Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
1.
Am J Respir Cell Mol Biol ; 69(2): 182-196, 2023 08.
Article in English | MEDLINE | ID: mdl-37098022

ABSTRACT

Asthma is a heterogeneous chronic airway disease with an unmet need for improved therapeutics in uncontrolled severe disease. The calcium-sensing receptor (CaSR) is a G protein-coupled receptor upregulated in asthma. The CaSR agonist, spermine, is also increased in asthmatic airways and contributes to bronchoconstriction. CaSR negative allosteric modulators (NAMs) oppose chronic airway inflammation, remodeling, and hyperresponsiveness in murine and guinea pig asthma models, but whether CaSR NAMs are effective acute bronchodilators compared with standard of care has not yet been established. Furthermore, the ability of different classes of NAMs to inhibit spermine-induced CaSR signaling or methacholine (MCh)-induced airway contraction has not been quantified. Here, we show CaSR NAMs differentially inhibit spermine-induced intracellular calcium mobilization and inositol monophosphate accumulation in HEK293 cells stably expressing the CaSR. NAMs reverse MCh-mediated airway contraction in mouse precision-cut lung slices with similar maximal relaxation compared with the standard treatment, salbutamol. Of note, the bronchodilator effects of CaSR NAMs are maintained under conditions of ß2-adrenergic receptor desensitization when salbutamol efficacy is abolished. Furthermore, overnight treatment with some, but not all, CaSR NAMs prevents MCh-mediated bronchoconstriction. These findings further support the CaSR as a putative drug target and NAMs as alternative or adjunct bronchodilators in asthma.


Subject(s)
Asthma , Bronchodilator Agents , Mice , Humans , Animals , Guinea Pigs , Bronchodilator Agents/pharmacology , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism , HEK293 Cells , Spermine/therapeutic use , Asthma/drug therapy , Asthma/metabolism , Albuterol/pharmacology , Methacholine Chloride/pharmacology
2.
Pharmacol Rev ; 72(3): 558-604, 2020 07.
Article in English | MEDLINE | ID: mdl-32467152

ABSTRACT

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.


Subject(s)
Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/antagonists & inhibitors , Animals , Binding Sites , GTP-Binding Proteins/metabolism , Humans , Models, Molecular , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Signal Transduction , Small Molecule Libraries/pharmacology
3.
J Pharmacol Exp Ther ; 376(1): 51-63, 2021 01.
Article in English | MEDLINE | ID: mdl-33115824

ABSTRACT

Asthma is still an incurable disease, and there is a recognized need for novel small-molecule therapies for people with asthma, especially those poorly controlled by current treatments. We previously demonstrated that calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs), calcilytics, uniquely suppress both airway hyperresponsiveness (AHR) and inflammation in human cells and murine asthma surrogates. Here we assess the feasibility of repurposing four CaSR NAMs, which were originally developed for oral therapy for osteoporosis and previously tested in the clinic as a novel, single, and comprehensive topical antiasthma therapy. We address the hypotheses, using murine asthma surrogates, that topically delivered CaSR NAMs 1) abolish AHR; 2) are unlikely to cause unwanted systemic effects; 3) are suitable for topical application; and 4) inhibit airway inflammation to the same degree as the current standard of care, inhaled corticosteroids, and, furthermore, inhibit airway remodeling. All four CaSR NAMs inhibited poly-L-arginine-induced AHR in naïve mice and suppressed both AHR and airway inflammation in a murine surrogate of acute asthma, confirming class specificity. Repeated exposure to inhaled CaSR NAMs did not alter blood pressure, heart rate, or serum calcium concentrations. Optimal candidates for repurposing were identified based on anti-AHR/inflammatory activities, pharmacokinetics/pharmacodynamics, formulation, and micronization studies. Whereas both inhaled CaSR NAMs and inhaled corticosteroids reduced airways inflammation, only the former prevented goblet cell hyperplasia in a chronic asthma model. We conclude that inhaled CaSR NAMs are likely a single, safe, and effective topical therapy for human asthma, abolishing AHR, suppressing airways inflammation, and abrogating some features of airway remodeling. SIGNIFICANCE STATEMENT: Calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs) reduce airway smooth muscle hyperresponsiveness, reverse airway inflammation as efficiently as topical corticosteroids, and suppress airway remodeling in asthma surrogates. CaSR NAMs, which were initially developed for oral therapy of osteoporosis proved inefficacious for this indication despite being safe and well tolerated. Here we show that structurally unrelated CaSR NAMs are suitable for inhaled delivery and represent a one-stop, steroid-free approach to asthma control and prophylaxis.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Indans/therapeutic use , Naphthalenes/therapeutic use , Phenylpropionates/therapeutic use , Quinazolinones/therapeutic use , Receptors, Calcium-Sensing/agonists , Allosteric Regulation , Animals , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/pharmacology , Bronchi/drug effects , Bronchi/metabolism , Drug Repositioning , HEK293 Cells , Humans , Indans/adverse effects , Indans/pharmacology , Male , Mice , Mice, Inbred BALB C , Naphthalenes/adverse effects , Naphthalenes/pharmacology , Phenylpropionates/adverse effects , Phenylpropionates/pharmacology , Quinazolinones/adverse effects , Quinazolinones/pharmacology , Receptors, Calcium-Sensing/metabolism
4.
BMC Nephrol ; 22(1): 275, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34372813

ABSTRACT

BACKGROUND: Parathyroid hormone (PTH) acts on bone to indirectly increase the number and activity of osteoclasts. Thus, PTH has a stimulatory effect on bone resorption and upregulates bone turnover. However, the responsiveness of bone to PTH varies widely among patients receiving dialysis. In fact, relative to the serum PTH level, the level of serum tartrate-resistant acid phosphatase-5b (TRACP-5b), a bone resorption marker derived from osteoclasts, varies as well. This study aimed to examine factors related to bone responsiveness to PTH in patients undergoing chronic hemodialysis (HD). METHODS: This study included patients receiving chronic HD in Kawasaki Municipal Tama Hospital (Kanagawa, Japan) and Yonaha Medical Clinic (Okinawa, Japan) and excluded patients who received HD for less than 6 months, those who received a combination of HD and peritoneal dialysis, and those who had cancer bone metastases or myeloma. The TRACP-5b/intact PTH (iPTH) ratio was created as an index of bone responsiveness to PTH, categorized into tertiles (low, medium, and high), and a cross-sectional study was conducted. P < 0.05 indicated statistically significant differences. RESULTS: One hundred and six patients were analyzed. Age (P = 0.010), body mass index (BMI) (P = 0.003), use of calcium-sensing receptor (CaSR) agonists (P = 0.008), use of vitamin D receptor activators (VDRAs) (P = 0.012), plasma iPTH level (P < 0.001), serum 1,25(OH)2D level (P = 0.003), and serum TRACP-5b level (P < 0.001) were significantly different among the three categories. In the single linear regression analysis, age (P = 0.016), corrected serum calcium level (P = 0.007), and ln [1,25(OH)2D] (P = 0.044) showed a significant positive correlation with ln [TRACP-5b/iPTH], whereas BMI (P = 0.026), use of CaSR agonists (P = 0.001), use of VDRAs (P = 0.009), and serum phosphorus level (P = 0.018) showed a significant negative correlation. Upon conducting multiple linear regression analysis incorporating significant variables in the single linear regression analysis, a significant negative correlation was observed between the TRACP-5b/iPTH ratio and intravenous administration of a CaSR agonist (etelcalcetide) and/or a VDRA (calcitriol or maxacalcitol) in all the adjusted models. CONCLUSIONS: Bone responsiveness to PTH is negatively correlated with the intravenous administration of a CaSR agonist and/or a VDRA in patients undergoing chronic HD.


Subject(s)
Bone Remodeling , Bone Resorption , Kidney Failure, Chronic , Parathyroid Hormone , Receptors, Calcitriol/metabolism , Receptors, Calcium-Sensing/agonists , Renal Dialysis , Tartrate-Resistant Acid Phosphatase , Bone Remodeling/drug effects , Bone Remodeling/physiology , Bone Resorption/metabolism , Bone Resorption/prevention & control , Cross-Sectional Studies , Female , Humans , Japan/epidemiology , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Male , Middle Aged , Osteoclasts/drug effects , Osteoclasts/physiology , Parathyroid Hormone/blood , Parathyroid Hormone/metabolism , Peritoneal Dialysis/adverse effects , Peritoneal Dialysis/methods , Renal Dialysis/adverse effects , Renal Dialysis/methods , Tartrate-Resistant Acid Phosphatase/blood , Tartrate-Resistant Acid Phosphatase/metabolism
5.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670622

ABSTRACT

The Hippo pathway is involved in human tumorigenesis and tissue repair. Here, we investigated the Hippo coactivator Yes-associated protein 1 (YAP1) and the kinase large tumor suppressor 1/2 (LATS1/2) in tumors of the parathyroid glands, which are almost invariably associated with primary hyperparathyroidism. Compared with normal parathyroid glands, parathyroid adenomas (PAds) and carcinomas show variably but reduced nuclear YAP1 expression. The kinase LATS1/2, which phosphorylates YAP1 thus promoting its degradation, was also variably reduced in PAds. Further, YAP1 silencing reduces the expression of the key parathyroid oncosuppressor multiple endocrine neoplasia type 1(MEN1), while MEN1 silencing increases YAP1 expression. Treatment of patient-derived PAds-primary cell cultures and Human embryonic kidney 293A (HEK293A) cells expressing the calcium-sensing receptor (CASR) with the CASR agonist R568 induces YAP1 nuclear accumulation. This effect was prevented by the incubation of the cells with RhoA/Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitors Y27632 and H1152. Lastly, CASR activation increased the expression of the YAP1 gene targets CYR61, CTGF, and WNT5A, and this effect was blunted by YAP1 silencing. Concluding, here we provide preliminary evidence of the involvement of the Hippo pathway in human tumor parathyroid cells and of the existence of a CASR-ROCK-YAP1 axis. We propose a tumor suppressor role for YAP1 and LATS1/2 in parathyroid tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Parathyroid Glands/metabolism , Parathyroid Neoplasms/genetics , Receptors, Calcium-Sensing/genetics , Transcription Factors/genetics , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Amides/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression/drug effects , HEK293 Cells , Humans , Parathyroid Neoplasms/metabolism , Phenethylamines/pharmacology , Propylamines/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pyridines/pharmacology , RNA Interference , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
6.
Mol Pharmacol ; 97(1): 35-45, 2020 01.
Article in English | MEDLINE | ID: mdl-31704718

ABSTRACT

Current operational models of agonism and allosterism quantify ligand actions at receptors where agonist concentration-response relationships are nonhyperbolic by introduction of a transducer slope that relates receptor occupancy to response. However, for some receptors nonhyperbolic concentration-response relationships arise from multiple endogenous agonist molecules binding to a receptor in a cooperative manner. Thus, we developed operational models of agonism in systems with cooperative agonist binding and evaluated the models by simulating data describing agonist effects. The models were validated by analyzing experimental data demonstrating the effects of agonists and allosteric modulators at receptors where agonist binding follows hyperbolic (M4 muscarinic acetylcholine receptors) or nonhyperbolic relationships (metabotropic glutamate receptor 5 and calcium-sensing receptor). For hyperbolic agonist concentration-response relationships, no differences in estimates of ligand affinity, efficacy, or cooperativity were observed when the slope was assigned to either a transducer slope or agonist binding slope. In contrast, for receptors with nonhyperbolic agonist concentration-response relationships, estimates of ligand affinity, efficacy, or cooperativity varied depending on the assignment of the slope. The extent of this variation depended on the magnitude of the slope value and agonist efficacy, and for allosteric modulators on the magnitude of cooperativity. The modified operational models described herein are well suited to analyzing agonist and modulator interactions at receptors that bind multiple orthosteric agonists in a cooperative manner. Accounting for cooperative agonist binding is essential to accurately quantify agonist and drug actions. SIGNIFICANCE STATEMENT: Some orthosteric agonists bind to multiple sites on a receptor, but current analytical methods to characterize such interactions are limited. Herein, we develop and validate operational models of agonism and allosterism for receptors with multiple orthosteric binding sites, and demonstrate that such models are essential to accurately quantify agonist and drug actions. These findings have important implications for the discovery and development of drugs targeting receptors such as the calcium-sensing receptor, which binds at least five calcium ions.


Subject(s)
Binding Sites/drug effects , Calcium Ionophores/pharmacology , Drug Agonism , Models, Biological , Receptors, Calcium-Sensing/agonists , Allosteric Regulation/drug effects , Calcium/metabolism , Computer Simulation , Dose-Response Relationship, Drug , Drug Synergism , HEK293 Cells , Humans , Ligands , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/chemistry , Receptor, Metabotropic Glutamate 5/metabolism , Receptor, Muscarinic M4/agonists , Receptor, Muscarinic M4/chemistry , Receptor, Muscarinic M4/metabolism , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism
7.
Biochem Biophys Res Commun ; 521(1): 227-231, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31635806

ABSTRACT

In addition to the maintenance of Ca2+ homeostasis, the calcium-sensing receptor (CaSR) is involved in many diverse physiological functions in the mammalian body. The receptor works as a kokumi taste receptor in taste buds and as a nutrient sensor in the gut, where it regulates the secretion of glycemic response and appetite-related hormones. To identify novel human CaSR (hCaSR) activators from food ingredients, we conducted a screening using a cell-based hCaSR assay. Hen egg-white lysozyme, which is a sweet protein, was found to be a novel orthosteric agonist of hCaSR with an EC50 value of 592 µM. Lysozyme hydrolysate was not able to activate hCaSR, thus suggesting that the protein structure of lysozyme is necessary for hCaSR activation. Thaumatin, which is another sweet protein, also activated hCaSR with an EC50 value of 71 µM. This is the first report that shows hCaSR activation by proteins with molecular weights exceeding 10,000 Da. These results provide a new avenue for the development of hCaSR activators, which could be applicable in food or drugs that modulate taste perception, appetite, or glucose tolerance, in addition to Ca2+ homeostasis.


Subject(s)
Muramidase/metabolism , Plant Proteins/metabolism , Receptors, Calcium-Sensing/agonists , Calcium/analysis , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydrolysis , Receptors, Calcium-Sensing/metabolism , Static Electricity
8.
J Pharmacol Sci ; 142(2): 41-49, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31831259

ABSTRACT

Diabetic nephropathy (DN) is the most serious end-stage renal disease which characterized by renal glomerular sclerosis including glomerular hypertrophy, glomerular basement membrane (GBM) thickening, mesangial expansion and renal fibrosis. TGF-ß/Smads signal pathway plays a crucial role in the development of renal fibrosis. In this study, we found that GdCl3 which was an agonist of Calcium-sensing receptor (CaSR) could repress the activation of TGF-ß/Smads signal pathway induced by TGF-ß1 or high glucose and then alleviated the accumulation of extracellular matrix (ECM) in mesangial cells and the kidney of type1 diabetic rats. Further study indicated that GdCl3 could induce the binding of CaSR and TßR II and then both of these two receptors translocated from cell membrane to cytoplasm, in this case, TßR II on the cell membrane was decreased and then desensitized to the stimulation of its ligand TGF-ß1, so that the activation of its downstream factors such as Smad2 and Smad3 were blocked, finally, ECM expression in mesangial cells were inhibited. We concluded that GdCl3 could alleviate the accumulation of ECM in mesangial cells via antagonizing TGF-ß/Smads signal pathway in diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/drug therapy , Gadolinium/pharmacology , Kidney Glomerulus/drug effects , Receptors, Calcium-Sensing/agonists , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Kidney Glomerulus/pathology , Male , Mesangial Cells/metabolism , Mesangial Cells/pathology , Rats , Rats, Sprague-Dawley , Sclerosis , Signal Transduction , Streptozocin
9.
Am J Physiol Renal Physiol ; 316(5): F1006-F1015, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30838870

ABSTRACT

316: F1006-F1015, 2019. First published March 6, 2019; doi: 10.1152/ajprenal.00413.2018 .-Experimental studies have shown that pharmacological activation of calcium-sensing receptor (CaSR) attenuates renal fibrosis in some animal models beyond modification of bone and mineral homeostasis; however, its underlying mechanisms remain largely unknown. Since excessive collagen deposition is the key feature of fibrosis, the present study aimed to examine whether CaSR was involved in the regulation of collagen expression in rats with adenine diet-induced renal fibrosis and in profibrotic transforming growth factor (TGF)-ß1-treated renal proximal tubular epithelial cells (PTECs). The results showed that the CaSR agonist cinacalcet significantly attenuated renal collagen accumulation and tubular injury in adenine diet-fed rats. Additionally, the in vitro experiment showed that profibrotic TGF-ß1 significantly increased the expression of collagen and decreased CaSR expression at the mRNA and protein levels in a concentration- and time-dependent manner. Furthermore, the CaSR CRISPR activation plasmid and cinacalcet partially abrogated the upregulation of collagen induced by TGF-ß1 treatment. Blockade of CaSR by the CRISPR/Cas9 KO plasmid or the pharmacological antagonist Calhex231 further enhanced TGF-ß1-induced collagen expression. Mechanistic experiments found that Smad2 phosphorylation and Snail expression were markedly increased in PTECs treated with TGF-ß1, whereas the CaSR CRISPR activation plasmid and cinacalcet substantially suppressed this induction. In summary, this study provides evidence for a direct renal tubular epithelial protective effect of CaSR activation in renal fibrosis, possibly through suppression of collagen expression in PTECs.


Subject(s)
Calcimimetic Agents/pharmacology , Cinacalcet/pharmacology , Collagen/metabolism , Epithelial Cells/drug effects , Kidney Diseases/prevention & control , Kidney Tubules, Proximal/drug effects , Receptors, Calcium-Sensing/agonists , Adenine , Animals , Benzamides/pharmacology , CRISPR-Cas Systems , Cells, Cultured , Cyclohexylamines/pharmacology , Disease Models, Animal , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Humans , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Phosphorylation , Rats, Wistar , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Smad2 Protein/metabolism , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology
10.
Am J Physiol Renal Physiol ; 317(2): F240-F253, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31042050

ABSTRACT

To garner insights into the renal regulation of Ca2+ homeostasis, we performed an mRNA microarray on kidneys from mice treated with the Ca2+-sensing receptor (CaSR) agonist cinacalcet. This revealed decreased gene expression of Na+/H+ exchanger isoform 8 (NHE8) in response to CaSR activation. These results were confirmed by quantitative real-time PCR. Moreover, administration of vitamin D also decreased NHE8 mRNA expression. In contrast, renal NHE8 protein expression from the same samples was increased. To examine the role of NHE8 in transmembrane Ca2+ fluxes, we used the normal rat kidney (NRK) cell line. Cell surface biotinylation and confocal immunofluorescence microscopy demonstrated NHE8 apical expression. Functional experiments found 5-(N-ethyl-N-isopropyl)amiloride (EIPA)-inhibitable NHE activity in NRK cells at concentrations minimally attenuating NHE1 activity in AP-1 cells. To determine how NHE8 might regulate Ca2+ balance, we measured changes in intracellular Ca2+ uptake by live cell Ca2+ imaging with the fluorophore Fura-2 AM. Inhibition of NHE8 with EIPA or by removing extracellular Na+-enhanced Ca2+ influx into NRK cells. Ca2+ influx was mediated by a voltage-dependent Ca2+ channel rather than directly via NHE8. NRK cells express Cav1.3 and display verapamil-sensitive Ca2+ influx and NHE8 inhibition-augmented Ca2+ influx via a voltage-dependent Ca2+ channel. Finally, proximal tubules perused ex vivo demonstrated increased Ca2+ influx in the presence of luminal EIPA at a concentration that would inhibit NHE8. The results of the present study are consistent with NHE8 regulating Ca2+ uptake into the proximal tubule epithelium.


Subject(s)
Calcium Signaling , Calcium/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , CHO Cells , Calcimimetic Agents/pharmacology , Calcium Channels/metabolism , Cinacalcet/pharmacology , Cricetulus , Epithelial Cells/drug effects , Homeostasis , Kidney Tubules, Proximal/drug effects , Mutation , Rats , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism , Sodium-Hydrogen Exchanger 1/genetics , Sodium-Hydrogen Exchanger 1/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics
11.
Arch Biochem Biophys ; 657: 15-22, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30217510

ABSTRACT

Long-term high-calcium intake and intestinal calcium hyperabsorption are hazardous to the body. It is hypothesized that enterocytes possess mechanisms for preventing superfluous calcium absorption, including secretion of negative regulators of calcium absorption and utilization of calcium-sensing receptor (CaSR) to detect luminal calcium. Herein, Caco-2 monolayers were treated with high doses of 1,25(OH)2D3 to induce calcium hyperabsorption or directly exposed to high apical calcium. The expression of counterregulatory factor of calcium absorption, fibroblast growth factor (FGF)-23, was also investigated in the intestine of lactating rats, which physiologically exhibit calcium hyperabsorption. We found that FGF-23 expression was enhanced in all intestinal segments of lactating rats. In Caco-2 monolayers, high apical calcium and 1,25(OH)2D3 induced FGF-23 secretion into culture media. FGF-23 antagonized 1,25(OH)2D3-induced calcium transport and led to a significant, but small, change in paracellular permeability. Furthermore, high-dose 1,25(OH)2D3 upregulated FGF-23 expression, which was prevented by CaSR inhibitors. Activation of apical CaSR by cinacalcet and AC-265347 abolished 1,25(OH)2D3-induced calcium transport in a dose-dependent manner. In conclusion, the intestinal FGF-23 expression was upregulated in conditions with calcium hyperabsorption, presumably to help protect against excessive calcium absorption, while CaSR probably monitored calcium in the lumen and induced FGF-23 production for preventing superfluous calcium uptake.


Subject(s)
Benzothiazoles/pharmacology , Calcitriol/metabolism , Calcium/metabolism , Cinacalcet/pharmacology , Intestinal Absorption/drug effects , Receptors, Calcium-Sensing/agonists , Animals , Caco-2 Cells , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Intestinal Mucosa/metabolism , Lactation/metabolism , Pregnancy , Rats, Sprague-Dawley , Up-Regulation
12.
Bioorg Med Chem Lett ; 28(11): 2055-2060, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29724589

ABSTRACT

The calcium-sensing receptor (CaSR) plays an important role in sensing extracellular calcium ions and regulating parathyroid hormone secretion by parathyroid gland cells, and the receptor is a suitable target for the treatment of hyperparathyroidism. Cinacalcet hydrochloride is a representative CaSR agonist which widely used for the hyperparathyroidism. However, it has several issues to clinical use, such as nausea/vomiting and strong inhibition of CYP2D6. We tried to improve these issues of cinacalcet for a new pharmaceutical agent as a preferable CaSR agonist. Optimization from cinacalcet resulted in the identification of pyrrolidine compounds and successfully led to the discovery of evocalcet as an oral allosteric CaSR agonist. Evocalcet, which exhibited highly favorable profiles such as CaSR agonistic activity and good DMPK profiles, will provide a novel therapeutic option for secondary hyperparathyroidism.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Drug Discovery , Hyperparathyroidism/drug therapy , Pyrrolidines/pharmacology , Receptors, Calcium-Sensing/agonists , Animals , Cytochrome P-450 Enzyme Inhibitors/chemical synthesis , Cytochrome P-450 Enzyme Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Rats , Structure-Activity Relationship
13.
Sheng Li Xue Bao ; 70(3): 269-280, 2018 Jun 25.
Article in Zh | MEDLINE | ID: mdl-29926068

ABSTRACT

The present study was to investigate the role of the interaction between canonical transient receptor potential channel 1 (TRPC1) and calcium release-activated calcium modulator 1 (Orai1) in extracellular Ca2+-sensing receptor (CaR)-induced extracellular Ca2+ influx and nitric oxide (NO) production. Human umbilical vein endothelial cells (HUVECs) were incubated with CaR agonist Spermine [activating store-operated calcium channels (SOC) and receptor-operated calcium channels (ROC)] alone or in combination with the following reagents: CaR negative allosteric modulator Calhex231 plus ROC analogue TPA (activating ROC and blocking SOC), Ro31-8220 (PKC inhibitor that activates SOC and blocks ROC) or Go6967 (PKCs and PKCµ inhibitor that activates SOC and blocks ROC). The protein expressions and co-localization of TRPC1 and Orai1 were determined using immunofluorescent staining. The interaction between TRPC1 and Orai1 was examined by co-immunoprecipitation. We silenced the expressions of their genes in the HUVECs by transfection of constructed TRPC1 and Orai1 shRNA plasmids. Intracellular Ca2+ concentration ([Ca2+]i) was detected using Ca2+ indicator Fura-2/AM, and NO production was determined by DAF-FM staining. The results showed that TRPC1 and Orai1 protein expressions were co-located on the cell membrane of the HUVECs. Compared with Spermine+Ca2+ group, Calhex231+ TPA+Spermine+Ca2+, Ro31-8220+Spermine+Ca2+ and Go6976+Spermine+Ca2+ groups exhibited down-regulated protein expressions of TRPC1 and Orai1 in cytoplasm and decreased co-localization on the cell membrane. Co-immunoprecipitation results showed that the interaction between TRPC1 and Orai1 was reduced by Calhex231 plus TPA, Ro31-8220 or Go6976 addition in the Spermine-stimulated HUVECs. Double knockdown of Trpc1 and Orai1 genes significantly decreased [Ca2+]i level and NO production in all of the Spermine+Ca2+, Calhex231+TPA+Spermine+Ca2+, Ro31-8220+Spermine+Ca2+ and Go6976+Spermine+Ca2+ groups. These results suggest that TRPC1/Orai1 may form a complex that mediates Ca2+ influx and No production via SOC and ROC activation.


Subject(s)
Calcium/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Nitric Oxide/metabolism , ORAI1 Protein/metabolism , TRPC Cation Channels/metabolism , Benzamides/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling , Cell Membrane , Cyclohexylamines/pharmacology , Gene Silencing , Humans , Indoles/pharmacology , RNA, Small Interfering , Receptors, Calcium-Sensing/agonists , Spermine/pharmacology
14.
Am J Physiol Cell Physiol ; 313(1): C27-C41, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28381520

ABSTRACT

Melamine causes renal tubular cell injury through inflammation, fibrosis, and apoptosis. Although melamine affects the rise in intracellular Ca2+ concentration ([Ca2+]i), reactive oxygen species (ROS) production, and proapoptotic pathway activation, the mechanism of upstream Ca2+ signaling is unknown. Because melamine has some structural similarities with l-amino acids, which endogenously activate Ca2+-sensing receptors (CSR), we examined the effect of melamine on CSR-induced Ca2+ signaling and apoptotic cell death. We show here that melamine activates CSR, causing a sustained Ca2+ entry in the renal epithelial cell line, LLC-PK1. Moreover, such CSR stimulation resulted in a rise in [Ca2+]i, leading to enhanced ROS production. Furthermore, melamine-induced elevated [Ca2+]i and ROS production caused a dose-dependent increase in apoptotic (by DAPI staining, DNA laddering, and annexin V assay) and necrotic (propidium iodide staining) cell death. Upon examining the downstream mechanism, we found that transforming growth factor ß1 (TGF-ß1), which increases extracellular matrix genes and proapoptotic signaling, was also upregulated at lower doses of melamine, which could be due to an early event inducing apoptosis. Additionally, cells exposed to melamine displayed a rise in pERK activation and lactate dehydrogenase release resulting in cytotoxicity. These results offer a novel insight into the molecular mechanisms by which melamine exerts its effect on CSR, causing a sustained elevation of [Ca2+]i, leading to ROS generation, fibronectin production, proapoptotic pathway activation, and renal cell damage. Together, these results thus suggest that melamine-induced apoptosis and/or necrosis may subsequently result in acute kidney injury and promote kidney stone formation.


Subject(s)
Calcium/metabolism , Environmental Pollutants/toxicity , Epithelial Cells/drug effects , Kidney Tubules, Proximal/drug effects , Receptors, Calcium-Sensing/agonists , Triazines/toxicity , Animals , Apoptosis/drug effects , Cell Line , DNA Fragmentation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Signal Transduction , Spectrometry, Fluorescence , Swine , Time-Lapse Imaging , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
15.
Amino Acids ; 49(9): 1587-1599, 2017 09.
Article in English | MEDLINE | ID: mdl-28623466

ABSTRACT

In-feed antibiotics have been commonly used to promote the growth performance of piglets. The antibiotics can increase protein utilization, but the underlying mechanism is largely unknown. The present study investigated the effects of in-feed antibiotics on intestinal AA transporters and receptors to test the hypothesis that the alteration of circulating AA profiles may be concomitant with the change of intestinal AA transporters and receptors. Sixteen litters of piglets at day 7 started to receive creep feed with (Antibiotic) or without (Control) antibiotic. Piglets were weaned at day 23 after birth, and fed the same diets until day 42. In-feed antibiotics did not affect the BW of 23-day-old (P = 0.248), or 42-day-old piglets (P = 0.089), but increased the weight gain to feed ratio from day 23 to 42 (P = 0.020). At day 42 after birth, antibiotic treatment increased the concentrations of most AAs in serum (P < 0.05), and decreased the concentrations of most AAs in jejunal and ileal digesta. Antibiotics upregulated (P < 0.05) the mRNA expression levels for jejunal AAs transporters (CAT1, EAAC1, ASCT2, y+LAT1), peptide transporters (PepT1), and Na+-K+-ATPase (ATP1A1), and ileal AA transporters (ASCT2, y+LAT1, b0,+AT, and B0AT1), and ATP1A1. The antibiotics also upregulated the mRNA expression of jejunal AAs receptors T1R3 and CaSR, and ileal T1R3. Protein expression levels for jejunal AA transporters (EAAC1, b0,+AT, and ASCT2) and PepT1 were also upregulated. Correlation analysis revealed that the alterations of AA profiles in serum after the in-feed antibiotics were correlated with the upregulations of mRNA expression levels for key AA transporters and receptors in the small intestine. In conclusion, the in-feed antibiotics increased serum level of most AAs and decreased most AAs in the small intestine. These changes correlated with the upregulations of mRNA expression levels for key AA transporters and receptors in the small intestine. The findings provide further insights into the mechanism of in-feed antibiotics, which may provide new framework for designing alternatives to antibiotics in animal feed in the future.


Subject(s)
Amino Acids/blood , Anti-Bacterial Agents/pharmacology , Gene Expression Regulation/drug effects , Weight Gain/drug effects , Amino Acid Transport System ASC/agonists , Amino Acid Transport System ASC/genetics , Amino Acid Transport System ASC/metabolism , Animal Feed/analysis , Animals , Animals, Newborn , Biological Transport/drug effects , Excitatory Amino Acid Transporter 3/agonists , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Kitasamycin/pharmacology , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Oxytetracycline/pharmacology , Peptide Transporter 1/agonists , Peptide Transporter 1/genetics , Peptide Transporter 1/metabolism , Quinoxalines/pharmacology , RNA, Messenger/agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Swine , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Weaning
16.
Nephrol Dial Transplant ; 32(10): 1723-1730, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28057872

ABSTRACT

BACKGROUND: Secondary hyperparathyroidism (SHPT) is a major complication associated with chronic kidney disease. We evaluated the efficacy and safety of etelcalcetide (ONO-5163/AMG 416), a novel intravenous calcimimetic, in Japanese haemodialysis patients with SHPT. METHODS: In this phase 3, multicentre, randomized, double-blind, placebo-controlled, parallel-group study, etelcalcetide was administered three times per week at an initial dose of 5 mg, and subsequently adjusted to doses between 2.5 and 15 mg at 4-week intervals for 12 weeks. A total of 155 SHPT patients with serum intact parathyroid hormone (iPTH) levels ≥300 pg/mL were assigned to receive etelcalcetide (n = 78) or placebo (n = 77). The primary endpoint was the proportion of patients with decreased serum iPTH to the target range proposed by the Japanese Society for Dialysis Therapy (60-240 pg/mL). The major secondary endpoint was the proportion of patients with ≥30% reductions in serum iPTH from baseline. RESULTS: The proportion of patients meeting the primary endpoint was significantly higher for etelcalcetide (59.0%) versus placebo (1.3%). Similarly, the proportion of patients meeting the major secondary endpoint was significantly higher for etelcalcetide (76.9%) versus placebo (5.2%). Serum albumin-corrected calcium, phosphorus and intact fibroblast growth factor-23 levels were decreased in the etelcalcetide group. Nausea, vomiting and symptomatic hypocalcaemia were mild with etelcalcetide. Serious adverse events related to etelcalcetide were not observed. CONCLUSIONS: This study demonstrated the efficacy and safety of etelcalcetide. As the only available intravenous calcium-sensing receptor agonist, etelcalcetide is likely to provide a new treatment option for SHPT in haemodialysis patients.


Subject(s)
Hyperparathyroidism, Secondary/drug therapy , Peptides/therapeutic use , Receptors, Calcium-Sensing/agonists , Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/complications , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Calcium/blood , Double-Blind Method , Female , Humans , Hyperparathyroidism, Secondary/etiology , Male , Middle Aged , Parathyroid Hormone/blood , Phosphorus/blood , Young Adult
17.
Diabetes Obes Metab ; 19 Suppl 1: 54-62, 2017 09.
Article in English | MEDLINE | ID: mdl-28880472

ABSTRACT

Glucose is a primary stimulator of insulin secretion. It has been thought that glucose exerts its effect by a mechanism solely dependent on glucose metabolism. We show here that glucose induces rapid Ca2+ and cyclic AMP signals in ß-cells. These rapid signals are independent of glucose-metabolism and are reproduced by non-metabolizable glucose analogues. These results led us to postulate that glucose activates a cell-surface receptor, namely the glucose-sensing receptor. Rapid signals induced by glucose are blocked by inhibition of a sweet taste receptor subunit T1R3 and a calcium-sensing receptor subunit CaSR. In accordance with these observations, T1R3 and CaSR form a heterodimer. In addition, a heterodimer of T1R3 and CaSR is activated by glucose. These results suggest that a heterodimer of T1R3 and CaSR is a major component of the glucose-sensing receptor. When the glucose-sensing receptor is blocked, glucose-induced insulin secretion is inhibited. Also, ATP production is significantly attenuated by the inhibition of the receptor. Conversely, stimulation of the glucose-sensing receptor by either artificial sweeteners or non-metabolizable glucose analogue increases ATP. Hence, the glucose-sensing receptor signals promote glucose metabolism. Collectively, glucose activates the cell-surface glucose-sensing receptor and promotes its own metabolism. Glucose then enters the cells and is metabolized through already activated metabolic pathways. The glucose-sensing receptor is a key molecule regulating the action of glucose in ß-cells.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Models, Biological , Receptors, Cell Surface/agonists , Animals , Calcium Signaling , Cyclic AMP/metabolism , Dimerization , Enzyme Activation , Gene Expression Regulation , Humans , Insulin Secretion , Insulin-Secreting Cells/enzymology , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Multimerization , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Second Messenger Systems
18.
Bioorg Med Chem Lett ; 27(20): 4673-4677, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28916340

ABSTRACT

Modulation of gastrointestinal nutrient sensing pathways provides a promising a new approach for the treatment of metabolic diseases including diabetes and obesity. The calcium-sensing receptor has been identified as a key receptor involved in mineral and amino acid nutrient sensing and thus is an attractive target for modulation in the intestine. Herein we describe the optimization of gastrointestinally restricted calcium-sensing receptor agonists starting from a 3-aminopyrrolidine-containing template leading to the identification of GI-restricted agonist 19 (GSK3004774).


Subject(s)
Receptors, Calcium-Sensing/agonists , Animals , Calcium/metabolism , Cell Membrane Permeability/drug effects , Dogs , Gastrointestinal Tract/metabolism , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Pyrrolidines/chemistry , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Structure-Activity Relationship
19.
Drug Metab Dispos ; 44(8): 1319-31, 2016 08.
Article in English | MEDLINE | ID: mdl-26895981

ABSTRACT

AMG 416 (etelcalcetide) is a novel synthetic peptide agonist of the calcium-sensing receptor composed of a linear chain of seven d-amino acids (referred to as the d-amino acid backbone) with a d-cysteine linked to an l-cysteine via a disulfide bond. AMG 416 contains four basic d-arginine residues and is a +4 charged peptide at physiologic pH with a mol. wt. of 1048.3 Da. The pharmacokinetics (PK), disposition, and potential of AMG 416 to cause drug-drug interaction were investigated in nonclinical studies with two single (14)C-labels placed either at a potentially metabolically labile acetyl position or on the d-alanine next to d-cysteine in the interior of the d-amino acid backbone. After i.v. dosing, the PK and disposition of AMG 416 were similar in male and female rats. Radioactivity rapidly distributed to most tissues in rats with intact kidneys, and renal elimination was the predominant clearance pathway. No strain-dependent differences were observed. In bilaterally nephrectomized rats, minimal radioactivity (1.2%) was excreted via nonrenal pathways. Biotransformation occurred primarily via disulfide exchange with endogenous thiol-containing molecules in whole blood rather than metabolism by enzymes, such as proteases or cytochrome P450s; the d-amino acid backbone remained unaltered. A substantial proportion of the plasma radioactivity was covalently conjugated to albumin. AMG 416 presents a low risk for P450 or transporter-mediated drug-drug interactions because it showed no interactions in vitro. These studies demonstrated a (14)C label on either the acetyl or the d-alanine in the d-amino acid backbone would be appropriate for clinical studies.


Subject(s)
Calcimimetic Agents/pharmacokinetics , Peptides/pharmacokinetics , Receptors, Calcium-Sensing/agonists , Administration, Intravenous , Animals , Biotransformation , Calcimimetic Agents/administration & dosage , Calcimimetic Agents/blood , Calcimimetic Agents/toxicity , Cytochrome P-450 Enzyme System/metabolism , Dogs , Drug Interactions , Female , HEK293 Cells , Humans , Kidney/metabolism , Liver/metabolism , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Molecular Structure , Peptides/administration & dosage , Peptides/blood , Peptides/toxicity , Protein Binding , Rats, Inbred BN , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Renal Elimination , Risk Assessment , Serum Albumin/metabolism , Structure-Activity Relationship , Tissue Distribution , Transfection
20.
Calcif Tissue Int ; 98(4): 341-58, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26319799

ABSTRACT

The actions of extracellular Ca(2+) in regulating parathyroid gland and kidney functions are mediated by the extracellular calcium receptor (CaR), a G protein-coupled receptor. The CaR is one of the essential molecules maintaining systemic Ca(2+) homeostasis and is a molecular target for drugs useful in treating bone and mineral disorders. Ligands that activate the CaR are termed calcimimetics and are classified as either agonists (type I) or positive allosteric modulators (type II); calcimimetics inhibit the secretion of parathyroid hormone (PTH). Cinacalcet is a type II calcimimetic that is used to treat secondary hyperparathyroidism in patients receiving dialysis and to treat hypercalcemia in some forms of primary hyperparathyroidism. The use of cinacalcet among patients with secondary hyperparathyroidism who are managed with dialysis effectively lowers circulating PTH levels, reduces serum phosphorus and FGF23 concentrations, improves bone histopathology, and may diminish skeletal fracture rates and the need for parathyroidectomy. A second generation type II calcimimetic (AMG 416) is currently under regulatory review. Calcilytics are CaR antagonists that stimulate the secretion of PTH. Several calcilytic compounds have been evaluated as orally active anabolic therapies for postmenopausal osteoporosis but clinical development of all of them has been abandoned because they lacked clinical efficacy. Calcilytics might be repurposed for new indications like autosomal dominant hypocalcemia or other disorders beyond those involving systemic Ca(2+) homeostasis.


Subject(s)
Calcimimetic Agents/therapeutic use , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/antagonists & inhibitors , Hypercalcemia/drug therapy , Hyperparathyroidism/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL