Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 626(8001): 1141-1148, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326620

ABSTRACT

The calcium-sensing receptor (CaSR) is a family C G-protein-coupled receptor1 (GPCR) that has a central role in regulating systemic calcium homeostasis2,3. Here we use cryo-electron microscopy and functional assays to investigate the activation of human CaSR embedded in lipid nanodiscs and its coupling to functional Gi versus Gq proteins in the presence and absence of the calcimimetic drug cinacalcet. High-resolution structures show that both Gi and Gq drive additional conformational changes in the activated CaSR dimer to stabilize a more extensive asymmetric interface of the seven-transmembrane domain (7TM) that involves key protein-lipid interactions. Selective Gi and Gq coupling by the receptor is achieved through substantial rearrangements of intracellular loop 2 and the C terminus, which contribute differentially towards the binding of the two G-protein subtypes, resulting in distinct CaSR-G-protein interfaces. The structures also reveal that natural polyamines target multiple sites on CaSR to enhance receptor activation by zipping negatively charged regions between two protomers. Furthermore, we find that the amino acid L-tryptophan, a well-known ligand of CaSR extracellular domains, occupies the 7TM bundle of the G-protein-coupled protomer at the same location as cinacalcet and other allosteric modulators. Together, these results provide a framework for G-protein activation and selectivity by CaSR, as well as its allosteric modulation by endogenous and exogenous ligands.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Receptors, Calcium-Sensing , Humans , Allosteric Regulation/drug effects , Cinacalcet/pharmacology , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Ligands , Lipids , Nanostructures/chemistry , Polyamines/metabolism , Protein Conformation/drug effects , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/ultrastructure , Substrate Specificity , Tryptophan/metabolism , Calcium/metabolism
2.
J Antimicrob Chemother ; 79(4): 903-917, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38412335

ABSTRACT

BACKGROUND: MDR Staphylococcus aureus infections, along with the severity of biofilm-associated infections, continue to threaten human health to a great extent. It necessitates the urgent development of novel antimicrobial and antibiofilm agents. OBJECTIVES: To reveal the mechanism and target of cinacalcet as an antibacterial and antimicrobial agent for S. aureus. METHODS: Screening of non-antibiotic drugs for antibacterial and antibiofilm properties was conducted using a small-molecule drug library. In vivo efficacy was assessed through animal models, and the antibacterial mechanism was studied using quantitative proteomics, biochemical assays, LiP-SMap, BLI detection and gene knockout techniques. RESULTS: Cinacalcet, an FDA-approved drug, demonstrated antibacterial and antibiofilm activity against S. aureus, with less observed development of bacterial resistance. Importantly, cinacalcet significantly improved survival in a pneumonia model and bacterial clearance in a biofilm infection model. Moreover, the antibacterial mechanism of cinacalcet mainly involves the destruction of membrane-targeted structures, alteration of energy metabolism, and production of reactive oxygen species (ROS). Cinacalcet was found to target IcaR, inhibiting biofilm formation through the negative regulation of IcaADBC. CONCLUSIONS: The findings suggest that cinacalcet has potential for repurposing as a therapeutic agent for MDR S. aureus infections and associated biofilms, warranting further investigation.


Subject(s)
Anti-Infective Agents , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Humans , Staphylococcus aureus , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Iron-Dextran Complex/therapeutic use , Drug Repositioning , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Cell Membrane , Biofilms , Microbial Sensitivity Tests
3.
FASEB J ; 37(8): e23094, 2023 08.
Article in English | MEDLINE | ID: mdl-37462513

ABSTRACT

Little is known about the effect of the recently developed calcimimetic evocalcet (Evo) on parathyroid calcium-sensing receptor (CaSR) and vitamin D receptor (VDR) expression. We examined the effects of Evo and cinacalcet (Cina) on CaSR and VDR expression in 5/6 nephrectomized Sprague-Dawley rats fed a high-phosphorus diet for 4 weeks to develop secondary hyperparathyroidism (SHPT). These uremic rats were divided into 4 groups-baseline control (Nx4W) and groups with additional treatment with either the Vehicle, Evo, or Cina for 2 weeks; normal rats were used as normal controls (NC). Blood parameters and parathyroid tissue were analyzed. CaSR and VDR expression levels were determined using immunohistochemistry. The degree of kidney injury and hyperphosphatemia was similar in the uremic groups (Nx4W, Vehicle, Cina, and Evo). Serum parathyroid hormone levels were significantly higher in the Nx4W and Vehicle groups than in the NC group. This increase was significantly suppressed in the Cina and Evo groups compared with that in the Vehicle group. Serum calcium levels were significantly and equally lower in the Cina and Evo groups relative to those in the Vehicle group. CaSR expression was significantly lower in the Nx4W and Vehicle groups than in the NC group. This downregulation was of an equally lesser magnitude in the Cina and Evo groups. A similar trend was observed for VDR expression. These results indicate that Evo and Cina treatment can increase parathyroid CaSR and VDR expression in uremic rats with SHPT, which could provide better control of mineral and bone disorder markers.


Subject(s)
Hyperparathyroidism, Secondary , Receptors, Calcitriol , Rats , Animals , Receptors, Calcitriol/metabolism , Receptors, Calcium-Sensing/metabolism , Rats, Sprague-Dawley , Parathyroid Glands/metabolism , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/complications , Hyperparathyroidism, Secondary/metabolism , Parathyroid Hormone/metabolism , Cinacalcet/pharmacology , Cinacalcet/metabolism
4.
J Vet Pharmacol Ther ; 47(4): 274-279, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38563476

ABSTRACT

Cinacalcet is an oral calcimimetic that has potential to non-invasively treat primary hyperparathyroidism in dogs (Canis lupis familiaris). There is minimal data assessing its efficacy in dogs. This study aimed to determine whether a single dose of cinacalcet decreases serum ionized calcium (iCa), total calcium (tCa), and parathyroid hormone (PTH) concentrations. Twelve dogs received a median dose of 0.49 mg/kg (range 0.30-0.69 mg/kg) cinacalcet per os. Venous blood samples were collected at time 0 (before cinacalcet administration), 3, 8, and 24 h following cinacalcet administration. PTH, iCa, and tCa concentrations were measured at each time point and compared to 0 hour concentrations. A significant (50%) decrease in serum PTH occurred at 3 h with a median PTH of 4.6 pmol/L (range 2.7-10.8) at baseline and 1.65 pmol/L (range 0.5-14.7) at 3 h; p = .005. A significant, but not clinically relevant, decrease in serum iCa from a median baseline of 1.340 mmol/L (range 1.32-1.41) to a 3 h median of 1.325 mmol/L (range 1.26-1.39), p = .043, was also observed. tCa concentrations were not different. This study showed that a single dose of cinacalcet leads to transient decreases in iCa and PTH concentrations in healthy dogs.


Subject(s)
Calcium , Cinacalcet , Parathyroid Hormone , Animals , Dogs/blood , Parathyroid Hormone/blood , Cinacalcet/administration & dosage , Cinacalcet/pharmacology , Calcium/blood , Male , Female , Administration, Oral , Calcimimetic Agents/administration & dosage , Calcimimetic Agents/pharmacology
5.
Hum Mol Genet ; 30(10): 880-892, 2021 05 29.
Article in English | MEDLINE | ID: mdl-33729479

ABSTRACT

Adaptor protein 2 (AP2), a heterotetrameric complex comprising AP2α, AP2ß2, AP2µ2 and AP2σ2 subunits, is ubiquitously expressed and involved in endocytosis and trafficking of membrane proteins, such as the calcium-sensing receptor (CaSR), a G-protein coupled receptor that signals via Gα11. Mutations of CaSR, Gα11 and AP2σ2, encoded by AP2S1, cause familial hypocalciuric hypercalcaemia types 1-3 (FHH1-3), respectively. FHH3 patients have heterozygous AP2S1 missense Arg15 mutations (p.Arg15Cys, p.Arg15His or p.Arg15Leu) with hypercalcaemia, which may be marked and symptomatic, and occasional hypophosphataemia and osteomalacia. To further characterize the phenotypic spectrum and calcitropic pathophysiology of FHH3, we used CRISPR/Cas9 genome editing to generate mice harboring the AP2S1 p.Arg15Leu mutation, which causes the most severe FHH3 phenotype. Heterozygous (Ap2s1+/L15) mice were viable, and had marked hypercalcaemia, hypermagnesaemia, hypophosphataemia, and increases in alkaline phosphatase activity and fibroblast growth factor-23. Plasma 1,25-dihydroxyvitamin D was normal, and no alterations in bone mineral density or bone turnover were noted. Homozygous (Ap2s1L15/L15) mice invariably died perinatally. Co-immunoprecipitation studies showed that the AP2S1 p.Arg15Leu mutation impaired protein-protein interactions between AP2σ2 and the other AP2 subunits, and also with the CaSR. Cinacalcet, a CaSR positive allosteric modulator, decreased plasma calcium and parathyroid hormone concentrations in Ap2s1+/L15 mice, but had no effect on the diminished AP2σ2-CaSR interaction in vitro. Thus, our studies have established a mouse model that is representative for FHH3 in humans, and demonstrated that the AP2S1 p.Arg15Leu mutation causes a predominantly calcitropic phenotype, which can be ameliorated by treatment with cinacalcet.


Subject(s)
Adaptor Protein Complex 2/genetics , Adaptor Protein Complex sigma Subunits/genetics , Fibroblast Growth Factor-23/genetics , Hypercalcemia/genetics , Receptors, Calcium-Sensing/genetics , Animals , Bone Density/genetics , CRISPR-Cas Systems/genetics , Calcium/metabolism , Cinacalcet/pharmacology , Disease Models, Animal , Gene Editing , Humans , Hypercalcemia/drug therapy , Hypercalcemia/metabolism , Hypercalcemia/pathology , Mice , Mutation/genetics , Phenotype
6.
Clin Endocrinol (Oxf) ; 98(4): 516-526, 2023 04.
Article in English | MEDLINE | ID: mdl-36316798

ABSTRACT

OBJECTIVES: Human physiology and epidemiology studies have demonstrated complex interactions between the renin-angiotensin-aldosterone system, parathyroid hormone and calcium homeostasis. Several of these studies have suggested that aldosterone inhibition may lower parathyroid hormone (PTH) levels. The objective of this study was to assess the effect of 4 weeks of maximally tolerated mineralocorticoid receptor antagonist therapy with eplerenone on PTH levels in patients with primary hyperparathyroidism (P-HPT) when compared to amiloride and placebo. We also investigated the synergistic effect of these interventions when combined with cinacalcet for an additional 2 weeks. DESIGN: Randomized, double-blinded, three parallel-group, placebo-controlled trial. PATIENTS: Patients with P-HPT. RESULTS: Most patients were women (83%) and White (76%). Maximally tolerated doses of eplerenone and amiloride induced significant reductions in blood pressure and increases in renin and aldosterone production; however, despite these physiologic changes, neither intervention induced significant changes in PTH or calcium levels when compared to the placebo. Both eplerenone and amiloride therapy induced significant reductions in procollagen type 1 N-terminal propeptide levels when compared to placebo. When cinacalcet therapy was added, PTH and calcium levels were markedly reduced in all groups; however, there was no significant difference in PTH or serum calcium reductions between groups. CONCLUSIONS: Although maximally tolerated therapy with eplerenone and amiloride induced expected changes in renin, aldosterone and blood pressure, there were no meaningful changes in PTH or serum calcium levels in P-HPT patients. These results suggest that inhibition of aldosterone action does not have a clinically meaningful role in medical therapy for P-HPT.


Subject(s)
Amiloride , Hyperparathyroidism, Primary , Humans , Female , Male , Eplerenone/therapeutic use , Cinacalcet/pharmacology , Amiloride/therapeutic use , Aldosterone , Calcium , Renin , Parathyroid Hormone
7.
Calcif Tissue Int ; 112(4): 452-462, 2023 04.
Article in English | MEDLINE | ID: mdl-36754901

ABSTRACT

Patients with type 2 diabetes mellitus (T2DM) experience a higher risk of fractures despite paradoxically exhibiting normal to high bone mineral density (BMD). This has drawn into question the applicability to T2DM of conventional fracture reduction treatments that aim to retain BMD. In a primary human osteoblast culture system, high glucose levels (25 mM) impaired cell proliferation and matrix mineralization compared to physiological glucose levels (5 mM). Treatment with parathyroid hormone (PTH, 10 nM), a bone anabolic agent, and cinacalcet (CN, 1 µM), a calcimimetic able to target the Ca2+-sensing receptor (CaSR), were tested for their effects on proliferation and differentiation. Strikingly, CN+PTH co-treatment was shown to promote cell growth and matrix mineralization under both physiological and high glucose conditions. CN+PTH reduced apoptosis by 0.9-fold/0.4-fold as measured by Caspase-3 activity assay, increased alkaline phosphatase (ALP) expression by 1.5-fold/twofold, increased the ratio of nuclear factor κ-B ligand (RANKL) to osteoprotegerin (OPG) by 2.1-fold/1.6-fold, and increased CaSR expression by 1.7-fold/4.6-fold (physiological glucose/high glucose). Collectively, these findings indicate a potential for CN+PTH combination therapy as a method to ameliorate the negative impact of chronic high blood glucose on bone remodeling.


Subject(s)
Diabetes Mellitus, Type 2 , Parathyroid Hormone , Humans , Cinacalcet/pharmacology , Cinacalcet/metabolism , Diabetes Mellitus, Type 2/metabolism , Osteoblasts/metabolism , Osteoprotegerin/metabolism , Glucose/metabolism , RANK Ligand/metabolism , Cells, Cultured
8.
Kidney Int ; 101(6): 1186-1199, 2022 06.
Article in English | MEDLINE | ID: mdl-35271934

ABSTRACT

Calcimimetic agents allosterically increase the calcium ion sensitivity of the calcium-sensing receptor (CaSR), which is expressed in the tubular system and to a lesser extent in podocytes. Activation of this receptor can reduce glomerular proteinuria and structural damage in proteinuric animal models. However, the precise role of the podocyte CaSR remains unclear. Here, a CaSR knockdown in cultured murine podocytes and a podocyte-specific CaSR knockout in BALB/c mice were generated to study its role in proteinuria and kidney function. Podocyte CaSR knockdown abolished the calcimimetic R-568 mediated calcium ion-influx, disrupted the actin cytoskeleton, and reduced cellular attachment and migration velocity. Adriamycin-induced proteinuria enhanced glomerular CaSR expression in wild-type mice. Albuminuria, podocyte foot process effacement, podocyte loss and glomerular sclerosis were significantly more pronounced in adriamycin-treated podocyte-specific CaSR knockout mice compared to wild-type littermates. Co-treatment of wild-type mice with adriamycin and the calcimimetic cinacalcet reduced proteinuria in wild-type, but not in podocyte-specific CaSR knockout mice. Additionally, four children with nephrotic syndrome, whose parents objected to glucocorticoid therapy, were treated with cinacalcet for one to 33 days. Proteinuria declined transiently by up to 96%, serum albumin increased, and edema resolved. Thus, activation of podocyte CaSR regulates key podocyte functions in vitro and reduced toxin-induced proteinuria and glomerular damage in mice. Hence, our findings suggest a potential novel role of CaSR signaling in control of glomerular disease.


Subject(s)
Kidney Diseases , Podocytes , Animals , Calcium/metabolism , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Doxorubicin/toxicity , Humans , Kidney Diseases/metabolism , Mice , Mice, Knockout , Podocytes/metabolism , Proteinuria/chemically induced , Proteinuria/genetics , Proteinuria/metabolism , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism
9.
Kidney Int ; 101(6): 1110-1112, 2022 06.
Article in English | MEDLINE | ID: mdl-35597590

ABSTRACT

Calcimimetics allosterically increase the calcium ion sensitivity of the calcium-sensing receptor (CaSR). Using a CaSR knockdown in podocytes and a podocyte-specific CaSR knockout in mice, Mühlig et al. uncovered a stabilizing role for actin cytoskeleton and cell adhesion. Short-term alleviation of albuminuria and proteinuria was observed in 4 children treated with cinacalcet. Here we discuss the potential mechanisms whereby CaSR displays a favorable effect in podocytes and the context in which calcimimetics may alleviate nephrotic syndrome.


Subject(s)
Nephrotic Syndrome , Podocytes , Animals , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Mice , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/metabolism , Podocytes/metabolism , Proteinuria/drug therapy , Proteinuria/metabolism , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism
10.
Med Sci Monit ; 28: e937338, 2022 Aug 09.
Article in English | MEDLINE | ID: mdl-35941808

ABSTRACT

BACKGROUND Cinacalcet is a calcium-sensing receptor agonist that is clinically approved for the treatment of secondary hyperparathyroidism in chronic kidney disease and hypercalcemia in patients with parathyroid carcinoma. This study aimed to use quantitative mass spectrometry-based label-free proteomics to evaluate the effects of cinacalcet on protein expression in rat brains and livers. MATERIAL AND METHODS We randomly assigned 18 Wistar rats to 2 groups: an untreated control group (n=6) and a group treated with cinacalcet at a dose corresponding to the maximum dose used in humans (2 mg/kg/body weight, 5 days/week) divided into 7-day (n=6) and 21-day (n=6) treatment subgroups. A mass-spectrometry-based label-free quantitative proteomics approach using peptides peak area calculation was used to evaluate the changes in protein expression in examined tissues. Bioinformatics analysis of quantitative proteomics data was done using MaxQuant and Perseus environment. RESULTS No changes in protein expression were revealed in the 7-day treatment subgroup. We detected 10 upregulated and 3 downregulated proteins in the liver and 1 upregulated protein in the brain in the 21-day treatment subgroup compared to the control group. Based on Gene Ontology classification, all identified differentially expressed proteins were indicated as molecular functions involved in the enzyme regulator activity (36%), binding (31%), and catalytic activity (19%). CONCLUSIONS These findings indicate that long-term cinacalcet therapy can impair phase II of enzymatic detoxication and can cause disturbances in blood hemostasis, lipid metabolism, and inflammatory mediators or contribute to the acceleration of cognitive dysfunction; therefore, appropriate patient monitoring should be considered.


Subject(s)
Proteomics , Receptors, Calcium-Sensing , Animals , Brain/metabolism , Calcium , Cinacalcet/pharmacology , Cinacalcet/therapeutic use , Humans , Liver/metabolism , Mass Spectrometry , Naphthalenes , Parathyroid Hormone , Rats , Rats, Wistar , Receptors, Calcium-Sensing/metabolism
11.
Chin J Physiol ; 65(6): 277-281, 2022.
Article in English | MEDLINE | ID: mdl-36588353

ABSTRACT

Ca2+-sensing receptors (CaSRs) are G protein-coupled receptors activated by elevated concentrations of extracellular Ca2+. In our previous works, we showed protein and functional expression of CaSR in mouse cerebral endothelial cell (EC) (bEND.3); the CaSR response (high Ca2+-elicited cytosolic [Ca2+] elevation) was unaffected by suppression of phospholipase C but in part involved Ca2+ influx through transient receptor potential V1 (TRPV1) channels. In this work, we investigated if extracellular acidity affected CaSR-mediated Ca2+ influx triggered by high (3 mM) Ca2+ (CaSR agonist), 3 mM spermine (CaSR agonist), and 10 mM cinacalcet (positive allosteric modulator of CaSR). Extracellular acidosis (pH 6.8 and pH 6.0) strongly suppressed cytosolic [Ca2+] elevation triggered by high Ca2+, spermine, and cinacalcet; acidosis also inhibited Mn2+ influx stimulated by high Ca2+ and cinacalcet. Purinoceptor-triggered Ca2+ response, however, was not suppressed by acidosis. Extracellular acidity also did not affect membrane potential, suggesting suppressed CaSR-mediated Ca2+ influx in acidity did not result from the reduced electrical driving force for Ca2+. Our results suggest Ca2+ influx through a putative CaSR-TRP complex in bEND.3 EC was sensitive to extracellular pH.


Subject(s)
Calcium Signaling , Endothelial Cells , Mice , Animals , Endothelial Cells/metabolism , Cinacalcet/pharmacology , Cinacalcet/metabolism , Spermine/pharmacology , Spermine/metabolism , Membrane Potentials , Calcium/metabolism
12.
Int J Mol Sci ; 23(8)2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35457141

ABSTRACT

Neuroblastoma is the most common extracranial solid tumor of childhood, with heterogeneous clinical manifestations ranging from spontaneous regression to aggressive metastatic disease. The calcium-sensing receptor (CaSR) is a G protein-coupled receptor (GPCR) that senses plasmatic fluctuation in the extracellular concentration of calcium and plays a key role in maintaining calcium homeostasis. We have previously reported that this receptor exhibits tumor suppressor properties in neuroblastoma. The activation of CaSR with cinacalcet, a positive allosteric modulator of CaSR, reduces neuroblastoma tumor growth by promoting differentiation, endoplasmic reticulum (ER) stress and apoptosis. However, cinacalcet treatment results in unmanageable hypocalcemia in patients. Based on the bias signaling shown by calcimimetics, we aimed to identify a new drug that might exert tumor-growth inhibition similar to cinacalcet, without affecting plasma calcium levels. We identified a structurally different calcimimetic, AC-265347, as a promising therapeutic agent for neuroblastoma, since it reduced tumor growth by induction of differentiation, without affecting plasma calcium levels. Microarray analysis suggested biased allosteric modulation of the CaSR signaling by AC-265347 and cinacalcet towards distinct intracellular pathways. No upregulation of genes involved in calcium signaling and ER stress were observed in patient-derived xenografts (PDX) models exposed to AC-265347. Moreover, the most significant upregulated biological pathways promoted by AC-265347 were linked to RHO GTPases signaling. AC-265347 upregulated cancer testis antigens (CTAs), providing new opportunities for CTA-based immunotherapies. Taken together, this study highlights the importance of the biased allosteric modulation when targeting GPCRs in cancer. More importantly, the capacity of AC-265347 to promote differentiation of malignant neuroblastoma cells provides new opportunities, alone or in combination with other drugs, to treat high-risk neuroblastoma patients.


Subject(s)
Hypocalcemia , Neuroblastoma , Calcium/metabolism , Cinacalcet/pharmacology , Humans , Male , Neuroblastoma/drug therapy , Receptors, Calcium-Sensing/metabolism
13.
Toxicol Appl Pharmacol ; 421: 115533, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33848515

ABSTRACT

Decreased activity of AMP-activated protein kinase (AMPK) is implicated in the pathogenesis of diabetic cardiomyopathy (DCM). Recent evidence suggests a crosstalk between cinacalcet and AMPK activation. This study investigated the effects of cinacalcet on cardiac remodeling and dysfunction in type 2 diabetic rats (T2DM). High fat diet for 4 weeks combined with single intraperitoneal injection of streptozotocin (30 mg/kg) was used to induce type 2 diabetes in rats. Diabetic rats were either orally treated with vehicle, 5 or 10 mg/kg cinacalcet for 4 weeks. Control rats were fed standard chow diet and intraperitoneally injected with citrate buffer. T2DM rats showed lower body weight (BW), hyperglycemia and dyslipidemia, along with increased heart weight (HW) and HW/BW ratio. Masson's trichrome stained cardiac sections revealed massive fibrosis in T2DM rats. There were increased TGF-ß1 and hydroxyproline levels, coupled with up-regulation of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) in hearts of T2DM rats. These alterations were associated with redox imbalance and impaired cardiac functions. Decreased phosphorylation of AMPK at threonine172 residue was found in T2DM hearts. Cinacalcet for 4 weeks significantly activated AMPK and alleviated cardiac remodeling and dysfunction in a dose-dependent manner, without affecting blood glucose, serum calcium and phosphorus levels. Cinacalcet increased the mitochondrial DNA content, and expressions of PGC-1α, UCP-3, beclin-1 and LC3-II/LC3-I ratio. Cinacalcet decreased the pro-apoptotic Bax, while increased the anti-apoptotic Bcl-2 in cardiac tissue of T2DM rats. These findings might highlight cinacalcet as an alternative therapy to combat the development and progression of DCM.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Cinacalcet/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/prevention & control , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Ventricular Remodeling/drug effects , Animals , Apoptosis/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Fibrosis , Hemodynamics/drug effects , Male , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats, Wistar , Signal Transduction , Streptozocin
14.
Pharmazie ; 76(10): 511-514, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34620281

ABSTRACT

The lack of effective strategies remains a pivotal challenge for hepatocellular carcinoma (HCC) treatment. YAP/ TAZ is a promising target for effective drugs against HCC. In this study, we profiled the regulatory effect of 98 drugs on transcriptional activity of YAP/TAZ and identified the calcimimetic agent cinacalcet as a potent YAP inhibitor. Cinacalcet inhibited YAP expression in HCC models at both transcriptional and protein levels, and ultimately arrested cell proliferation of HCC. Overexpression of YAP weakened the anticancer efficacy of cinacalcet, indicating that YAP was responsible for the antineoplastic activity of cinacalcet. Collectively, this study suggested cinacalcet as a feasible anticancer drug for HCC via its inhibition on YAP/TAZ.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/pathology , Cinacalcet/pharmacology , Humans , Liver Neoplasms/pathology , Signal Transduction , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
15.
Am J Physiol Renal Physiol ; 318(3): F639-F646, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31961714

ABSTRACT

Little is known about changes in parathyroid cells when calcimimetics are withdrawn. We examined the response of parathyroid glands to cinacalcet (Cina) withdrawal in uremic Sprague-Dawley rats fed a high-phosphate diet to develop secondary hyperparathyroidism and divided into groups treated with vehicle (UC), Cina, and Cina and maxacalcitol (Maxa), a vitamin D receptor activator (CiNa + Maxa). After 2 wk of treatment, vehicle and Cina were withdrawn and Maxa was continued. Rats were analyzed immediately (day 0) and 7 days (day 7) after withdrawal. The Cina and CiNa + Maxa groups had significantly lower parathyroid hormone (PTH) than the UC group on day 0, although PTH in the Cina group reached UC levels on day 7. On day 0, there were significantly more proliferating cell nuclear antigen-positive cells in the UC group compared with normal controls, and this increase was significantly suppressed in the Cina and CiNa + Maxa groups. On day 7, the Cina group, but not the CiNa + Maxa group, showed a significant increase in proliferating cell nuclear antigen-positive cells compared with the UC group. This increase was related to parathyroid cell diameter regression to UC levels, whereas combination treatment maintained diameter suppression. These results indicate that parathyroid growth activity is stimulated by Cina withdrawal, although the PTH level was not further increased. Continuous administration of Cina may be required for optimal control of secondary hyperparathyroidism, and simultaneous use of a vitamin D receptor activator may be advisable during Cina withdrawal.


Subject(s)
Cinacalcet/pharmacology , Parathyroid Glands/drug effects , Renal Insufficiency/drug therapy , Renal Insufficiency/etiology , Animals , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cinacalcet/administration & dosage , Hyperparathyroidism, Secondary/chemically induced , Hyperparathyroidism, Secondary/drug therapy , Male , Nephrectomy , Rats , Rats, Sprague-Dawley
16.
Hum Mol Genet ; 27(5): 901-911, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29325022

ABSTRACT

Mutations of the sigma subunit of the heterotetrameric adaptor-related protein complex 2 (AP2σ) impair signalling of the calcium-sensing receptor (CaSR), and cause familial hypocalciuric hypercalcaemia type 3 (FHH3). To date, FHH3-associated AP2σ mutations have only been identified at one residue, Arg15. We hypothesized that additional rare AP2σ variants may also be associated with altered CaSR function and hypercalcaemia, and sought for these by analysing >111 995 exomes (>60 706 from ExAc and dbSNP, and 51 289 from the Geisinger Health System-Regeneron DiscovEHR dataset, which also contains clinical data). This identified 11 individuals to have 9 non-synonymous AP2σ variants (Arg3His, Arg15His (x3), Ala44Thr, Phe52Tyr, Arg61His, Thr112Met, Met117Ile, Glu122Gly and Glu142Lys) with 3 of the 4 individuals who had Arg15His and Met117Ile AP2σ variants having mild hypercalcaemia, thereby indicating a prevalence of FHH3-associated AP2σ mutations of ∼7.8 per 100 000 individuals. Structural modelling of the novel eight AP2σ variants (Arg3His, Ala44Thr, Phe52Tyr, Arg61His, Thr112Met, Met117Ile, Glu122Gly and Glu142Lys) predicted that the Arg3His, Thr112Met, Glu122Gly and Glu142Lys AP2σ variants would disrupt polar contacts within the AP2σ subunit or affect the interface between the AP2σ and AP2α subunits. Functional analyses of all eight AP2σ variants in CaSR-expressing cells demonstrated that the Thr112Met, Met117Ile and Glu142Lys variants, located in the AP2σ α4-α5 helical region that forms an interface with AP2α, impaired CaSR-mediated intracellular calcium (Cai2+) signalling, consistent with a loss of function, and this was rectified by treatment with the CaSR positive allosteric modulator cinacalcet. Thus, our studies demonstrate another potential class of FHH3-causing AP2σ mutations located at the AP2σ-AP2α interface.


Subject(s)
Adaptor Protein Complex alpha Subunits/metabolism , Adaptor Protein Complex sigma Subunits/genetics , Mutation , Receptors, Calcium-Sensing/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex sigma Subunits/metabolism , Cinacalcet/pharmacology , Databases, Genetic , Exome , Female , Humans , Hypercalcemia/drug therapy , Hypercalcemia/genetics , Male , Middle Aged , Models, Molecular , Protein Conformation , Signal Transduction , Exome Sequencing
17.
Biol Pharm Bull ; 43(10): 1583-1590, 2020.
Article in English | MEDLINE | ID: mdl-32999168

ABSTRACT

Chronic kidney disease (CKD) patients with secondary hyperparathyroidism (SHPT) have an increased risk of cardiovascular disease (CVD). Cinacalcet is a calcimimetic that permits impaired endothelial functions to be recovered via inhibiting parathyroid hormone (PTH) production in SHPT patients. However, the underlying mechanism for its action remains unknown. The purpose of this study was to examine the effect of cinacalcet on the redox state of human serum albumin (HSA), a reliable marker for assessing endothelial oxidative damage in SHPT patients who were receiving hemodialysis. Cinacalcet was administered to six SHPT patients for a period of 8 weeks. After 4 weeks of treatment, cinacalcet significantly decreased the oxidized albumin ratio which is a ratio of reduced and oxidized forms of HSA via increasing reduced form of HSA. Moreover, the radical scavenging abilities of HSA that was isolated from SHPT patients were increased by cinacalcet, suggesting the recovery of the impaired vascular anti-oxidant ability. Interestingly, the oxidized albumin ratio in SHPT patients was significantly higher than that in hemodialysis patients. In addition, the changes of intact PTH levels were significantly correlated with the oxidized albumin ratio. It therefore appears that PTH may induce oxidative stress in SHPT patients. In fact, an active analogue of PTH increased the production of reactive oxygen species in human endothelial cells. Thus, cinacalcet exhibits anti-oxidative activity through its pharmacological action. Additionally, cinacalcet itself showed radical scavenging activity. In conclusion, cinacalcet improves the redox status of HSA by inhibiting PTH production and partially by its radical scavenging action.


Subject(s)
Antioxidants/therapeutic use , Cinacalcet/therapeutic use , Hyperparathyroidism, Secondary/blood , Hyperparathyroidism, Secondary/drug therapy , Renal Dialysis/trends , Serum Albumin, Human/metabolism , Adult , Aged , Antioxidants/pharmacology , Calcium-Regulating Hormones and Agents/pharmacology , Calcium-Regulating Hormones and Agents/therapeutic use , Cinacalcet/pharmacology , Female , Humans , Male , Middle Aged , Oxidation-Reduction/drug effects , Parathyroid Hormone/antagonists & inhibitors , Parathyroid Hormone/blood , Renal Dialysis/adverse effects , Treatment Outcome
18.
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
19.
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
20.
Kidney Blood Press Res ; 44(6): 1327-1338, 2019.
Article in English | MEDLINE | ID: mdl-31747666

ABSTRACT

OBJECTIVE: To assess the long-term effects including all-cause mortality, cardiovascular mortality, and fracture incidence, of cinacalcet on secondary hyperparathyroidism (SHPT) in patients on dialysis. METHODS: PubMed, Embase, and the Cochrane Central Register of Controlled Trials were searched from their inception to October 2018. Randomized controlled trials (RCTs) and cohort design prospective observational studies assessing cinacalcet for the treatment of SHPT in dialysis patients were included. Data extraction was independently completed by 2 authors who determined the methodological quality of the studies and extracted data in duplicate. Study-specific risk estimates were tested by using a fixed effects model. RESULTS: A total of 14 articles with 38,219 participants were included, of which 10 RCTs with 7,471 participants and 4 prospective observational studies with 30,748 participants fulfilled the eligibility criteria. Compared with no cinacalcet, cinacalcet administration reduced all-cause mortality (relative risk [RR] 0.91, 95% CI 0.89-0.94, p < 0.001) and cardiovascular mortality (RR 0.92, 95% CI 0.89-0.95, p < 0.001), but it did not significantly reduce the incidence of fractures (RR 0.93, 95% CI 0.87-1.00, p = 0.05). CONCLUSIONS: The results of this meta-analysis indicated that the treatment of SHPT with cinacalcet may in fact reduce all-cause mortality and cardiovascular mortality among patients receiving maintenance dialysis.


Subject(s)
Cinacalcet/therapeutic use , Hyperparathyroidism, Secondary/drug therapy , Kidney Failure, Chronic/therapy , Survival Rate , Calcimimetic Agents/pharmacology , Calcimimetic Agents/therapeutic use , Cardiovascular Diseases/etiology , Cardiovascular Diseases/mortality , Cinacalcet/pharmacology , Fractures, Bone/prevention & control , Humans , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/mortality , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/mortality , Renal Dialysis/mortality
SELECTION OF CITATIONS
SEARCH DETAIL