic and analytical improvement. Albeit resolution of vitamin D deficiency, the patient relapsed with mild and intermittent lower limb paresthesias. Pseudohypoparathyroidism was confirmed by molecular identification of the 3-kb STX16 deletion. The treatment was readjusted, and one year later, symptomatic remission was attained. Clinical and biochemical features, and their respective course, along with lack of distinctive features of Albright hereditary osteodystrophy pointed to pseudohypoparathyroidism type 1B. A careful follow-up is needed to avoid complications and recurrence. Once correction of hypocalcemia and hyperphosphatemia is achieved, with no reported complications and recurrence, a good prognosis is anticipated, comparable to the general population.


Assuntos
Humanos , Feminino , Pseudo-Hipoparatireoidismo , Pseudo-Hipoparatireoidismo/diagnóstico , Pseudo-Hipoparatireoidismo/genética , Deficiência de Vitamina D , Hipocalcemia , Hipocalcemia/genética , Hormônio Paratireóideo
10.
J Clin Endocrinol Metab ; 106(4): e1775-e1792, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33340048

RESUMO

CONTEXT: The calcium-sensing receptor (CaSR) is essential to maintain a stable calcium concentration in serum. Spermatozoa are exposed to immense changes in concentrations of CaSR ligands such as calcium, magnesium, and spermine during epididymal maturation, in the ejaculate, and in the female reproductive environment. However, the role of CaSR in human spermatozoa is unknown. OBJECTIVE: This work aimed to investigate the role of CaSR in human spermatozoa. METHODS: We identified CaSR in human spermatozoa and characterized the response to CaSR agonists on intracellular calcium, acrosome reaction, and 3',5'-cyclic adenosine 5'-monophosphate (cAMP) in spermatozoa from men with either loss-of-function or gain-of-function mutations in CASR and healthy donors. RESULTS: CaSR is expressed in human spermatozoa and is essential for sensing extracellular free ionized calcium (Ca2+) and Mg2+. Activators of CaSR augmented the effect of sperm-activating signals such as the response to HCO3- and the acrosome reaction, whereas spermatozoa from men with a loss-of-function mutation in CASR had a diminished response to HCO3-, lower progesterone-mediated calcium influx, and were less likely to undergo the acrosome reaction in response to progesterone or Ca2+. CaSR activation increased cAMP through soluble adenylyl cyclase (sAC) activity and increased calcium influx through CatSper. Moreover, external Ca2+ or Mg2+ was indispensable for HCO3- activation of sAC. Two male patients with a CASR loss-of-function mutation in exon 3 presented with normal sperm counts and motility, whereas a patient with a loss-of-function mutation in exon 7 had low sperm count, motility, and morphology. CONCLUSION: CaSR is important for the sensing of Ca2+, Mg2+, and HCO3- in spermatozoa, and loss-of-function may impair male sperm function.


Assuntos
Bicarbonatos/metabolismo , Cálcio/metabolismo , Receptores de Detecção de Cálcio/fisiologia , Espermatozoides/metabolismo , Reação Acrossômica/efeitos dos fármacos , Reação Acrossômica/genética , Adulto , Bicarbonatos/farmacologia , Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Estudos de Casos e Controles , Feminino , Humanos , Hipercalcemia/congênito , Hipercalcemia/genética , Hipercalcemia/metabolismo , Hipercalcemia/patologia , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipercalciúria/patologia , Hipocalcemia/genética , Hipocalcemia/metabolismo , Hipocalcemia/patologia , Hipoparatireoidismo/congênito , Hipoparatireoidismo/genética , Hipoparatireoidismo/metabolismo , Hipoparatireoidismo/patologia , Rim/metabolismo , Rim/patologia , Magnésio/metabolismo , Magnésio/farmacologia , Masculino , Mutação , Receptores de Detecção de Cálcio/genética , Motilidade dos Espermatozoides/efeitos dos fármacos , Motilidade dos Espermatozoides/genética , Espermatozoides/efeitos dos fármacos , Espermatozoides/fisiologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia
11.
Am J Med Genet A ; 182(12): 3029-3034, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33010201

RESUMO

Kenny-Caffey syndrome (KCS) is a rare hereditary skeletal disorder involving hypoparathyroidism. The autosomal dominant form (KCS2), caused by heterozygous pathogenic variants in the FAM111A gene, is distinguished from the autosomal recessive form (KCS1) and Sanjad-Sakati syndrome (SSS), both caused by pathogenic variants in the tubulin folding cofactor E (TBCE) gene, by the absence of microcephaly and intellectual disability. We present a patient with KCS2 caused by a de novo pathogenic variant c.1706G>A (p.Arg569His) in FAM111A gene, presenting intellectual disability and microcephaly, which are considered to be typical signs of SSS. We suggest that KCS1, KCS2, and SSS may not represent mutually exclusive clinical entities, but possibly an overlapping spectrum.


Assuntos
Anormalidades Múltiplas/patologia , Nanismo/patologia , Transtornos do Crescimento/patologia , Hiperostose Cortical Congênita/patologia , Hipocalcemia/patologia , Hipoparatireoidismo/patologia , Deficiência Intelectual/patologia , Mutação , Osteocondrodisplasias/patologia , Fenótipo , Receptores Virais/genética , Convulsões/patologia , Anormalidades Múltiplas/genética , Adolescente , Nanismo/complicações , Nanismo/genética , Transtornos do Crescimento/complicações , Transtornos do Crescimento/genética , Humanos , Hiperostose Cortical Congênita/complicações , Hiperostose Cortical Congênita/genética , Hipocalcemia/complicações , Hipocalcemia/genética , Hipoparatireoidismo/complicações , Hipoparatireoidismo/genética , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Masculino , Osteocondrodisplasias/complicações , Osteocondrodisplasias/genética , Convulsões/complicações , Convulsões/genética
12.
Zhonghua Er Ke Za Zhi ; 58(11): 917-922, 2020 Nov 02.
Artigo em Chinês | MEDLINE | ID: mdl-33120464

RESUMO

Objective: To analyze the clinical and genetic characteristics of primary hypoparathyroidism in children. Methods: The clinical data including age, symptoms, laboratory examination and cranial CT of 13 children with primary hypoparathyroidism diagnosed in the Capital Institute of Pediatrics from May 2017 to December 2019 were collected and analyzed retrospectively. These children and their parents also had gene detected by whole exome sequencing and (or) copy number variation sequencing. Results: Among the 13 patients, 7 were male and 6 female. The onset age was 3 years (1 day-12 years) old. The time from onset to confirmed diagnosis was 2 months (2 days-10 years). The clinical manifestations included convulsion (9 cases), tetany (2 cases), muscle pain (1 case), mental retardation (5 cases), deafness (1 case), and initially misdiagnosed epilepsy (5 cases). The lab examination showed average blood calcium level of (1.7±0.3) mmol/L, blood phosphorus of (2.8±0.4) mmol/L, and parathyroid hormone of 8.2 (3.9-28.7)ng/L. Head CT found 7 cases of ectopic calcification. Among the 7 cases who had genetic abnormalities according to the gene detection, 5 had heterozygous deletion of 22q11.2 region, and only one of whom was diagnosed with typical DiGeorge syndrome. As for the rest 2 cases, one had autosomal dominant hypocalcemia caused by novel heterozygous variation of CaSR gene c.2495T>G (p.F832C), and the other was hypoparathyroidism-deafness-renal dysplasia syndrome caused by GATA3 c.708dupC (p.S237Qfs*66) novel heterozygous variation. Conclusions: Primary hypoparathyroidism in children is mainly characterized by hypocalcemia and usually accompanied with diverse symptoms which may indicate genetic disorders. The detection of large fragment deletion should be considered to exclude 22q11.2 deletion syndrome.


Assuntos
Hipocalcemia , Hipoparatireoidismo , Anormalidades Urogenitais , Criança , Variações do Número de Cópias de DNA , Feminino , Humanos , Hipocalcemia/diagnóstico , Hipocalcemia/genética , Hipoparatireoidismo/diagnóstico , Hipoparatireoidismo/genética , Masculino , Estudos Retrospectivos
13.
Endocrine ; 67(2): 466-472, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31939093

RESUMO

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


Assuntos
Hipocalcemia , Pseudo-Hipoparatireoidismo , Adolescente , Cromograninas/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Hipocalcemia/genética , Masculino , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Pseudo-Hipoparatireoidismo/genética
14.
Proc Natl Acad Sci U S A ; 116(8): 3294-3299, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30718391

RESUMO

The parathyroid hormone (PTH) and its related peptide (PTHrP) activate PTH receptor (PTHR) signaling, but only the PTH sustains GS-mediated adenosine 3',5'-cyclic monophosphate (cAMP) production after PTHR internalization into early endosomes. The mechanism of this unexpected behavior for a G-protein-coupled receptor is not fully understood. Here, we show that extracellular Ca2+ acts as a positive allosteric modulator of PTHR signaling that regulates sustained cAMP production. Equilibrium and kinetic studies of ligand-binding and receptor activation reveal that Ca2+ prolongs the residence time of ligands on the receptor, thus, increasing both the duration of the receptor activation and the cAMP signaling. We further find that Ca2+ allostery in the PTHR is strongly affected by the point mutation recently identified in the PTH (PTHR25C) as a new cause of hypocalcemia in humans. Using high-resolution and mass accuracy mass spectrometry approaches, we identified acidic clusters in the receptor's first extracellular loop as key determinants for Ca2+ allosterism and endosomal cAMP signaling. These findings coupled to defective Ca2+ allostery and cAMP signaling in the PTHR by hypocalcemia-causing PTHR25C suggest that Ca2+ allostery in PTHR signaling may be involved in primary signaling processes regulating calcium homeostasis.


Assuntos
AMP Cíclico/genética , Hipocalcemia/genética , Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Regulação Alostérica/genética , Animais , Células COS , Sinalização do Cálcio/genética , Chlorocebus aethiops , AMP Cíclico/metabolismo , Humanos , Hipocalcemia/metabolismo , Hipocalcemia/patologia , Cinética , Ligantes , Hormônio Paratireóideo/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/genética , Mutação Puntual/genética , Ligação Proteica/genética , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
15.
J Bone Miner Res ; 34(4): 661-668, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30496603

RESUMO

The role of the calcium-sensing receptor (CaSR) as a regulator of parathyroid hormone secretion is well established, but its function in bone is less well defined. In an effort to elucidate the CaSR's skeletal role, bone tissue and material characteristics from patients with autosomal dominant hypocalcemia (ADH), a genetic form of primary hypoparathyroidism caused by CASR gain-of-function mutations, were compared to patients with postsurgical hypoparathyroidism (PSH). Bone structure and formation/resorption indices and mineralization density distribution (BMDD), were examined in transiliac biopsy samples from PSH (n = 13) and ADH (n = 6) patients by histomorphometry and quantitative backscatter electron imaging, respectively. Bone mineral density (BMD by DXA) and biochemical characteristics were measured at the time of the biopsy. Because both study groups comprised children and adults, all measured biopsy parameters and BMD outcomes were converted to Z-scores for comparison. Histomorphometric indices were normal and not different between ADH and PSH, with the exception of mineral apposition rate Z-score, which was higher in the ADH group. Similarly, average BMD Z-scores were normal and not different between ADH and PSH. Significant differences were observed for the BMDD: average Z-scores of mean and typical degree of mineralization (CaMean, CaPeak, respectively) were lower (p = 0.02 and p = 0.03, respectively), whereas the heterogeneity of mineralization (CaWidth) and percentage of lower mineralized areas (CaLow) were increased in ADH versus PSH (p = 0.01 and p = 0.002, respectively). The BMDD outcomes point toward a direct, PTH-independent role of the CaSR in the regulation of bone mineralization. © 2018 American Society for Bone and Mineral Research.


Assuntos
Densidade Óssea , Mutação com Ganho de Função , Hipercalciúria , Hipocalcemia , Hipoparatireoidismo/congênito , Complicações Pós-Operatórias , Receptores de Detecção de Cálcio , Adolescente , Adulto , Criança , Feminino , Humanos , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipercalciúria/patologia , Hipocalcemia/genética , Hipocalcemia/metabolismo , Hipocalcemia/patologia , Hipoparatireoidismo/genética , Hipoparatireoidismo/metabolismo , Hipoparatireoidismo/patologia , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/genética , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/patologia , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo
16.
Nat Rev Endocrinol ; 15(1): 33-51, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30443043

RESUMO

The Ca2+-sensing receptor (CaSR) is a dimeric family C G protein-coupled receptor that is expressed in calcitropic tissues such as the parathyroid glands and the kidneys and signals via G proteins and ß-arrestin. The CaSR has a pivotal role in bone and mineral metabolism, as it regulates parathyroid hormone secretion, urinary Ca2+ excretion, skeletal development and lactation. The importance of the CaSR for these calcitropic processes is highlighted by loss-of-function and gain-of-function CaSR mutations that cause familial hypocalciuric hypercalcaemia and autosomal dominant hypocalcaemia, respectively, and also by the fact that alterations in parathyroid CaSR expression contribute to the pathogenesis of primary and secondary hyperparathyroidism. Moreover, the CaSR is an established therapeutic target for hyperparathyroid disorders. The CaSR is also expressed in organs not involved in Ca2+ homeostasis: it has noncalcitropic roles in lung and neuronal development, vascular tone, gastrointestinal nutrient sensing, wound healing and secretion of insulin and enteroendocrine hormones. Furthermore, the abnormal expression or function of the CaSR is implicated in cardiovascular and neurological diseases, as well as in asthma, and the CaSR is reported to protect against colorectal cancer and neuroblastoma but increase the malignant potential of prostate and breast cancers.


Assuntos
Calcimiméticos/uso terapêutico , Hipercalcemia/congênito , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/congênito , Nefrolitíase/genética , Receptores de Detecção de Cálcio/genética , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença/epidemiologia , Humanos , Hipercalcemia/tratamento farmacológico , Hipercalcemia/genética , Hipercalcemia/fisiopatologia , Hipercalciúria/tratamento farmacológico , Hipercalciúria/fisiopatologia , Hipocalcemia/tratamento farmacológico , Hipocalcemia/fisiopatologia , Hipoparatireoidismo/tratamento farmacológico , Hipoparatireoidismo/genética , Hipoparatireoidismo/fisiopatologia , Incidência , Masculino , Mutação/genética , Nefrolitíase/tratamento farmacológico , Nefrolitíase/fisiopatologia , Prognóstico , Receptores de Detecção de Cálcio/efeitos dos fármacos , Medição de Risco , Resultado do Tratamento
17.
J Clin Endocrinol Metab ; 103(8): 2879-2888, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29846619

RESUMO

Context: Autosomal dominant hypocalcemia type 1 (ADH1) is caused by heterozygous activating mutations in the calcium-sensing receptor gene (CASR). Whether polymorphisms that are benign in the heterozygous state pathologically alter receptor function in the homozygous state is unknown. Objective: To identify the genetic defect in an adolescent female with a history of surgery for bilateral cataracts and seizures. The patient has hypocalcemia, hyperphosphatemia, and low serum PTH level. The parents of the proband are healthy. Methods: Mutation testing of PTH, GNA11, GCM2, and CASR was done on leukocyte DNA of the proband. Functional analysis in transfected cells was conducted on the gene variant identified. Public single nucleotide polymorphism (SNP) databases were searched for the presence of the variant allele. Results: No mutations were identified in PTH, GNA11, and GCM2 in the proband. However, a germline homozygous variant (c.1631G>A; p.R544Q) in exon 6 of the CASR was identified. Both parents are heterozygous for the variant. The variant allele frequency was near 0.1% in SNP databases. By in vitro functional analysis, the variant was significantly more potent in stimulating both the Ca2+i and MAPK signaling pathways than wild type when transfected alone (P < 0.05) but not when transfected together with wild type. The overactivity of the mutant CaSR is due to loss of a critical structural cation-π interaction. Conclusions: The patient's hypoparathyroidism is due to homozygosity of a variant in the CASR that normally has weak or no phenotypic expression in heterozygosity. Although rare, this has important implications for genetic counseling and clinical management.


Assuntos
Hipocalcemia/genética , Hipoparatireoidismo/genética , Polimorfismo de Nucleotídeo Único , Receptores de Detecção de Cálcio/genética , Substituição de Aminoácidos , Arginina/genética , Feminino , Ácido Glutâmico/genética , Homozigoto , Humanos , Hipocalcemia/complicações , Hipoparatireoidismo/complicações , Adulto Jovem
18.
Mol Cell Endocrinol ; 456: 9-15, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27816765

RESUMO

Parathyroid glands regulate calcium homeostasis through synthesis and secretion of parathormone (PTH). They sense the extracellular calcium concentration through the G-protein coupled calcium sensing receptor (CASR) and release PTH in order to preserve calcium concentration in the physiological range. Tumors of the parathyroid glands are common endocrine neoplasia associated with primary or secondary/tertiary hyperparathyroidisms. Small non-coding RNAs are regulators of gene expression able to modulate hormone synthesis, hormone release and endocrine cell proliferation. In this scenario, microRNA (miRNA) expression profiles have been investigated in parathyroid tumors, while miRNAs are involved in hypocalcemia and uremia-induced PTH release from normal parathyroid cells. Here we reviewed data about the role of miRNAs in the regulation of: 1) PTH synthesis and secretion; 2) CASR expression; 3) parathyroid cell tumorigenesis. Though studies about miRNAs in parathyroid gland pathophysiology are limited, they contribute in elucidating regulatory pathways involved in PTH release and parathyroid cell tumorigenesis.


Assuntos
Regulação Neoplásica da Expressão Gênica , Hiperparatireoidismo/genética , Hipocalcemia/genética , MicroRNAs/genética , Glândulas Paratireoides/metabolismo , Neoplasias das Paratireoides/genética , Animais , Cálcio/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Endócrinas/metabolismo , Células Endócrinas/patologia , Homeostase , Humanos , Hiperparatireoidismo/metabolismo , Hiperparatireoidismo/fisiopatologia , Hipocalcemia/metabolismo , Hipocalcemia/fisiopatologia , MicroRNAs/metabolismo , Glândulas Paratireoides/fisiopatologia , Hormônio Paratireóideo/genética , Hormônio Paratireóideo/metabolismo , Neoplasias das Paratireoides/metabolismo , Neoplasias das Paratireoides/fisiopatologia , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo , Transdução de Sinais
19.
Eur J Endocrinol ; 175(3): 211-8, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27334330

RESUMO

OBJECTIVE: Autosomal dominant hypocalcemia (ADH) is characterized by hypocalcemia and inappropriately low PTH concentrations. ADH type 1 is caused by activating mutations in the calcium-sensing receptor (CASR), a G-protein-coupled receptor signaling through α11 (Gα11) and αq (Gαq) subunits. Heterozygous activating mutations in GNA11, the gene encoding Gα11, underlie ADH type 2. This study describes disease characteristics in a family with ADH caused by a gain-of-function mutation in GNA11. DESIGN: A three-generation family with seven members (3 adults, 4 children) presenting with ADH. METHODS: Biochemical parameters of calcium metabolism, clinical, genetic and brain imaging findings were analyzed. RESULTS: Sanger sequencing revealed a heterozygous GNA11 missense mutation (c.1018G>A, p.V340M) in all seven hypocalcemic subjects, but not in the healthy family members (n=4). The adult patients showed clinical symptoms of hypocalcemia, while the children were asymptomatic. Plasma ionized calcium ranged from 0.95 to 1.14mmol/L, yet plasma PTH was inappropriately low for the degree of hypocalcemia. Serum 25OHD was normal. Despite hypocalcemia 1,25(OH)2D and urinary calcium excretion were inappropriately in the reference range. None of the patients had nephrocalcinosis. Two adults and one child (of the two MRI scanned children) had distinct intracranial calcifications. All affected subjects had short stature (height s.d. scores ranging from -3.4 to -2.3 vs -0.5 in the unaffected children). CONCLUSIONS: The identified GNA11 mutation results in biochemical abnormalities typical for ADH. Additional features, including short stature and early intracranial calcifications, cosegregated with the mutation. These findings may indicate a wider role for Gα11 signaling besides calcium regulation.


Assuntos
Calcinose/genética , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/congênito , Adulto , Calcinose/sangue , Calcinose/diagnóstico por imagem , Cálcio/sangue , Criança , Feminino , Humanos , Hipercalciúria/sangue , Hipercalciúria/diagnóstico por imagem , Hipocalcemia/sangue , Hipocalcemia/diagnóstico por imagem , Hipoparatireoidismo/sangue , Hipoparatireoidismo/diagnóstico por imagem , Hipoparatireoidismo/genética , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Linhagem , Tomografia Computadorizada por Raios X
20.
J Biol Chem ; 291(20): 10876-85, 2016 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-26994139

RESUMO

Germline loss- and gain-of-function mutations of G-protein α-11 (Gα11), which couples the calcium-sensing receptor (CaSR) to intracellular calcium (Ca(2+) i) signaling, lead to familial hypocalciuric hypercalcemia type 2 (FHH2) and autosomal dominant hypocalcemia type 2 (ADH2), respectively, whereas somatic Gα11 mutations mediate uveal melanoma development by constitutively up-regulating MAPK signaling. Cinacalcet and NPS-2143 are allosteric CaSR activators and inactivators, respectively, that ameliorate signaling disturbances associated with CaSR mutations, but their potential to modulate abnormalities of the downstream Gα11 protein is unknown. This study investigated whether cinacalcet and NPS-2143 may rectify Ca(2+) i alterations associated with FHH2- and ADH2-causing Gα11 mutations, and evaluated the influence of germline gain-of-function Gα11 mutations on MAPK signaling by measuring ERK phosphorylation, and assessed the effect of NPS-2143 on a uveal melanoma Gα11 mutant. WT and mutant Gα11 proteins causing FHH2, ADH2 or uveal melanoma were transfected in CaSR-expressing HEK293 cells, and Ca(2+) i and ERK phosphorylation responses measured by flow-cytometry and Alphascreen immunoassay following exposure to extracellular Ca(2+) (Ca(2+) o) and allosteric modulators. Cinacalcet and NPS-2143 rectified the Ca(2+) i responses of FHH2- and ADH2-associated Gα11 loss- and gain-of-function mutations, respectively. ADH2-causing Gα11 mutations were demonstrated not to be constitutively activating and induced ERK phosphorylation following Ca(2+) o stimulation only. The increased ERK phosphorylation associated with ADH2 and uveal melanoma mutants was rectified by NPS-2143. These findings demonstrate that CaSR-targeted compounds can rectify signaling disturbances caused by germline and somatic Gα11 mutations, which respectively lead to calcium disorders and tumorigenesis; and that ADH2-causing Gα11 mutations induce non-constitutive alterations in MAPK signaling.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hipercalcemia/metabolismo , Hipocalcemia/metabolismo , Mutação de Sentido Incorreto , Receptores de Detecção de Cálcio/metabolismo , Transdução de Sinais , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/genética , Substituição de Aminoácidos , Cinacalcete/farmacologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Hipercalcemia/genética , Hipocalcemia/genética , Naftalenos/farmacologia , Receptores de Detecção de Cálcio/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA