Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Am Soc Nephrol ; 33(1): 201-212, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34732508

RESUMEN

BACKGROUND: Phosphorus levels in the range seen clinically among patients undergoing dialysis have been reported to attenuate calcium receptor activation and modify parathyroid hormone (PTH) release from isolated parathyroid glands in vitro. Some clinicians and providers of dialysis thus have suggested that calcimimetic agents are ineffective and should not be used to manage secondary hyperparathyroidism among those undergoing dialysis when serum phosphorus concentrations exceed certain threshold levels. METHODS: To determine whether hyperphosphatemia diminishes the therapeutic response to calcimimetic agents, we used data from large clinical trials to analyze the effects of etelcalcetide and cinacalcet to lower plasma PTH levels in individuals on hemodialysis who had secondary hyperparathyroidism and varying degrees of hyperphosphatemia. RESULTS: Plasma PTH levels declined progressively during 26 weeks of treatment with either etelcalcetide or cinacalcet without regard to the degree of hyperphosphatemia at baseline. However, with each calcimimetic agent, the decreases in PTH from baseline were less at each interval of follow-up during the trials among participants with serum phosphorus levels above one of three prespecified threshold values compared with those with serum phosphorus levels below these thresholds. CONCLUSIONS: These in vivo findings are the first in humans to support the idea that hyperphosphatemia attenuates calcium receptor activation by calcium ions and by calcimimetic agents. The effect of hyperphosphatemia on the responsiveness to calcimimetic agents appears relatively modest, however, and unlikely to be significant therapeutically. The efficacy of treatment with calcimimetic agents for lowering plasma PTH levels among those with secondary hyperparathyroidism remains robust despite substantial elevations in serum phosphorus.


Asunto(s)
Calcimiméticos/uso terapéutico , Hiperparatiroidismo Secundario/tratamiento farmacológico , Hiperfosfatemia/complicaciones , Diálisis Renal , Insuficiencia Renal Crónica/complicaciones , Anciano , Cinacalcet/uso terapéutico , Femenino , Humanos , Hiperparatiroidismo Secundario/sangre , Hiperparatiroidismo Secundario/complicaciones , Hiperfosfatemia/sangre , Masculino , Persona de Mediana Edad , Hormona Paratiroidea/sangre , Péptidos/uso terapéutico , Fósforo/sangre , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/terapia , Estudios Retrospectivos
2.
J Am Soc Nephrol ; 33(7): 1323-1340, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35581010

RESUMEN

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.


Asunto(s)
Receptores Sensibles al Calcio , Calcificación Vascular , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Klotho , Ratones , Ratones Noqueados , Minerales/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Calcificación Vascular/etiología
3.
Pharmacol Rev ; 72(3): 558-604, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32467152

RESUMEN

The calcium-sensing receptor (CaSR) is a class C G protein-coupled receptor that responds to multiple endogenous agonists and allosteric modulators, including divalent and trivalent cations, L-amino acids, γ-glutamyl peptides, polyamines, polycationic peptides, and protons. The CaSR plays a critical role in extracellular calcium (Ca2+ o) homeostasis, as demonstrated by the many naturally occurring mutations in the CaSR or its signaling partners that cause Ca2+ o homeostasis disorders. However, CaSR tissue expression in mammals is broad and includes tissues unrelated to Ca2+ o homeostasis, in which it, for example, regulates the secretion of digestive hormones, airway constriction, cardiovascular effects, cellular differentiation, and proliferation. Thus, although the CaSR is targeted clinically by the positive allosteric modulators (PAMs) cinacalcet, evocalcet, and etelcalcetide in hyperparathyroidism, it is also a putative therapeutic target in diabetes, asthma, cardiovascular disease, and cancer. The CaSR is somewhat unique in possessing multiple ligand binding sites, including at least five putative sites for the "orthosteric" agonist Ca2+ o, an allosteric site for endogenous L-amino acids, two further allosteric sites for small molecules and the peptide PAM, etelcalcetide, and additional sites for other cations and anions. The CaSR is promiscuous in its G protein-coupling preferences, and signals via Gq/11, Gi/o, potentially G12/13, and even Gs in some cell types. Not surprisingly, the CaSR is subject to biased agonism, in which distinct ligands preferentially stimulate a subset of the CaSR's possible signaling responses, to the exclusion of others. The CaSR thus serves as a model receptor to study natural bias and allostery. SIGNIFICANCE STATEMENT: The calcium-sensing receptor (CaSR) is a complex G protein-coupled receptor that possesses multiple orthosteric and allosteric binding sites, is subject to biased signaling via several different G proteins, and has numerous (patho)physiological roles. Understanding the complexities of CaSR structure, function, and biology will aid future drug discovery efforts seeking to target this receptor for a diversity of diseases. This review summarizes what is known to date regarding key structural, pharmacological, and physiological features of the CaSR.


Asunto(s)
Receptores Sensibles al Calcio/agonistas , Receptores Sensibles al Calcio/antagonistas & inhibidores , Animales , Sitios de Unión , Proteínas de Unión al GTP/metabolismo , Humanos , Modelos Moleculares , Receptores Sensibles al Calcio/química , Receptores Sensibles al Calcio/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/farmacología
4.
Int J Mol Sci ; 22(14)2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34298895

RESUMEN

Skeletal muscle has an outstanding capacity for regeneration in response to injuries, but there are disorders in which this process is seriously impaired, such as sarcopenia. Pharmacological treatments to restore muscle trophism are not available, therefore, the identification of suitable therapeutic targets that could be useful for the treatment of skeletal reduced myogenesis is highly desirable. In this in vitro study, we explored the expression and function of the calcium-sensing receptor (CaSR) in human skeletal muscle tissues and their derived satellite cells. The results obtained from analyses with various techniques of gene and protein CaSR expression and of its secondary messengers in response to calcium (Ca2+) and CaSR drugs have demonstrated that this receptor is not present in human skeletal muscle tissues, neither in the established satellite cells, nor during in vitro myogenic differentiation. Taken together, our data suggest that, although CaSR is a very important drug target in physiology and pathology, this receptor probably does not have any physiological role in skeletal muscle in normal conditions.


Asunto(s)
Calcio/metabolismo , Músculo Esquelético/metabolismo , Receptores Sensibles al Calcio/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Células HEK293 , Humanos , Desarrollo de Músculos/fisiología , Mioblastos/metabolismo , Regeneración/fisiología , Sarcopenia/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal/fisiología
5.
Adv Exp Med Biol ; 1131: 1031-1063, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31646544

RESUMEN

The calcium-sensing receptor (CaSR) is a G protein-coupled receptor that plays a key role in calcium homeostasis, by sensing free calcium levels in blood and regulating parathyroid hormone secretion in response. The CaSR is highly expressed in parathyroid gland and kidney where its role is well characterised, but also in other tissues where its function remains to be determined. The CaSR can be activated by a variety of endogenous ligands, as well as by synthetic modulators such as Cinacalcet, used in the clinic to treat secondary hyperparathyroidism in patients with chronic kidney disease. The CaSR couples to multiple G proteins, in a tissue-specific manner, activating several signalling pathways and thus regulating diverse intracellular events. The multifaceted nature of this receptor makes it a valuable therapeutic target for calciotropic and non-calciotropic diseases. It is therefore essential to understand the complexity behind the pharmacology, trafficking, and signalling characteristics of this receptor. This review provides an overview of the latest knowledge about the CaSR and discusses future hot topics in this field.


Asunto(s)
Calcio , Hiperparatiroidismo Secundario , Receptores Sensibles al Calcio , Calcio/metabolismo , Cinacalcet/uso terapéutico , Humanos , Hiperparatiroidismo Secundario/tratamiento farmacológico , Hiperparatiroidismo Secundario/etiología , Riñón/metabolismo , Glándulas Paratiroides/metabolismo , Receptores Sensibles al Calcio/química , Receptores Sensibles al Calcio/metabolismo , Insuficiencia Renal Crónica/complicaciones
6.
Mol Pharmacol ; 96(2): 204-211, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189667

RESUMEN

The calcium-sensing receptor (CaS) is the principal controller of extracellular calcium (Ca2+ o) homeostasis and is inhibited in vitro and in vivo by protein kinase C (PKC)-mediated phosphorylation at CaST888 However, PKC inhibition enhances signaling even in CaSs lacking Thr-888, suggesting that an additional inhibitory site exists. An apparently equivalent PKC regulatory site in metabotropic glutamate receptor 5 (Ser-839) aligns not with CaST888 but instead with CaSS875, which was not previously considered to be a PKC site. CaSS875A (nonphosphorylatable) exhibited significantly enhanced Ca2+ o sensitivity of both intracellular Ca2+ mobilization and extracellular signal-regulated kinase 1/2 activation, whereas the phosphomimetic CaSS875D mutant exhibited a loss of function. The CaSS875A/T888A double mutant exhibited even greater Ca2+ o sensitivity than CaST888A alone, a response no longer enhanced by PKC inhibition. Finally, when expressed in CaS lacking its extracellular domain, the CaSS875A/T888A double mutation elicited maximal activation even under control conditions, but remained sensitive to negative allosteric modulation [N-(2-hydroxy-3-(2-cyano-3-chlorophenoxy)propyl)-1,1-dimethyl-2-(2-nephthyl)ethylamine] or Ca2+ o removal. Therefore, we have now identified CaSS875 as the missing PKC phosphorylation site that, together with CaST888, shapes the CaS signaling that underpins Ca2+ o homeostasis. Together with the inactive form of the CaS extracellular domain, these sites attenuate Ca2+ o sensitivity to attain appropriate physiologic Ca2+ o sensing. SIGNIFICANCE STATEMENT: Serine-875 represents the missing inhibitory PKC phosphorlyation site in CaS that in tandem with Thr-888 controls receptor activity.


Asunto(s)
Mutación , Proteína Quinasa C/metabolismo , Receptores Sensibles al Calcio/química , Serina/metabolismo , Calcio/metabolismo , Señalización del Calcio , Células HEK293 , Humanos , Fosforilación , Dominios Proteicos , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores Sensibles al Calcio/genética , Receptores Sensibles al Calcio/metabolismo , Treonina/metabolismo
7.
J Am Soc Nephrol ; 26(9): 2163-71, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25556167

RESUMEN

The calcium-sensing receptor (CaR) modulates renal calcium reabsorption and parathyroid hormone (PTH) secretion and is involved in the etiology of secondary hyperparathyroidism in CKD. Supraphysiologic changes in extracellular pH (pHo) modulate CaR responsiveness in HEK-293 (CaR-HEK) cells. Therefore, because acidosis and alkalosis are associated with altered PTH secretion in vivo, we examined whether pathophysiologic changes in pHo can significantly alter CaR responsiveness in both heterologous and endogenous expression systems and whether this affects PTH secretion. In both CaR-HEK and isolated bovine parathyroid cells, decreasing pHo from 7.4 to 7.2 rapidly inhibited CaR-induced intracellular calcium (Ca(2+)i) mobilization, whereas raising pHo to 7.6 potentiated responsiveness to extracellular calcium (Ca(2+)o). Similar pHo effects were observed for Ca(2+)o-induced extracellular signal-regulated kinase phosphorylation and actin polymerization and for L-Phe-induced Ca(2+)i mobilization. Intracellular pH was unaffected by acute 0.4-unit pHo changes, and the presence of physiologic albumin concentrations failed to attenuate the pHo-mediated effects. None of the individual point mutations created at histidine or cysteine residues in the extracellular domain of CaR attenuated pHo sensitivity. Finally, pathophysiologic pHo elevation reversibly suppressed PTH secretion from perifused human parathyroid cells, and acidosis transiently increased PTH secretion. Therefore, pathophysiologic pHo changes can modulate CaR responsiveness in HEK-293 and parathyroid cells independently of extracellular histidine residues. Specifically, pathophysiologic acidification inhibits CaR activity, thus permitting PTH secretion, whereas alkalinization potentiates CaR activity to suppress PTH secretion. These findings suggest that acid-base disturbances may affect the CaR-mediated control of parathyroid function and calcium metabolism in vivo.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glándulas Paratiroides/metabolismo , Hormona Paratiroidea/metabolismo , Receptores Sensibles al Calcio/metabolismo , Acidosis/metabolismo , Alcalosis/metabolismo , Animales , Bovinos , Cisteína/genética , Cisteína/metabolismo , Células HEK293 , Histidina/genética , Histidina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Fosforilación
8.
Am J Physiol Cell Physiol ; 306(7): C679-86, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24477236

RESUMEN

Statins and aspirin deliver well-established cardiovascular benefits resulting in their increased use as combined polypills to decrease risk of stroke and heart disease. However, the direct endothelial effect of combined statin/aspirin cotreatment remains unclear. Histamine is an inflammatory mediator that increases vascular permeability, and so we examined the effect of treating human umbilical vein endothelial cells (HUVECs) for 24 h with 1 µM simvastatin and 100 µM aspirin on histamine responsiveness. Subsequent histamine (1 µM) challenge increased intracellular calcium (Ca(2+)i) concentration, an effect that was significantly inhibited by combined simvastatin/aspirin pretreatment but not when then the compounds were given separately, even at 10-fold higher concentrations. In contrast, the Ca(2+)i mobilization response to ATP challenge (10 µM) was not inhibited by combined simvastatin/aspirin pretreatment. The H1 receptor antagonist pyrilamine significantly inhibited both histamine-induced Ca(2+)i mobilization and extracellular signal-regulated kinase (ERK) activation, whereas ranitidine (H2 receptor antagonist) was without effect. However, combined simvastatin/aspirin pretreatment failed to decrease H1 receptor protein expression ruling out receptor downregulation as the mechanism of action. Histamine-induced ERK activation was also inhibited by atorvastatin pretreatment, while simvastatin further inhibited histamine-induced vascular endothelial cadherin phosphorylation as well as altered HUVEC morphology and inhibited actin polymerization. Therefore, in addition to the known therapeutic benefits of statins and aspirin, here we provide initial cellular evidence that combined statin/aspirin treatment inhibits histamine responsiveness in HUVECs.


Asunto(s)
Aspirina/farmacología , Fármacos Cardiovasculares/farmacología , Agonistas de los Receptores Histamínicos/farmacología , Histamina/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simvastatina/farmacología , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Antagonistas de los Receptores Histamínicos/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Fosforilación , Receptores Histamínicos H1/efectos de los fármacos , Receptores Histamínicos H1/metabolismo , Factores de Tiempo
9.
Am J Physiol Cell Physiol ; 304(7): C666-72, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23344160

RESUMEN

Low blood concentrations of 25-hydroxyvitamin D(3) are associated with increased mortality, while some studies suggest improved cardiovascular outcomes with vitamin D(3) supplementation in chronic kidney disease. However, the physiological effects of vitamin D(3) on the cardiovascular system remain poorly understood making it difficult to determine whether vitamin D(3) supplementation might provide cardiovascular benefit or even cause harm. Thus here we investigated the effects of chronic 1,25-dihydroxyvitamin D(3) treatment on intracellular signaling in human coronary artery smooth muscle cells (HCASMCs) and found that 1,25-dihydroxyvitamin D(3) significantly potentiated endothelin (ET-1) signaling. Specifically, 1,25-dihydroxyvitamin D(3) (24-h pretreatment) caused a more than threefold enhancement in both ET-1-induced intracellular calcium mobilization and extracellular signal-regulated kinase (ERK) activation. This 1,25-dihydroxyvitamin D(3)-elicited signaling enhancement was not observed for either vasopressin or carbachol. With the use of endothelin receptor (ETR) isoform-selective antagonists, ETRA was found to be primarily responsible for the 1,25-dihydroxyvitamin D(3)-induced ET-1 responsiveness and yet ETRA mRNA expression and protein abundance were unaltered following 1,25-dihydroxyvitamin D(3) treatment. While there was an increase in ETRB mRNA expression in response to 1,25-dihydroxyvitamin D(3), the protein abundance of ETRB was again unchanged. Finally, ETRA/ETRB heterodimerization was not detected in HCASMCs in either the absence or presence of 1,25-dihydroxyvitamin D(3). Together, these data show for the first time that 1,25-dihydroxyvitamin D(3) enhances endothelin responsiveness in HCASMCs and that the effect is mediated through ETRA.


Asunto(s)
Calcitriol/farmacología , Vasos Coronarios/citología , Endotelina-1/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Calcio/metabolismo , Células Cultivadas , Endotelina-1/genética , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Fosforilación , Receptores de Endotelina/metabolismo , Transducción de Señal
10.
Front Physiol ; 14: 1154374, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064904

RESUMEN

As both a sensor of extracellular calcium (Ca2+ o) concentration and a key controller of Ca2+ o homeostasis, one of the most interesting properties of the calcium-sensing receptor (CaR) is its sensitivity to, and modulation by, ions and small ligands other than Ca2+. There is emerging evidence that extracellular phosphate can act as a partial, non-competitive CaR antagonist to modulate parathyroid hormone (PTH) secretion, thus permitting the CaR to integrate mineral homeostasis more broadly. Interestingly, phosphorylation of certain intracellular CaR residues can also inhibit CaR responsiveness. Thus, negatively charged phosphate can decrease CaR activity both extracellularly (via association with arginine) and intracellularly (via covalent phosphorylation).

11.
J Biol Chem ; 285(19): 14170-7, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20233724

RESUMEN

The calcium-sensing receptor (CaR) elicits oscillatory Ca(2+)(i) mobilization associated with dynamic, inhibitory protein kinase C-mediated phosphorylation of CaR(T888). While modest CaR stimulation elicits Ca(2+)(i) oscillations, greater stimulation either increases oscillation frequency or elicits sustained responses by an unknown mechanism. Here, moderate CaR stimulation (2.5 mm Ca(2+)(o), 10 min) increased CaR(T888) phosphorylation (160-kDa mature receptor) 5-fold in CaR stably transfected HEK-293 cells, whereas 3-5 mm Ca(2+)(o) treatments were without apparent effect. Treatment with 2 mm Ca(2+)(o) caused sustained CaR(T888) phosphorylation (> or = 20 min) and oscillatory Ca(2+)(i) mobilization. However, 5 mm Ca(2+)(o) increased CaR(T888) phosphorylation only briefly while eliciting sustained Ca(2+)(i) mobilization, suggesting that greater CaR activation induces rapid CaR(T888) dephosphorylation, thus permitting sustained Ca(2+)(i) responses. Indeed, 5 mm Ca(2+)(o) stimulated protein phosphatase 2A activity and induced CaR(T888) dephosphorylation following acute phorbol ester pretreatment, the latter effect being mimicked by CaR-positive allosteric modulators (NPS-R467 and l-Phe). Finally, the phosphatase inhibitor calyculin-A reversed CaR-induced inhibition of parathyroid hormone secretion from bovine parathyroid slices and normal human parathyroid cells, demonstrating the physiological importance of phosphorylation status on parathyroid function. Therefore, high Ca(2+)(o)-stimulated protein kinase C acts in concert with high Ca(2+)(o)-induced phosphatase activity to generate and maintain CaR-induced Ca(2+)(i) oscillations via the dynamic phosphorylation and dephosphorylation of CaR(T888).


Asunto(s)
Calcio/metabolismo , Glándulas Paratiroides/metabolismo , Hormona Paratiroidea/metabolismo , Proteína Quinasa C/metabolismo , Receptores Sensibles al Calcio/metabolismo , Animales , Señalización del Calcio , Bovinos , Células Cultivadas , Humanos , Immunoblotting , Riñón/citología , Riñón/metabolismo , Fosforilación , Proteína Fosfatasa 2/metabolismo , Transducción de Señal
12.
J Endocr Soc ; 5(9): bvab057, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34337274

RESUMEN

25-hydroxyvitamin D 1α-hydroxylase (encoded by CYP27B1), which catalyzes the synthesis of 1,25-dihydroxyvitamin D3, is subject to negative or positive modulation by extracellular Ca2+ (Ca2+ o) depending on the tissue. However, the Ca2+ sensors and underlying mechanisms are unidentified. We tested whether calcium-sensing receptors (CaSRs) mediate Ca2+ o-dependent control of 1α-hydroxylase using HEK-293 cells stably expressing the CaSR (HEK-CaSR cells). In HEK-CaSR cells, but not control HEK-293 cells, cotransfected with reporter genes for CYP27B1-Photinus pyralis (firefly) luciferase and control Renilla luciferase, an increase in Ca2+ o from 0.5mM to 3.0mM induced a 2- to 3-fold increase in firefly luciferase activity as well as mRNA and protein levels. Surprisingly, firefly luciferase was specifically suppressed at Ca2+ o ≥ 5.0mM, demonstrating biphasic Ca2+ o control. Both phases were mediated by CaSRs as revealed by positive and negative modulators. However, Ca2+ o induced simple monotonic increases in firefly luciferase and endogenous CYP27B1 mRNA levels, indicating that the inhibitory effect of high Ca2+ o was posttranscriptional. Studies with inhibitors and the CaSR C-terminal mutant T888A identified roles for protein kinase C (PKC), phosphorylation of T888, and extracellular regulated protein kinase (ERK)1/2 in high Ca2+ o-dependent suppression of firefly luciferase. Blockade of both PKC and ERK1/2 abolished Ca2+ o-stimulated firefly luciferase, demonstrating that either PKC or ERK1/2 is sufficient to stimulate the CYP27B1 promoter. A key CCAAT box (-74 bp to -68 bp), which is regulated downstream of PKC and ERK1/2, was required for both basal transcription and Ca2+ o-mediated transcriptional upregulation. The CaSR mediates Ca2+ o-dependent transcriptional upregulation of 1α-hydroxylase and an additional CaSR-mediated mechanism is identified by which Ca2+ o can promote luciferase and possibly 1α-hydroxylase breakdown.

13.
Biochim Biophys Acta ; 1782(3): 188-95, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18261471

RESUMEN

The aminoglycoside antibiotic gentamicin elicits proximal tubular toxicity and cell death. In calcium-sensing receptor (CaR)-transfected HEK-293 (CaR-HEK) cells and CaR-expressing proximal tubule-derived opossum kidney (OK) cells, chronic gentamicin treatment elicits dose-dependent, caspase-mediated apoptotic cell death. Here we investigated whether the renal cell toxicity of the CaR agonist gentamicin could be prevented by CaR antagonism or by lithium cotreatment which may interfere with receptor-mediated signalling. Chronic treatment of OK and CaR-HEK cells with low concentrations of gentamicin elicited cell death, an effect that was ameliorated by cotreatment with the CaR negative allosteric modulator (calcilytic) NPS-89636. This calcilytic also attenuated CaR agonist-induced ERK activation in these cells. In addition, 1 mM LiCl, equivalent to its therapeutic plasma concentration, also inhibited gentamicin-induced toxicity in both cell types. This protective effect of lithium was not due to the disruption of phosphatidylinositol-mediated gentamicin uptake as the cellular entry of Texas red-conjugated gentamicin into OK and CaR-HEK cells was unchanged by lithium treatment. However, the protective effect of lithium was mimicked by glycogen synthase 3beta inhibition. Together, these data implicate CaR activation and a lithium-inhibitable signalling pathway in the induction of cell death by gentamicin in renal epithelial cells in culture.


Asunto(s)
Aminoglicósidos/farmacología , Muerte Celular/efectos de los fármacos , Gentamicinas/farmacología , Riñón/efectos de los fármacos , Compuestos de Litio/farmacología , Receptores Sensibles al Calcio/antagonistas & inhibidores , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Humanos , Zarigüeyas
14.
Biochim Biophys Acta ; 1778(10): 2318-24, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18573233

RESUMEN

The calcium-sensing receptor (CaR) is expressed on intestinal epithelial serosal membrane and in Caco-2 cells. In renal epithelium, CaR expressed on the basolateral membrane acts to limit excess tubular Ca2+ reabsorption. Therefore, here we investigated whether extracellular calcium (Ca(o)2+) can regulate active or passive 45Ca2+ transport across differentiated Caco-2 monolayers via CaR-dependent or CaR-independent mechanisms. Raising the Ca(o)2+ concentration from 0.8 to 1.6 mM increased transepithelial electrical resistance (TER) and decreased passive Ca2+ permeability but failed to alter active Ca2+ transport. The Ca(o)2+ effect on TER was rapid, sustained and concentration-dependent. Increasing basolateral Mg2+ concentration increased TER and inhibited both passive and active Ca2+ transport, whereas spermine and the CaR-selective calcimimetic NPS R-467 were without effect. We conclude that small increases in divalent cation concentration elicit CaR-independent increases in TER and inhibit passive Ca2+ transport across Caco-2 monolayers, most probably through a direct effect on tight junction permeability. Whilst it is known that the complete removal of Ca(o)2+ lowers TER, here we show that Ca(o)2+ addition actually increases TER in a concentration-dependent manner. Therefore, such Ca(o)2+-sensitivity could modulate intestinal solute transport including the limiting of excess Ca2+ absorption.


Asunto(s)
Células CACO-2/metabolismo , Calcio/metabolismo , Magnesio/metabolismo , Regulación Alostérica , Transporte Biológico/fisiología , Células CACO-2/citología , Colecalciferol/metabolismo , Humanos , Receptores Sensibles al Calcio/metabolismo
15.
Nat Commun ; 10(1): 4693, 2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31619668

RESUMEN

Extracellular phosphate regulates its own renal excretion by eliciting concentration-dependent secretion of parathyroid hormone (PTH). However, the phosphate-sensing mechanism remains unknown and requires elucidation for understanding the aetiology of secondary hyperparathyroidism in chronic kidney disease (CKD). The calcium-sensing receptor (CaSR) is the main controller of PTH secretion and here we show that raising phosphate concentration within the pathophysiologic range for CKD significantly inhibits CaSR activity via non-competitive antagonism. Mutation of residue R62 in anion binding site-1 abolishes phosphate-induced inhibition of CaSR. Further, pathophysiologic phosphate concentrations elicit rapid and reversible increases in PTH secretion from freshly-isolated human parathyroid cells consistent with a receptor-mediated action. The same effect is seen in wild-type murine parathyroid glands, but not in CaSR knockout glands. By sensing moderate changes in extracellular phosphate concentration, the CaSR represents a phosphate sensor in the parathyroid gland, explaining the stimulatory effect of phosphate on PTH secretion.


Asunto(s)
Glándulas Paratiroides/metabolismo , Hormona Paratiroidea/metabolismo , Fosfatos/metabolismo , Receptores Sensibles al Calcio/metabolismo , Animales , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Hiperparatiroidismo Secundario/etiología , Hiperparatiroidismo Secundario/metabolismo , Ratones , Mutación , Receptores Sensibles al Calcio/genética , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo
16.
J Bone Miner Res ; 34(9): 1609-1618, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31063613

RESUMEN

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.


Asunto(s)
Compuestos de Calcio/uso terapéutico , Hipercalciuria/tratamiento farmacológico , Hipocalcemia/tratamiento farmacológico , Hipoparatiroidismo/congénito , Adulto , Área Bajo la Curva , Compuestos de Calcio/efectos adversos , Compuestos de Calcio/farmacocinética , Línea Celular , Femenino , Genotipo , Humanos , Hipercalciuria/genética , Hipocalcemia/genética , Hipoparatiroidismo/tratamiento farmacológico , Hipoparatiroidismo/genética , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
17.
Cell Calcium ; 44(2): 210-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18221783

RESUMEN

In this study, the presence of GPRC6A receptors in rat mesenteric artery was investigated. In artery homogenates, GPRC6A mRNA was detected and Western blotting showed the presence of GPRC6A protein. Immunohistochemical studies revealed GPRC6A in both endothelial cells and myocytes. In whole vessel segments, the GPRC6A activators, 300 microM l-ornithine and 100 microM Al(3+), induced endothelium-dependent myocyte hyperpolarizations sensitive to 10 microM TRAM-34, a blocker of intermediate conductance, Ca(2+)-sensitive K(+) channels (IK(Ca)). Activation of IK(Ca) with calindol (300 nM; a positive allosteric Ca(2+)-sensing receptor - CaR - modulator) was inhibited by 500 nM ouabain (inhibition of rat type 2 and type 3 Na(+)/K(+)-ATPases) but unaffected by 30 microM Ba(2+) (blockade of inwardly rectifying K(+) channels). Neither l-ornithine nor Al(3+) activated CaRs heterologously expressed in CHO or HEK293 cells. In the presence of 300 microM l-ornithine or 100 microM Al(3+), myocyte hyperpolarizations to calindol were potentiated whereas this potentiation and hyperpolarizations to l-ornithine were lost following incubation with an anti-GPRC6A antibody. It is concluded that GPRC6A receptors are present on mesenteric artery endothelial cells and myocytes and that their activation selectively opens IK(Ca) channels. This triggers a ouabain-sensitive myocyte hyperpolarization suggesting a close functional relationship between GPRC6A, the IK(Ca) channel and type 2 and/or type 3 Na(+)/K(+)-ATPases.


Asunto(s)
Calcio/metabolismo , Vasos Coronarios/metabolismo , Endotelio Vascular/metabolismo , Arterias Mesentéricas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Western Blotting , Células CHO , Cardiotónicos/farmacología , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Cricetinae , Cricetulus , Electrofisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Indoles/farmacología , Inositol/metabolismo , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Arterias Mesentéricas/citología , Arterias Mesentéricas/efectos de los fármacos , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Naftalenos/farmacología , Ornitina/farmacología , Ouabaína/farmacología , Fosforilación/efectos de los fármacos , Canales de Potasio Calcio-Activados/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Porcinos
18.
Circ Res ; 97(4): 391-8, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-16037572

RESUMEN

Small increases in extracellular Ca2+ dilate isolated blood vessels. In the present study, the possibility that a vascular, extracellular Ca2+-sensing receptor (CaSR) could mediate these vasodilator actions was investigated. Novel ligands that interact with the CaSR were used in microelectrode recordings from rat isolated mesenteric and porcine coronary arteries. The major findings were that (1) raising extracellular Ca2+ or adding calindol, a CaSR agonist, produced concentration-dependent hyperpolarizations of vascular myocytes, actions attenuated by Calhex 231, a negative allosteric modulator of CaSR. (2) Calindol-induced hyperpolarizations were inhibited by the intermediate conductance, Ca2+-sensitive K+ (IKCa) channel inhibitors, TRAM-34, and TRAM-39. (3) The effects of calindol were not observed in the absence of endothelium. (4) CaSR mRNA and protein were present in rat mesenteric arteries and in porcine coronary artery endothelial cells. (5) CaSR and IKCa proteins were restricted to caveolin-poor membrane fractions. We conclude that activation of vascular endothelial CaSRs opens endothelial cell IKCa channels with subsequent myocyte hyperpolarization. The endothelial cell CaSR may have a physiological role in the control of arterial blood pressure.


Asunto(s)
Benzamidas/farmacología , Ciclohexilaminas/farmacología , Células Endoteliales/fisiología , Receptores Sensibles al Calcio/fisiología , Animales , Bencimidazoles/farmacología , Presión Sanguínea , Calcio/metabolismo , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiología , Masculino , Arterias Mesentéricas/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Péptidos/farmacología , Fenilefrina/farmacología , Canales de Potasio/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Sensibles al Calcio/análisis , Porcinos
19.
Sci Transl Med ; 7(284): 284ra60, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25904744

RESUMEN

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.


Asunto(s)
Asma/patología , Asma/fisiopatología , Hiperreactividad Bronquial/metabolismo , Hipersensibilidad/patología , Receptores Sensibles al Calcio/antagonistas & inhibidores , Alérgenos/química , Animales , Asma/metabolismo , Biopsia , Bronquios/metabolismo , Bronquios/patología , Líquido del Lavado Bronquioalveolar , Broncoconstricción , Cationes , Células HEK293 , Homeostasis , Humanos , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Cell Calcium ; 35(3): 217-28, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15200145

RESUMEN

As a G protein-coupled receptor (GPCR), the extracellular calcium-sensing receptor (CaR) responds to changes in extracellular free calcium concentration by inducing intracellular signalling. These CaR-induced signals then specifically modulate cellular functions such as parathyroid hormone secretion from the parathyroid glands and calcium reabsorption in the kidney and thus to understand how the CaR functions one must understand how it signals. CaR-induced signalling involves intracellular Ca2+ mobilisation/oscillations as well as the activation of various phospholipases and protein kinases and the suppression of cAMP formation. This review will detail the intracellular pathways by which the CaR is believed to elicit its physiological functions and summarises the evidence for cell- and agonist-specific differential signalling.


Asunto(s)
Receptores Sensibles al Calcio/fisiología , Transducción de Señal/fisiología , Animales , Huesos/citología , Huesos/metabolismo , Calcio/metabolismo , Caveolas/metabolismo , Proteínas Contráctiles/metabolismo , Células Epiteliales/metabolismo , Espacio Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Filaminas , Proteínas de Unión al GTP/metabolismo , Humanos , Espacio Intracelular/metabolismo , Riñón/citología , Riñón/metabolismo , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Fosfolipasas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Quinasas/metabolismo , Receptores Sensibles al Calcio/agonistas , Receptores Sensibles al Calcio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA