Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
2.
J Bone Miner Res ; 37(10): 1926-1935, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35879818

RESUMO

Autosomal dominant hypocalcemia type 1 (ADH1) is a rare form of hypoparathyroidism due to activating variants of the calcium-sensing receptor gene (CASR). Inherited or de novo activating variants of the CASR alter the set point for extracellular calcium, resulting in inadequate parathyroid hormone (PTH) secretion and inappropriate renal calcium excretion leading to hypocalcemia and hypercalciuria. Conventional therapy includes calcium and activated vitamin D, which can worsen hypercalciuria, resulting in renal complications. A systematic literature review, using published reports from 1994 to 2021, was conducted to catalog CASR variants, to define the ADH1 clinical spectrum, and to determine the effect of treatment on patients with ADH1. There were 113 unique CASR variants reported, with a general lack of genotype/phenotype correlation. Clinical data were available in 191 patients; 27% lacked symptoms, 32% had mild/moderate symptoms, and 41% had severe symptoms. Seizures, the most frequent clinical presentation, occurred in 39% of patients. In patients with blood and urine chemistries available at the time of diagnosis (n = 91), hypocalcemia (99%), hyperphosphatemia (59%), low PTH levels (57%), and hypercalciuria (34%) were observed. Blood calcium levels were significantly lower in patients with severe symptoms compared with asymptomatic patients (6.8 ± 0.7 versus 7.6 ± 0.7 mg/dL [mean ± SD]; p < 0.0001), and the age of presentation was significantly lower in severely symptomatic patients (9.1 ± 15.0 versus 19.3 ± 19.4 years; p < 0.01). Assessments for complications including nephrocalcinosis, nephrolithiasis, renal impairment, and brain calcifications in 57 patients on conventional therapy showed that 75% had at least one complication. Hypercalciuria was associated with nephrocalcinosis, nephrolithiasis, renal impairment, or brain calcifications (odds ratio [OR] = 9.3; 95% confidence interval [CI] 2.4-37.2; p < 0.01). In 27 patients with urine calcium measures before and after starting conventional therapy, the incidence of hypercalciuria increased by 91% (p < 0.05) after therapy initiation. ADH1 is a condition often associated with severe symptomatology at presentation with an increase in the risk of renal complications after initiation of conventional therapy. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Assuntos
Hipocalcemia , Hipoparatireoidismo , Nefrocalcinose , Nefrolitíase , Humanos , Hipercalciúria/genética , Hipercalciúria/tratamento farmacológico , Hipocalcemia/genética , Hipocalcemia/tratamento farmacológico , Receptores de Detecção de Cálcio/genética , Cálcio , Nefrocalcinose/genética , Hipoparatireoidismo/tratamento farmacológico , Hipoparatireoidismo/genética , Hormônio Paratireóideo/uso terapêutico , Vitamina D/uso terapêutico
3.
J Am Soc Nephrol ; 33(1): 201-212, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34732508

RESUMO

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.


Assuntos
Calcimiméticos/uso terapêutico , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperfosfatemia/complicações , Diálise Renal , Insuficiência Renal Crônica/complicações , Idoso , Cinacalcete/uso terapêutico , Feminino , Humanos , Hiperparatireoidismo Secundário/sangue , Hiperparatireoidismo Secundário/complicações , Hiperfosfatemia/sangue , Masculino , Pessoa de Meia-Idade , Hormônio Paratireóideo/sangue , Peptídeos/uso terapêutico , Fósforo/sangue , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/terapia , Estudos Retrospectivos
4.
JBMR Plus ; 4(10): e10402, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33103030

RESUMO

Calcilytics are calcium-sensing receptor (CaSR) antagonists that reduce the sensitivity of the CaSR to extracellular calcium. Calcilytics have the potential to treat autosomal dominant hypocalcemia type 1 (ADH1), which is caused by germline gain-of-function CaSR mutations and leads to symptomatic hypocalcemia, inappropriately low PTH concentrations, and hypercalciuria. To date, only one calcilytic compound, NPSP795, has been evaluated in patients with ADH1: Doses of up to 30 mg per patient have been shown to increase PTH concentrations, but did not significantly alter ionized blood calcium concentrations. The aim of this study was to further investigate NPSP795 for the treatment of ADH1 by undertaking in vitro and in vivo studies involving Nuf mice, which have hypocalcemia in association with a gain-of-function CaSR mutation, Leu723Gln. Treatment of HEK293 cells stably expressing the mutant Nuf (Gln723) CaSR with 20nM NPSP795 decreased extracellular Ca2+-mediated intracellular calcium and phosphorylated ERK responses. An in vivo dose-ranging study was undertaken by administering a s.c. bolus of NPSP795 at doses ranging from 0 to 30 mg/kg to heterozygous (Casr +/Nuf ) and to homozygous (Casr Nuf/Nuf ) mice, and measuring plasma PTH responses at 30 min postdose. NPSP795 significantly increased plasma PTH concentrations in a dose-dependent manner with the 30 mg/kg dose causing a maximal (≥10-fold) rise in PTH. To determine whether NPSP795 can rectify the hypocalcemia of Casr +/Nuf and Casr Nuf/Nuf mice, a submaximal dose (25 mg/kg) was administered, and plasma adjusted-calcium concentrations measured over a 6-hour period. NPSP795 significantly increased plasma adjusted-calcium in Casr +/Nuf mice from 1.87 ± 0.03 mmol/L to 2.16 ± 0.06 mmol/L, and in Casr Nuf/Nuf mice from 1.70 ± 0.03 mmol/L to 1.89 ± 0.05 mmol/L. Our findings show that NPSP795 elicits dose-dependent increases in PTH and ameliorates the hypocalcemia in an ADH1 mouse model. Thus, calcilytics such as NPSP795 represent a potential targeted therapy for ADH1. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

5.
Nat Commun ; 10(1): 4693, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31619668

RESUMO

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.


Assuntos
Glândulas Paratireoides/metabolismo , Hormônio Paratireóideo/metabolismo , Fosfatos/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Animais , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Hiperparatireoidismo Secundário/etiologia , Hiperparatireoidismo Secundário/metabolismo , Camundongos , Mutação , Receptores de Detecção de Cálcio/genética , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo
6.
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
7.
Prog Med Chem ; 57(1): 1-86, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29680147

RESUMO

The extracellular calcium receptor (CaR) is a G protein-coupled receptor (GPCR) and the pivotal molecule regulating systemic Ca2+ homeostasis. The CaR was a challenging target for drug discovery because its physiological ligand is an inorganic ion (Ca2+) rather than a molecule so there was no structural template to guide medicinal chemistry. Nonetheless, small molecules targeting this receptor were discovered. Calcimimetics are agonists or positive allosteric modulators of the CaR, while calcilytics are antagonists and all to date are negative allosteric modulators. The calcimimetic cinacalcet was the first allosteric modulator of a GPCR to achieve regulatory approval and is a first-in-class treatment for secondary hyperparathyroidism in patients on dialysis, and for hypercalcemia in some forms of primary hyperparathyroidism. It is also useful in treating some rare genetic diseases that cause hypercalcemia. Two other calcimimetics are now on the market (etelcalcetide) or under regulatory review (evocalcet). Calcilytics stimulate the secretion of parathyroid hormone and were initially developed as treatments for osteoporosis. Three different calcilytics of two different chemotypes failed in clinical trials due to lack of efficacy. Calcilytics are now being repurposed and might be useful in treating hypoparathyroidism and several rare genetic diseases causing hypocalcemia. The challenges ahead for medicinal chemists are to design compounds that select conformations of the CaR that preferentially target a particular signalling pathway and/or that affect the CaR in a tissue-selective manner.


Assuntos
Calcimiméticos/farmacologia , Cálcio/metabolismo , Cálcio/farmacologia , Descoberta de Drogas , Receptores de Detecção de Cálcio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Calcimiméticos/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Detecção de Cálcio/genética
8.
Best Pract Res Clin Endocrinol Metab ; 27(3): 373-84, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23856266

RESUMO

The calcium-sensing receptor (CaSR) plays a pivotal role in regulating systemic Ca(2+) homeostasis and is a target for drugs designed to treat certain disorders of bone and mineral metabolism. Calcimimetics are agonists or positive allosteric modulators of the CaSR; they inhibit parathyroid hormone (PTH) secretion and stimulate renal Ca(2+) excretion. The first calcimimetic drug is cinacalcet, a positive allosteric modulator of the CaSR that is approved for treating secondary hyperparathyroidism (HPT) in patients on renal replacement therapy and for some forms of primary HPT characterized by clinically significant hypercalcemia. Cinacalcet is also being investigated as a therapy for other hypercalcemic conditions and certain hypophosphatemic disorders. Calcilytics are CaSR inhibitors that stimulate the secretion of PTH and decrease renal excretion of Ca(2+). Although calcilytics have failed thus far as anabolic therapies for osteoporosis, they are currently being evaluated as novel therapies for new indications involving hypocalcemia and/or hypercalciuria.


Assuntos
Calcimiméticos/uso terapêutico , Hipercalcemia/tratamento farmacológico , Hipocalcemia/tratamento farmacológico , Osteoporose/tratamento farmacológico , Receptores de Detecção de Cálcio/metabolismo , Humanos , Hipercalcemia/metabolismo , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperparatireoidismo Secundário/metabolismo , Hipocalcemia/metabolismo , Hipofosfatemia/tratamento farmacológico , Hipofosfatemia/metabolismo , Osteoporose/metabolismo , Receptores de Detecção de Cálcio/agonistas
9.
J Biol Chem ; 286(47): 40922-33, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21969374

RESUMO

The calcium-sensing receptor (CaSR) is a family C G protein-coupled receptor that is activated by elevated levels of extracellular divalent cations. The CaSR couples to members of the G(q) family of G proteins, and in the endocrine system this receptor is instrumental in regulating the release of parathyroid hormone from the parathyroid gland and calcitonin from thyroid cells. Here, we demonstrate that in medullary thyroid carcinoma cells, the CaSR promotes cellular adhesion and migration via coupling to members of the integrin family of extracellular matrix-binding proteins. Immunopurification and mass spectrometry, co-immunoprecipitation, and co-localization studies showed that the CaSR and ß1-containing integrins are components of a macromolecular protein complex. In fibronectin-based cell adhesion and migration assays, the CaSR-positive allosteric modulator NPS R-568 induced a concentration-dependent increase in cell adhesion and migration; both of these effects were blocked by a specific CaSR-negative allosteric modulator. These effects were mediated by integrins because they were blocked by a peptide inhibitor of integrin binding to fibronectin and ß1 knockdown experiments. An analysis of intracellular signaling pathways revealed a key role for CaSR-induced phospholipase C activation and the release of intracellular calcium. These results demonstrate for the first time that an ion-sensing G protein-coupled receptor functionally couples to the integrins and, in conjunction with intracellular calcium release, promotes cellular adhesion and migration in tumor cells. The significance of this interaction is further highlighted by studies implicating the CaSR in cancer metastasis, axonal growth, and stem cell attachment, functions that rely on integrin-mediated cell adhesion.


Assuntos
Movimento Celular , Integrinas/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Regulação Alostérica/efeitos dos fármacos , Compostos de Anilina/farmacologia , Animais , Cálcio/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Fibronectinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Cadeias beta de Integrinas/metabolismo , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Oligopeptídeos/farmacologia , Fenetilaminas , Propilaminas , Transporte Proteico/efeitos dos fármacos , Ratos , Receptores de Detecção de Cálcio/química , Transdução de Sinais/efeitos dos fármacos
10.
Bone ; 46(2): 534-42, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19786130

RESUMO

Daily subcutaneous administration of exogenous parathyroid hormone (PTH) promotes bone formation in patients with osteoporosis. Here we describe two novel, short-acting calcium-sensing receptor antagonists (SB-423562 and its orally bioavailable precursor, SB-423557) that elicit transient PTH release from the parathyroid gland in several preclinical species and in humans. In an ovariectomized rat model of bone loss, daily oral administration of SB-423557 promoted bone formation and improved parameters of bone strength at lumbar spine, proximal tibia and midshaft femur. Chronic administration of SB-423557 did not increase parathyroid cell proliferation in rats. In healthy human volunteers, single doses of intravenous SB-423562 and oral SB-423557 elicited transient elevations of endogenous PTH concentrations in a profile similar to that observed with subcutaneously administered PTH. Both agents were well tolerated in humans. Transient increases in serum calcium, an expected effect of increased parathyroid hormone concentrations, were observed post-dose at the higher doses of SB-423557 studied. These data constitute an early proof of principle in humans and provide the basis for further development of this class of compound as a novel, orally administered bone-forming treatment for osteoporosis.


Assuntos
Etanolaminas/farmacologia , Naftalenos/farmacologia , Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/sangue , Fenilpropionatos/farmacologia , Receptores de Detecção de Cálcio/antagonistas & inibidores , Administração Oral , Animais , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Cálcio/sangue , Proliferação de Células/efeitos dos fármacos , Cães , Esquema de Medicação , Etanolaminas/administração & dosagem , Etanolaminas/química , Etanolaminas/farmacocinética , Haplorrinos , Humanos , Masculino , Naftalenos/administração & dosagem , Naftalenos/química , Naftalenos/farmacocinética , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Glândulas Paratireoides/citologia , Glândulas Paratireoides/efeitos dos fármacos , Fenilpropionatos/administração & dosagem , Fenilpropionatos/química , Fenilpropionatos/farmacocinética , Ratos , Ratos Sprague-Dawley
11.
J Med Chem ; 52(21): 6599-605, 2009 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19821575

RESUMO

When administered as a single agent to rats, the previously reported calcium receptor antagonist 3 elicited a sustained elevation of plasma PTH resulting in no increase in overall bone mineral density. The lack of a bone building effect for analogue 3 was attributed to the large volume of distribution (V(dss)(rat) = 11 L/kg), producing a protracted plasma PTH profile. Incorporation of a carboxylic acid functionality into the amino alcohol template led to the identification of 12 with a lower volume of distribution (V(dss)(12) = 1.18 L/kg) and a shorter half-life. The zwitterionic nature of antagonist 12 necessitated the utility of an ester prodrug approach to increase overall permeability. Antagonist 12 elicited a rapid and transient increase in circulating levels of PTH following oral dosing of the ester prodrug 11 in the dog. The magnitude and duration of the increases in plasma levels of PTH would be expected to stimulate new bone formation.


Assuntos
Amino Álcoois/síntese química , Hormônio Paratireóideo/sangue , Fenilpropionatos/síntese química , Pró-Fármacos/síntese química , Propanolaminas/síntese química , Receptores de Detecção de Cálcio/antagonistas & inibidores , Administração Oral , Amino Álcoois/farmacocinética , Amino Álcoois/farmacologia , Animais , Disponibilidade Biológica , Cálcio/metabolismo , Linhagem Celular , Citoplasma/metabolismo , Cães , Ésteres , Humanos , Hormônio Paratireóideo/metabolismo , Permeabilidade , Fenilpropionatos/farmacocinética , Fenilpropionatos/farmacologia , Pró-Fármacos/farmacocinética , Pró-Fármacos/farmacologia , Propanolaminas/farmacocinética , Propanolaminas/farmacologia , Ensaio Radioligante , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
12.
J Med Chem ; 52(13): 3982-93, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19492813

RESUMO

Functional screening of the former SmithKline Beecham compound collection against the human calcium receptor (CaR) resulted in the identification of the amino alcohol-based hit 2 (IC(50) = 11 microM). Structure-activity studies of 2 focused on the optimization of the right- and left-hand side aromatic moieties as well as the amino alcohol linker region. Critical to the optimization of this antagonist template was the discovery that the chirality of the C-2 secondary alcohol played a key role in enhancing both CaR potency as well as selectivity over the beta-adrenergic receptor subtypes. These SAR studies ultimately led to the identification of 38 (NPS 2143; SB-262470A), a potent and orally active CaR antagonist. Pharmacokinetic characterization of 38 in the rat revealed that this molecule had a large volume of distribution (11 L/kg), which resulted in a prolonged systemic exposure, protracted increases in the plasma levels of PTH, and an overall lack of net bone formation effect in a rodent model of osteoporosis.


Assuntos
Amino Álcoois/química , Amino Álcoois/farmacocinética , Hormônio Paratireóideo/sangue , Receptores de Detecção de Cálcio/antagonistas & inibidores , Administração Oral , Animais , Humanos , Osteoporose/tratamento farmacológico , Ratos , Relação Estrutura-Atividade , Distribuição Tecidual
13.
IDrugs ; 11(11): 827-40, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18988128

RESUMO

ZT-031 is a cyclic 31-amino acid analog of parathyroid hormone (PTH) that is in development by Zelos Therapeutics Inc for the treatment of osteoporosis and other bone-related disorders. ZT-031 activates the PTH type 1 receptor - the molecular target of the currently marketed osteogenic peptides PTH and PTH(1-34). Daily subcutaneous injections of ZT-031 prevented bone loss and replaced bone that had already been lost in an ovariectomized rat model of osteoporosis. Daily subcutaneous injections of ZT-031 in gonad-intact monkeys increased bone mineral density (BMD) at cortical and cancellous bone sites and increased serum levels of bone formation markers. The daily subcutaneous administration of ZT-031 to postmenopausal women with osteoporosis elicited a dose-dependent increase in BMD of the lumbar spine, proximal femur and total hip area. Plasma levels of bone formation markers were significantly increased above baseline after 1 month of dosing, and prior to increases in bone resorption markers. ZT-031 was demonstrated to be safe and well tolerated; episodes of hypercalcemia were infrequent and observed with a frequency greater than with placebo only at the highest doses tested for the drug. Although available data are limited, the results obtained following treatment with ZT-031 have generally been at least as favorable as those obtained with other osteogenic PTH peptides. A novel dosing paradigm has been planned for the drug.


Assuntos
Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/química , Conservadores da Densidade Óssea/uso terapêutico , Osteoporose/tratamento farmacológico , Hormônio Paratireóideo/química , Hormônio Paratireóideo/uso terapêutico , Peptídeos Cíclicos/uso terapêutico , Anabolizantes/farmacologia , Animais , Conservadores da Densidade Óssea/farmacocinética , Conservadores da Densidade Óssea/toxicidade , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Pessoa de Meia-Idade , Hormônio Paratireóideo/efeitos adversos , Hormônio Paratireóideo/síntese química , Hormônio Paratireóideo/farmacologia , Patentes como Assunto , Peptídeos Cíclicos/efeitos adversos , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/farmacologia , Primatas , Ratos , Relação Estrutura-Atividade
14.
J Cell Biochem ; 96(2): 428-38, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16052472

RESUMO

Since NO production by NOS-2 made by astrocytes activated by proinflammatory cytokines contributes to the killing of neurons in variously damaged human brains, knowing the mechanisms responsible for NOS-2 expression should contribute to developing effective therapeutics. The expression and activation of NOS-2 in normal adult human cerebral cortical astrocytes treated with three proinflammatory cytokines, IL-1beta, TNF-alpha, and IFN-gamma, are driven by two separable mechanisms. NOS-2 expression requires a burst of p38 MAPK activity, while the activation of the resulting enzyme protein requires MEK/ERK-dependent BH4 (tetrahydrobiopterin) synthesis between 24 and 24.5 h after adding the cytokines to the culture medium. Here we show that NOS-2 expression in the activated astrocytes requires that the culture medium contain 1.8 mM Ca2+, but it is unaffected by inhibiting calcium-sensing receptors (CASRs) with NPS 89636. However, NOS-2 activation is inhibited by NPS 89626 during the MEK/ERK-dependent stage between 24 and 24.5 h after adding the cytokines, and this inhibition can be overridden by exogenous BH4. Therefore, NOS-2 expression and the subsequent BH4-dependent NOS-2-activation in human astrocytes need 1.8 mM Ca2+ to be in the culture medium, while NOS-2 activation also needs functional CASRs between 24 and 24.5 h after cytokine addition. These findings raise the possibility that calcilytic drugs prevent NO-induced damage and death of human neurons.


Assuntos
Astrócitos/efeitos dos fármacos , Biopterinas/análogos & derivados , Cálcio/farmacologia , Citocinas/farmacologia , Regulação Enzimológica da Expressão Gênica , Receptores de Detecção de Cálcio/metabolismo , Adulto , Envelhecimento/fisiologia , Astrócitos/citologia , Astrócitos/metabolismo , Biopterinas/farmacologia , Cálcio/química , Cátions Bivalentes/química , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Óxido Nítrico/metabolismo
15.
J Pharmacol Sci ; 97(3): 355-60, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15781990

RESUMO

Circulating levels of calcium ion (Ca(2+)) are maintained within a narrow physiological range mainly by the action of parathyroid hormone (PTH) secreted from parathyroid cells. Parathyroid cells can sense small fluctuations in plasma Ca(2+) levels by virtue of a cell surface Ca(2+) receptor (CaR) that belongs to the superfamily of G-protein-coupled receptors. Calcimimetics are positive allosteric modulators that activate the CaR on parathyroid cells and thereby immediately suppress PTH secretion. Pre-clinical studies with NPS R-568, a first generation calcimimetic compound, have demonstrated that daily oral administration inhibits the elevation of plasma PTH levels and parathyroid gland hyperplasia and ameliorates impaired bone qualities in rats with chronic renal insufficiency. The results of clinical trials with cinacalcet hydrochloride, a second generation calcimimetic compound, have shown that calcimimetics possess lowering effects not only on serum PTH levels but also on serum calcium x phosphorus product levels, a hallmark of an increased risk for cardiovascular death in dialysis patients with end-stage renal disease (ESRD). Thus, calcimimetics have considerable potential as an innovative medical approach to manage secondary hyperparathyroidism associated with ESRD. Indeed, cinacalcet hydrochloride has been approved in several countries and is the first positive allosteric modulator of any G protein-coupled receptor to reach the market.


Assuntos
Cálcio/metabolismo , Desenho de Fármacos , Hiperparatireoidismo Secundário/tratamento farmacológico , Receptores de Detecção de Cálcio/efeitos dos fármacos , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Animais , Cálcio/agonistas , Cinacalcete , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Hiperparatireoidismo Secundário/sangue , Hiperparatireoidismo Secundário/etiologia , Hiperparatireoidismo Secundário/metabolismo , Falência Renal Crônica/complicações , Modelos Biológicos , Estrutura Molecular , Naftalenos/uso terapêutico , Glândulas Paratireoides/citologia , Glândulas Paratireoides/metabolismo , Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/metabolismo , Fenetilaminas , Propilaminas , Ratos , Receptores de Detecção de Cálcio/genética
16.
J Biol Chem ; 279(50): 51739-44, 2004 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-15579475

RESUMO

We previously demonstrated that the human calcium-sensing receptor (CaR) is allosterically activated by L-amino acids (Conigrave, A. D., Quinn, S. J., and Brown, E. M. (2000) Proc. Natl. Acad. Sci. U. S. A. 97, 4814-4819). However, the domain-based location of amino acid binding has been uncertain. We now show that the Venus Fly Trap (VFT) domain of CaR, but none of its other major domains, is required for amino acid sensing. Several constructs were informative when expressed in HEK293 cells. First, the wild-type CaR exhibited allosteric activation by L-amino acids as previously observed. Second, two CaR-mGlu chimeric receptor constructs that retained the VFT domain of CaR, one containing the extracellular Cys-rich region of CaR and the other containing the Cys-rich region of the rat metabotropic glutamate type-1 (mGlu-1) receptor, together with the rat mGlu-1 transmembrane region and C-terminal tail, retained amino acid sensing. Third, a CaR lacking residues 1-599 of the N-terminal extracellular head but retaining an intact CaR transmembrane region and a functional but truncated C terminus (headless-T903 CaR) failed to respond to L-amino acids but retained responsiveness to the type-II calcimimetic NPS R-467. Finally, a T903 CaR control that retained an intact N terminus also retained L-amino acid sensing. Taken together, the data indicate that the VFT domain of CaR is necessary for L-amino acid sensing and are consistent with the hypothesis that the VFT domain is the site of L-amino acid binding. The findings support the concept that the mGlu-1 amino acid binding site for L-glutamate is conserved as an L-amino acid binding site in its homolog, the CaR.


Assuntos
Aminoácidos/metabolismo , Receptores de Detecção de Cálcio/química , Receptores de Detecção de Cálcio/metabolismo , Aminoácidos/química , Aminoácidos/farmacologia , Compostos de Anilina/farmacologia , Animais , Sítios de Ligação , Cálcio/agonistas , Cálcio/metabolismo , Linhagem Celular , Humanos , Técnicas In Vitro , Fenilalanina/química , Fenilalanina/farmacologia , Estrutura Terciária de Proteína , Ratos , Receptores de Detecção de Cálcio/genética , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Estereoisomerismo , Transfecção
17.
Proc Natl Acad Sci U S A ; 101(14): 5140-5, 2004 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-15051872

RESUMO

We investigated the direct effects of changes in free ionized extracellular calcium concentrations ([Ca2+]o) on osteoblast function and the involvement of the calcium-sensing receptor (CaR) in mediating these responses. CaR mRNA and protein were detected in osteoblast models, freshly isolated fetal rat calvarial cells and murine clonal osteoblastic 2T3 cells, and in freshly frozen, undecalcified preparations of human mandible and rat femur. In fetal rat calvarial cells, elevating [Ca2+]o and treatment with gadolinium, a nonpermeant CaR agonist, resulted in phosphorylation of the extracellular signal-regulated kinases 1 and 2, Akt, and glycogensynthase kinase 3beta, consistent with signals of cell survival and proliferation. In agreement, cell number was increased under these conditions. Expression of the osteoblast differentiation markers core binding factor alpha1, osteocalcin, osteopontin, and collagen I mRNAs was increased by high [Ca2+]o, as was mineralized nodule formation. Alkaline phosphatase activity was maximal for [Ca2+]o between 1.2 and 1.8 mM. Inhibition of CaR by NPS 89636 blocked responses to the CaR agonists. In conclusion, we show that small deviations of [Ca2+]o from physiological values have a profound impact on bone cell fate, by means of the CaR and independently of systemic calciotropic peptides.


Assuntos
Cálcio/metabolismo , Osteoblastos/fisiologia , Animais , Sequência de Bases , Osso e Ossos/metabolismo , Divisão Celular , Primers do DNA , Espaço Extracelular/metabolismo , Expressão Gênica , Humanos , Imuno-Histoquímica , Osteoblastos/metabolismo , Ratos , Ratos Sprague-Dawley
18.
J Pharmacol Exp Ther ; 308(2): 627-35, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14593085

RESUMO

Calcimimetic compounds, which activate the parathyroid cell Ca(2+) receptor (CaR) and inhibit parathyroid hormone (PTH) secretion, are under experimental study as a treatment for hyperparathyroidism. This report describes the salient pharmacodynamic properties, using several test systems, of a new calcimimetic compound, cinacalcet HCl. Cinacalcet HCl increased the concentration of cytoplasmic Ca(2+) ([Ca(2+)](i)) in human embryonic kidney 293 cells expressing the human parathyroid CaR. Cinacalcet HCl (EC(50) = 51 nM) in the presence of 0.5 mM extracellular Ca(2+) elicited increases in [Ca(2+)](i) in a dose- and calcium-dependent manner. Similarly, in the presence of 0.5 mM extracellular Ca(2+), cinacalcet HCl (IC(50) = 28 nM) produced a concentration-dependent decrease in PTH secretion from cultured bovine parathyroid cells. Using rat medullary thyroid carcinoma 6-23 cells expressing the CaR, cinacalcet HCl (EC(50) = 34 nM) produced a concentration-dependent increase in calcitonin secretion. In vivo studies in rats demonstrated cinacalcet HCl is orally bioavailable and displays approximately linear pharmacokinetics over the dose range of 1 to 36 mg/kg. Furthermore, this compound suppressed serum PTH and blood-ionized Ca(2+) levels and increased serum calcitonin levels in a dose-dependent manner. Cinacalcet was about 30-fold more potent at lowering serum levels of PTH than it was at increasing serum calcitonin levels. The S-enantiomer of cinacalcet (S-AMG 073) was at least 75-fold less active in these assay systems. The present findings provide compelling evidence that cinacalcet HCl is a potent and stereoselective activator of the parathyroid CaR and, as such, might be beneficial in the treatment of hyperparathyroidism.


Assuntos
Calcitonina/metabolismo , Naftalenos/farmacologia , Glândulas Paratireoides/efeitos dos fármacos , Hormônio Paratireóideo/metabolismo , Animais , Calcitonina/sangue , Cálcio/sangue , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Cinacalcete , Humanos , Masculino , Naftalenos/farmacocinética , Glândulas Paratireoides/metabolismo , Hormônio Paratireóideo/sangue , Fósforo/sangue , Ratos , Ratos Sprague-Dawley
19.
J Biol Chem ; 277(37): 33736-41, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12114500

RESUMO

The extracellular calcium (Ca(2+)(o))-sensing receptor (CaR) can be potentiated by allosteric activators including calcimimetics and l-amino acids. In this study, we found that many mutations had differential effects on the functional modulation of the CaR by these two allosteric activators, supporting the idea that these modulators act through distinct sites. 10 mm l-phenylalanine and 1 microm NPS R-467, submaximal doses of the two agents, each elicited similar modulation of R185Q. However, there are different relative potencies for these two modulators with some receptors being more responsive to l-phenylalanine and others being more responsive to NPS R-467. The responsiveness of the CaR to Ca(2+)(o) appears to be essential to observe the potentiating action of l-phenylalanine but not of NPS R-467 on the receptor. NPS R-467 reduces the Hill coefficients of the wild-type as well as mutant receptors, suggesting that engagement of all Ca(2+) binding sites is not required when the receptor is activated by NPS R-467. In contrast, l-phenylalanine has little effect on the Hill coefficients of mutant receptors. The two-site model is further supported by the observation that these two classes of modulators exert a synergistic effect on CaRs with inactivating mutations that are responsive to both modulators.


Assuntos
Compostos de Anilina/farmacologia , Fenilalanina/farmacologia , Receptores de Superfície Celular/efeitos dos fármacos , Regulação Alostérica , Sítios de Ligação , Cálcio/farmacologia , Linhagem Celular , Sinergismo Farmacológico , Humanos , Receptores de Detecção de Cálcio , Receptores de Superfície Celular/química , Receptores de Superfície Celular/fisiologia
20.
J Am Soc Nephrol ; 11(5): 903-911, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10770968

RESUMO

The secondary hyperparathyroidism that develops in rats with chronic renal insufficiency (CRI) can be totally prevented by activation of the parathyroid Ca(2+) receptor with a calcimimetic compound, when treatment is initiated before parathyroid cell hyperplasia and increased circulating parathyroid hormone levels develop. In clinical practice, however, secondary hyperparathyroidism is usually manifest by the time CRI is diagnosed. This study examined the effects of daily oral gavage or continuous subcutaneous infusion for 8 wk of the calcimimetic NPS R-568 on the progression of established mild or moderate-to-severe secondary hyperparathyroidism in rats with CRI induced by 5/6 nephrectomy. Both oral and infused NPS R-568 completely prevented further hyperplasia but did not reduce total parathyroid cell number below that present at the initiation of treatment. This prevention of cellular proliferation occurred despite increases in plasma phosphate and decreases in Ca(2+) and 1, 25-dihydroxyvitamin D levels, and supports the view that the Ca(2+) receptor is the dominant regulator of parathyroid cell hyperplasia in addition to parathyroid hormone secretion. The clinical implications of these findings suggest that controlling Ca(2+) receptor activity with calcimimetic compounds could be sufficient to manage secondary hyperparathyroidism in CRI.


Assuntos
Compostos de Anilina/uso terapêutico , Proteínas de Ligação ao Cálcio/efeitos dos fármacos , Cálcio/agonistas , Hiperparatireoidismo Secundário/tratamento farmacológico , Uremia/complicações , Análise de Variância , Compostos de Anilina/administração & dosagem , Animais , Nitrogênio da Ureia Sanguínea , Cálcio/sangue , Proteínas de Ligação ao Cálcio/metabolismo , Creatinina/sangue , Hiperparatireoidismo Secundário/sangue , Hiperparatireoidismo Secundário/patologia , Hiperplasia , Glândulas Paratireoides/patologia , Hormônio Paratireóideo/sangue , Fenetilaminas , Fosfatos/sangue , Propilaminas , Ratos , Ratos Sprague-Dawley , Esteroide Hidroxilases/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA