Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
1.
Braz. J. Pharm. Sci. (Online) ; 58: e20007, 2022. tab, graf
Article in English | LILACS | ID: biblio-1394052

ABSTRACT

Abstract The prolonged entry of large amounts of calcium into the mitochondria through the mitochondrial calcium uniporter complex (MCUC) may cause the permeability transition pore (mPTP) to open, which contributes to the pathogenesis of several diseases. Tissue-specific differences in mPTP opening due to variable expression of MCUC components may contribute to disease outcomes. We designed this study to determine differential mPTP opening in mitochondria isolated from different regions of mouse brain and kidney and to compare it with the expression of MCUC components. mPTP opening was measured using mitochondria isolated from the left/right brain hemispheres (LH/RH, respectively) and from kidney cortex/medulla, while the expression level of MCUC components was assessed from total cellular RNA. Interestingly, LH mitochondria showed less calcium-induced mPTP opening as compared to RH mitochondria at two different calcium concentrations. Conversely, mPTP opening was similar in the renal cortex and renal medulla mitochondria. However, the kidney mitochondria demonstrated bigger and faster mPTP opening as compared to the brain mitochondria. Furthermore, asymmetric mPTP opening in the LH and RH mitochondria was not associated with the expression of MCUC components. In brief, this study demonstrates thus far unreported asymmetric mPTP opening in mouse brain hemispheres that is not associated with the mRNA levels of MCUC components.


Subject(s)
Animals , Male , Female , Mice , Brain , Calcium/agonists , Cerebrum/abnormalities , Mitochondrial Permeability Transition Pore/analysis , Mice , Mitochondria , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Kidney Cortex
2.
Braz. J. Pharm. Sci. (Online) ; 58: e20101, 2022. tab, graf
Article in English | LILACS | ID: biblio-1403732

ABSTRACT

Abstract Ligustrazine is widely used for the treatment of cardiovascular diseases in traditional Chinese medication. It has been reported that Ligustrazine decreases the concentration of intracellular calcium ions (Ca2+); however, the underlying mechanism remains unknown. In the present study, the effect of Ligustrazine on adenosine diphosphate (ADP)-induced platelet aggregation was evaluated using a turbidimetric approach. The changes in concentration of intracellular Ca2+ stimulated by ADP was measured using fluo-4, a fluorescent Ca2+ indicator dye. The mRNA expression of stromal interaction molecule l (STIM1) and Orai1, calcium sensor, was determined using real-time PCR. In addition, the protein expression of STIM1, Orai1, and serum/glucocorticoid-regulated protein kinase 1 (SGK1) was determined using Western blot analysis. The data demonstrated that Ligustrazine significantly suppressed platelet aggregation in a dose-dependent manner and reduced the concentration of intracellular Ca2+ triggered by ADP. Our data showed that Ligustrazine treatment inhibited the expression of STIM1 and Orai1 induced by ADP at both mRNA and protein levels, and suppressed the protein expression of SGK1. Taken together, our data indicated that Ligustrazine suppressed platelet aggregation by partly inhibiting the activities of calcium sensors, thereby suggesting that Ligustrazine may be a promising candidate for the treatment of platelet aggregation.


Subject(s)
Animals , Male , Rats , Protein Kinases , Cardiovascular Diseases/pathology , Platelet Aggregation , Adenosine Diphosphate/pharmacology , Blotting, Western/methods , Calcium/agonists , Asian People/classification , Stromal Interaction Molecules
3.
Cell Rep ; 29(6): 1524-1538.e6, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693893

ABSTRACT

Exercise engages signaling networks to control the release of circulating factors beneficial to health. However, the nature of these networks remains undefined. Using high-throughput phosphoproteomics, we quantify 20,249 phosphorylation sites in skeletal muscle-like myotube cells and monitor their responses to a panel of cell stressors targeting aspects of exercise signaling in vivo. Integrating these in-depth phosphoproteomes with the phosphoproteome of acute aerobic exercise in human skeletal muscle suggests that co-administration of ß-adrenergic and calcium agonists would activate complementary signaling relevant to this exercise context. The phosphoproteome of cells treated with this combination reveals a surprising divergence in signaling from the individual treatments. Remarkably, only the combination treatment promotes multisite phosphorylation of SERBP1, a regulator of Serpine1 mRNA stability, a pro-fibrotic secreted protein. Secretome analysis reveals that the combined treatments decrease secretion of SERPINE1 and other deleterious factors. This study provides a framework for dissecting phosphorylation-based signaling relevant to acute exercise.


Subject(s)
Exercise/physiology , Muscle, Skeletal/metabolism , Phosphoproteins/metabolism , Protein Kinases/metabolism , Proteome/metabolism , Signal Transduction/physiology , Stress, Physiological/genetics , AMP-Activated Protein Kinase Kinases , Adrenergic beta-Agonists/metabolism , Animals , Aripiprazole/metabolism , Aripiprazole/pharmacology , Calcium/agonists , Calcium/metabolism , Drug Interactions , Humans , Isoproterenol/metabolism , Isoproterenol/pharmacology , Mass Spectrometry , Mice , Phosphoproteins/chemistry , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Translocation Systems/genetics , Protein Translocation Systems/metabolism , Proteome/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Stress, Physiological/physiology , Thapsigargin/metabolism , Thapsigargin/pharmacology
4.
JCI Insight ; 52019 04 23.
Article in English | MEDLINE | ID: mdl-31013259

ABSTRACT

Plasma calcium (Ca2+) is maintained by amending the release of parathyroid hormone and through direct effects of the Ca2+ sensing receptor (CaSR) in the renal tubule. Combined, these mechanisms alter intestinal Ca2+ absorption by modulating 1,25-dihydroxy vitamin D3 production, bone resorption, and renal Ca2+ excretion. The CaSR is a therapeutic target in the treatment of secondary hyperparathyroidism and hypocalcemia a common complication of calcimimetic therapy. The CaSR is also expressed in intestinal epithelium, however, a direct role in regulating local intestinal Ca2+ absorption is unknown. Chronic CaSR activation decreased expression of genes involved in Ca2+ absorption. In Ussing chambers, increasing extracellular Ca2+ or basolateral application of the calcimimetic cinacalcet decreased net Ca2+ absorption across intestinal preparations acutely. Conversely, Ca2+ absorption increased with decreasing extracellular Ca2+ concentration. These responses were absent in mice expressing a non-functional TRPV6, TRPV6D541A. Cinacalcet also attenuated Ca2+ fluxes through TRPV6 in Xenopus oocytes when co-expressed with the CaSR. Moreover, the phospholipase C inhibitor, U73122, prevented cinacalcet-mediated inhibition of Ca2+ flux. These results reveal a regulatory pathway whereby activation of the CaSR in the basolateral membrane of the intestine directly attenuates local Ca2+ absorption via TRPV6 to prevent hypercalcemia and help explain how calcimimetics induce hypocalcemia.


Subject(s)
Calcimimetic Agents/adverse effects , Calcium Channels/metabolism , Calcium/metabolism , Intestinal Mucosa/metabolism , Receptors, Calcium-Sensing/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/agonists , Calcium/blood , Calcium Channels/genetics , Cinacalcet/adverse effects , Disease Models, Animal , Estrenes/pharmacology , Female , Gene Knock-In Techniques , Humans , Hypercalcemia/chemically induced , Hypercalcemia/prevention & control , Hyperparathyroidism, Secondary/chemically induced , Hyperparathyroidism, Secondary/drug therapy , Hypocalcemia/chemically induced , Hypocalcemia/drug therapy , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Kidney Tubules/metabolism , Male , Mice , Mice, Transgenic , Oocytes , Parathyroid Hormone/metabolism , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Receptors, Calcium-Sensing/agonists , TRPV Cation Channels/genetics , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Xenopus
5.
Cell Microbiol ; 21(6): e13011, 2019 06.
Article in English | MEDLINE | ID: mdl-30673152

ABSTRACT

Apicomplexan parasites including Toxoplasma gondii and Plasmodium spp. manufacture a complex arsenal of secreted proteins used to interact with and manipulate their host environment. These proteins are organised into three principle exocytotic compartment types according to their functions: micronemes for extracellular attachment and motility, rhoptries for host cell penetration, and dense granules for subsequent manipulation of the host intracellular environment. The order and timing of these events during the parasite's invasion cycle dictates when exocytosis from each compartment occurs. Tight control of compartment secretion is, therefore, an integral part of apicomplexan biology. Control of microneme exocytosis is best understood, where cytosolic intermediate molecular messengers cGMP and Ca2+ act as positive signals. The mechanisms for controlling secretion from rhoptries and dense granules, however, are virtually unknown. Here, we present evidence that dense granule exocytosis is negatively regulated by cytosolic Ca2+ , and we show that this Ca2+ -mediated response is contingent on the function of calcium-dependent protein kinases TgCDPK1 and TgCDPK3. Reciprocal control of micronemes and dense granules provides an elegant solution to the mutually exclusive functions of these exocytotic compartments in parasite invasion cycles and further demonstrates the central role that Ca2+ signalling plays in the invasion biology of apicomplexan parasites.


Subject(s)
Calcium/metabolism , Cytoplasmic Vesicles/metabolism , Organelles/metabolism , Protein Kinases/metabolism , Toxoplasma/metabolism , Calcium/agonists , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cytoplasm/metabolism , Exocytosis/genetics , Fibroblasts/parasitology , Humans , Protein Kinases/genetics , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Toxoplasma/genetics , Toxoplasma/pathogenicity
6.
Free Radic Res ; 52(5): 605-615, 2018 May.
Article in English | MEDLINE | ID: mdl-29580121

ABSTRACT

Chlorogenic acid (CGA) is a phenolic compound with various health-promoting properties, including antioxidant effects and a wide range of antibacterial activities. However, the antibacterial mechanism remains unclear. We investigated the underlying mode of action of CGA against Escherichia coli, which shows bacterial apoptosis-like death. Cells treated with CGA showed apoptotic features such as membrane depolarisation, caspase-like protein expression, increased intracellular Ca2+ levels, phosphatidylserine externalisation, and DNA fragmentation. In contrast to common bacterial apoptosis-like death, which is caused by reactive oxygen species (ROS) accumulation, CGA depleted intracellular ROS. Because ROS are important intracellular signalling molecules, and ROS depletion may affect bacterial intracellular signalling pathways, leading to cell death. To determine whether deficiencies in intracellular ROS cause apoptosis-like death, the cells were treated with H2O2 after CGA treatment. H2O2 restored depleted intracellular ROS levels to similar levels as in untreated cells, and cell viability was increased compared to CGA-treated cells. Moreover, apoptotic features were attenuated in H2O2 post-treated cells. These results demonstrate that CGA induces bacterial apoptosis in E. coli and intracellular ROS depletion is a core regulator in the progression of bacterial apoptosis-like death.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chlorogenic Acid/pharmacology , Escherichia coli Proteins/genetics , Escherichia coli/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Calcium/agonists , Calcium/metabolism , Caspases/genetics , Caspases/metabolism , DNA Fragmentation/drug effects , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression , Hydrogen Peroxide/pharmacology , Microbial Sensitivity Tests , Microbial Viability/drug effects , Molecular Mimicry , Phosphatidylserines/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors
7.
Mol Hum Reprod ; 23(11): 786-802, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29040664

ABSTRACT

STUDY QUESTION: Does coumestrol inhibit proliferation of human placental choriocarcinoma cells? SUMMARY ANSWER: Coumestrol promotes cell death in the choriocarcinoma cells by regulating ERK1/2 MAPK and JNK MAPK signaling pathways and through disruption of Ca2+ and ROS homeostasis. WHAT IS KNOWN ALREADY: A number of patients who suffer from choriocarcinomas fail to survive due to delayed diagnosis or a recurrent tumor and resistance to traditional chemotherapy using platinum-based agents and methotrexate. To overcome these limitations, it is important to discover novel compounds which have no adverse effects yet can inhibit the expression of a target molecule to develop, as a novel therapeutic for prevention and/or treatment of choriocarcinomas. STUDY DESIGN, SIZE, DURATION: Effects of coumestrol on human placental choriocarcinoma cell lines, JAR and JEG3, were assessed in diverse assays in a dose- and time-dependent manner. PARTICIPCANTS/MATERIALS, SETTING, METHODS: Effects of coumestrol on cell proliferation, apoptosis (annexin V expression, propidium iodide staining, TUNEL and invasion assays), mitochondria-mediated apoptosis, production of reactive oxygen species (ROS), lipid peroxidation, glutathione levels and endoplasmic reticulum (ER) stress proteins in JAR and JEG3 cells were determined. Signal transduction pathways in JAR and JEG3 cells in response to coumestrol were determined by western blot analyses. MAIN RESULTS AND THE ROLE OF CHANCE: Results of the present study indicated that coumestrol suppressed proliferation and increased apoptosis in JAR and JEG3 cells by inducing pro-apoptotic proteins, Bax and Bak. In addition, coumestrol increased ROS production, as well as lipid peroxidation and glutathione levels in JAR and JEG3 cells. Moreover, coumestrol-induced depolarization of mitochondrial membrane potential (MMP) and increased cytosolic and mitochondrial Ca2+ levels in JAR and JEG3 cells. Consistent with those results, treatment of JAR and JEG3 cells with a Ca2+ chelator and an inhibitor of IP3 receptor decreased coumestrol-induced depolarization of MMP and increased proliferation in JAR and JEG3 cells. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: A lack of in vivo animal studies is a major limitation of this research. The effectiveness of coumestrol to induce apoptosis of human placental choriocarcinoma cells requires further investigation. WIDER IMPLICATIONS OF THE FINDINGS: Our results indicate that coumestrol induces apoptotic effects on placental choriocarcinoma cells by regulating cell signaling and mitochondrial-mediated functions, with a potential to impair progression of the cancer. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by grants from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (No. HI15C0810 awarded to G.S. and HI17C0929 awarded to W.L.).


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Coumestrol/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Mitochondria/drug effects , Phytoestrogens/pharmacology , Apoptosis/genetics , Calcium/agonists , Calcium/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chorion/drug effects , Chorion/metabolism , Chorion/pathology , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glutathione/metabolism , Humans , Lipid Peroxidation , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , 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 , Signal Transduction , bcl-2 Homologous Antagonist-Killer Protein/agonists , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
10.
Biochim Biophys Acta ; 1863(12): 3096-3105, 2016 12.
Article in English | MEDLINE | ID: mdl-27725190

ABSTRACT

OBJECTIVES: Differentiation of embryonic stem (ES) cells may be regulated by mechanical strain. Herein, signaling molecules underlying mechanical stimulation of vasculogenesis and expression of angiogenesis guidance cues were investigated in ES cell-derived embryoid bodies. METHODS AND RESULTS: Treatment of embryoid bodies with 10% static mechanical strain using a Flexercell strain system significantly increased CD31-positive vascular structures and the angiogenesis guidance molecules plexinB1, ephrin B2, neuropilin1 (NRP1), semaphorin 4D (sem4D) and robo4 as well as vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2) and platelet-derived growth factor-BB (PDGF-BB) as evaluated by Western blot and real time RT-PCR. In contrast ephrin type 4 receptor B (EphB4) expression was down-regulated upon mechanical strain, indicating an arterial-type differentiation. Robo1 protein expression was modestly increased with no change in mRNA expression. Mechanical strain increased intracellular calcium as well as reactive oxygen species (ROS) and nitric oxide (NO). Mechanical strain-induced vasculogenesis was abolished by the NOS inhibitor L-NAME, the NADPH oxidase inhibitor VAS2870, upon chelation of intracellular calcium by BAPTA as well as upon siRNA inactivation of ephrin B2, NRP1 and robo4. BAPTA blunted the strain-induced expression of angiogenic growth factors, the increase in NO and ROS as well as the expression of NRP1, sem4D and plexinB1, whereas ephrin B2, EphB4 as well as robo1 and robo4 expression were not impaired. CONCLUSIONS: Mechanical strain stimulates vasculogenesis of ES cells by the intracellular messengers ROS, NO and calcium as well as by upregulation of angiogenesis guidance molecules and the angiogenic growth factors VEGF, FGF-2 and PDGF-BB.


Subject(s)
Calcium/metabolism , Embryoid Bodies/metabolism , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Becaplermin , Benzoxazoles/pharmacology , Biomechanical Phenomena , Calcium/agonists , Cell Differentiation , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Embryo, Mammalian , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Ephrin-B2/antagonists & inhibitors , Ephrin-B2/genetics , Ephrin-B2/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/genetics , Neuropilin-1/metabolism , Nitric Oxide/agonists , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Semaphorins/genetics
11.
Cell Physiol Biochem ; 39(4): 1404-20, 2016.
Article in English | MEDLINE | ID: mdl-27606689

ABSTRACT

BACKGROUND/AIMS: Resveratrol and its derivate piceatannol are known to induce cancer cell-specific cell death. While multiple mechanisms of actions have been described including the inhibition of ATP synthase, changes in mitochondrial membrane potential and ROS levels, the exact mechanisms of cancer specificity of these polyphenols remain unclear. This paper is designed to reveal the molecular basis of the cancer-specific initiation of cell death by resveratrol and piceatannol. METHODS: The two cancer cell lines EA.hy926 and HeLa, and somatic short-term cultured HUVEC were used. Cell viability and caspase 3/7 activity were tested. Mitochondrial, cytosolic and endoplasmic reticulum Ca2+ as well as cytosolic and mitochondrial ATP levels were measured using single cell fluorescence microscopy and respective genetically-encoded sensors. Mitochondria-ER junctions were analyzed applying super-resolution SIM and ImageJ-based image analysis. RESULTS: Resveratrol and piceatannol selectively trigger death in cancer but not somatic cells. Hence, these polyphenols strongly enhanced mitochondrial Ca2+ uptake in cancer exclusively. Resveratrol and piceatannol predominantly affect mitochondrial but not cytosolic ATP content that yields in a reduced SERCA activity. Decreased SERCA activity and the strongly enriched tethering of the ER and mitochondria in cancer cells result in an enhanced MCU/Letm1-dependent mitochondrial Ca2+ uptake upon intracellular Ca2+ release exclusively in cancer cells. Accordingly, resveratrol/piceatannol-induced cancer cell death could be prevented by siRNA-mediated knock-down of MCU and Letm1. CONCLUSIONS: Because their greatly enriched ER-mitochondria tethering, cancer cells are highly susceptible for resveratrol/piceatannol-induced reduction of SERCA activity to yield mitochondrial Ca2+ overload and subsequent cancer cell death.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Calcium/agonists , Endoplasmic Reticulum/drug effects , Mitochondria/drug effects , Stilbenes/pharmacology , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Endoplasmic Reticulum/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Ion Transport/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Organ Specificity , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Resveratrol , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
12.
Molecules ; 21(7)2016 Jul 08.
Article in English | MEDLINE | ID: mdl-27399663

ABSTRACT

The human enteroendocrine L cell line NCI-H716, expressing taste receptors and taste signaling elements, constitutes a unique model for the studies of cellular responses to glucose, appetite regulation, gastrointestinal motility, and insulin secretion. Targeting these gut taste receptors may provide novel treatments for diabetes and obesity. However, NCI-H716 cells are cultured in suspension and tend to form multicellular aggregates, preventing high-throughput calcium imaging due to interferences caused by laborious immobilization and stimulus delivery procedures. Here, we have developed an automated microfluidic platform that is capable of trapping more than 500 single cells into microwells with a loading efficiency of 77% within two minutes, delivering multiple chemical stimuli and performing calcium imaging with enhanced spatial and temporal resolutions when compared to bath perfusion systems. Results revealed the presence of heterogeneity in cellular responses to the type, concentration, and order of applied sweet and bitter stimuli. Sucralose and denatonium benzoate elicited robust increases in the intracellular Ca(2+) concentration. However, glucose evoked a rapid elevation of intracellular Ca(2+) followed by reduced responses to subsequent glucose stimulation. Using Gymnema sylvestre as a blocking agent for the sweet taste receptor confirmed that different taste receptors were utilized for sweet and bitter tastes. This automated microfluidic platform is cost-effective, easy to fabricate and operate, and may be generally applicable for high-throughput and high-content single-cell analysis and drug screening.


Subject(s)
High-Throughput Screening Assays/methods , Lab-On-A-Chip Devices , Receptors, G-Protein-Coupled/metabolism , Single-Cell Analysis/methods , Taste Perception/drug effects , Time-Lapse Imaging/methods , Calcium/agonists , Calcium/metabolism , Calcium Signaling/drug effects , Cell Line , Enteroendocrine Cells/cytology , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Glucose/antagonists & inhibitors , Glucose/pharmacology , Gymnema sylvestre/chemistry , High-Throughput Screening Assays/instrumentation , Humans , Models, Biological , Plant Extracts/chemistry , Plant Extracts/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Single-Cell Analysis/instrumentation , Sucrose/analogs & derivatives , Sucrose/pharmacology , Taste/drug effects , Taste/physiology , Taste Perception/physiology , Time-Lapse Imaging/instrumentation
13.
J Biochem Mol Toxicol ; 30(11): 539-547, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27252039

ABSTRACT

The effect of protriptyline on Ca2+ physiology in human hepatoma is unclear. This study explored the effect of protriptyline on [Ca2+ ]i and cytotoxicity in HepG2 human hepatoma cells. Protriptyline (50-150 µM) evoked [Ca2+ ]i rises. The Ca2+ entry was inhibited by removal of Ca2+ . Protriptyline-induced Ca2+ entry was confirmed by Mn2+ -induced quench of fura-2 fluorescence. Except nifedipine, econazole, SKF96365, GF109203X, and phorbol 12-myristate 13 acetate did not inhibit Ca2+ entry. Treatment with the endoplasmic reticulum Ca2+ pump inhibitor 2,5-di-tert-butylhydroquinone (BHQ) inhibited 40% of protriptyline-induced response. Treatment with protriptyline abolished BHQ-induced response. Inhibition of phospholipase C (PLC) suppressed protriptyline-evoked response by 70%. At 20-40 µM, protriptyline killed cells which was not reversed by the Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM). Together, in HepG2 cells, protriptyline induced [Ca2+ ]i rises that involved Ca2+ entry through nifedipine-sensitive Ca2+ channels and PLC-dependent Ca2+ release from endoplasmic reticulum. Protriptyline induced Ca2+ -independent cell death.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Calcium/metabolism , Cell Death/drug effects , Protriptyline/pharmacology , Calcium/agonists , Cations, Divalent , Econazole/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Fluorescent Dyes , Fura-2 , Hep G2 Cells , Humans , Hydroquinones/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Ion Transport/drug effects , Kinetics , Maleimides/pharmacology , Manganese/pharmacology , Nifedipine/pharmacology , Protriptyline/antagonists & inhibitors , Spectrometry, Fluorescence , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
14.
PLoS One ; 10(11): e0143243, 2015.
Article in English | MEDLINE | ID: mdl-26600013

ABSTRACT

The uterine myometrium (UT-myo) is a therapeutic target for preterm labor, labor induction, and postpartum hemorrhage. Stimulation of intracellular Ca2+-release in UT-myo cells by oxytocin is a final pathway controlling myometrial contractions. The goal of this study was to develop a dual-addition assay for high-throughput screening of small molecular compounds, which could regulate Ca2+-mobilization in UT-myo cells, and hence, myometrial contractions. Primary murine UT-myo cells in 384-well plates were loaded with a Ca2+-sensitive fluorescent probe, and then screened for inducers of Ca2+-mobilization and inhibitors of oxytocin-induced Ca2+-mobilization. The assay exhibited robust screening statistics (Z´ = 0.73), DMSO-tolerance, and was validated for high-throughput screening against 2,727 small molecules from the Spectrum, NIH Clinical I and II collections of well-annotated compounds. The screen revealed a hit-rate of 1.80% for agonist and 1.39% for antagonist compounds. Concentration-dependent responses of hit-compounds demonstrated an EC50 less than 10µM for 21 hit-antagonist compounds, compared to only 7 hit-agonist compounds. Subsequent studies focused on hit-antagonist compounds. Based on the percent inhibition and functional annotation analyses, we selected 4 confirmed hit-antagonist compounds (benzbromarone, dipyridamole, fenoterol hydrobromide and nisoldipine) for further analysis. Using an ex vivo isometric contractility assay, each compound significantly inhibited uterine contractility, at different potencies (IC50). Overall, these results demonstrate for the first time that high-throughput small-molecules screening of myometrial Ca2+-mobilization is an ideal primary approach for discovering modulators of uterine contractility.


Subject(s)
Calcium/metabolism , Drug Discovery , High-Throughput Screening Assays , Myometrium/metabolism , Uterine Contraction , Uterus/metabolism , Uterus/physiology , Animals , Calcium/agonists , Calcium Channel Blockers/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Mice , Myometrium/cytology , Myometrium/drug effects , Oxytocin/pharmacology , Pregnancy , Primary Cell Culture , Reproducibility of Results , Uterus/drug effects
15.
PLoS One ; 10(9): e0138384, 2015.
Article in English | MEDLINE | ID: mdl-26375676

ABSTRACT

Idiopathic pulmonary arterial hypertension (IPAH) is a rare and progressive disease of unknown pathogenesis. Vascular remodeling due to excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs) is a critical pathogenic event that leads to early morbidity and mortality. The excessive cell proliferation is closely linked to the augmented Ca2+ signaling in PASMCs. More recently, we have shown by an siRNA knockdown method that the Ca2+-sensing receptor (CaSR) is upregulated in PASMCs from IPAH patients, involved in the enhanced Ca2+ response and subsequent excessive cell proliferation. In this study, we examined whether pharmacological blockade of CaSR attenuated the excessive proliferation of PASMCs from IPAH patients by MTT assay. The proliferation rate of PASMCs from IPAH patients was much higher (~1.5-fold) than that of PASMCs from normal subjects and patients with chronic thromboembolic pulmonary hypertension (CTEPH). Treatment with NPS2143, an antagonist of CaSR or calcilytic, clearly suppressed the cell proliferation in a concentration-dependent manner (IC50 = 2.64 µM) in IPAH-PASMCs, but not in normal and CTEPH PASMCs. Another calcilytic, Calhex 231, which is structurally unrelated to NPS2143, also concentration-dependently inhibited the excessive proliferation of IPAH-PASMCs (IC50 = 1.89 µM). In contrast, R568, an activator of CaSR or calcimimetic, significantly facilitated the proliferation of IPAH-PASMCs (EC50 = 0.33 µM). Similar results were obtained by BrdU incorporation assay. These results reveal that the excessive PASMC proliferation was modulated by pharmacological tools of CaSR, showing us that calcilytics are useful for a novel therapeutic approach for pulmonary arterial hypertension.


Subject(s)
Cell Proliferation/drug effects , Familial Primary Pulmonary Hypertension/pathology , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology , Pulmonary Embolism/pathology , Receptors, Calcium-Sensing/antagonists & inhibitors , Thromboembolism/pathology , Aniline Compounds/pharmacology , Benzamides/pharmacology , Calcium/agonists , Case-Control Studies , Cells, Cultured , Chronic Disease , Cyclohexylamines/pharmacology , Familial Primary Pulmonary Hypertension/drug therapy , Familial Primary Pulmonary Hypertension/metabolism , Humans , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Naphthalenes/pharmacology , Phenethylamines , Propylamines , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Embolism/drug therapy , Pulmonary Embolism/metabolism , Thromboembolism/drug therapy , Thromboembolism/metabolism
16.
Peptides ; 65: 34-45, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25645492

ABSTRACT

Ghrelin, a gut and brain peptide, has recently been shown to be involved in motivated behavior and regulation of the sleep and wakefulness cycle. The laterodorsal tegmental nucleus (LDT) is involved in appetitive behavior and control of the arousal state of an organism, and accordingly, behavioral actions of ghrelin could be mediated by direct cellular actions within this nucleus. Consistent with this interpretation, postsynaptically mediated depolarizing membrane actions of ghrelin on LDT neurons have been reported. Direct actions were ascribed solely to closure of a potassium conductance however this peptide has been shown in other cell types to lead to rises in calcium via release of calcium from intracellular stores. To determine whether ghrelin induced intracellular calcium rises in mouse LDT neurons, we conducted calcium imaging studies in LDT brain slices loaded with the calcium binding dye, Fura-2AM. Ghrelin elicited TTX-insensitive changes in dF/F indicative of rises in calcium, and a portion of these rises were independent of membrane depolarization, as they persisted in conditions of high extracellular potassium solutions and were found to involve SERCA-pump mediated intracellular calcium stores. Involvement of the ghrelin receptor (GHR-S) in these actions was confirmed. Taken together with other studies, our data suggest that ghrelin has multiple cellular actions on LDT cells. Ghrelin's induction of calcium via intracellular release in the LDT could play a role in behavioral actions of this peptide as the LDT governs processes involved in stimulation of motivated behavior and control of cortical arousal.


Subject(s)
Action Potentials/drug effects , Calcium/metabolism , Ghrelin/pharmacology , Neurons/drug effects , Tegmentum Mesencephali/drug effects , Action Potentials/physiology , Animals , Animals, Newborn , Appetite/physiology , Arousal/physiology , Calcium/agonists , Fluorescent Dyes , Fura-2/analogs & derivatives , Mice , Microtomy , Molecular Imaging , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Potassium/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Tegmentum Mesencephali/cytology , Tegmentum Mesencephali/metabolism , Tetrodotoxin/pharmacology , Tissue Culture Techniques
17.
Neuromolecular Med ; 16(4): 645-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24948534

ABSTRACT

The excess vascular endothelial growth factor (VEGF) produced in the Alzheimer's disease (AD) brain can harm neurons, blood vessels, and other components of the neurovascular units (NVUs). But could astrocytes partaking in networks of astrocyte-neuron teams and connected to blood vessels of NVUs contribute to VEGF production? We have shown with cultured cerebral cortical normal (i.e., untransformed) adult human astrocytes (NAHAs) that exogenous amyloid-ß peptides (Aßs) stimulate the astrocytes to make and secrete large amounts of Aßs and nitric oxide by a mechanism mediated through the calcium-sensing receptor (CaSR). Here, we report that exogenous Aßs stimulate the NAHAs to produce and secrete even VEGF-A through a CaSR-mediated mechanism. This is indicated by the ability of Aßs to specifically bind the CaSR, and the capability of a CaSR activator, the "calcimimetic" NPS R-568, to imitate, and of the CaSR antagonist, "calcilytic" NPS 2143, to inhibit, the Aßs stimulation of VEGF-A production and secretion by the NAHAs. Thus, Aßs that accumulate in the AD brain may make the astrocytes that envelop and functionally collaborate with neurons into multi-agent AD-driving "machines" via a CaSR signaling mechanism(s). These observations suggest the possibility that CaSR allosteric antagonists such as NPS 2143 might impede AD progression.


Subject(s)
Amyloid beta-Peptides/pharmacology , Astrocytes/drug effects , Peptide Fragments/pharmacology , Receptors, Calcium-Sensing/physiology , Vascular Endothelial Growth Factor A/biosynthesis , Adult , Allosteric Regulation , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Aniline Compounds/pharmacology , Astrocytes/metabolism , Calcium/agonists , Cell Communication , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Naphthalenes/pharmacology , Neurons/metabolism , Nitric Oxide/metabolism , Phenethylamines , Propylamines , Protein Binding , Receptors, Calcium-Sensing/antagonists & inhibitors , Temporal Lobe/cytology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
Am J Physiol Cell Physiol ; 306(3): C298-306, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24336654

ABSTRACT

To clarify the mechanism(s) underlying intracellular Ca(2+) concentration ([Ca(2+)]i) oscillations induced by an elevation in extracellular Ca(2+) concentration ([Ca(2+)]e) via the extracellular Ca(2+)-sensing receptor (CaR), we analyzed the pattern of [Ca(2+)]i response in multiple (2,303) individual HEK-293 cells transfected with the human CaR. An increase in the [Ca(2+)]e from 1.5 to 3 mM produced oscillatory fluctuations in [Ca(2+)]i in 70% of the cell population. To determine the role of PKC in the generation of [Ca(2+)]i oscillations, cells were exposed to increasing concentrations (0.5-5 µM) of the preferential PKC inhibitor Ro-31-8220 before stimulation by extracellular Ca(2+). Ro-31-8220 at 3-5 µM completely eliminated the [Ca(2+)]e-evoked [Ca(2+)]i oscillations and transformed the pattern to a peak and sustained plateau response. Treatment with other broad PKC inhibitors, including GFI or Gö6983, produced an identical response. Similarly, treatment with Ro-31-8220 or GFI eliminated [Ca(2+)]e-evoked [Ca(2+)]i oscillations in colon-derived SW-480 cells expressing the CaR. Treatment with inhibitors targeting classic PKCs, including Gö6976 and Ro-32-0432 as well as small interfering RNA-mediated knockdown of PKCα, strikingly reduced the proportion of cell displaying [Ca(2+)]e-evoked [Ca(2+)]i oscillations. Furthermore, none of the cells analyzed expressing a CaR mutant in which the major PKC phosphorylation site Thr(888) was converted to alanine (CaRT888A) showed [Ca(2+)]i oscillations after CaR activation. Our results show that [Ca(2+)]i oscillations induced by activation of the CaR in response to an increase in extracellular Ca(2+) or exposure to the calcimimetic R-568 result from negative feedback involving PKCα-mediated phosphorylation of the CaR at Thr(888).


Subject(s)
Calcium Signaling , Calcium/metabolism , Protein Kinase C-alpha/metabolism , Receptors, Calcium-Sensing/metabolism , Aniline Compounds/pharmacology , Calcium/agonists , Cell Line , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Indoles/pharmacology , Ion Transport , Maleimides/pharmacology , Phenethylamines , Phosphorylation , Propylamines , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering
19.
PLoS One ; 8(9): e73816, 2013.
Article in English | MEDLINE | ID: mdl-24040082

ABSTRACT

Amniotic fluid-derived stem (AFS) cells have been identified as a promising source for cell therapy applications in bone traumatic and degenerative damage. Calcium Sensing Receptor (CaSR), a G protein-coupled receptor able to bind calcium ions, plays a physiological role in regulating bone metabolism. It is expressed in different kinds of cells, as well as in some stem cells. The bone CaSR could potentially be targeted by allosteric modulators, in particular by agonists such as calcimimetic R-568, which may potentially be helpful for the treatment of bone disease. The aim of our study was first to investigate the presence of CaSR in ovine Amniotic Fluid Mesenchymal Stem Cells (oAFMSCs) and then the potential role of calcimimetics in in vitro osteogenesis. oAFMSCs were isolated, characterized and analyzed to examine the possible presence of CaSR by western blotting and flow cytometry analysis. Once we had demonstrated CaSR expression, we worked out that 1 µM R-568 was the optimal and effective concentration by cell viability test (MTT), cell number, Alkaline Phosphatase (ALP) and Alizarin Red S (ARS) assays. Interestingly, we observed that basal diffuse CaSR expression in oAFMSCs increased at the membrane when cells were treated with R-568 (1 µM), potentially resulting in activation of the receptor. This was associated with significantly increased cell mineralization (ALP and ARS staining) and augmented intracellular calcium and Inositol trisphosphate (IP3) levels, thus demonstrating a potential role for calcimimetics during osteogenic differentiation. Calhex-231, a CaSR allosteric inhibitor, totally reversed R-568 induced mineralization. Taken together, our results demonstrate for the first time that CaSR is expressed in oAFMSCs and that calcimimetic R-568, possibly through CaSR activation, can significantly improve the osteogenic process. Hence, our study may provide useful information on the mechanisms regulating osteogenesis in oAFMSCs, perhaps prompting the use of calcimimetics in bone regenerative medicine.


Subject(s)
Amniotic Fluid/cytology , Aniline Compounds/pharmacology , Cell Differentiation/drug effects , Mesenchymal Stem Cells/metabolism , Receptors, Calcium-Sensing/metabolism , Alkaline Phosphatase/metabolism , Aniline Compounds/chemistry , Animals , Benzamides/pharmacology , Blotting, Western , Calcium/agonists , Calcium/metabolism , Cell Membrane/metabolism , Cell Survival/drug effects , Cells, Cultured , Cyclohexylamines/pharmacology , Dose-Response Relationship, Drug , Female , Flow Cytometry , Inositol 1,4,5-Trisphosphate/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Phenethylamines , Propylamines , Receptors, Calcium-Sensing/antagonists & inhibitors , Sheep , Stereoisomerism
20.
Pharmacol Res ; 76: 106-18, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23911580

ABSTRACT

Although calcimimetics were developed to block parathyroid hormone synthesis, some reports suggest that they may also reduce blood pressure by unknown mechanisms. Calcimimetic-induced changes in the synthesis of endothelial vasoactive factors could be involved. Wistar rats were treated with the calcimimetic R-568, and systolic blood pressure (SBP) was registered with a tail-cuff sphygmomanometer, the content of endothelial nitric oxide synthase (eNOS) and endothelin-converting enzyme (ECE-1) in tissue was evaluated by immunohistochemistry and Western blot, circulating levels of endothelin-1 (ET-1) were measured by ELISA. R-568 reduced SBP and circulating levels of ET-1, without changes in eNOS expression. In contrast, R-568 increased the lung and vascular content of ECE-1. In order to analyze the mechanisms involved, we studied the effect of R-568 on human endothelial cells. R-568 did not modify neither eNOS protein content nor pre-pro-ET-1 mRNA expression, but increased ECE-1 protein content, and decreased ET-1 synthesis and ECE-1 activity. The inhibition of ECE-1 activity was very strong, similar to the classic ECE inhibitor phosphoramidon, the addition of exogenous zinc restored enzymatic activity. Moreover, the amount of zinc in immunoprecipitated ECE from R-568 treated cells was 3-fold less than in control cells. In conclusion, R-568 inhibits ECE by expelling zinc from the enzyme, with the subsequent decrease in enzymatic activity and reducing circulating levels of ET-1, which may be responsible for the lower SBP observed in R-568-treated rats. This descent would be partially compensated by the increased synthesis of the ECE-1 itself, and by other homeostatic mechanisms that regulate SBP.


Subject(s)
Aniline Compounds/pharmacology , Aspartic Acid Endopeptidases/metabolism , Calcium/agonists , Metalloendopeptidases/metabolism , Animals , Aspartic Acid Endopeptidases/analysis , Blood Pressure/drug effects , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelin-1/blood , Endothelin-1/metabolism , Endothelin-Converting Enzymes , Humans , Male , Metalloendopeptidases/analysis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/metabolism , Phenethylamines , Propylamines , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...