Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Antimicrob Chemother ; 79(4): 903-917, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38412335

RESUMO

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.


Assuntos
Anti-Infecciosos , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Animais , Humanos , Staphylococcus aureus , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Complexo Ferro-Dextran/uso terapêutico , Reposicionamento de Medicamentos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Anti-Infecciosos/uso terapêutico , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Membrana Celular , Biofilmes , Testes de Sensibilidade Microbiana
2.
Nature ; 626(8001): 1141-1148, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38326620

RESUMO

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.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP , Receptores de Detecção de Cálcio , Humanos , Regulação Alostérica/efeitos dos fármacos , Cinacalcete/farmacologia , Microscopia Crioeletrônica , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Ligantes , Lipídeos , Nanoestruturas/química , Poliaminas/metabolismo , Conformação Proteica/efeitos dos fármacos , Receptores de Detecção de Cálcio/química , Receptores de Detecção de Cálcio/metabolismo , Receptores de Detecção de Cálcio/ultraestrutura , Especificidade por Substrato , Triptofano/metabolismo , Cálcio/metabolismo
3.
Transl Res ; 265: 17-25, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37990828

RESUMO

ADPKD is characterized by progressive cyst formation and enlargement leading to kidney failure. Tolvaptan is currently the only FDA-approved treatment for ADPKD; however, it can cause serious adverse effects including hepatotoxicity. There remains an unmet clinical need for effective and safe treatments for ADPKD. The extracellular Ca2+-sensing receptor (CaSR) is a regulator of epithelial ion transport. FDA-approved CaSR activator cinacalcet can reduce cAMP-induced Cl- and fluid secretion in various epithelial cells by activating phosphodiesterases (PDE) that hydrolyze cAMP. Since elevated cAMP is a key mechanism of ADPKD progression by promoting cell proliferation, cyst formation and enlargement (via Cl- and fluid secretion), here we tested efficacy of cinacalcet in cell and animal models of ADPKD. Cinacalcet treatment reduced cAMP-induced Cl- secretion and CFTR activity in MDCK cells as suggested by ∼70 % lower short-circuit current (Isc) changes in response to forskolin and CFTRinh-172, respectively. Cinacalcet treatment inhibited forskolin-induced cAMP elevation by 60 % in MDCK cells, and its effect was completely reversed by IBMX (PDE inhibitor). In MDCK cells treated with forskolin, cinacalcet treatment concentration-dependently reduced cell proliferation, cyst formation and cyst enlargement by up to 50 % without affecting cell viability. Cinacalcet treatment (20 mg/kg/day for 7 days, subcutaneous) reduced renal cyst index in a mouse model of ADPKD (Pkd1flox/flox;Ksp-Cre) by 20 %. Lastly, cinacalcet treatment reduced cyst enlargement and cell proliferation in human ADPKD cells by 60 %. Considering its efficacy as shown here, and favorable safety profile including extensive post-approval data, cinacalcet can be repurposed as a novel ADPKD treatment.


Assuntos
Cistos , Rim Policístico Autossômico Dominante , Camundongos , Animais , Humanos , Rim Policístico Autossômico Dominante/tratamento farmacológico , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Receptores de Detecção de Cálcio , Colforsina/farmacologia , Reposicionamento de Medicamentos , Células Cultivadas , Rim
4.
Transl Res ; 263: 45-52, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37678755

RESUMO

Cyclic nucleotide elevation in intestinal epithelial cells is the key pathology causing intestinal fluid loss in secretory diarrheas such as cholera. Current secretory diarrhea treatment is primarily supportive, and oral rehydration solution is the mainstay of cholera treatment. There is an unmet need for safe, simple and effective diarrhea treatments. By promoting cAMP hydrolysis, extracellular calcium-sensing receptor (CaSR) is a regulator of intestinal fluid transport. We studied the antidiarrheal mechanisms of FDA-approved CaSR activator cinacalcet and tested its efficacy in clinically relevant human cell, mouse and intestinal organoid models of secretory diarrhea. By using selective inhibitors, we found that cAMP agonists-induced secretory short-circuit currents (Isc) in human intestinal T84 cells are mediated by collective actions of apical membrane cystic fibrosis transmembrane conductance regulator (CFTR) and Clc-2 Cl- channels, and basolateral membrane K+ channels. 30 µM cinacalcet pretreatment inhibited all 3 components of forskolin and cholera toxin-induced secretory Isc by ∼75%. In mouse jejunal mucosa, cinacalcet inhibited forskolin-induced secretory Isc by ∼60% in wild type mice, with no antisecretory effect in intestinal epithelia-specific Casr knockout mice (Casr-flox; Vil1-cre). In suckling mouse model of cholera induced by oral cholera toxin, single dose (30 mg/kg) oral cinacalcet treatment reduced intestinal fluid accumulation by ∼55% at 20 hours. Lastly, cinacalcet inhibited forskolin-induced secretory Isc by ∼75% in human colonic and ileal organoids. Our findings suggest that CaSR activator cinacalcet has antidiarrheal efficacy in distinct human cell, organoid and mouse models of secretory diarrhea. Considering its excellent clinical safety profile, cinacalcet can be repurposed as a treatment for cyclic nucleotide-mediated secretory diarrheas including cholera.


Assuntos
Antidiarreicos , Cólera , Camundongos , Humanos , Animais , Antidiarreicos/metabolismo , Antidiarreicos/farmacologia , Antidiarreicos/uso terapêutico , Cólera/tratamento farmacológico , Cólera/metabolismo , Cólera/patologia , Toxina da Cólera/metabolismo , Toxina da Cólera/farmacologia , Toxina da Cólera/uso terapêutico , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Cinacalcete/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Receptores de Detecção de Cálcio/uso terapêutico , Nucleotídeos Cíclicos/metabolismo , Nucleotídeos Cíclicos/farmacologia , Nucleotídeos Cíclicos/uso terapêutico , Colforsina/metabolismo , Colforsina/farmacologia , Colforsina/uso terapêutico , Diarreia/tratamento farmacológico , Diarreia/metabolismo , Mucosa Intestinal/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/uso terapêutico , Camundongos Knockout
5.
Eur J Pharmacol ; 956: 175936, 2023 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-37541363

RESUMO

Upacicalcet (formerly SK-1403/AJT240) is a novel non-peptide calcimimetic agent that acts as a calcium-sensing receptor (CaSR) agonist for the treatment of secondary hyperparathyroidism (SHPT) in chronic kidney disease (CKD). We compared upacicalcet with other calcimimetics (etelcalcetide or cinacalcet) and examined its in vitro and in vivo characteristics in terms of its human CaSR agonistic activity, its efficacy in normal and CKD rats after a single administration, and its effect on gastric emptying in rats. Upacicalcet activated human CaSR depending on the extracellular calcium (Ca2+) concentration without exhibiting an agonistic action when the extracellular Ca2+ level was below the physiological level. On the other hand, etelcalcetide had an agonistic activity even in the absence of physiological levels of extracellular Ca2+. The intravenous administration of upacicalcet to normal and double-nephrectomized rats dose-dependently (0.03-3mg/kg and 0.3-30mg/kg, respectively) decreased the serum intact parathyroid hormone (iPTH) and serum Ca2+ levels; however, the effect of upacicalcet on the reduction in serum Ca2+ disappeared at extracellular Ca2+ levels below the physiologically range, even when administered at a dose higher (100-fold) than the effective dose. Furthermore, upacicalcet did not affect gastric emptying in normal rats when administered up to a dose of 10mg/kg (300-fold higher than the dose affecting serum iPTH levels), while the administration of cinacalcet significantly slowed gastric emptying by approximately 50%. These findings suggest that upacicalcet has potential as an alternative calcimimetic agent with good pharmacological properties and a lower risk of hypocalcemia and gastrointestinal complications.


Assuntos
Hiperparatireoidismo Secundário , Insuficiência Renal Crônica , Humanos , Ratos , Animais , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Receptores de Detecção de Cálcio/agonistas , Hormônio Paratireóideo , Cálcio , Calcimiméticos/farmacologia , Calcimiméticos/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Diálise Renal/efeitos adversos
6.
FASEB J ; 37(8): e23094, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37462513

RESUMO

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.


Assuntos
Hiperparatireoidismo Secundário , Receptores de Calcitriol , Ratos , Animais , Receptores de Calcitriol/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Ratos Sprague-Dawley , Glândulas Paratireoides/metabolismo , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperparatireoidismo Secundário/complicações , Hiperparatireoidismo Secundário/metabolismo , Hormônio Paratireóideo/metabolismo , Cinacalcete/farmacologia , Cinacalcete/metabolismo
7.
J Med Chem ; 66(14): 9418-9444, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37442941

RESUMO

The calcium sensing receptor (CaSR) plays an important role in maintaining calcium homeostasis. The use of calcimimetic cinacalcet has been established to activate CaSR and normalize hypercalcemia. However, cinacalcet has limitations due to its high cLogP and pKa. A systematic optimization of cinacalcet to reduce its cLogP and pKa yielded compound 23a (LNP1892). Compound 23a showed excellent potency and a favorable pharmacokinetics profile, and lacked the liabilities of cinacalcet, making it a highly differentiated precision calcimimetic. In adenine-diet-induced chronic kidney disease (CKD) models, 23a demonstrated robust and dose-dependent efficacy, as measured by plasma parathyroid hormone (PTH) levels. It also showed an excellent safety profile in animal studies. Phase 1 clinical trials with 23a in healthy volunteers confirmed its excellent safety, tolerability, and effectiveness in lowering PTH levels in a dose-dependent manner, without causing symptomatic hypocalcaemia. Encouraged by these promising results, LNP1892 was taken to a Phase 2 study in CKD patients.


Assuntos
Hiperparatireoidismo Secundário , Insuficiência Renal Crônica , Animais , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Naftalenos/farmacologia , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperparatireoidismo Secundário/etiologia , Hormônio Paratireóideo/uso terapêutico , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico , Cálcio
8.
Calcif Tissue Int ; 112(4): 452-462, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36754901

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2 , Hormônio Paratireóideo , Humanos , Cinacalcete/farmacologia , Cinacalcete/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Osteoblastos/metabolismo , Osteoprotegerina/metabolismo , Glucose/metabolismo , Ligante RANK/metabolismo , Células Cultivadas
9.
Clin Endocrinol (Oxf) ; 98(4): 516-526, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36316798

RESUMO

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.


Assuntos
Amilorida , Hiperparatireoidismo Primário , Humanos , Feminino , Masculino , Eplerenona/uso terapêutico , Cinacalcete/farmacologia , Amilorida/uso terapêutico , Aldosterona , Cálcio , Renina , Hormônio Paratireóideo
10.
Med Sci Monit ; 28: e937338, 2022 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-35941808

RESUMO

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.


Assuntos
Proteômica , Receptores de Detecção de Cálcio , Animais , Encéfalo/metabolismo , Cálcio , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Humanos , Fígado/metabolismo , Espectrometria de Massas , Naftalenos , Hormônio Paratireóideo , Ratos , Ratos Wistar , Receptores de Detecção de Cálcio/metabolismo
11.
Eur Rev Med Pharmacol Sci ; 26(8): 3010-3024, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35503601

RESUMO

OBJECTIVE: The aim of the study was to improve the bioavailability of Cinacalcet hydrochloride (CLC) and enhance its efficacy by the nanoemulsion drug delivery system. MATERIALS AND METHODS: First, cinacalcet hydrochloride-nanoemulsion (CLC-NE) was prepared and optimized through the pseudo ternary phase diagram and central composite design response surface methodology (CCD). The release of CLC-NE in vitro was investigated with four different dissolution media, and the bioavailability of CLC-NE in vivo was studied through beagle dogs. Finally, the pharmacodynamics of CLC-NE was evaluated by the rat model of uremia. RESULTS: Oleic acid, op-10, and PEG-200 were selected as oil phase, emulsifier, and co-emulsifier, respectively. The optimum ratio of oleic acid, op-10, PEG-200, and water was 9.87%, 38.33%, 12.78%, and 39.02%. CLC-NE has similar dissolution rates in different pH media, and the relative bioavailability of CLC-NE was 166.5%. The uremia model showed that CLC-NE could enhance renal function and reduce the excessive phosphorus (P), serum creatinine (Scr), and urea nitrogen (Urea) of model rats, as well as the inhibited increase of fibroblast growth factor-23 (FGF23) and parathyroid hormone (PTH). CONCLUSIONS: The solubility, bioavailability, and pharmacodynamics of CLC can be significantly improved through the nanoemulsion drug delivery system.


Assuntos
Nanopartículas , Uremia , Animais , Disponibilidade Biológica , Cinacalcete/farmacologia , Cães , Sistemas de Liberação de Medicamentos/métodos , Emulsões/química , Feminino , Humanos , Masculino , Nanopartículas/química , Ácido Oleico , Tamanho da Partícula , Ratos , Solubilidade , Ureia
12.
Kidney Int ; 101(6): 1110-1112, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35597590

RESUMO

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.


Assuntos
Síndrome Nefrótica , Podócitos , Animais , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Camundongos , Síndrome Nefrótica/tratamento farmacológico , Síndrome Nefrótica/metabolismo , Podócitos/metabolismo , Proteinúria/tratamento farmacológico , Proteinúria/metabolismo , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo
13.
Int J Mol Sci ; 23(8)2022 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-35457141

RESUMO

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.


Assuntos
Hipocalcemia , Neuroblastoma , Cálcio/metabolismo , Cinacalcete/farmacologia , Humanos , Masculino , Neuroblastoma/tratamento farmacológico , Receptores de Detecção de Cálcio/metabolismo
14.
Iran J Kidney Dis ; 16(2): 135-146, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35489082

RESUMO

INTRODUCTION: Secondary hyperparathyroidism may cause an increase in blood pressure among maintenance hemodialysis (MHD) patients. The objective of this study were to observe the effects of different treatment modalities of hyperparathyroidism on blood pressure among MHD patients with secondary hyperparathyroidism. METHODS: This retrospective cohort study was conducted on 69 patients divided into three groups, based on the therapeutic strategies (parathyroidectomy, n = 22; cinacalcet, n = 14; calcitriol, n = 33). Changes in systolic blood pressure (SBP) and diastolic blood pressure (DBP) from pre- to post-treatment visits at 1st, 3rd, 6th, and 12th month were analyzed by mixed-effects repeated-measures model. Serum levels of the renin-angiotensin system (RAS) mediators (renin and aldosterone), endothelin, and echocardiography were compared before and after one year of treatment within the three groups. RESULTS: Changes in blood pressure were significantly different among the three groups (SBP: P for group < 0.05; DBP: P for group < .05; both P for group × time interaction < .05). SBP and DBP showed a significant downward trend in the surgery group (P for change in SBP < .05, P for change in DBP < .001, adjusted mean change of SBP = -12.16 (-19.70 to -4.62) mmHg and of DBP = -6.82 (-10.58 to -3.06) mmHg in the surgery group on the 12th month). Diastolic BP showed a significant upward trend in the cinacalcet group (P for change in DBP < .05, adjusted mean change of DBP = 6.03 (2.08 to 9.98) mmHg in cinacalcet group in the 12th month). No significant change in BP was observed in the calcitriol group. The levels of serum RAS mediators, endothelin, or cardiac ultrasonography didn't change and almost remained consistent during the treatment course. CONCLUSION: Blood pressure decreased significantly over a year in patients with parathyroidectomy, while DBP increased significantly over time by cinacalcet treatment.  DOI: 10.52547/ijkd.6686.


Assuntos
Hiperparatireoidismo Secundário , Hipertensão , Pressão Sanguínea , Calcitriol , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Humanos , Hiperparatireoidismo Secundário/tratamento farmacológico , Hiperparatireoidismo Secundário/etiologia , Hiperparatireoidismo Secundário/cirurgia , Paratireoidectomia , Diálise Renal/efeitos adversos , Estudos Retrospectivos
15.
J Mol Endocrinol ; 69(1): 243-257, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35318962

RESUMO

Loss-of-function calcium-sensing receptor (CASR) mutations cause mineral metabolism disorders, familial hypocalciuric hypercalcemia, or neonatal severe hyperparathyroidism and increase the risk of femoral fracture, chronic kidney disease, coronary heart disease, and other diseases. In severe cases, CaSR mutations are lethal. Off-label use of the CaSR-positive allosteric modulator (PAM), cinacalcet, corrects hypercalcemia in some patients with CaSR mutations. However, other patients remain unresponsive to cinacalcet, attesting to the need for novel treatments. Here, we compared the effects of cinacalcet to two other clinically approved synthetic CaSR activators, evocalcet and etelcalcetide, as well as a novel PAM, 1-(2,4-dimethylphenyl)-1-(4,5-dimethylthiazol-2-yl)ethan-1-ol (MIPS-VD-836-108) on clinically relevant CaSR mutations. We assessed the compounds in CaSR-expressing HEK293 cells for correction of mutation-induced impairments in intracellular calcium (Ca2+i) mobilization and cell surface expression. While cinacalcet, MIPS-VD-836-108 and evocalcet rescued the signaling of cell surface-expressed mutants, albeit to varying degrees, etelcalcetide was ineffective. Cinacalcet and evocalcet, but not MIPS-VD-836-108 or etelcalcetide, restored the expression of a R680H mutant. However, no compound rescued expression of I81K and C582R mutants or a receptor missing 77 amino acids in the extracellular domain mimicking deletion of CASRexon 5, which impairs CaSR function. These data suggest specific compounds may be clinically effective in some patients with CaSR mutations, but other patients will remain refractory to treatment with currently available CaSR-targeting activators, highlighting the need for new generation drugs to rescue both the signaling and expression of mutant CaSRs.


Assuntos
Hipercalcemia , Hiperparatireoidismo Primário , Cálcio/metabolismo , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Células HEK293 , Humanos , Hipercalcemia/tratamento farmacológico , Hipercalcemia/genética , Hipercalcemia/metabolismo , Hiperparatireoidismo Primário/tratamento farmacológico , Hiperparatireoidismo Primário/genética , Recém-Nascido , Mutação , Medicina de Precisão , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo
16.
Kidney Int ; 101(6): 1186-1199, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35271934

RESUMO

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.


Assuntos
Nefropatias , Podócitos , Animais , Cálcio/metabolismo , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Doxorrubicina/toxicidade , Humanos , Nefropatias/metabolismo , Camundongos , Camundongos Knockout , Podócitos/metabolismo , Proteinúria/induzido quimicamente , Proteinúria/genética , Proteinúria/metabolismo , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo
17.
Chin J Physiol ; 65(6): 277-281, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36588353

RESUMO

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.


Assuntos
Sinalização do Cálcio , Células Endoteliais , Camundongos , Animais , Células Endoteliais/metabolismo , Cinacalcete/farmacologia , Cinacalcete/metabolismo , Espermina/farmacologia , Espermina/metabolismo , Potenciais da Membrana , Cálcio/metabolismo
18.
J Bone Miner Res ; 37(2): 215-225, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34633122

RESUMO

The possible mechanisms underlying the quantitative and qualitative effects of cinacalcet on bone were explored in a chronic kidney disease-mineral and bone disorder (CKD-MBD) mouse model in relation to the influence of the interactions among the osteoclast (OC) endoplasmic reticulum (ER) stress, autophagy and apoptosis pathways on OC differentiation. Body weight and biochemical parameters improved significantly in the CKD + cinacalcet groups compared to the CKD group. Micro-computed tomography (µCT) revealed both cortical and trabecular parameters deteriorated significantly in the CKD group and were reversed by cinacalcet in a dose-dependent manner. Nanoindentation analysis of bone quality proved that both cortical hardness and elastic modulus improved significantly with high dose cinacalcet treatment. In vitro studies revealed that cinacalcet inhibited receptor activator of NF-κB ligand (RANKL)/receptor activator of NF-κB (RANK)-induced OC differentiation in a concentration-dependent manner through a close interaction between activation of caspase-related apoptosis, reversal of OC autophagy through the protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and adenosine monophosphate-activated protein kinase (AMPK) pathways, and attenuation of the OC ER stress/CREBH/NFATc1 signaling pathway. Cinacalcet improves both bone quantity and bone quality in CKD mouse model and inhibits OC differentiation through regulation of the interactions among the apoptosis, ER stress, and autophagy pathways within OCs. © 2021 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Distúrbio Mineral e Ósseo na Doença Renal Crônica , Osteoclastos , Animais , Autofagia , Diferenciação Celular , Distúrbio Mineral e Ósseo na Doença Renal Crônica/tratamento farmacológico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/metabolismo , Cinacalcete/farmacologia , Cinacalcete/uso terapêutico , Estresse do Retículo Endoplasmático , Camundongos , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Microtomografia por Raio-X
19.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34940833

RESUMO

CONTEXT: Parathyroid hormone (PTH) replacement is a promising approach in the management of hypoparathyroidism but long-acting analogues need to be developed. To date, animal models for testing PTH required parathyroidectomy by surgery. We have developed a nonsurgical rodent hypoparathyroid model and tested a delayed-clearance PTH molecule (DC-PTH). OBJECTIVE: The aim of this study was to use cinacalcet to suppress calcium levels in normal rats and to reverse these effects with the administration of PTH or PTH analogues. METHODS: Male Wistar rats were gavaged with either 30 mg/kg cinacalcet-HCl (cinacalcet) or vehicle only. Animals were then dosed with either single or repeated subcutaneous doses of PTH 1-34 or a DC-PTH at 20 nmol/kg. Control animals received vehicle only. Serum samples were analyzed for ionized calcium (iCa), phosphate, PTH, and DC-PTH. A pharmacokinetic-pharmacodynamic (PK-PD) model was built for cinacalcet, PTH 1-34, and DC-PTH using Phoenix64. RESULTS: Cinacalcet reduced iCa levels between 2 and 24 hours, returning to baseline by 72 hours post dose with nadir at 8 hours (analysis of variance P < .001), associated with a fall in rat PTH. For phosphate there was a variable biphasic response. Single-dose PTH abrogated the cinacalcet-induced fall in iCa for up to 2 hours. DC-PTH prevented the fall in iCa from 4 hours post dose and gave a prolonged response, with iCa levels quicker to return to baseline than controls. DC-PTH has a half-life of 11.5 hours, approximately 44 times longer than human PTH 1-34. The PK-PD models defined the reproducible effect of cinacalcet on iCa and that DC-PTH had prolonged biological activity. CONCLUSION: The administration of cinacalcet provides a robust and reproducible nonsurgical animal model of hypoparathyroidism. DC-PTH holds promise for the treatment of hypoparathyroidism in the future.


Assuntos
Cinacalcete/farmacologia , Hipoparatireoidismo/fisiopatologia , Hormônio Paratireóideo/sangue , Animais , Células CHO , Cálcio/química , Cálcio/metabolismo , Cricetulus , Modelos Animais de Doenças , Masculino , Glândulas Paratireoides/fisiopatologia , Hormônio Paratireóideo/química , Paratireoidectomia , Fosfatos/química , Ratos , Ratos Wistar , Resultado do Tratamento
20.
Pharmazie ; 76(10): 511-514, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34620281

RESUMO

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.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Hepatocelular/patologia , Cinacalcete/farmacologia , Humanos , Neoplasias Hepáticas/patologia , Transdução de Sinais , Transativadores , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA