Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 576: 73-79, 2021 10 22.
Article in English | MEDLINE | ID: mdl-34482026

ABSTRACT

OBJECTIVES: MicroRNA-199b-3p (miR-199b-3p) plays a crucial role in the malignant development of various cancers, but little known in prostate cancer (PCa). The aim of our study was to demonstrate the function of miR-199b-3p in PCa. METHODS: Quantitative real-time polymerase chain reaction (RT-qPCR) was used to detect miR-199b-3p expression in PCa and benign prostatic hyperplasia (BPH) tissue samples. In addition, we examined the relationship between the poor prognosis in PCa and miR-199b-3p. Western blot was used to analyze the expression of Phospholipase Cε (PLCε). CCK8 and colony-forming assays were applied to detect the proliferation of PCa. EdU assay is used to detect PCa cells uptake of EdU. Luciferase reporter assay was applied to analyze the binding between miR-199b-3p and PLCε. RESULTS: It has been shown that miR-199b-3p in PCa was significantly lower than that in benign prostatic hyperplasia and correlated with poor prognosis. Meanwhile, upregulation of miR-199b-3p can prominently inhibit the proliferation of PCa cells, while its down-regulation triggered opposite result. PLCε was identified as the downstream binding target gene and negatively associated with that of miR-199b-3p. CONCLUSION: miR-199b-3p suppresses malignant proliferation by inhibiting PLCε in prostate cancer in vitro and vivo.


Subject(s)
MicroRNAs/genetics , Phosphoinositide Phospholipase C/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Down-Regulation , Heterografts , Humans , Male , Mice , Mice, Nude , MicroRNAs/metabolism , Middle Aged , Phosphoinositide Phospholipase C/genetics , Phosphoinositide Phospholipase C/metabolism , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
2.
Neuropharmacology ; 187: 108492, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33582153

ABSTRACT

In this work, modulation by orexin-A of the release of glutamate and GABA from bipolar and amacrine cells respectively was studied by examining the effects of the neuropeptide on miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of rat retinal ganglion cells (GCs). Using RNAscope in situ hybridization in combination with immunohistochemistry, we showed positive signals for orexin receptor-1 (OX1R) mRNA in the bipolar cell terminals and those for orexin receptor-2 (OX2R) mRNA in the amacrine cell terminals. With whole-cell patch-clamp recordings in rat retinal slices, we demonstrated that application of orexin-A reduced the interevent interval of mEPSCs of GCs through OX1R. However, it increased the interevent interval of mIPSCs, mediated by GABAA receptors, through OX2R. Furthermore, orexin-A-induced reduction of mEPSC interevent interval was abolished by the application of PI-PLC inhibitors or PKC inhibitors. In contrast, orexin-A-induced increase of GABAergic mIPSC interevent interval was mimicked by 8-Br-cAMP or an adenylyl cyclase activator, but was eliminated by PKA antagonists. Finally, application of nimodipine, an L-type Ca2+ channel blocker, increased both mEPSC and mIPSC interevent interval, and co-application of orexin-A no longer changed the mEPSCs and mIPSCs. We conclude that orexin-A increases presynaptic glutamate release onto GCs by activating L-type Ca2+ channels in bipolar cells, a process that is mediated by an OX1R/PI-PLC/PKC signaling pathway. However, orexin-A decreases presynaptic GABA release onto GCs by inhibiting L-type Ca2+ channels in amacrine cells, a process that is mediated by an OX2R/cAMP-PKA signaling pathway.


Subject(s)
Amacrine Cells/metabolism , Excitatory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/genetics , Orexin Receptors/genetics , Orexins/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism , Synaptic Transmission/genetics , Amacrine Cells/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Orexin Receptors/metabolism , Orexins/pharmacology , Patch-Clamp Techniques , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Retinal Bipolar Cells/drug effects , Retinal Ganglion Cells/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
3.
J Cell Biochem ; 120(6): 10678-10687, 2019 06.
Article in English | MEDLINE | ID: mdl-30652343

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the leading pathologic type in China. miR-145 has been reported to be downregulated in multiple tumors. This study was aimed to investigate the role of miR-145 in ESCC. miR-145 expression was investigated in 65 ESCC samples as well as four ESCC cell lines by quantitative real-time polymerase chain reaction (qRT-PCR). Targetscan 6.2 website (http://www.targetscan.org/) was used to predict the targets of miR-145. Expression of phospholipase C epsilon 1 (PLCE1) messenger RNA and protein was detected by qRT-PCR or Western blot. MTT and wound healing assay were conducted to explore the effects of miR-145 on the proliferation and migration of ESCC cell lines, respectively. miR-145 was significantly decreased in ESCC tissues. An inverse correlation between miR-145 and invasion depth and TNM stage were observed. PLCE1 was a direct target of miR-145, and the expression of PLCE1 was inversely correlated with miR-145 expression in ESCC tissues. In addition, overexpression of miR-145 suppressed cell proliferation and migration in ESCC cells. The enforced expression of PLCE1 partially reversed the suppressive effect of miR-145. These results prove that miR-145 may perform as a tumor suppressor in ESCC by targeting PLCE1.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Phosphoinositide Phospholipase C/genetics , Aged , Base Sequence , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/surgery , Female , Humans , Lymphatic Metastasis , Male , MicroRNAs/metabolism , Middle Aged , Neoplasm Staging , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
4.
J Biol Chem ; 292(24): 9967-9974, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28442571

ABSTRACT

Beta adrenergic receptors (ßARs) are G-protein-coupled receptors essential for physiological responses to the hormones/neurotransmitters epinephrine and norepinephrine which are found in the nervous system and throughout the body. They are the targets of numerous widely used drugs, especially in the case of the most extensively studied ßAR, ß2AR, whose ligands are used for asthma and cardiovascular disease. ßARs signal through Gαs G-proteins and via activation of adenylyl cyclase and cAMP-dependent protein kinase, but some alternative downstream pathways have also been proposed that could be important for understanding normal physiological functioning of ßAR signaling and its disruption in disease. Using fluorescence-based Ca2+ flux assays combined with pharmacology and gene knock-out methods, we discovered a previously unrecognized endogenous pathway in HEK-293 cells whereby ß2AR activation leads to robust Ca2+ mobilization from intracellular stores via activation of phospholipase C and opening of inositol trisphosphate (InsP3) receptors. This pathway did not involve cAMP, Gαs, or Gαi or the participation of the other members of the canonical ß2AR signaling cascade and, therefore, constitutes a novel signaling mechanism for this receptor. This newly uncovered mechanism for Ca2+ mobilization by ß2AR has broad implications for adrenergic signaling, cross-talk with other signaling pathways, and the effects of ßAR-directed drugs.


Subject(s)
Calcium Signaling , Endoplasmic Reticulum/metabolism , Epinephrine/metabolism , Inositol 1,4,5-Trisphosphate Receptors/agonists , Norepinephrine/metabolism , Phosphoinositide Phospholipase C/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-Agonists/pharmacology , Boron Compounds/pharmacology , CRISPR-Cas Systems , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Isoproterenol/pharmacology , Kinetics , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/chemistry , Pyrrolidinones/pharmacology , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/pharmacology
5.
Toxicol Lett ; 258: 237-248, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27313092

ABSTRACT

Sulfite is a compound commonly used as preservative in foods and pharmaceuticals. Many studies have examined the neurotoxicity of sulfite, but its effect on neuronal calcium homeostasis has not yet been reported. Here, we observed the effect of sulfite on the cytosolic free calcium concentration ([Ca(2+)]i) in cultured cortical neurons using Fura-2/AM based calcium imaging technique. Sulfite (250-1000µM) caused a sustained increase in [Ca(2+)]i in the neurons via a dose-dependent manner. In Ca(2+)-free solution, sulfite failed to increase [Ca(2+)]i. After the depletion of the intracellular calcium store, the effect of sulfite on the [Ca(2+)]i was largely abolished. Pharmacological inhibition of phospholipase C (PLC)-inositol 1,4,5-triphosphate (IP3) signaling pathway blocked sulfite-induced increase of [Ca(2+)]i. Interestingly, antioxidants such as trolox and dithiothreitol, abolished the increase of [Ca(2+)]i induced by sulfite. Exposure to sulfite triggered generation of sulfur- and oxygen-centered free radicals in neurons and increased oxidative stress both in the cultured cortical neurons and the prefrontal cortex of rats. Furthemore, sulfite decreased cell viability in cultured cortical neurons via a calcium-dependent manner. Thus, our current study suggests that the redox-dependent calcium overload triggered by sulfite in cortical neuronsmay be involved in its neurotoxicity.


Subject(s)
Calcium Signaling , Cerebral Cortex/metabolism , Excipients/adverse effects , Food Preservatives/adverse effects , Neurons/metabolism , Oxidative Stress , Sulfites/adverse effects , Animals , Antioxidants/adverse effects , Antioxidants/metabolism , Antioxidants/pharmacology , Calcium Signaling/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Enzyme Inhibitors/pharmacology , Excipients/metabolism , Food Preservatives/metabolism , Free Radicals/agonists , Free Radicals/antagonists & inhibitors , Free Radicals/metabolism , Inositol 1,4,5-Trisphosphate/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate/metabolism , Male , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Oxidants/adverse effects , Oxidants/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats, Sprague-Dawley , Sulfites/metabolism , Tissue Distribution
6.
Tumour Biol ; 36(12): 9723-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26156799

ABSTRACT

Phospholipase Cε (PLCε) is a multifunctional enzyme implicated in inflammatory functions. There are limited data, however, on how PLCε can alter inflammatory cytokine by affecting downstream pathways. Recent studies suggest that inflammation is likely to have an important role in transitional cell carcinoma of bladder (TCCB) and cancer disease progression. Here, we showed that PLCε and p-STAT3 expression were both elevated in TCCB tissues compared to adjacent tissues, and the increase of PLCε level was associated with the increase of p-STAT3 level. Then, knockdown of PLCε using adenovirus-shPLCε significantly decreased inflammatory cytokine (IL-6, TNF-α, IL-1ß) expression and inflammation-associated gene (TLR4, MyD88, p-STAT3) expression. Furthermore, we demonstrated that PLCε knockdown blocked LPS-induced inflammatory cytokine and p-STAT3 expression. Additionally, we found that combined treatment of STAT3 inhibitor S3I-201 with adenovirus-shPLCε exhibited synergistic inhibitory effects on expression of p-STAT3. Our results suggested that STAT3 phosphorylation is involved in PLCε-mediated inflammatory cytokine release. Our research is of potential importance in drug development programs using PLCε as a therapeutic target for TCCB.


Subject(s)
Carcinoma, Transitional Cell/genetics , Inflammation/genetics , Phosphoinositide Phospholipase C/genetics , STAT3 Transcription Factor/biosynthesis , Urinary Bladder Neoplasms/genetics , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Inflammation/pathology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/biosynthesis , Phosphorylation/genetics , STAT3 Transcription Factor/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Urinary Bladder Neoplasms/pathology
7.
Ukr Biochem J ; 87(6): 104-12, 2015.
Article in English | MEDLINE | ID: mdl-27025064

ABSTRACT

The effects of functional interplay of calcium ions, reactive oxygen species (ROS) and nitric oxide (NO) in the cells of wheat plantlets roots (Triticum aestivum L.) at the induction of their heat resistance by a short-term influence of hyperthermia (heating at the temperature of 42 degrees C during 1 minute) have been investigated. The transitional increase of NO and H2O2 content, invoked by heating, was suppressed by the treatment of plantlets with the antagonists of calcium EGTA (chelator of exocellular calcium), lanthanum chloride (blocker of calcium channels of various types) and neomycin (inhibitor of phosphatidylinositol-dependent phospholipase C). The rise of hydrogen peroxide content, caused by hardening, was partially suppressed by the action of inhibitors of nitrate reductase (sodium wolframate) and NO-synthase (N(G)-nitro-L-arginine methyl ester--L-NAME), and the increasing of nitric oxide content was suppressed by the treatment of plants with the antioxidant ionol and with the scavenger of hydrogen peroxide (dimethylthiourea). These compounds and antagonists of calcium also partially removed the effect of the rise of plantlets' heat resistance, invoked by hardening heating. The conclusion on calcium's role in the activation of enzymatic systems, generating reactive oxygen species and nitric oxide, and on the functional interplay of these signal mediators at the induction of heat resistance of plantlets by hardening heating is made.


Subject(s)
Calcium Signaling , Calcium/metabolism , Plant Roots/drug effects , Triticum/drug effects , Adaptation, Physiological , Antioxidants/pharmacology , Butylated Hydroxytoluene/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Chelating Agents/pharmacology , Cations, Divalent , Egtazic Acid/pharmacology , Hot Temperature , Hydrogen Peroxide/metabolism , Lanthanum/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Neomycin/pharmacology , Nitrate Reductase/antagonists & inhibitors , Nitrate Reductase/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , Plant Roots/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Seedlings/drug effects , Seedlings/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology , Triticum/metabolism , Tungsten Compounds/pharmacology
8.
PLoS One ; 9(8): e105061, 2014.
Article in English | MEDLINE | ID: mdl-25121594

ABSTRACT

Plant phosphoinositide-specific phospholipases C (PI-PLCs) function in several essential plant processes associated with either development or environmental stress. In this report, we examined the expression patterns of TaPLC1 under drought and high salinity stress at the transcriptional and post-transcriptional levels. TaPLC1 mRNA was expressed in all wheat organs examined. U73122 and edelfosine, the PLC inhibitor, impaired seedling growth and enhanced seedling sensitivity to drought and high salinity stress. Though TaPLC1 expression in wheat was lowest at the seedling stage, it was strongly induced under conditions of stress. When 6-day-old wheat seedlings were treated with 200 mM NaCl or 20% (w/v) PEG 6000 for 6 or 12 h, respectively, the TaPLC1 transcript level increased by 16-fold compared to the control. Western blotting showed that the TaPLC protein concentration was also maintained at a high level from 24 to 48 h during stress treatment. Together, our results indicate the possible biological functions of TaPLC1 in regulating seedling growth and the response to drought and salinity stress.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Plant , Phosphoinositide Phospholipase C/genetics , Stress, Physiological , Triticum/genetics , Base Sequence , DNA Primers , Genes, Plant , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phylogeny , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Triticum/enzymology
9.
Biochem Biophys Res Commun ; 448(4): 409-13, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24796667

ABSTRACT

Phospholipase C epsilon 1 (PLCε1) has been recently identified as a novel potential biomarker for gastric cancer because of its critical role in inflammation and tumorigenesis. Until now, there are no further reports to investigate the feasibility of gene therapy by suppressing PLCε1 expression for gastric cancer. In this study, a small interfering RNA (shRNA) targeting PLCε1 was firstly transfected into gastric cancer cells in order to silence PLCε1 expression. Both mRNA and protein expression of PLCε1 in gastric cancer cells significantly reduced by RT-PCR and Western blotting analysis. Moreover, subsequent results revealed that PLCε1 shRNA depressed the in vitro and in vivo growth of gastric cancer cells by using MTT assay and tumor xenograft experiment. Furthermore, after PLCε1 shRNA transfection, the expression of proinflammatory molecules including tumor necrosis factor-α (TNF-α), cyclooxygenase 2 (COX-2), interleukin (IL)-6 and chemokine (C-X-C motif) ligand (CXCL)-1 were unaffected, but only chemokine (C-C motif) ligand (CCL)-2 expression decreased in the gastric cancer cells. It is implied that PLCε1 may inhibit the growth of gastric cancer cells via CCL-2 protein mediated pathway. These results suggest that PLCε1 might be an alternative molecular target for gastric cancer gene therapy.


Subject(s)
Genetic Therapy/methods , Phosphoinositide Phospholipase C/antagonists & inhibitors , RNA Interference , Stomach Neoplasms/enzymology , Stomach Neoplasms/therapy , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Down-Regulation , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphoinositide Phospholipase C/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Xenograft Model Antitumor Assays
10.
Cell Biochem Funct ; 32(3): 294-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24357048

ABSTRACT

The pathogenesis of lung cancer is to be further investigated. Recent reports indicate that phospholipase C ε-1 (PLCE1) is a critical molecule involved in tumour growth. This study aims to investigate the role of PLCE1 in the regulation of apoptosis in lung cancer cells. In this study, the surgically removed non-small-cell lung cancer (NSCLC) tissue was collected from 36 patients. Single NSCLC cells were prepared from the tissue, in which immune cells of CD3(+) , CD11c(+) , CD19(+) , CD68(+) and CD14(+) were eliminated by magnetic cell sorting. The expression of PLCE1 and p53 was assessed by quantitative real-time polymerase chain reaction and Western blotting. Apoptosis of NSCLC cells was analysed by flow cytometry. The results showed that, in cultured NSCLC cells, high levels of PLCE1 and low levels p53 were detected; the two molecules showed a negative correlation (p < 0.01). The addition of anti-PLCE1 antibody increased the expression of p53 in NSCLC cells, which increased the frequency of apoptotic NSCLC cells. We conclude that NSCLC cells express high levels of PLCE1, which suppresses the expression of p53 in NSCLC cells. PLCE1 can be a therapeutic target of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phosphoinositide Phospholipase C/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/genetics , Tumor Suppressor Protein p53/genetics
11.
Yakugaku Zasshi ; 132(6): 699-704, 2012.
Article in Japanese | MEDLINE | ID: mdl-22687728

ABSTRACT

Human bronchial mucins, such as MUC5AC, have traditionally been defined as a family of high-molecular weight glycoproteins. Changes in the contents of sugar chains on MUC5AC are among the fundamental features in inflammatory respiratory disease. The changes have been shown to lead to unfavorable alterations in the viscosity of mucus, resulting in impairment of mucociliary transport, vulnerability to viral/bacterial infection as sugar chains play an important role in adhesion of some viruses and bacteria to the epithelium, and finally inflammatory cell infiltration in the airway. Recently, we found that expression of some glycosyltransferases associated with the contents and structure of sugar chains is regulated by phosphatidylinositol-phospholipase (PI-PL) C signaling in cells. L-Carbocisteine, a mucoregulatory drug, normalized or balanced fucosylated and sialylated sugar chains, such as sialyl Lewis x through inhibition of PI-PL C signaling. We prepared MUC5AC fusion protein with tandem repeats associated with MUC5AC, and confirmed that L-carbocisteine inhibited the increases in viscosity associated with sialyl Lewis x expression levels. In addition, the clinical study (2008) noted that L-carbocisteine reduced the frequency of common colds and exacerbation of symptoms in patients with COPD. These favorable effects in patients may be due to normalization of sugar chain contents on mucins. We suggest that the inhibitory effect on infection of airway epithelial cells by rhinoviruses, respiratory syncytial virus, and influenza viruses by treatment with L-carbocisteine may also be based on the regulation of sugar chain contents or structures on mucins.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Carbocysteine/pharmacology , Expectorants/pharmacology , Mucin 5AC/chemistry , Mucus/chemistry , Phosphoinositide Phospholipase C/antagonists & inhibitors , Respiratory Tract Infections/etiology , Animals , Anti-Infective Agents, Local/therapeutic use , Carbocysteine/therapeutic use , Carbohydrate Conformation , Expectorants/therapeutic use , Humans , Mucin 5AC/physiology , Mucus/metabolism , Mucus/physiology , Oligosaccharides/metabolism , Phosphoinositide Phospholipase C/physiology , Rats , Respiratory Tract Infections/drug therapy , Sialyl Lewis X Antigen , Signal Transduction/physiology , Viscosity
12.
Chem Pharm Bull (Tokyo) ; 60(1): 137-43, 2012.
Article in English | MEDLINE | ID: mdl-22223385

ABSTRACT

We describe the total synthesis and structural determination of (+)-akaterpin (1), an inhibitor of phosphatidylinositol-specific phospholipase C (PI-PLC). The key features of the synthetic strategy include the resolution of ß,γ-unsaturated ketone (±)-2a with chiral sulfoximine 6. The absolute stereochemistry was determined by comparison of the specific optical rotation data of (+)-1 and (-)-1 with that of natural akaterpin.


Subject(s)
Enzyme Inhibitors/chemistry , Triterpenes/chemistry , Enzyme Inhibitors/chemical synthesis , Ketones/chemistry , Molecular Conformation , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , Stereoisomerism , Triterpenes/chemical synthesis
13.
Cell Biol Int ; 35(10): 1031-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21121899

ABSTRACT

PLCε (phospholipase Cε), one of effectors belonging to the small GTPase superfamily, has been suggested to play a crucial role in carcinogenesis. However, its bio-function in bladder cancer has never been demonstrated. In our previous study, we found that PLCε mRNA was highly expressed in bladder cancer tissues. In the present study, we silenced the PLCε gene by shRNA (small-hairpin RNA) in the bladder cancer cell line BIU-87. The results showed that it significantly inhibited cell proliferation and arrested the cell cycle at G0/G1-phase. The regulation of cell characteristics has been related to PKCα (protein kinase Cα) activity. Further study showed that knockdown of the PLCε gene down-regulated oncogenes c-fos and c-jun. These results indicate that PLCε plays a crucial role in bladder cancer, and PLCε may be a key molecule regulating the signal pathway of bladder cancer proliferation.


Subject(s)
Phosphoinositide Phospholipase C/metabolism , Protein Kinase C-alpha/metabolism , Signal Transduction , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , G1 Phase Cell Cycle Checkpoints , Humans , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/genetics , Protein Kinase C-alpha/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
14.
Br J Pharmacol ; 160(6): 1293-4, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20590620

ABSTRACT

In this issue MacMillan and McCarron in 2010 demonstrated that the phospholipase C (PLC) inhibitor U-73122 can potently inhibit Ca(2+) release from isolated smooth muscle cells independent of its effect on PLC. Their data suggest that the PLC inhibitor can block the sarcoplasmic/endoplasmic reticulum calcium ATPase pump in smooth muscle and cast doubt on the reliability of U-73122 as the main pharmacological tool to assess the role of the phosphotidyl inositol-PLC pathway in cellular signalling.


Subject(s)
Estrenes/pharmacology , Phosphoinositide Phospholipase C/antagonists & inhibitors , Pyrrolidinones/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Animals , Calcium/metabolism , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction
15.
Br J Pharmacol ; 160(6): 1295-301, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20590621

ABSTRACT

BACKGROUND AND PURPOSE: The sarcoplasmic reticulum (SR) releases Ca(2+) via inositol 1,4,5-trisphosphate receptors (IP(3)R) in response to IP(3)-generating agonists. Ca(2+) release subsequently propagates as Ca(2+) waves. To clarify the role of IP(3) production in wave generation, the contribution of a key enzyme in the production of IP(3) was examined using a phosphoinositide-specific phospholipase C (PI-PLC) inhibitor, U-73122. EXPERIMENTAL APPROACH: Single colonic myocytes were voltage-clamped in whole-cell configuration and cytosolic Ca(2+) concentration ([Ca(2+)](cyto)) measured using fluo-3. SR Ca(2+) release was evoked either by activation of IP(3)Rs (by carbachol or photolysis of caged IP(3)) or ryanodine receptors (RyRs; by caffeine). KEY RESULTS: U-73122 inhibited carbachol-evoked [Ca(2+)](cyto) transients. The drug also inhibited [Ca(2+)](cyto) increases, evoked by direct IP(3)R activation (by photolysis of caged IP(3)) and RyR activation (by caffeine), which do not require PI-PLC activation. U-73122 also increased steady-state [Ca(2+)](cyto) and slowed the rate of Ca(2+) removal from the cytoplasm. An inactive analogue of U-73122, U-73343, was without effect on either IP(3)R- or RyR-mediated Ca(2+) release. CONCLUSIONS AND IMPLICATIONS: U-73122 inhibited carbachol-evoked [Ca(2+)](cyto) increases. However, the drug also reduced Ca(2+) release when evoked by direct activation of IP(3)R or RyR, slowed Ca(2+) removal and increased steady-state [Ca(2+)](cyto). These results suggest U-73122 reduces IP(3)-evoked Ca(2+) transients by inhibiting the SR Ca(2+) pump to deplete the SR of Ca(2+) rather than by inhibiting PI-PLC.


Subject(s)
Calcium/metabolism , Estrenes/pharmacology , Phosphoinositide Phospholipase C/antagonists & inhibitors , Pyrrolidinones/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Aniline Compounds/chemistry , Animals , Caffeine/pharmacology , Carbachol/pharmacology , Colon/cytology , Cytosol/drug effects , Cytosol/metabolism , Guinea Pigs , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Myocytes, Smooth Muscle/drug effects , Patch-Clamp Techniques , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Xanthenes/chemistry
16.
J Biol Chem ; 285(30): 22748-52, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20519514

ABSTRACT

Protein kinase C and protein kinase D are potently activated by agonist-evoked increases in diacylglycerol. Using live cell-imaging probes for kinase activity, we have shown that both kinases are robustly activated at the Golgi following stimulation of G(q)-coupled receptor signaling pathways, displaying activation signatures at the Golgi that are distinct from those at the plasma membrane. Here we report that Ca(2+) is the mediator that allows signals received at the plasma membrane to activate these two protein kinases at the Golgi. Specifically, using fluorescence resonance energy transfer-based reporters to image diacylglycerol production, we show that Ca(2+) is necessary and sufficient to elevate diacylglycerol levels at the Golgi. First, raising intracellular Ca(2+) by treating cells with thapsigargin induces diacylglycerol production at the Golgi. Second, chelation of intracellular Ca(2+) prevents UTP-stimulated increases in diacylglycerol at the Golgi. Thus, agonist-evoked increases in intracellular Ca(2+) cause increases in Golgi diacylglycerol, allowing this intracellular membrane to serve as a platform for signaling by protein kinases C and D.


Subject(s)
Calcium Signaling , Cell Membrane/metabolism , Diglycerides/biosynthesis , Golgi Apparatus/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , COS Cells , Cell Membrane/drug effects , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Fluorescence Resonance Energy Transfer , Golgi Apparatus/drug effects , Phosphatidate Phosphatase/antagonists & inhibitors , Phosphoinositide Phospholipase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Structure, Tertiary
17.
Exp Parasitol ; 124(4): 436-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20045694

ABSTRACT

In the present paper we studied the involvement of the phosphatidylinositol-specific PLC (PI-PLC)/protein kinase C (PKC) pathway in (Na(+)+K(+))ATPase stimulation by heme in Leishmania amazonensis promastigotes. Heme stimulated the PKC-like activity with a concentration of 50nM. Interestingly, the maximal stimulation of the PKC-like activity promoted by phorbol ester was of the same magnitude promoted by heme. However, the stimulatory effect of heme is completely abolished by ET-18-OCH(3) and U73122, specific inhibitors of PI-PLC. (Na(+)+K(+))ATPase activity is increased in the presence of increased concentrations of heme, being maximally affected at 50nM. This effect was completely reversed by 10nM calphostin C, an inhibitor of PKC. Thus, the effect of 50nM heme on (Na(+)+K(+))ATPase activity is completely abolished by ET-18-OCH(3) and U73122. Taken together, these results demonstrate that the heme receptor mediates the stimulatory effect of heme on the (Na(+)+K(+))ATPase activity through a PI-PLC/PKC signaling pathway.


Subject(s)
Heme/pharmacology , Leishmania mexicana/drug effects , Phosphoinositide Phospholipase C/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Estrenes/pharmacology , Leishmania mexicana/enzymology , Phosphodiesterase Inhibitors/pharmacology , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phospholipid Ethers/pharmacology , Pyrrolidinones/pharmacology , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/drug effects
18.
Pharmacol Res ; 61(2): 162-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19782752

ABSTRACT

In this study, we examined the role of phosphatidylcholine-phospholipase C (PC-PLC) and protein kinase C (PKC) in peptidoglycan (PGN)-induced nuclear factor-kappaB (NF-kappaB) activation and cyclooxygenase-2 (COX-2) expression in RAW 264.7 macrophages. PGN-induced COX-2 expression was attenuated by a PC-PLC inhibitor (D609) and by PKC inhibitors (Go 6976 and Ro 31-8220), but not by a phosphatidylinositol-PLC (PI-PLC) inhibitor (U-73122). PGN caused an increase in PKC activity, and this effect was inhibited by D609, Go 6976, and Ro 31-8220, but not by U-73122. Furthermore, the PGN-mediated increases in kappaB-luciferase activity were also inhibited by D609 and Ro 31-8220. Our data demonstrate that PGN activates PC-PLC which induces PKC activation; this in turn initiates NF-kappaB activation, and ultimately induces COX-2 expression in RAW 264.7 macrophages.


Subject(s)
Cyclooxygenase 2/metabolism , Macrophages/drug effects , NF-kappa B/metabolism , Peptidoglycan/pharmacology , Protein Kinase C/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism , Animals , Bridged-Ring Compounds/pharmacology , Carbazoles/pharmacology , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation , Estrenes/pharmacology , Indoles/pharmacology , Macrophages/enzymology , Mice , NF-kappa B/genetics , Norbornanes , Phosphodiesterase Inhibitors/pharmacology , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Thiocarbamates , Thiones/pharmacology , Time Factors , Transfection , Type C Phospholipases/antagonists & inhibitors
19.
Planta ; 230(5): 925-33, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19672622

ABSTRACT

Phosphoinositide-specific phospholipase C (PI-PLC) activities are involved in mediating plant cell responses to environmental stimuli. Two variants of PI-PLC have been partially purified from the roots of oat seedlings; one cytosolic and one particulate. Although the cytosolic enzyme was significantly purified, the activity still co-migrated with a number of other proteins on heparin HPLC and also on size-exclusion chromatography. The partially purified PI-PLC was tested by Western blotting, and we found that actin and actin-binding proteins, profilin and tropomyosin, co-purified with cytosolic phospholipase C. After a non-ionic detergent (Triton X-100) treatment, PI-PLC activities still remained with the actin cytoskeleton. The effects of phalloidin and F-buffer confirmed this association; these conditions, which favor actin polymerization, decreased the release of PI-PLC from the cytoskeleton. The treatments of latrunculin and G-buffer, the conditions that favor actin depolymerization, increased the release of PI-PLC from the cytoskeleton. These results suggest that oat PI-PLC associates with the actin cytoskeleton.


Subject(s)
Actins/metabolism , Avena/enzymology , Cytoskeleton/enzymology , Phosphoinositide Phospholipase C/metabolism , Plant Roots/enzymology , Actin Cytoskeleton/metabolism , Animals , Avena/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cattle , Cell Membrane/drug effects , Cell Membrane/enzymology , Cross Reactions/drug effects , Cytoskeleton/drug effects , Detergents/pharmacology , Immune Sera/pharmacology , Immunoprecipitation , Phalloidine/metabolism , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/isolation & purification , Phospholipase C beta/metabolism , Plant Roots/drug effects , Profilins/metabolism , Protein Binding/drug effects , Solubility/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Thiazolidines/pharmacology
20.
Vascul Pharmacol ; 51(2-3): 147-53, 2009.
Article in English | MEDLINE | ID: mdl-19539781

ABSTRACT

In rabbit intrapulmonary arteries, substance P (SP) has been reported to induce endothelium-dependent relaxation (EDR) and endothelium-dependent contraction (EDC) via tachykinin NK(1) receptors, and endothelium-independent contraction (EIC) via tachykinin NK(2) receptors. The present study pharmacologically examined whether these opposite responses (EDR and EDC) are mediated by the same NK(1) receptor. Five tachykinin agonists, including septide, a reportedly atypical NK(1) agonist, caused concentration-dependent EDR in the presence of NK2 antagonist (SR-48968) + TXA2 synthetase inhibitor (ozagrel), which blocked EIC and EDC, in pre-contracted arteries, and concentration-dependent EDC in the presence of NK2 antagonist (SR-48968) + nitric oxide synthase inhibitor (l-N(G)-nitro-arginine methyl ester), which blocked EIC and EDR, in non-contracted arteries. The EC(50) values of these agonists for EDR were smaller than those for EDC, indicating that the affinities of NK(1) agonists to NK(1) receptors are different between EDR and EDC. However, the rank order of their potency for EDR and EDC was the same: SP = septide > SP methyl ester (SPME) > neurokinin A > neurokinin B. [Ala(5), beta-Ala(8)]-alpha-neurokinin fragment 4-10 (NK2 agonist) and senktide (NK3 agonist) caused no responses. Two structurally different NK(1) antagonists, CP-99994 and SR-140333, shifted the concentration-EDC and -EDR curves of SPME, a selective NK(1) agonist, and septide rightward and suppressed their maximal responses in a similar concentration-dependent manner, indicating that the affinities of NK(1) antagonists to NK1 receptors are similar between EDR and EDC. U-73122, a phospholipase C inhibitor, and thapsigargin, 2,5-di-tert-butylhydroquinone, and ruthenium red, all intracellular Ca2+ release blockers, inhibited SP-induced EDR and EDC. Effective concentrations of ionomycin (Ca2+ ionophore) causing EDR were also lower than those causing EDC. Taken together, SP-induced EDR and EDC are mediated by activation of the same NK1 receptor followed by an increase in intracellular Ca2+, and sensitivity to Ca2+ may be higher in the EDR than EDC pathway.


Subject(s)
Calcium Signaling/physiology , Endothelium, Vascular/physiology , Pulmonary Artery/physiology , Receptors, Neurokinin-1/physiology , Vasoconstriction/physiology , Vasodilation/physiology , Animals , Calcium Channel Blockers/pharmacology , Inhibitory Concentration 50 , Ionophores , Male , Neurokinin A/metabolism , Neurokinin A/pharmacology , Neurokinin B/metabolism , Neurokinin B/pharmacology , Neurokinin-1 Receptor Antagonists , Nitric Oxide Synthase/antagonists & inhibitors , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Phosphoinositide Phospholipase C/antagonists & inhibitors , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism , Pyrrolidonecarboxylic Acid/pharmacology , Rabbits , Receptors, Neurokinin-1/agonists , Receptors, Neurokinin-2/agonists , Receptors, Neurokinin-2/antagonists & inhibitors , Receptors, Neurokinin-3/agonists , Substance P/analogs & derivatives , Substance P/metabolism , Substance P/pharmacology , Thromboxane-A Synthase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL