Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Am Soc Nephrol ; 33(7): 1323-1340, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35581010

RESUMO

BACKGROUND: Impaired mineral ion metabolism is a hallmark of CKD-metabolic bone disorder. It can lead to pathologic vascular calcification and is associated with an increased risk of cardiovascular mortality. Loss of calcium-sensing receptor (CaSR) expression in vascular smooth muscle cells exacerbates vascular calcification in vitro. Conversely, vascular calcification can be reduced by calcimimetics, which function as allosteric activators of CaSR. METHODS: To determine the role of the CaSR in vascular calcification, we characterized mice with targeted Casr gene knockout in vascular smooth muscle cells ( SM22α CaSR Δflox/Δflox ). RESULTS: Vascular smooth muscle cells cultured from the knockout (KO) mice calcified more readily than those from control (wild-type) mice in vitro. However, mice did not show ectopic calcifications in vivo but they did display a profound mineral ion imbalance. Specifically, KO mice exhibited hypercalcemia, hypercalciuria, hyperphosphaturia, and osteopenia, with elevated circulating fibroblast growth factor 23 (FGF23), calcitriol (1,25-D3), and parathyroid hormone levels. Renal tubular α-Klotho protein expression was increased in KO mice but vascular α-Klotho protein expression was not. Altered CaSR expression in the kidney or the parathyroid glands could not account for the observed phenotype of the KO mice. CONCLUSIONS: These results suggest that, in addition to CaSR's established role in the parathyroid-kidney-bone axis, expression of CaSR in vascular smooth muscle cells directly contributes to total body mineral ion homeostasis.


Assuntos
Receptores de Detecção de Cálcio , Calcificação Vascular , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Klotho , Camundongos , Camundongos Knockout , Minerais/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo , Calcificação Vascular/etiologia
2.
J Biol Chem ; 294(44): 16337-16350, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31527082

RESUMO

The calcium-sensing receptor (CaSR) is critical for skeletal development, but its mechanism of action in osteoblasts is not well-characterized. In the central nervous system (CNS), Homer scaffolding proteins form signaling complexes with two CaSR-related members of the G protein-coupled receptor (GPCR) family C, metabotropic glutamate receptor 1 (mGluR1) and mGluR5. Here, we show that CaSR and Homer1 are co-expressed in mineralized mouse bone and also co-localize in primary human osteoblasts. Co-immunoprecipitation experiments confirmed that Homer1 associates with CaSR in primary human osteoblasts. The CaSR-Homer1 protein complex, whose formation was increased in response to extracellular Ca2+, was bound to mechanistic target of rapamycin (mTOR) complex 2 (mTORC2), a protein kinase that phosphorylates and activates AKT Ser/Thr kinase (AKT) at Ser473 siRNA-based gene-silencing assays with primary osteoblasts revealed that both CaSR and Homer1 are required for extracellular Ca2+-stimulated AKT phosphorylation and thereby inhibit apoptosis and promote AKT-dependent ß-catenin stabilization and cellular differentiation. To confirm the role of the CaSR-Homer1 complex in AKT initiation, we show that in HEK-293 cells, co-transfection with both Homer1c and CaSR, but neither with Homer1c nor CaSR alone, establishes sensitivity of AKT-Ser473 phosphorylation to increases in extracellular Ca2+ concentrations. These findings indicate that Homer1 mediates CaSR-dependent AKT activation via mTORC2 and thereby stabilizes ß-catenin in osteoblasts.


Assuntos
Proteínas de Arcabouço Homer/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Detecção de Cálcio/metabolismo , beta Catenina/metabolismo , Animais , Apoptose/fisiologia , Cálcio/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Receptores de Detecção de Cálcio/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
3.
Biochim Biophys Acta ; 1833(7): 1732-44, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23267858

RESUMO

The calcium sensing receptor (CaSR) is a class C G-protein-coupled receptor that is crucial for the feedback regulation of extracellular free ionised calcium homeostasis. While extracellular calcium (Ca(2+)o) is considered the primary physiological ligand, the CaSR is activated physiologically by a plethora of molecules including polyamines and l-amino acids. Activation of the CaSR by different ligands has the ability to stabilise unique conformations of the receptor, which may lead to preferential coupling of different G proteins; a phenomenon termed 'ligand-biased signalling'. While mutations of the CaSR are currently not linked with any malignancies, altered CaSR expression and function are associated with cancer progression. Interestingly, the CaSR appears to act both as a tumour suppressor and an oncogene, depending on the pathophysiology involved. Reduced expression of the CaSR occurs in both parathyroid and colon cancers, leading to loss of the growth suppressing effect of high Ca(2+)o. On the other hand, activation of the CaSR might facilitate metastasis to bone in breast and prostate cancer. A deeper understanding of the mechanisms driving CaSR signalling in different tissues, aided by a systems biology approach, will be instrumental in developing novel drugs that target the CaSR or its ligands in cancer. This article is part of a Special Issue entitled: 12th European Symposium on Calcium.


Assuntos
Antineoplásicos/uso terapêutico , Cálcio/metabolismo , Neoplasias/patologia , Receptores de Detecção de Cálcio/metabolismo , Animais , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptores de Detecção de Cálcio/antagonistas & inibidores , Transdução de Sinais
4.
Br J Nutr ; 111 Suppl 1: S16-22, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24382107

RESUMO

The extracellular Ca-sensing receptor (CaSR) is a sensor for a number of key nutrients within the body, including Ca ions (Ca²âº) and L-amino acids. The CaSR is expressed in a number of specialised cells within the gastrointestinal (GI) tract, and much work has been done to examine CaSR's role as a nutrient sensor in this system. This review article examines two emerging roles for the CaSR within the GI tract--as a mediator of kokumi taste modulation in taste cells and as a regulator of dietary hormone release in response to L-amino acids in the intestine.


Assuntos
Aminoácidos/metabolismo , Cálcio/metabolismo , Hormônios Gastrointestinais/metabolismo , Trato Gastrointestinal/metabolismo , Estado Nutricional , Receptores de Detecção de Cálcio/metabolismo , Paladar , Animais , Humanos , Mucosa Intestinal/metabolismo , Papilas Gustativas/metabolismo , Percepção Gustatória
5.
Front Physiol ; 14: 1117352, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36818436

RESUMO

Whether GPCRs support the sensing of temperature as well as other chemical and physical modalities is not well understood. Introduction: Extracellular Ca2+ concentration (Ca2+ o) modulates core body temperature and the firing rates of temperature-sensitive CNS neurons, and hypocalcemia provokes childhood seizures. However, it is not known whether these phenomena are mediated by Ca2+ o-sensing GPCRs, including the calcium-sensing receptor (CaSR). In favor of the hypothesis, CaSRs are expressed in hypothalamic regions that support core temperature regulation, and autosomal dominant hypocalcemia, due to CaSR activating mutations, is associated with childhood seizures. Methods: Herein, we tested whether CaSR-dependent signaling is temperature sensitive using an established model system, CaSR-expressing HEK-293 cells. Results: We found that the frequency of Ca2+ o-induced Ca2+ i oscillations but not the integrated response was linearly dependent on temperature in a pathophysiologically relevant range. Chimeric receptor analysis showed that the receptor's C-terminus is required for temperature-dependent modulation and experiments with the PKC inhibitor GF109203X and CaSR mutants T888A and T888M, which eliminate a key phosphorylation site, demonstrated the importance of repetitive phosphorylation and dephosphorylation. Discussion and Conclusion: CaSRs mediate temperature-sensing and the mechanism, dependent upon repetitive phosphorylation and dephosphorylation, suggests that GPCRs more generally contribute to temperature-sensing.

6.
Org Biomol Chem ; 10(39): 7922-7, 2012 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-22940747

RESUMO

Three selectively fluorinated cinacalcet analogues are prepared and their activity as calcium-sensing receptor (CaR) agonists is assessed. Individual (2R,1'R)-2 and (2S,1'R)-3 fluorocinacalcet diastereoisomers were prepared using the MacMillan asymmetric fluorination reaction. Assays with the recombinant human CaR revealed that both diastereoisomers have a similar potency to each other although slightly lower (75-80%) than that of cinacalcet 1. The SF(5)-cinacalcet analogue 4 was prepared from meta-pentafluorosulfanyl benzyl alcohol and has ~75% agonist activity relative to cinacalcet 1 indicating that the SF(5) group can replace the CF(3) group and retain significant bioactivity.


Assuntos
Fluoretos/química , Flúor/química , Naftalenos/farmacologia , Receptores de Detecção de Cálcio/agonistas , Compostos de Enxofre/química , Cinacalcete , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Naftalenos/síntese química , Naftalenos/química , Relação Estrutura-Atividade
8.
Eur Biophys J ; 39(1): 149-65, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19495742

RESUMO

Oscillations of the intracellular concentration of Ca(2+) in cultured HEK-293 cells, which heterologously expressed the calcium-sensing receptor, were recorded with the fluorophore Fura-2 using fluorescence microscopy. HEK-293 cells are extremely sensitive to small perturbations in extracellular calcium concentrations. Resting cells were attached to cover slips and perifused with saline solution containing physiologically relevant extracellular Ca(2+) concentrations in the range 0.5-5 mM. Acquired digitized images of the cells showed oscillatory fluctuations in the intracellular Ca(2+) concentration over the time course, and were processed as a function of the change in Fura-2 excitation ratio and frequency at 12-37 degrees C. Newly developed data processing techniques with wavelet analysis were used to estimate the frequency at which the rectified sinusoidal oscillations occurred; we estimated ~4 min(-1) under normal conditions. Temperature variations revealed an Arrhenius relationship in oscillation frequency. A critical Ca(2+) concentration of ~2 mM was estimated, below which oscillations did not occur. These data were used to develop a kinetic model of the system that was simulated using Mathematica; kinetic parameter values were adjusted to match the experimentally observed oscillations of intracellular Ca(2+) concentration as a function of extracellular Ca(2+) concentration, and temperature; and from these, limit cycles were obtained and control coefficients were estimated for all parameters.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Simulação por Computador , Modelos Biológicos , Linhagem Celular , Humanos , Cinética , Receptores de Detecção de Cálcio/metabolismo , Temperatura
9.
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
10.
Sci Rep ; 6: 21975, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26911344

RESUMO

Optimal fetal lung growth requires anion-driven fluid secretion into the lumen of the developing organ. The fetus is hypercalcemic compared to the mother and here we show that in the developing human lung this hypercalcaemia acts on the extracellular calcium-sensing receptor, CaSR, to promote fluid-driven lung expansion through activation of the cystic fibrosis transmembrane conductance regulator, CFTR. Several chloride channels including TMEM16, bestrophin, CFTR, CLCN2 and CLCA1, are also expressed in the developing human fetal lung at gestational stages when CaSR expression is maximal. Measurements of Cl(-)-driven fluid secretion in organ explant cultures show that pharmacological CaSR activation by calcimimetics stimulates lung fluid secretion through CFTR, an effect which in humans, but not mice, was also mimicked by fetal hypercalcemic conditions, demonstrating that the physiological relevance of such a mechanism appears to be species-specific. Calcimimetics promote CFTR opening by activating adenylate cyclase and we show that Ca(2+)-stimulated type I adenylate cyclase is expressed in the developing human lung. Together, these observations suggest that physiological fetal hypercalcemia, acting on the CaSR, promotes human fetal lung development via cAMP-dependent opening of CFTR. Disturbances in this process would be expected to permanently impact lung structure and might predispose to certain postnatal respiratory diseases.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Maturidade dos Órgãos Fetais , Pulmão/embriologia , Pulmão/metabolismo , Organogênese , Receptores de Detecção de Cálcio/metabolismo , Adenilil Ciclases/metabolismo , Animais , Anoctamina-1 , Bestrofinas , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Espaço Extracelular , Proteínas do Olho/metabolismo , Feto , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipercalcemia/genética , Hipercalcemia/metabolismo , Imuno-Histoquímica , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Camundongos , Modelos Biológicos
11.
Elife ; 52016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27434672

RESUMO

Human calcium-sensing receptor (CaSR) is a G-protein-coupled receptor (GPCR) that maintains extracellular Ca(2+) homeostasis through the regulation of parathyroid hormone secretion. It functions as a disulfide-tethered homodimer composed of three main domains, the Venus Flytrap module, cysteine-rich domain, and seven-helix transmembrane region. Here, we present the crystal structures of the entire extracellular domain of CaSR in the resting and active conformations. We provide direct evidence that L-amino acids are agonists of the receptor. In the active structure, L-Trp occupies the orthosteric agonist-binding site at the interdomain cleft and is primarily responsible for inducing extracellular domain closure to initiate receptor activation. Our structures reveal multiple binding sites for Ca(2+) and PO4(3-) ions. Both ions are crucial for structural integrity of the receptor. While Ca(2+) ions stabilize the active state, PO4(3-) ions reinforce the inactive conformation. The activation mechanism of CaSR involves the formation of a novel dimer interface between subunits.


Assuntos
Cálcio/metabolismo , Receptores de Detecção de Cálcio/agonistas , Receptores de Detecção de Cálcio/química , Triptofano/química , Triptofano/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Fosfatos/metabolismo , Ligação Proteica , Conformação Proteica , Multimerização Proteica
12.
Endocrinology ; 156(4): 1330-42, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25607893

RESUMO

Calcium-sensing receptors (CaSRs) are class C G protein-coupled receptors that respond to physiological activators, including extracellular Ca2+ (Cao2+) and L-amino acids as well as the pharmaceutical calcimimetic, cinacalcet. Unlike Cao2+, which is an orthosteric agonist, L-amino acids and cinacalcet are positive allosteric modulators. CaSR expression levels vary considerably between tissues, but the physiological significance of these differences in expression for the effects of its activators is unknown. To investigate the impact of receptor expression on CaSR-mediated signaling we used a tetracycline-inducible expression system and focused on intracellular Ca2+ (Cai2+) responses in single cells and considered both population and single-cell behavior. Increased receptor expression positively modulated CaSR-mediated Cai2+ mobilization in response to elevated Cao2+, the amino acid L-phenylalanine, or the calcimimetic cinacalcet. It lowered threshold concentrations for the initiation of Cai2+ oscillations and for their transformation to sustained Cai2+ elevations, and it increased the proportions of responding cells. It also positively modulated the frequency of Cai2+ oscillations with the order of effectiveness: cinacalcet equal to or greater than Cao2+ greater than L-phenylalanine. The results indicate that receptor expression modulates key characteristics of the Cai2+ response at the single-cell level as well as the amplitude of whole-tissue CaSR-mediated responses by recruiting quiescent cells into the active pool of responding cells. By lowering the threshold concentrations for Cao2+- and L-amino acid-induced responses below the physiological levels of these nutrients in plasma, mechanisms that up-regulate receptor expression can control tissue function in the absence of dynamic changes in ligand concentration.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Cálcio/farmacologia , Cinacalcete , Citoplasma/metabolismo , Células HEK293 , Humanos , Naftalenos/farmacologia , Fenilalanina/farmacologia , Receptores de Detecção de Cálcio/genética
13.
Sci Transl Med ; 7(284): 284ra60, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904744

RESUMO

Airway hyperresponsiveness and inflammation are fundamental hallmarks of allergic asthma that are accompanied by increases in certain polycations, such as eosinophil cationic protein. Levels of these cations in body fluids correlate with asthma severity. We show that polycations and elevated extracellular calcium activate the human recombinant and native calcium-sensing receptor (CaSR), leading to intracellular calcium mobilization, cyclic adenosine monophosphate breakdown, and p38 mitogen-activated protein kinase phosphorylation in airway smooth muscle (ASM) cells. These effects can be prevented by CaSR antagonists, termed calcilytics. Moreover, asthmatic patients and allergen-sensitized mice expressed more CaSR in ASMs than did their healthy counterparts. Indeed, polycations induced hyperreactivity in mouse bronchi, and this effect was prevented by calcilytics and absent in mice with CaSR ablation from ASM. Calcilytics also reduced airway hyperresponsiveness and inflammation in allergen-sensitized mice in vivo. These data show that a functional CaSR is up-regulated in asthmatic ASM and targeted by locally produced polycations to induce hyperresponsiveness and inflammation. Thus, calcilytics may represent effective asthma therapeutics.


Assuntos
Asma/patologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/metabolismo , Hipersensibilidade/patologia , Receptores de Detecção de Cálcio/antagonistas & inibidores , Alérgenos/química , Animais , Asma/metabolismo , Biópsia , Brônquios/metabolismo , Brônquios/patologia , Líquido da Lavagem Broncoalveolar , Broncoconstrição , Cátions , Células HEK293 , Homeostase , Humanos , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Best Pract Res Clin Endocrinol Metab ; 27(3): 443-53, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23856271

RESUMO

In fetal mammals, serum levels of both total and ionized calcium significantly exceed those in the adult. This relative fetal hypercalcemia is crucial for skeletal development and is maintained irrespectively of maternal serum calcium levels. Elegant studies by Kovacs and Kronenberg have previously addressed the role of the CaSR in creating and maintaining this relative fetal hypercalcemia, through the regulation of parathyroid hormone-related peptide secretion. More recently we have shown that the CaSR is widely distributed throughout the developing fetus, where the receptor plays major, unexpected roles in ensuring growth and maturation of several organs. In this article, we present evidence for a role of the CaSR in the control of skeletal development, and how fetal hypercalcemia, acting through the CaSR, regulates lung development.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Desenvolvimento Fetal/fisiologia , Receptores de Detecção de Cálcio/fisiologia , Animais , Desenvolvimento Ósseo/fisiologia , Cartilagem/metabolismo , Feminino , Humanos , Hipercalcemia/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Gravidez
15.
PLoS One ; 8(11): e80294, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24282533

RESUMO

Airway branching morphogenesis in utero is essential for optimal postnatal lung function. In the fetus, branching morphogenesis occurs during the pseudoglandular stage (weeks 9-17 of human gestation, embryonic days (E)11.5-16.5 in mouse) in a hypercalcaemic environment (~1.7 in the fetus vs. ~1.1-1.3 mM for an adult). Previously we have shown that fetal hypercalcemia exerts an inhibitory brake on branching morphogenesis via the calcium-sensing receptor. In addition, earlier studies have shown that nifedipine, a selective blocker of L-type voltage-gated Ca(2+) channels (VGCC), inhibits fetal lung growth, suggesting a role for VGCC in lung development. The aim of this work was to investigate the expression of VGCC in the pseudoglandular human and mouse lung, and their role in branching morphogenesis. Expression of L-type (CaV1.2 and CaV1.3), P/Q type (CaV2.1), N-type (CaV2.2), R-type (CaV2.3), and T-type (CaV3.2 and CaV3.3) VGCC was investigated in paraffin sections from week 9 human fetal lungs and E12.5 mouse embryos. Here we show, for the first time, that Cav1.2 and Cav1.3 are expressed in both the smooth muscle and epithelium of the developing human and mouse lung. Additionally, Cav2.3 was expressed in the lung epithelium of both species. Incubating E12.5 mouse lung rudiments in the presence of nifedipine doubled the amount of branching, an effect which was partly mimicked by the Cav2.3 inhibitor, SNX-482. Direct measurements of changes in epithelial cell membrane potential, using the voltage-sensitive fluorescent dye DiSBAC2(3), demonstrated that cyclic depolarisations occur within the developing epithelium and coincide with rhythmic occlusions of the lumen, driven by the naturally occurring airway peristalsis. We conclude that VGCC are expressed and functional in the fetal human and mouse lung, where they play a role in branching morphogenesis. Furthermore, rhythmic epithelial depolarisations evoked by airway peristalsis would allow for branching to match growth and distension within the developing lung.


Assuntos
Canais de Cálcio/fisiologia , Cálcio/sangue , Pulmão/embriologia , Morfogênese , Animais , Epitélio/metabolismo , Humanos , Camundongos , Músculo Liso/metabolismo
16.
Eur J Endocrinol ; 164(3): 429-35, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21135065

RESUMO

OBJECTIVE: The calcium-sensing receptor (CASR) is a key controller of calcium homeostasis by regulating parathyroid hormone (PTH) secretion and renal calcium reabsorption. CASR(T888) is a protein kinase C (PKC) phosphorylation site in the receptor's intracellular domain that has previously been identified as a critical negative regulator of CASR downstream signaling in vitro, but whose importance in vivo is unknown. CASE REPORT: The proband presented with mild symptomatic hypocalcemia following treatment for nephrotic syndrome due to minimal change glomerulonephropathy. Laboratory tests revealed inappropriately normal PTH concentrations and relative hypercalciuria typical of autosomal dominant hypocalcemia. His asymptomatic father had similar laboratory test results. DESIGN AND METHODS: The CASR gene was sequenced. To investigate the molecular consequences of CASR(T888M) mutation, site-directed mutagenesis was used to modify the wild-type (wt)-CASR gene, with the resulting mutant being transfected transiently into HEK-293 cells. RESULTS: A novel CASR missense mutation, T888M, was identified in both cases. The CASR(T888M) mutant exhibited enhanced sensitivity to extracellular calcium concentration, both for intracellular calcium (Ca(2+)(i)) mobilization and for ERK phosphorylation, despite having unaltered levels of cell surface expression. Furthermore, CASR(T888M) elicited sustained Ca(2+)(i) mobilization rather than high frequency Ca(2+)(i) oscillations, and, unlike the wt-CASR, the response was resistant to acute inhibition by the PKC activator, phorbol 12-myristate 13-acetate. CONCLUSIONS: The clinical and functional data provide the first genotype-phenotype correlation for a mutation at T888, indicating its critical physiological importance in CASR signaling. Thus, CASR(T888) represents a functionally important, inhibitory phosphorylation site that contributes to the control of PTH secretion.


Assuntos
Hipocalcemia/genética , Mutação , Proteína Quinase C/metabolismo , Receptores de Detecção de Cálcio/genética , Adulto , Humanos , Masculino , Fosforilação/genética , Adulto Jovem
17.
Curr Pharm Biotechnol ; 10(3): 270-81, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19355937

RESUMO

The extracellular calcium-sensing receptor (CaR) is a class III G-protein coupled receptor that coordinates cellular responses to changes in extracellular free Ca(2+) or amino acid concentrations as well as ionic strength and pH. It regulates signalling cascades via recruiting and controlling the activities of various heterotrimeric G-proteins, including G(q/11), G(i/0), and G(12/13), even G(s) in some "unusual" circumstances, thereby inducing changes in the metabolism of membrane lipids, the phosphorylation state of protein kinases and their targets, the activation state of monomeric G-proteins and the levels of intracellular second messengers including cAMP, Ca(2+) ions, fatty acids and other small molecules. According to its site(s) of expression and available signalling pathways, the CaR modulates cell proliferation and survival, differentiation, peptide hormone secretion, ion and water transport and various other processes. In this article we consider the complex intracellular mechanisms by which the CaR elicits its cellular functions. We also consider some of the better understood CaR-regulated cell functions and the nature of the signalling mechanisms that support them.


Assuntos
Sinalização do Cálcio/fisiologia , Espaço Extracelular/fisiologia , Receptores de Detecção de Cálcio/fisiologia , Animais , Proliferação de Células , Sobrevivência Celular/fisiologia , Epitélio/metabolismo , Humanos , Metabolismo dos Lipídeos/fisiologia , Fenômenos Fisiológicos da Nutrição , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/fisiologia , Proteínas Quinases/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA