Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 499-503, 2008.
Article in English | WPRIM | ID: wpr-634933

ABSTRACT

In order to investigate the effects of aconitine on [Ca2+] oscillation patterns in cultured myocytes of neonatal rats, fluorescent Ca2+ indicator Fluo-4 NW and laser scanning confocal microscope (LSCM) were used to detect the real-time changes of [Ca2+] oscillation patterns in the cultured myocytes before and after aconitine (1.0 micromol/L) incubation or antiarrhythmic peptide (AAP) and aconitine co-incubation. The results showed under control conditions, [Ca2+] oscillations were irregular but relatively stable, occasionally accompanied by small calcium sparks. After incubation of the cultures with aconitine, high frequency [Ca2+] oscillations emerged in both nuclear and cytoplasmic regions, whereas typical calcium sparks disappeared and the average [Ca2+] in the cytoplasm of the cardiomyocyte did not change significantly. In AAP-treated cultures, intracellular [Ca2+] oscillation also changed, with periodic frequency, increased amplitudes and prolonged duration of calcium sparks. These patterns were not altered significantly by subsequent aconitine incubation. The basal value of [Ca2+] in nuclear region was higher than that in the cytoplasmic region. In the presence or absence of drugs, the [Ca2+] oscillated synchronously in both the nuclear and cytoplasmic regions of the same cardiomyocyte. It was concluded that although oscillating strenuously at high frequency, the average [Ca2+] in the cytoplasm of cardiomyocyte did not change significantly after aconitine incubation, compared to the controls. The observations indicate that aconitine induces the changes in [Ca2+] oscillation frequency other than the Ca2+ overload.


Subject(s)
Aconitine/pharmacology , Animals, Newborn , Calcium Signaling/drug effects , Cells, Cultured , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats, Sprague-Dawley
2.
Biol. Res ; 41(2): 183-195, 2008. ilus, graf
Article in English | LILACS | ID: lil-495753

ABSTRACT

Neurons are a diverse cell type exhibiting hugely different morphologies and neurotransmitter specifications. Their distinctive phenotypes are established during differentiation from pluripotent precursor cells. The signalling pathways that specify the lineage down which neuronal precursor cells differentiate remain to be fully elucidated. Among the many signáis that impinge on the differentiation of neuronal cells, cytosolic calcium (Ca2+) has an important role. However, little is known about the nature of the Ca2+ signáis involved in fate choice in neuronal precursor cells, or their sources. In this study, we show that activation of either muscarinic or platelet-derived growth factor (PDGF) receptors induces a biphasic increase in cytosolic Ca2+ that consists of reléase from intracellular stores followed by sustained entry across the plasma membrane. For both agonists, the prolonged Ca2+ entry occurred via a store-operated pathway that was pharmacologically indistinguishable from Ca2+ entry initiated by thapsigargin. However, muscarinic receptor-activated Ca2+ entry was inhibited by siRNA-mediated knockdown of TRPC6, whereas Ca2+ entry evoked by PDGF was not. These data provide evidence for agonist-specific activation of molecularly distinct store-operated Ca2+ entry pathways, and raise the possibility of privileged communication between these Ca2+ entry pathways and downstream processes.


Subject(s)
Humans , Calcium Channels/drug effects , Methacholine Chloride/pharmacology , Muscarinic Agonists/pharmacology , Neurons/drug effects , Platelet-Derived Growth Factor/pharmacology , Cells, Cultured , Calcium Channels/metabolism , Calcium Signaling/drug effects , Immunoblotting , Neurons/cytology , Neurons/metabolism , Time Factors
3.
Yonsei Medical Journal ; : 311-321, 2008.
Article in English | WPRIM | ID: wpr-30669

ABSTRACT

PURPOSE: The sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA), encoded by ATP2A2, is an essential component for G-protein coupled receptor (GPCR)-dependent Ca(2+) signaling. However, whether the changes in Ca(2+) signaling and Ca(2+) signaling proteins in parotid acinar cells are affected by a partial loss of SERCA2 are not known. MATERIALS AND METHODS: In SERCA2(+/-) mouse parotid gland acinar cells, Ca(2+) signaling, expression levels of Ca(2+) signaling proteins, and amylase secretion were investigated. RESULTS: SERCA2(+/-) mice showed decreased SERCA2 expression and an upregulation of the plasma membrane Ca(2+) ATPase. A partial loss of SERCA2 changed the expression level of 1, 4, 5-tris-inositolphosphate receptors (IP(3)Rs), but the localization and activities of IP3Rs were not altered. In SERCA2(+/-) mice, muscarinic stimulation resulted in greater amylase release, and the expression of synaptotagmin was increased compared to wild type mice. CONCLUSION: These results suggest that a partial loss of SERCA2 affects the expression and activity of Ca(2+) signaling proteins in the parotid gland acini, however, overall Ca(2+) signaling is unchanged.


Subject(s)
Animals , Mice , Amylases/metabolism , Blotting, Western , Calcium/metabolism , Calcium Signaling/drug effects , Carbachol/pharmacology , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice, Knockout , Parotid Gland/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Signal Transduction/drug effects
4.
Journal of Korean Medical Science ; : 290-297, 2007.
Article in English | WPRIM | ID: wpr-148950

ABSTRACT

Normal human epidermal keratinocytes (NHEK) respond to the autocrine activated extracellular signal-regulated kinase (ERK) signaling pathway, which contributes to the survival of keratinocytes. However, during the condition of calcium-induced differentiation, how the autocrine ERK signaling is regulated and affected is poorly understood. The purpose of this study was to understand and to obtain clues to the possible function of the autocrine ERK activation during the calcium-induced differentiation of NHEK. We demonstrated that the autocrine activated ERK was not interrupted by calcium triggering and that it was sustained for at least one day after changing the medium. We also found that the autocrine ERK activation was associated with the expression of cyclin D1 and the cell cycle regulation at the early stage of calcium triggering by treating the cells with the mitogen-activated protein kinase inhibitor PD98059. However, the PD98059 treatment did not have a significant influence on the expression of involucrin and loricrin. In addition, we demonstrated that autocrine ERK activation was associated with protein kinase C and p38MAPK signaling. We suggest that the activation of autocrine ERK is not interrupted by calcium triggering and it might participate in cell growth during the early stage of calcium-induced differentiation in NHEK.


Subject(s)
Humans , Keratinocytes/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Enzyme Activation/drug effects , Dose-Response Relationship, Drug , Cells, Cultured , Cell Differentiation/drug effects , Cell Cycle/drug effects , Calcium Signaling/drug effects , Calcium/administration & dosage , Autocrine Communication/drug effects
5.
Experimental & Molecular Medicine ; : 129-138, 2007.
Article in English | WPRIM | ID: wpr-90620

ABSTRACT

9-cis-retinoic acid (9CRA) plays an important role in the immune response; this includes cytokine production and cell migration. We have previously demonstrated that 9CRA increases expression of chemokine receptors CCR1 and CCR2 in human monocytes. To better understand how 9CRA induces CCR1 and CCR2 expression, we examined the contribution of signaling proteins in human monocytic THP-1 cells. The mRNA and surface protein up-regulation of CCR1 and CCR2 in 9CRA-stimulated cells were weakly blocked by the pretreatment of SB202190, a p38 MAPK inhibitor, and PD98059, an upstream ERK inhibitor. Activation of p38 MAPK and ERK1/2 was induced in both a time and dose-dependent manner after 9CRA stimulation. Both p38 MAPK and ERK1/2 phosphorylation peaked at 2 h after a 100 nM 9CRA treatment. 9CRA increased calcium influx and chemotactic activity in response to CCR1-dependent chemokines, Lkn-1/CCL15, MIP-1alpha/CCL3, and RANTES/CCL5, and the CCR2-specific chemokine, MCP-1/CCL2. Both SB202190 and PD98059 pretreatment diminished the increased calcium mobilization and chemotactic ability due to 9CRA. SB202190 inhibited the expression and functional activities of CCR1 and CCR2 more effectively than did PD98059. Therefore, our results demonstrate that 9CRA transduces the signal through p38 MAPK and ERK1/2 for CCR1 and CCR2 up-regulation, and may regulate the pro-inflammatory process through the p38 MAPK and ERK-dependent signaling pathways.


Subject(s)
Humans , Calcium Signaling/drug effects , Cell Line , Chemokines/pharmacology , Chemotaxis, Leukocyte/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/drug effects , Pyridines/pharmacology , RNA, Messenger/genetics , Receptors, CCR1 , Receptors, CCR2 , Receptors, Chemokine/genetics , Tretinoin/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Experimental & Molecular Medicine ; : 185-194, 2007.
Article in English | WPRIM | ID: wpr-90614

ABSTRACT

Phytosphingosine-1-phosphate (PhS1P) was found to stimulate an intracellular calcium increase via phospholipase C but not pertussis toxin (PTX)- sensitive G-proteins in L2071 mouse fibroblasts. PhS1P also activated ERK and p38 kinase, and these activations by PhS1P were inhibited by PTX. Moreover, PhS1P stimulated the chemotactic migration of L2071 cells via PTX-sensitive Gi protein(s). In addition, the PhS1P-induced chemotactic migration of L2071 cells was also dramatically inhibited by LY294002 and SB203580 (inhibitors of phosphoinositide 3-kinase and p38 kinase, respectively). L2071 cells are known to express four S1P receptors, i.e., S1P1, S1P2, S1P3, and S1P4, and pretreatment with an S1P1 and S1P3 antagonist (VPC 23019) did not affect on PhS1P-induced chemotaxis. This study demonstrates that PhS1P stimulates at least two different signaling cascades, one is a PTX-insensitive but phospholipase C dependent intracellular calcium increase, and the other is a PTX-sensitive chemotactic migration mediated by phosphoinositide 3-kinase and p38 kinase.


Subject(s)
Animals , Humans , Mice , Phosphatidylinositol 3-Kinase/metabolism , Calcium Signaling/drug effects , Chemotaxis/drug effects , Estrenes/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , GTP-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Pyrrolidinones/pharmacology , RNA, Messenger/genetics , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Biol. Res ; 39(3): 521-530, 2006. ilus
Article in English | LILACS | ID: lil-437384

ABSTRACT

An important pool of chelatable zinc is present in the synaptic vesicles of mossy fiber terminals from hippocampal CA3 area, being zinc released following single or repetitive electrical stimulation. Previous studies have suggested different synaptic roles for released mossy fiber zinc, including the inhibition of presynaptic calcium and of postsynaptic N-methyl-D-aspartate (NMDA) and gamma amino-butiric acid (GABA A) receptors. The effect of endogenously released zinc on mossy fiber long-term potentiation (LTP) induction also is not yet established. We have investigated the effect of the permeant zinc chelator N,N,N',N'-tetrakis(2-pyridylmethyl) ethylenediamine (TPEN) on mossy fiber calcium and on synaptic transmission, before and during the application of LTP-inducing stimulation. We have found, using the calcium indicator Fura-2, that single and tetanically-evoked mossy fiber calcium signals are both enhanced in the presence of 20 ìM TPEN, while the single field potentials are unaffected. As expected, no effect was observed on the single calcium signals or field potentials obtained at the CA3-CA1 synapses, from the CA1 area, which has a lower concentration of vesicular zinc. These results support the idea that at the hippocampal mossy fiber synapses, released zinc inhibits presynaptic calcium mechanisms. A higher concentration of TPEN (100 ìM) significantly reduced mossy fiber synaptic transmission but did not prevent the induction of mossy fiber LTP, suggesting that zinc is not required for the formation of this form of LTP.


Subject(s)
Animals , Rats , Calcium Signaling/drug effects , Chelating Agents/pharmacology , Ethylenediamines/pharmacology , Mossy Fibers, Hippocampal/drug effects , Synaptic Transmission/drug effects , Calcium Signaling/physiology , Electric Stimulation , Long-Term Potentiation , Rats, Wistar , Synaptic Transmission/physiology
8.
Biol. Res ; 39(3): 531-539, 2006. ilus
Article in English | LILACS | ID: lil-437385

ABSTRACT

Thymeleatoxin (TMX), an activator of Ca2+-sensitive protein kinase C (cPKC) isoforms, was used to assess the PKC isoform specificity of cholinergic potentiation of glucose (11 mM)-induced pulsatile 5-HT/insulin release (PIR) from single mouse pancreatic islets. TMX (100 nM) and carbachol (Cch, 50 mM) enhanced PIR ~ 3-fold while reducing the underlying [Ca2+]i oscillations (duration and amplitude) by ~ 40-50 percent. Both effects were ablated by the specific PKC inhibitor bisindolylmaleimide and chronic TMX pretreatment. Cch also evoked an initial transient [Ca2+]i rise and surge of 5-HT release, which remained unaffected by chronic TMX pretreatment. It is concluded that the immediate cholinergic responses are insensitive to cPKC. In contrast, specific activation of a cPKC isoform mediates sustained cholinergic potentiation of glucose-induced insulin secretion.


Subject(s)
Animals , Mice , Glucose/metabolism , Insulin , Islets of Langerhans , Phorbol Esters/pharmacology , Protein Kinase C/drug effects , Serotonin/metabolism , Calcium Signaling/drug effects , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Electrochemistry , Fluorometry , Islets of Langerhans/drug effects , Protein Kinase C/metabolism , Pulsatile Flow/drug effects
9.
Experimental & Molecular Medicine ; : 135-144, 2004.
Article in English | WPRIM | ID: wpr-37855

ABSTRACT

Dendritic cells (DCs) play a key role in activating the immune response against invading pathogens as well as dying cells or tumors. Although the immune response can be initiated by the phagocytic activity by DCs, the molecular mechanism involved in this process has not been fully investigated. Trp-Lys-Tyr-Met-Val-Met-NH2 (WKYMVM) stimulates the activation of phospholipase D (PLD) via Ca2+ increase and protein kinase C activation in mouse DC cell line, DC2.4. WKYMVM stimulates the phagocytic activity, which is inhibited in the presence of N-butanol but not t-butanol in DC2.4 cells. Furthermore, the addition of phosphatidic acid, an enzymatic product of PLD activity, enhanced the phagocytic activity in DC2.4 cells. Since at least two of formyl peptide receptor (FPR) family (FPR1 and FPR2) are expressed in DC2.4 as well as in mouse bone marrow-derived dendritic cells, this study suggests that the activation of FPR family by WKYMVM stimulates the PLD activity resulting in phagocytic activity in DC2.4 cells.


Subject(s)
Animals , Mice , 1-Butanol/pharmacology , Bone Marrow Cells/cytology , Calcium Signaling/drug effects , Cell Death/immunology , Cell Line , Communicable Diseases/immunology , Dendritic Cells/immunology , Neoplasms/immunology , Oligopeptides/pharmacology , Phagocytosis/drug effects , Phosphatidic Acids/pharmacology , Phospholipase D/metabolism , Receptors, Formyl Peptide/metabolism , tert-Butyl Alcohol/pharmacology
10.
The Korean Journal of Gastroenterology ; : 519-526, 2003.
Article in Korean | WPRIM | ID: wpr-96871

ABSTRACT

BACKGROUND/AIMS: Pancreatic acini of streptozotocin (STZ)-induced diabetic rats release amylase less than normal acini on cholecystokinin (CCK) stimulation. Pancreatic enzyme secretion has been closely related to the intracellular calcium concentration ([Ca2+]i) of the acinar cell. In the present study, sequential changes of the intracellular calcium signal which probably underlie the altered enzyme secretion in response to CCK-8 were investigated using pancreatic acini from diabetic rats. METHODS: Diabetic rats were prepared by single intravenous injection of STZ (70 mg/kg). Stimulating experiments with CCK-8 were performed 7 days later. Pancreatic acini were isolated by collagenase digestion. Amylase release and [Ca2+]i were measured by colorimethod and calcium imaging, respectively. The geometry of intracellular calcium signal was analyzed. RESULTS: Normal acini exhibited concentration-dependent [Ca2+]i increase and regular oscillatory calcium signal on CCK-8 stimulation. Amylase release was also concentration-dependent. However, diabetic acini showed significantly less [Ca2+]i increase, prolonged time to peak [Ca2+]i, decreased calcium spikes number, and decreased amylase release compared with normal acini. The decreased [Ca2+]i in diabetic acini was restored significantly by insulin treatment. CONCLUSIONS: Relatively decreased amylase release in diabetic pancreatic acini in response to CCK, appears to be associated with altered calcium signal due to insulin deficiency.


Subject(s)
Animals , Rats , Amylases/metabolism , Calcium Signaling/drug effects , Diabetes Mellitus, Experimental/physiopathology , Pancreas/cytology , Rats, Sprague-Dawley , Sincalide/pharmacology
11.
J Biosci ; 2000 Sep; 25(3): 243-52
Article in English | IMSEAR | ID: sea-111177

ABSTRACT

Aggregating Dictyostelium cells release protons when stimulated with cAMP. To find out whether the protons are generated by acidic vesicles or in the cytosol, we permeabilized the cells and found that this did not alter the cAMP-response. Proton efflux in intact cells was inhibited by preincubation with the V-type H(+) ATPase inhibitor concanamycin A and with the plasma membrane H(+) ATPase blocker miconazole. Surprisingly, miconazole also inhibited efflux in permeabilized cells, indicating that this type of H(+) ATPase is present on intracellular vesicles as well. Vesicular acidification was inhibited by miconazole and by concanamycin A, suggesting that the acidic vesicles contain both V-type and P-type H(+) ATPases. Moreover, concanamycin A and miconazole acted in concert, both in intact cells and in vesicles. The mechanism of cAMP-induced Ca2(+)-fluxes involves phospholipase A2 activity. Fatty acids circumvent the plasma membrane and stimulate vesicular Ca2(+)-efflux. Here we show that arachidonic acid elicited H(+)-efflux not only from intact cells but also from acidic vesicles. The target of regulation by arachidonic acid seemed to be the vesicular Ca2(+)-release channel.


Subject(s)
4-Chloro-7-nitrobenzofurazan/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Arachidonic Acid/pharmacology , Calcium Signaling/drug effects , Cyclic AMP/physiology , Dictyostelium/cytology , Fatty Acids/physiology , Filipin/pharmacology , Hydrogen/metabolism , Hydrogen-Ion Concentration , Ion Transport/drug effects , Macrolides , Membrane Proteins/antagonists & inhibitors , Miconazole/pharmacology , Models, Biological , Organelles/drug effects , Phospholipases A/physiology , Phospholipases A2 , Proton-Translocating ATPases/antagonists & inhibitors , Protons
12.
Experimental & Molecular Medicine ; : 137-144, 1998.
Article in English | WPRIM | ID: wpr-35392

ABSTRACT

Oxidative stress appears to be implicated in the pathogenesis of various diseases including alcoholic liver injury. In this study we investigated the mechanism of apoptosis induced by tert-butyl hydroperoxide (TBHP) in HepG2 human hepatoblastoma cells. Treatment with TBHP significantly reduced glutathione content and glutathione reductase activity, and increased glutathione peroxidase activity, indicating that TBHP induced oxidative stress in the HepG2 cells. TBHP also induced reduction of cell viability and DNA fragmentation, a hallmark of apoptosis, in a dose-dependent manner. In addition, TBHP induced a sustained increase in intracellular Ca2+ concentration, which was completely prevented by the extracellular Ca2+ chelation with EGTA. TBHP also induced Mn2+ influx. These results indicate that the intracellular Ca2+ increase by TBHP is exclusively due to Ca2+ influx from the extracellular site. Treatment with either an extracellular (EGTA) or an intracellular Ca2+ chelator (BAPTA/AM) significantly suppressed the TBHP-induced apoptosis. Taken together, these results suggest that TBHP induced the apoptotic cell death in the HepG2 cells and that Ca2+ influx may play an important role in the apoptosis induced by TBHP.


Subject(s)
Humans , Apoptosis/drug effects , Calcium Signaling/drug effects , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Egtazic Acid/analogs & derivatives , Hepatoblastoma/pathology , Hepatoblastoma/metabolism , Hepatoblastoma/drug therapy , Manganese/metabolism , Oxidative Stress/drug effects , Tumor Cells, Cultured , tert-Butylhydroperoxide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL