Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Curr Opin Nephrol Hypertens ; 33(4): 433-440, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38690798

RESUMO

PURPOSE OF REVIEW: Activation of the calcium-sensing receptor (CASR) in the parathyroid gland suppresses the release of parathyroid hormone (PTH). Furthermore, activation of the renal CASR directly increases the urinary excretion of calcium, by inhibiting transepithelial calcium transport in the nephron. Gain-of-function mutations in the CASR gene lead to autosomal dominant hypocalcemia 1 (ADH1), with inappropriately low PTH levels and hypocalcemia, indicative of excessive activation of the parathyroid CASR. However, hypercalciuria is not always observed. The reason why the manifestation of hypercalciuria is not uniform among ADH1 patients is not well understood. RECENT FINDINGS: Direct activation of the CASR in the kidney has been cumbersome to study, and an indirect measure to effectively estimate the degree of CASR activation following chronic hypercalcemia or genetic gain-of-function CASR activation has been lacking. Studies have shown that expression of the pore-blocking claudin-14 is strongly stimulated by the CASR in a dose-dependent manner. This stimulatory effect is abolished after renal Casr ablation in hypercalcemic mice, suggesting that claudin-14 abundance may gauge renal CASR activation. Using this marker has led to unexpected discoveries regarding renal CASR activation. SUMMARY: These new studies have informed on renal CASR activation thresholds and the downstream CASR-regulated calcium transport mechanisms.


Assuntos
Rim , Receptores de Detecção de Cálcio , Receptores de Detecção de Cálcio/metabolismo , Receptores de Detecção de Cálcio/genética , Humanos , Animais , Rim/metabolismo , Hipercalciúria/metabolismo , Hipercalciúria/genética , Cálcio/metabolismo , Hipercalcemia/metabolismo , Hipercalcemia/genética , Claudinas/metabolismo , Claudinas/genética , Hipocalcemia , Hipoparatireoidismo/congênito
2.
Intern Med J ; 54(6): 852-860, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38665051

RESUMO

Calcium-sensing receptors (CaSRs) are G protein-coupled receptors that help maintain Ca2+ concentrations, modulating calciotropic hormone release (parathyroid hormone (PTH), calcitonin and 1,25-dihydroxyvitamin D) by direct actions in the kidneys, gastrointestinal tract and bone. Variability in population calcium levels has been attributed to single nucleotide polymorphisms in CaSR genes, and several conditions affecting calcium and phosphate homeostasis have been attributed to gain- or loss-of-function mutations. An example is autosomal dominant hypercalciuric hypocalcaemia, because of a missense mutation at codon 128 of chromosome 3, as reported in our specific case and her family. As a consequence of treating symptomatic hypocalcaemia as a child, this female subject slowly developed progressive end-stage kidney failure because of nephrocalcinosis and nephrolithiasis. After kidney transplantation, she remains asymptomatic, with decreased vitamin D and elemental calcium requirements, stable fluid and electrolyte homeostasis during intercurrent illnesses and has normalised urinary calcium and phosphate excretion, reducing the likelihood of hypercalciuria-induced graft impairment. We review the actions of the CaSR, its role in regulating renal Ca2+ homeostasis along with the impact of a proven gain-of-function mutation in the CaSR gene resulting in autosomal dominant hypercalciuric hypocalcaemia before and after kidney transplantation.


Assuntos
Cálcio , Homeostase , Transplante de Rim , Receptores de Detecção de Cálcio , Humanos , Receptores de Detecção de Cálcio/genética , Feminino , Cálcio/metabolismo , Hipocalcemia/genética , Hipocalcemia/etiologia , Hipercalciúria/genética , Hipercalcemia/genética , Rim/metabolismo , Mutação de Sentido Incorreto , Nefrocalcinose/genética , Falência Renal Crônica/cirurgia , Hipoparatireoidismo/congênito
3.
J Physiol ; 602(13): 3207-3224, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38367250

RESUMO

High concentrations of urinary calcium counteract vasopressin action via the activation of the Calcium-Sensing Receptor (CaSR) expressed in the luminal membrane of the collecting duct cells, which impairs the trafficking of aquaporin-2 (AQP2). In line with these findings, we provide evidence that, with respect to wild-type mice, CaSR knock-in (KI) mice mimicking autosomal dominant hypocalcaemia, display a significant decrease in the total content of AQP2 associated with significantly higher levels of AQP2 phosphorylation at Ser261, a phosphorylation site involved in AQP2 degradation. Interestingly, KI mice also had significantly higher levels of phosphorylated p38MAPK, a downstream effector of CaSR and known to phosphorylate AQP2 at Ser261. Moreover, ATF1 phosphorylated at Ser63, a transcription factor downstream of p38MAPK, was significantly higher in KI. In addition, KI mice had significantly higher levels of AQP2-targeting miRNA137 consistent with a post-transcriptional downregulation of AQP2. In vivo treatment of KI mice with the calcilytic JTT-305, a CaSR antagonist, increased AQP2 expression and reduced AQP2-targeting miRNA137 levels in KI mice. Together, these results provide direct evidence for a critical role of CaSR in impairing both short-term vasopressin response by increasing AQP2-pS261, as well as AQP2 abundance, via the p38MAPK-ATF1-miR137 pathway. KEY POINTS: Calcium-Sensing Receptor (CaSR) activating mutations are the main cause of autosomal dominant hypocalcaemia (ADH) characterized by inappropriate renal calcium excretion leading to hypocalcaemia and hypercalciuria. Current treatments of ADH patients with parathyroid hormone, although improving hypocalcaemia, do not improve hypercalciuria or nephrocalcinosis. In vivo treatment with calcilytic JTT-305/MK-5442 ameliorates most of the ADH phenotypes of the CaSR knock-in mice including hypercalciuria or nephrocalcinosis and reverses the downregulation of the vasopressin-sensitive aquaporin-2 (AQP2) expression, providing direct evidence for a critical role of CaSR in impairing vasopressin response. The beneficial effect of calcilytic in reducing the risk of renal calcification may occur in a parathyroid hormone-independent action through vasopressin-dependent inhibition of cAMP synthesis in the thick ascending limb and in the collecting duct. The amelioration of most of the abnormalities in calcium metabolism including hypercalciuria, renal calcification, and AQP2-mediated osmotic water reabsorption makes calcilytic a good candidate as a novel therapeutic agent for ADH.


Assuntos
Aquaporina 2 , Regulação para Baixo , Receptores de Detecção de Cálcio , Vasopressinas , Animais , Aquaporina 2/metabolismo , Aquaporina 2/genética , Receptores de Detecção de Cálcio/metabolismo , Receptores de Detecção de Cálcio/genética , Camundongos , Vasopressinas/metabolismo , Técnicas de Introdução de Genes , Rim/metabolismo , Rim/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Masculino , Transdução de Sinais , Fenótipo , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipercalciúria/tratamento farmacológico , Cálcio/metabolismo , Fosforilação , Hipocalcemia , Hipoparatireoidismo/congênito
4.
Calcif Tissue Int ; 114(2): 110-118, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38078932

RESUMO

Familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHNNC) is a rare autosomal recessive renal tubulopathy disorder characterized by excessive urinary loss of calcium and magnesium, polyuria, polydipsia, bilateral nephrocalcinosis, progressive chronic kidney disease, and renal failure. Also, sometimes amelogenesis imperfecta and severe ocular abnormalities are involved. The CLDN-16 and CLDN-19 genes encode the tight junction proteins claudin-16 and claudin-19, respectively, in the thick ascending loop of Henle in the kidney, epithelial cells of the retina, dental enamel, etc. Loss of function of the CLDN-16 and/or CLDN-19 genes leads to FHHNC. We present a case of FHHNC type 1, which was first confused with autosomal dominant hypocalcaemia (ADH) due to the presence of a very low serum parathyroid hormone (PTH) concentration and other similar clinical features before the genetic investigations. After the exome sequencing, FHHNC type 1 was confirmed by uncovering a novel homozygous missense mutation in the CLDN-16 gene (Exon 2, c.374 T > C) which causes, altered protein structure with F55S. Associated clinical, biochemical, and imaging findings also corroborate final diagnosis. Our findings expand the spectrum of the CLDN-16 mutation, which will further help in the genetic diagnosis and management of FHNNC.


Assuntos
Hipocalcemia , Hipoparatireoidismo/congênito , Nefrocalcinose , Humanos , Magnésio , Mutação de Sentido Incorreto , Nefrocalcinose/complicações , Nefrocalcinose/diagnóstico , Nefrocalcinose/genética , Hipercalciúria/complicações , Hipercalciúria/diagnóstico , Hipercalciúria/genética , Hipocalcemia/complicações , Hipocalcemia/diagnóstico , Hipocalcemia/genética , Mutação , Claudinas/genética
5.
J Int Med Res ; 50(7): 3000605221110489, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35818129

RESUMO

Autosomal dominant hypocalcemia type 1 (ADH1) is a rare inherited disorder characterized by hypocalcemia with low parathyroid hormone (PTH) levels and high urinary calcium. Its clinical presentation varies from mild asymptomatic to severe hypocalcemia. It is caused by gain-of-function mutations in the calcium-sensing receptor gene (CASR) which affect PTH secretion from the parathyroid gland and calcium resorption in the kidney. Here, we describe a case who presented with symptoms of recurrent seizure caused by hypocalcemia with a novel CASR variant. We comprehensively analyzed the phenotypic features of this presentation and reviewed the current literature to better understand clinical manifestations and the genetic spectrum.


Assuntos
Hipercalcemia , Hipocalcemia , Hipoparatireoidismo , Cálcio , Humanos , Hipercalciúria , Hipocalcemia/genética , Hipoparatireoidismo/congênito , Hipoparatireoidismo/genética , Mutação , Receptores de Detecção de Cálcio/genética
6.
Ann Clin Lab Sci ; 52(3): 494-498, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35777808

RESUMO

Autosomal dominant hypocalcemia (ADH) is characterized by hypocalcemia and inappropriately low PTH concentrations. ADH type 2 (ADH2) is caused by a heterozygous gain-of-function mutation in GNA11 that encodes the subunit of G11, the principal G protein that transduces calcium-sensing receptor signaling in the parathyroid. Clinical features related to hypocalcemia in ADH2 range from asymptomatic to tetany and seizures. We report the clinical and molecular analysis of an infant with ADH2. Exome sequencing identified a de novo heterozygous missense variant, c. G548C (p. Arg183Pro) in GNA11. This is the youngest Korean case to be diagnosed with ADH 2. In addition, we summarized the literature related to eight mutations in GNA11 from 10 families.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP , Hipocalcemia , Hipoparatireoidismo , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipocalcemia/diagnóstico , Hipocalcemia/genética , Hipocalcemia/metabolismo , Hipoparatireoidismo/congênito , Hipoparatireoidismo/genética , Hipoparatireoidismo/metabolismo , Lactente
7.
EBioMedicine ; 78: 103947, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35313217

RESUMO

BACKGROUND: Parathyroid Ca2+-sensing receptor (CaSR) activation inhibits parathyroid hormone (PTH) release, while activation of renal CaSRs attenuates Ca2+ transport and increases expression of the pore-blocking claudin-14. Patients with autosomal dominant hypocalcemia 1 (ADH1), due to activating CASR mutations, exhibit hypocalcemia but not always hypercalciuria (elevated Ca2+ in urine). The latter promotes nephrocalcinosis and renal insufficiency. Although CaSRs throughout the body including the kidney harbor activating CASR mutations, it is not understood why only some ADH1 patients display hypercalciuria. METHODS: Activation of the CaSR was studied in mouse models and a ADH1 patient. In vitro CaSR activation was studied in HEK293 cells. FINDINGS: Cldn14 showed blood Ca2+ concentration-dependent regulation, which was absent in mice with kidney-specific Casr deletion, indicating Cldn14 is a suitable marker for chronic CaSR activation in the kidney. Mice with a gain-of-function mutation in the Casr (Nuf) were hypocalcemic with low plasma PTH levels. However, renal CaSRs were not activated at baseline but only after normalizing blood Ca2+ levels. Similarly, significant hypercalciuria was not observed in a ADH1 patient until blood Ca2+ was normalized. In vitro experiments indicate that increased CaSR expression in the parathyroid relative to the kidney could contribute to tissue-specific CaSR activation thresholds. INTERPRETATION: Our findings suggest that parathyroid CaSR overactivity can reduce plasma Ca2+ to levels insufficient to activate renal CaSRs, even when an activating mutation is present. These findings identify a conceptually new mechanism of CaSR-dependent Ca2+ balance regulation that aid in explaining the spectrum of hypercalciuria in ADH1 patients. FUNDING: Erasmus+ 2018/E+/4458087, the Canadian Institutes for Health research, the Novo Nordisk Foundation, the Beckett Foundation, the Carlsberg Foundation and Independent Research Fund Denmark.


Assuntos
Hipercalciúria , Hipocalcemia , Animais , Cálcio/metabolismo , Canadá , Células HEK293 , Humanos , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/congênito , Rim/metabolismo , Camundongos , Hormônio Paratireóideo , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo
8.
Medicine (Baltimore) ; 100(25): e26443, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34160437

RESUMO

RATIONALE: Autosomal dominant hypocalcaemia type 1 (ADH1) is a genetic disease characterized by benign hypocalcemia, inappropriately low parathyroid hormone levels and mostly hypercalciuria. It is caused by the activating mutations of the calcium-sensing receptor gene (CASR), which produces a left-shift in the set point for extracellular calcium. PATIENT CONCERNS: A 50-year-old man presenting with muscle spasms was admitted into the hospital. He has a positive familial history for hypocalcemia. Auxiliary examinations demonstrated hypocalcemia, hyperphosphatemia, normal parathyroid hormone level and nephrolithiasis. A missense heterozygous variant in CASR, c 613C > T (p. Arg205Cys) which has been reported in a familial hypocalciuric hypercalcemia type 1 patient was found in the patient's genotype. It is the first time that this variant is found associating with ADH1. The variant is predicted vicious by softwares and cosegregates with ADH1 in this pedigree. CASR Arg205Cys was deduced to be the genetic cause of ADH1 in the family. DIAGNOSIS: The patient was diagnosed with ADH1 clinically and genetically. INTERVENTIONS: Oral calcitriol, calcium and hydrochlorothiazide were prescribed to the patient. OUTCOMES: After the treatments for 1 week, the patient's symptom was improved and the re-examination revealed serum calcium in the normal range. A 3-month follow-up showed his symptom was mostly relieved. LESSONS: The variant of CASR Arg205Cys, responsible for ADH1 in this family, broadened the genetic spectrum of ADH1. Further and more studies are required to evaluate the correlation between genotype and phenotype in ADH1 patients.


Assuntos
Cálcio/administração & dosagem , Hipercalciúria/diagnóstico , Hipocalcemia/diagnóstico , Hipoparatireoidismo/congênito , Receptores de Detecção de Cálcio/genética , Calcitriol/administração & dosagem , Cálcio/sangue , Análise Mutacional de DNA , Quimioterapia Combinada/métodos , Feminino , Testes Genéticos , Heterozigoto , Humanos , Hidroclorotiazida/administração & dosagem , Hipercalciúria/sangue , Hipercalciúria/genética , Hipocalcemia/sangue , Hipocalcemia/genética , Hipoparatireoidismo/sangue , Hipoparatireoidismo/diagnóstico , Hipoparatireoidismo/genética , Masculino , Anamnese , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Linhagem , Resultado do Tratamento
9.
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
10.
BMJ Case Rep ; 13(6)2020 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-32513763

RESUMO

Autosomal dominant hypocalcaemia is a rare aetiology of hypocalcaemia, caused by gain-of-function mutations of the calcium-sensing receptor (CASR) gene. We present two cases of two asymptomatic women (50-year-old-case 1 and 25-year-old-case 2), referred to our endocrinology department for investigation of hypocalcaemia, hyperphosphatemia and inappropriately low parathormone. Both patients had relatives with the same laboratorial findings. At diagnosis, both patients presented basal ganglia calcifications. Genetic analysis was performed, identifying two novel heterozygous CASR variants: c.2269G>A (p.Glu757Lys) and c.2086C>G (p.Leu696Val), respectively, for case 1 and case 2. Affected individuals started oral calcium and vitamin D analogues, aiming to a low-normal calcium level. They remain under observation and are asymptomatic.


Assuntos
Gânglios da Base/diagnóstico por imagem , Calcinose , Hipercalciúria , Hiperfosfatemia , Hipocalcemia , Hipoparatireoidismo/congênito , Receptores de Detecção de Cálcio/genética , Conduta Expectante/métodos , Adulto , Doenças Assintomáticas , Calcinose/diagnóstico por imagem , Calcinose/etiologia , Sinalização do Cálcio , Feminino , Humanos , Hipercalciúria/sangue , Hipercalciúria/diagnóstico , Hiperfosfatemia/sangue , Hiperfosfatemia/etiologia , Hipocalcemia/sangue , Hipocalcemia/diagnóstico , Hipoparatireoidismo/sangue , Hipoparatireoidismo/diagnóstico , Pessoa de Meia-Idade , Mutação , Hormônio Paratireóideo/análise , Linhagem
11.
J Clin Endocrinol Metab ; 105(7)2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32421798

RESUMO

CONTEXT: Familial hypoparathyroidism has a heterogeneous presentation where patients usually have low parathyroid hormone (PTH) levels due to impaired production or secretion. This contrasts with pseudohypoparathyroidism, in which PTH resistance is usually associated with an elevated serum PTH. High levels of circulating PTH can also be due to bioinactive PTH, which is difficult to distinguish from pseudohypoparathyroidism on biochemical grounds. CASE DESCRIPTION: We report on 2 sisters from consanguineous parents who presented with tetany at birth and were diagnosed with congenital hypocalcemia. Serum PTH levels were normal for many years, but progressively increased in midadulthood to greater than 100x the upper limit of normal on multiple assays. Homozygosity mapping was performed on 1 sister that demonstrated loss of heterozygosity (LOH) around PTH. Sequencing revealed a previously unreported variant, c.94T>C, predicting a codon change of p.Ser32Pro that is biologically inactive. CONCLUSIONS: This case report shows a previously unreported unusual biochemical phenotype of a rising PTH in the context of a novel PTH mutation. This expands the evolving genotypes associated with hypoparathyroidism without established gene mutations.


Assuntos
Hipoparatireoidismo/sangue , Hipoparatireoidismo/genética , Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/genética , Feminino , Humanos , Hipocalcemia/sangue , Hipocalcemia/complicações , Hipoparatireoidismo/complicações , Hipoparatireoidismo/congênito , Pessoa de Meia-Idade , Mutação , Irmãos
12.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31820785

RESUMO

CONTEXT: Autosomal dominant hypocalcemia types 1 and 2 (ADH1 and ADH2) are caused by germline gain-of-function mutations of the calcium-sensing receptor (CaSR) and its signaling partner, the G-protein subunit α 11 (Gα 11), respectively. More than 70 different gain-of-function CaSR mutations, but only 6 different gain-of-function Gα 11 mutations are reported to date. METHODS: We ascertained 2 additional ADH families and investigated them for CaSR and Gα 11 mutations. The effects of identified variants on CaSR signaling were evaluated by transiently transfecting wild-type (WT) and variant expression constructs into HEK293 cells stably expressing CaSR (HEK-CaSR), and measuring intracellular calcium (Ca2+i) and MAPK responses following stimulation with extracellular calcium (Ca2+e). RESULTS: CaSR variants were not found, but 2 novel heterozygous germline Gα 11 variants, p.Gly66Ser and p.Arg149His, were identified. Homology modeling of these revealed that the Gly66 and Arg149 residues are located at the interface between the Gα 11 helical and GTPase domains, which is involved in guanine nucleotide binding, and this is the site of 3 other reported ADH2 mutations. The Ca2+i and MAPK responses of cells expressing the variant Ser66 or His149 Gα 11 proteins were similar to WT cells at low Ca2+e, but significantly increased in a dose-dependent manner following Ca2+e stimulation, thereby indicating that the p.Gly66Ser and p.Arg149His variants represent pathogenic gain-of-function Gα 11 mutations. Treatment of Ser66- and His149-Gα 11 expressing cells with the CaSR negative allosteric modulator NPS 2143 normalized Ca2+i and MAPK responses. CONCLUSION: Two novel ADH2-causing mutations that highlight the Gα 11 interdomain interface as a hotspot for gain-of-function Gα 11 mutations have been identified.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Mutação com Ganho de Função/genética , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/congênito , Receptores de Detecção de Cálcio/genética , Adulto , Criança , Feminino , Células HEK293 , Humanos , Hipoparatireoidismo/genética , Masculino , Linhagem
13.
J Bone Miner Res ; 34(9): 1609-1618, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31063613

RESUMO

Autosomal dominant hypocalcemia type 1 (ADH1) is a rare form of hypoparathyroidism caused by heterozygous, gain-of-function mutations of the calcium-sensing receptor gene (CAR). Individuals are hypocalcemic with inappropriately low parathyroid hormone (PTH) secretion and relative hypercalciuria. Calcilytics are negative allosteric modulators of the extracellular calcium receptor (CaR) and therefore may have therapeutic benefits in ADH1. Five adults with ADH1 due to four distinct CAR mutations received escalating doses of the calcilytic compound NPSP795 (SHP635) on 3 consecutive days. Pharmacokinetics, pharmacodynamics, efficacy, and safety were assessed. Parallel in vitro testing with subject CaR mutations assessed the effects of NPSP795 on cytoplasmic calcium concentrations (Ca2+i ), and ERK and p38MAPK phosphorylation. These effects were correlated with clinical responses to administration of NPSP795. NPSP795 increased plasma PTH levels in a concentration-dependent manner up to 129% above baseline (p = 0.013) at the highest exposure levels. Fractional excretion of calcium (FECa) trended down but not significantly so. Blood ionized calcium levels remained stable during NPSP795 infusion despite fasting, no calcitriol supplementation, and little calcium supplementation. NPSP795 was generally safe and well-tolerated. There was significant variability in response clinically across genotypes. In vitro, all mutant CaRs were half-maximally activated (EC50 ) at lower concentrations of extracellular calcium (Ca2+o ) compared to wild-type (WT) CaR; NPSP795 exposure increased the EC50 for all CaR activity readouts. However, the in vitro responses to NPSP795 did not correlate with any clinical parameters. NPSP795 increased plasma PTH levels in subjects with ADH1 in a dose-dependent manner, and thus, serves as proof-of-concept that calcilytics could be an effective treatment for ADH1. Albeit all mutations appear to be activating at the CaR, in vitro observations were not predictive of the in vivo phenotype or the response to calcilytics, suggesting that other parameters impact the response to the drug. © 2019 American Society for Bone and Mineral Research.


Assuntos
Compostos de Cálcio/uso terapêutico , Hipercalciúria/tratamento farmacológico , Hipocalcemia/tratamento farmacológico , Hipoparatireoidismo/congênito , Adulto , Área Sob a Curva , Compostos de Cálcio/efeitos adversos , Compostos de Cálcio/farmacocinética , Linhagem Celular , Feminino , Genótipo , Humanos , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/tratamento farmacológico , Hipoparatireoidismo/genética , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
14.
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
15.
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
16.
J Clin Endocrinol Metab ; 103(11): 4023-4032, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30137364

RESUMO

Context: Most cases of autosomal dominant isolated hypoparathyroidism are caused by gain-of-function mutations in CASR or GNA11 or dominant negative mutations in GCM2 or PTH. Objective: To identify the genetic etiology for dominantly transmitted isolated hypoparathyroidism in two multigenerational families with 14 affected family members. Methods: We performed whole exome sequencing of DNA from two families and examined the consequences of mutations by minigene splicing assay. Results: We discovered disease-causing mutations in both families. A splice-altering mutation in TBX1 (c.1009+1G>C) leading to skipping of exon 8 (101 bp) was identified in 10 affected family members and five unaffected subjects of family A, indicating reduced penetrance for this point mutation. In a second family from France (family B), we identified another splice-altering mutation (c.1009+2T>C) adjacent to the mutation identified in family A that results in skipping of the same exon; two subjects in family B had isolated hypoparathyroidism, whereas a third subject manifested the clinical triad of the 22q11.2 deletion syndrome, indicative of variable expressivity. Conclusions: We report evidence that heterozygous TBX1 mutations can cause isolated hypoparathyroidism. This study adds knowledge to the increasingly expanding list of causative and candidate genes in isolated hypoparathyroidism.


Assuntos
Síndrome de DiGeorge/genética , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/congênito , Proteínas com Domínio T/genética , Idoso , Síndrome de DiGeorge/sangue , Síndrome de DiGeorge/diagnóstico , Éxons/genética , Feminino , Heterozigoto , Humanos , Hipercalciúria/sangue , Hipercalciúria/diagnóstico , Hipocalcemia/sangue , Hipocalcemia/diagnóstico , Hipoparatireoidismo/sangue , Hipoparatireoidismo/diagnóstico , Hipoparatireoidismo/genética , Lactente , Masculino , Mutação , Linhagem , Penetrância , Sítios de Splice de RNA/genética , Sequenciamento do Exoma
17.
Hum Mol Genet ; 27(21): 3720-3733, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30052933

RESUMO

The calcium-sensing receptor (CaSR) is a homodimeric G-protein-coupled receptor that signals via intracellular calcium (Ca2+i) mobilisation and phosphorylation of extracellular signal-regulated kinase 1/2 (ERK) to regulate extracellular calcium (Ca2+e) homeostasis. The central importance of the CaSR in Ca2+e homeostasis has been demonstrated by the identification of loss- or gain-of-function CaSR mutations that lead to familial hypocalciuric hypercalcaemia (FHH) or autosomal dominant hypocalcaemia (ADH), respectively. However, the mechanisms determining whether the CaSR signals via Ca2+i or ERK have not been established, and we hypothesised that some CaSR residues, which are the site of both loss- and gain-of-function mutations, may act as molecular switches to direct signalling through these pathways. An analysis of CaSR mutations identified in >300 hypercalcaemic and hypocalcaemic probands revealed five 'disease-switch' residues (Gln27, Asn178, Ser657, Ser820 and Thr828) that are affected by FHH and ADH mutations. Functional expression studies using HEK293 cells showed disease-switch residue mutations to commonly display signalling bias. For example, two FHH-associated mutations (p.Asn178Asp and p.Ser820Ala) impaired Ca2+i signalling without altering ERK phosphorylation. In contrast, an ADH-associated p.Ser657Cys mutation uncoupled signalling by leading to increased Ca2+i mobilization while decreasing ERK phosphorylation. Structural analysis of these five CaSR disease-switch residues together with four reported disease-switch residues revealed these residues to be located at conformationally active regions of the CaSR such as the extracellular dimer interface and transmembrane domain. Thus, our findings indicate that disease-switch residues are located at sites critical for CaSR activation and play a role in mediating signalling bias.


Assuntos
Mutação com Ganho de Função , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/congênito , Mutação com Perda de Função , Receptores de Detecção de Cálcio/genética , Transdução de Sinais , Sequência de Aminoácidos , Sinalização do Cálcio , Análise Mutacional de DNA , Células HEK293 , Humanos , Hipercalciúria/metabolismo , Hipocalcemia/metabolismo , Hipoparatireoidismo/genética , Hipoparatireoidismo/metabolismo , Conformação Proteica , Receptores de Detecção de Cálcio/metabolismo , Alinhamento de Sequência
18.
BMJ Case Rep ; 20182018 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-29804071

RESUMO

A female patient with consanguineous parents presented at the age of 4 with isolated hypoparathyroidism due to a parathyroid hormone (PTH) gene mutation. She was managed with alfacalcidol and calcium supplements, and developed normally. Her consanguineous parents described symptoms suggestive of hypocalcaemia but had normal serum calcium and low normal PTH levels. A molecular diagnosis obtained in her adulthood revealed the presence of homozygous point mutation (c.68C>A) in exon 2 introducing a premature stop codon resulting in a non-functional precursor protein. This mutation has been reported only once before. Our patient remained on stable doses of alfacalcidol during pregnancy, but stopped all supplementation while breast feeding. This case confirms that alternative mechanisms (likely breast-derived parathyroid hormone-related protein) contribute to calcium homeostasis during breast feeding. Heterozygotes for the c.68C>A mutation may have latent hypoparathyroidism and maintain calcium homeostasis except during prolonged hypocalcaemia. This would suggest incomplete dominance, or a dose effect of the wild-type PTH allele.


Assuntos
Aleitamento Materno , Hipoparatireoidismo/congênito , Proteína Relacionada ao Hormônio Paratireóideo/genética , Mutação Puntual , Complicações na Gravidez/genética , Adulto , Códon , Consanguinidade , Éxons , Feminino , Homozigoto , Humanos , Hipoparatireoidismo/genética , Gravidez , Resultado da Gravidez
19.
J Mol Endocrinol ; 61(1): R1-R12, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29599414

RESUMO

The calcium-sensing receptor (CASR) is a class C G-protein-coupled receptor (GPCR) that detects extracellular calcium concentrations, and modulates parathyroid hormone secretion and urinary calcium excretion to maintain calcium homeostasis. The CASR utilises multiple heterotrimeric G-proteins to mediate signalling effects including activation of intracellular calcium release; mitogen-activated protein kinase (MAPK) pathways; membrane ruffling; and inhibition of cAMP production. By studying germline mutations in the CASR and proteins within its signalling pathway that cause hyper- and hypocalcaemic disorders, novel mechanisms governing GPCR signalling and trafficking have been elucidated. This review focusses on two recently described pathways that provide novel insights into CASR signalling and trafficking mechanisms. The first, identified by studying a CASR gain-of-function mutation that causes autosomal dominant hypocalcaemia (ADH), demonstrated a structural motif located between the third transmembrane domain and the second extracellular loop of the CASR that mediates biased signalling by activating a novel ß-arrestin-mediated G-protein-independent pathway. The second, in which the mechanism by which adaptor protein-2 σ-subunit (AP2σ) mutations cause familial hypocalciuric hypercalcaemia (FHH) was investigated, demonstrated that AP2σ mutations impair CASR internalisation and reduce multiple CASR-mediated signalling pathways. Furthermore, these studies showed that the CASR can signal from the cell surface using multiple G-protein pathways, whilst sustained signalling is mediated only by the Gq/11 pathway. Thus, studies of FHH- and ADH-associated mutations have revealed novel steps by which CASR mediates signalling and compartmental bias, and these pathways could provide new targets for therapies for patients with calcaemic disorders.


Assuntos
Cálcio/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Animais , Homeostase/fisiologia , Humanos , Hipercalciúria/metabolismo , Hipocalcemia/metabolismo , Hipoparatireoidismo/congênito , Hipoparatireoidismo/metabolismo , Receptores de Detecção de Cálcio/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
20.
Sci Signal ; 11(518)2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29463778

RESUMO

The calcium-sensing receptor (CaSR) is a G protein-coupled receptor (GPCR) that signals through Gq/11 and Gi/o to stimulate cytosolic calcium (Ca2+i) and mitogen-activated protein kinase (MAPK) signaling to control extracellular calcium homeostasis. Studies of loss- and gain-of-function CASR mutations, which cause familial hypocalciuric hypercalcemia type 1 (FHH1) and autosomal dominant hypocalcemia type 1 (ADH1), respectively, have revealed that the CaSR signals in a biased manner. Thus, some mutations associated with FHH1 lead to signaling predominantly through the MAPK pathway, whereas mutations associated with ADH1 preferentially enhance Ca2+i responses. We report a previously unidentified ADH1-associated R680G CaSR mutation, which led to the identification of a CaSR structural motif that mediates biased signaling. Expressing CaSRR680G in HEK 293 cells showed that this mutation increased MAPK signaling without altering Ca2+i responses. Moreover, this gain of function in MAPK activity occurred independently of Gq/11 and Gi/o and was mediated instead by a noncanonical pathway involving ß-arrestin proteins. Homology modeling and mutagenesis studies showed that the R680G CaSR mutation selectively enhanced ß-arrestin signaling by disrupting a salt bridge formed between Arg680 and Glu767, which are located in CaSR transmembrane domain 3 and extracellular loop 2, respectively. Thus, our results demonstrate CaSR signaling through ß-arrestin and the importance of the Arg680-Glu767 salt bridge in mediating signaling bias.


Assuntos
Membrana Celular/metabolismo , Hipercalciúria/fisiopatologia , Hipocalcemia/fisiopatologia , Hipoparatireoidismo/congênito , Sistema de Sinalização das MAP Quinases , Mutação , Receptores de Detecção de Cálcio/metabolismo , Sais/metabolismo , beta-Arrestinas/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Cálcio/metabolismo , Membrana Celular/química , Saúde da Família , Feminino , Humanos , Hipercalciúria/genética , Hipocalcemia/genética , Hipoparatireoidismo/genética , Hipoparatireoidismo/fisiopatologia , Masculino , Modelos Moleculares , Linhagem , Conformação Proteica , Receptores de Detecção de Cálcio/química , Receptores de Detecção de Cálcio/genética , Sais/química , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA