Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Neuroreport ; 32(17): 1379-1387, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34718250

ABSTRACT

OBJECTIVES: Paeoniflorin, an active component of Radix Paeoniae Alba, has a neuroprotective effect in Parkinson's animal models. However, its mechanism of action remains to be determined. METHODS: In this study, we hypothesized that the neuroprotective effect of paeoniflorin occurs through the α-synuclein/protein kinase C δ subtype (PKC-δ) signaling pathway. We tested our hypothesis in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced mouse model of Parkinson's disease. We evaluated the effects of paeoniflorin on the expression levels of signal components of the α-synuclein/PKC-δ pathway, cellular apoptosis and motor performance. RESULTS: Our results demonstrated that paeoniflorin restored the motor performance impairment caused by MPTP, inhibited apoptosis, and protected the ultrastructure of neurons. Paeoniflorin treatment also resulted in the dose-dependent upregulation of an antiapoptotic protein, B-cell lymphoma-2, at the mRNA and protein levels, similar to the effects of the positive control, selegiline. In contrast, paeoniflorin treatment downregulated the expression of pro-apoptotic proteins BCL2-Associated X2, α-synuclein, and PKC-δ at the mRNA and protein levels, as well as the level of the activated form of nuclear factor kappa B (p-NF-κB p65). CONCLUSIONS: Thus, our results showed that paeoniflorin exerts its neuroprotective effect by regulating the α-synuclein/PKC-δ signaling pathway to reduce neuronal apoptosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Glucosides/pharmacology , Monoterpenes/pharmacology , Parkinsonian Disorders/metabolism , Protein Kinase C-delta/drug effects , Substantia Nigra/drug effects , alpha-Synuclein/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Annexin A5/drug effects , Annexin A5/metabolism , Antiparkinson Agents/pharmacology , Disease Models, Animal , Mice , Microscopy, Electron, Transmission , Neurotoxins , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Protein Kinase C-delta/metabolism , Rotarod Performance Test , Selegiline/pharmacology , Substantia Nigra/metabolism , Substantia Nigra/pathology , alpha-Synuclein/metabolism
2.
Eur J Pharmacol ; 900: 174046, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33745958

ABSTRACT

This study is designed to investigate the role of novel protein kinases C (nPKC) in mediating pulmonary artery smooth muscle cells (PASMCs) proliferation in pulmonary hypertension (PH) and the underlying mechanisms. Mouse PASMCs was isolated using magnetic separation technology. The PASMCs were divided into 24 h group, 48 h group and 72 h group according to different hypoxia treatment time, then detected cell proliferation rate and nPKC expression level in each group. We treated PASMCs with agonists or inhibitors of PKCdelta (PKCδ) and PKCepsilon (PKCε) and exposed them to hypoxia or normoxia for 72 h, then measured the proliferation of PASMCs. We also constructed a lentiviral vector containing siRNA fragments for inhibiting PKCδ and PKCε to transfected PASMCs, then examined their proliferation. PASMCs isolated successfully by magnetic separation method and were in good condition. Hypoxia promoted the proliferation of PASMCs, and the treatment for 72 h had the most significant effect. Hypoxia upregulated the expression of PKCδ and PKCε in mouse PASMCs, leading to PASMCs proliferation. Moreover, Our study demonstrated that hypoxia induced upregulation of PKCδ and PKCε expression resulting to the proliferation of PASMCs via up-regulating the phosphorylation of AKT and ERK. Our study provides clear evidence that increased nPKC expression contributes to PASMCs proliferation and uncovers the correlation between AKT and ERK pathways and nPKC-mediated proliferation of PASMCs. These findings may provide novel targets for molecular therapy of pulmonary hypertension.


Subject(s)
Cell Hypoxia/physiology , Hypertension, Pulmonary/pathology , Myocytes, Smooth Muscle , Protein Kinase C/biosynthesis , Pulmonary Artery/pathology , Animals , Cell Proliferation , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Oncogene Protein v-akt/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta/drug effects , Protein Kinase C-epsilon/drug effects , Protein Kinase Inhibitors/pharmacology , Up-Regulation/physiology
3.
Nat Prod Res ; 34(15): 2151-2156, 2020 Aug.
Article in English | MEDLINE | ID: mdl-30835553

ABSTRACT

Neo-debromoaplysiatoxin C (1), a new member of the aplysiatoxin family, was isolated from the marine cyanobacterium Lyngbya sp. The structure of 1 was elucidated based on spectroscopic data, and its stereochemistry was determined from NOESY spectrum and biosynthetic considerations. This new compound presents an intriguing 10-membered lactone ring skeleton derived from debromoaplysiatoxin by structural rearrangement, which is the first example in the aplysiatoxin family. Its biological properties were evaluated for cytotoxicity, PKCδ activation and inhibitory effects on potassium channel.


Subject(s)
Cyanobacteria/chemistry , Lyngbya Toxins/chemistry , Cytotoxins/pharmacology , Lactones/chemistry , Lactones/pharmacology , Magnetic Resonance Spectroscopy , Molecular Structure , Potassium Channel Blockers/pharmacology , Protein Kinase C-delta/drug effects , Seaweed/chemistry
4.
Endocr Regul ; 53(3): 154-164, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31517630

ABSTRACT

OBJECTIVES: Aldosterone rapidly enhances protein kinase C (PKC) alpha and beta1 proteins in the rat kidney. The G protein-coupled receptor 30 (GPR30)-mediated PKC pathway is involved in the inhibition of the potassium channel in HEK-239 cells. GPR30 mediates rapid actions of aldosterone in vitro. There are no reports available regarding the aldosterone action on other PKC isoforms and GPR30 proteins in vivo. The aim of the present study was to examine rapid actions of aldosterone on protein levels of phosphorylated PKC (p-PKC) delta, p-PKC epsilon, and GPR30 simultaneously in the rat kidney. METHODS: Male Wistar rats were intraperitoneally injected with normal saline solution or aldosterone (150 µg/kg body weight). After 30 minutes, abundance and immunoreactivity of p-PKC delta, p-PKC epsilon, and GPR30 were determined by Western blot analysis and immunohisto-chemistry, respectively. RESULTS: Aldosterone administration significantly increased the renal protein abundance of p-PKC delta by 80% (p<0.01) and decreased p-PKC epsilon protein by 50% (p<0.05). Aldosterone injection enhanced protein immunoreactivity of p-PKC delta but suppressed p-PKC epsilon protein intensity in both kidney cortex and medulla. Protein abundance of GPR30 was elevated by aldosterone treatment (p<0.05), whereas the immunoreactivity was obviously changed in the kidney cortex and inner medulla. Aldosterone translocated p-PKC delta and GPR30 proteins to the brush border membrane of proximal convoluted tubules. CONCLUSIONS: This is the first in vivo study simultaneously demonstrating that aldosterone administration rapidly elevates protein abundance of p-PKC delta and GPR30, while p-PKC epsilon protein is suppressed in rat kidney. The stimulation of p-PKC delta protein levels by aldosterone may be involved in the activation of GPR30.


Subject(s)
Aldosterone/pharmacology , Kidney/drug effects , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Receptors, G-Protein-Coupled/metabolism , Aldosterone/blood , Aldosterone/urine , Animals , Kidney/metabolism , Male , Phosphorylation/drug effects , Protein Kinase C-delta/drug effects , Protein Kinase C-epsilon/drug effects , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/drug effects , Time Factors
5.
J Neurosci ; 39(3): 472-484, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30478032

ABSTRACT

Stress contributes to numerous psychiatric disorders. Corticotropin releasing factor (CRF) signaling and CRF neurons in the bed nucleus of the stria terminalis (BNST) drive negative affective behaviors, thus agents that decrease activity of these cells may be of therapeutic interest. Here, we show that acute restraint stress increases cFos expression in CRF neurons in the mouse dorsal BNST, consistent with a role for these neurons in stress-related behaviors. We find that activation of α2A-adrenergic receptors (ARs) by the agonist guanfacine reduced cFos expression in these neurons both in stressed and unstressed conditions. Further, we find that α- and ß-ARs differentially regulate excitatory drive onto these neurons. Pharmacological and channelrhodopsin-assisted mapping experiments suggest that α2A-ARs specifically reduce excitatory drive from parabrachial nucleus (PBN) afferents onto CRF neurons. Given that the α2A-AR is a Gi-linked GPCR, we assessed the impact of activating the Gi-coupled DREADD hM4Di in the PBN on restraint stress regulation of BNST CRF neurons. CNO activation of PBN hM4Di reduced stress-induced Fos in BNST Crh neurons. Further, using Prkcd as an additional marker of BNST neuronal identity, we uncovered a female-specific upregulation of the coexpression of Prkcd/Crh in BNST neurons following stress, which was prevented by ovariectomy. These findings show that stress activates BNST CRF neurons, and that α2A-AR activation suppresses the in vivo activity of these cells, at least in part by suppressing excitatory drive from PBN inputs onto CRF neurons.SIGNIFICANCE STATEMENT Stress is a major variable contributing to mood disorders. Here, we show that stress increases activation of BNST CRF neurons that drive negative affective behavior. We find that the clinically well tolerated α2A-AR agonist guanfacine reduces activity of these cells in vivo, and reduces excitatory PBN inputs onto these cells ex vivo Additionally, we uncover a novel sex-dependent coexpression of Prkcd with Crh in female BNST neurons after stress, an effect abolished by ovariectomy. These results demonstrate input-specific interactions between norepinephrine and CRF, and point to an action by which guanfacine may reduce negative affective responses.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Corticotropin-Releasing Hormone/physiology , Neurons/physiology , Parabrachial Nucleus/drug effects , Receptors, Adrenergic, alpha-2/drug effects , Septal Nuclei/drug effects , Animals , Female , Gene Expression/drug effects , Genes, fos/drug effects , Guanfacine/pharmacology , Male , Mice , Mice, Inbred C57BL , Norepinephrine/pharmacology , Ovariectomy , Patch-Clamp Techniques , Protein Kinase C-delta/drug effects , Receptors, G-Protein-Coupled/drug effects , Restraint, Physical , Stress, Psychological/physiopathology
6.
Neurochem Int ; 122: 144-148, 2019 01.
Article in English | MEDLINE | ID: mdl-30500461

ABSTRACT

Compelling evidence suggests that far-infrared ray (FIR) possesses beneficial effects on emotional disorders. However, the underlying mechanism conveyed by FIR remains unclear. Recently, we demonstrated that exposure to FIR induces antioxidant potentials via up-regulation of glutathione peroxidase (GPx)-1 gene. The antioxidant potentials might be important for the modulation on the neuropsychotoxic conditions. Exposure to FIR protects from methamphetamine (MA)-induced memory impairments via phosphorylation of ERK 1/2 signaling by positive modulation of protein kinase C δ (PKCδ), M1 muscarinic acetylcholine receptor (M1 mAChR), and nuclear factor E2-related factor 2 (Nrf2) transcription factor. In addition, exposure to FIR positively modulates MA-induced behavioral sensitization via attenuating mitochondrial dysfunction by down-regulation of dopamine D1 receptor. In this mini-review, we have discussed with the protective potentials mediated by FIR against MA-induced psychotoxic burdens.


Subject(s)
Methamphetamine/pharmacology , Mitochondria/drug effects , Protein Kinase C-delta/drug effects , Receptors, Dopamine D1/drug effects , Animals , Antioxidants/pharmacology , Humans , Infrared Rays/therapeutic use , Mitochondria/metabolism , Protein Kinase C-delta/metabolism , Receptors, Dopamine D1/metabolism
7.
Neurochem Int ; 116: 63-76, 2018 06.
Article in English | MEDLINE | ID: mdl-29572053

ABSTRACT

We demonstrated that activation of protein kinase Cδ (PKCδ) and inactivation of the glutathione peroxidase-1 (GPx-1)-dependent systems are critical for methamphetamine (MA)-induced recognition memory impairment. We also demonstrated that exposure to far-infrared rays (FIR) causes induction of the glutathione (GSH)-dependent system, including induction of the GPx-1 gene. Here, we investigated whether exposure to FIR rays affects MA-induced recognition memory impairment and whether it modulates PKC, cholinergic receptors, and the GSH-dependent system. Because the PKC activator bryostatin-1 mainly induces PKCα, PKCε, and PKCδ, we assessed expression of these proteins after MA treatment. MA treatment selectively increased PKCδ expression and its phosphorylation. Exposure to FIR rays significantly attenuated MA-induced increases in PKCδ phosphorylation. Importantly, bryostatin-1 potentiated MA-induced phosphorylation of PKCδ. MA treatment significantly decreased M1, M3, and M4 muscarinic acetylcholine receptors (mAChRs) and ß2 nicotinic acetylcholine receptor expression. Of these, the decrease was most pronounced in M1 mAChR. Exposure to FIR significantly attenuated MA-induced decreases in the M1 mAChR and phospho-ERK1/2, while it facilitated Nrf2-dependent GSH induction. Dicyclomine, an M1 mAChR antagonist, and l-buthionine-(S, R)-sulfoximine (BSO), an inhibitor of GSH synthesis, counteracted against the protective potentials mediated by FIR. More importantly, the memory-enhancing potential of FIR rays was significantly counteracted by bryostatin-1, dicyclomine, and BSO. Our results suggest that exposure to FIR rays attenuates MA-induced impairment in recognition memory via up-regulation of M1 mAChR, Nrf2-dependent GSH induction, and ERK1/2 phosphorylation by inhibiting PKCδ phosphorylation by bryostatin-1.


Subject(s)
Memory Disorders/drug therapy , NF-E2-Related Factor 2/drug effects , Protein Kinase C-delta/drug effects , Receptor, Muscarinic M1/drug effects , Animals , Glutathione Peroxidase , Memory Disorders/chemically induced , Methamphetamine/pharmacology , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Protein Kinase C-delta/metabolism , Up-Regulation , Glutathione Peroxidase GPX1
8.
Am J Physiol Gastrointest Liver Physiol ; 310(6): G399-409, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26744470

ABSTRACT

Cholecystokinin (CCK) is a gastrointestinal hormone that induces exocytotic amylase release in pancreatic acinar cells. The activation of protein kinase C (PKC) is involved in the CCK-induced pancreatic amylase release. Myristoylated alanine-rich C kinase substrate (MARCKS) is a ubiquitously expressed substrate of PKC. MARCKS has been implicated in membrane trafficking in several cell types. The phosphorylation of MARCKS by PKC results in the translocation of MARCKS from the membrane to the cytosol. Here, we studied the involvement of MARCKS in the CCK-induced amylase release in rat pancreatic acini. Employing Western blotting, we detected MARCKS protein in the rat pancreatic acini. CCK induced MARCKS phosphorylation. A PKC-δ inhibitor, rottlerin, inhibited the CCK-induced MARCKS phosphorylation and amylase release. In the translocation assay, we also observed CCK-induced PKC-δ activation. An immunohistochemistry study showed that CCK induced MARCKS translocation from the membrane to the cytosol. When acini were lysed by a detergent, Triton X-100, CCK partially induced displacement of the MARCKS from the GM1a-rich detergent-resistant membrane fractions (DRMs) in which Syntaxin2 is distributed. A MARCKS-related peptide inhibited the CCK-induced amylase release. These findings suggest that MARCKS phosphorylation by PKC-δ and then MARCKS translocation from the GM1a-rich DRMs to the cytosol are involved in the CCK-induced amylase release in pancreatic acinar cells.


Subject(s)
Amylases/metabolism , Cholecystokinin/pharmacology , Intracellular Signaling Peptides and Proteins/biosynthesis , Membrane Proteins/biosynthesis , Pancreas/metabolism , Protein Kinase C/metabolism , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholecystokinin/antagonists & inhibitors , Cytosol/drug effects , Cytosol/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Microdomains/metabolism , Membrane Proteins/genetics , Myristoylated Alanine-Rich C Kinase Substrate , Pancreas/drug effects , Pancreas/enzymology , Phosphorylation , Protein Kinase C-delta/drug effects , Protein Kinase C-delta/metabolism , Qa-SNARE Proteins/metabolism , Rats , Rats, Sprague-Dawley , Translocation, Genetic
9.
Pharmazie ; 71(10): 575-582, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-29441925

ABSTRACT

OBJECTIVE: This study was performed to evaluate the regulating effects of acetylpuerarin on inflammation in an Alzheimer's disease (AD) rat model and an inflammatory cell model. METHODS: Healthy female Wistar rats and mouse BV2 microglia cells were selected. AD rat models were established with the method of bilateral intrahippocampal amyloid-ß(Aß)1-42 injections and the inflammatory cell models were established using Aß25-35-induced mouse BV2 microglia cells. The cytotoxicity of acetylpuerarin on BV2 microglial cells was detected by MTT assay and the morphological changes of BV2 microglia cells were observed under inverted phase contrast microscope. As inflammatory parameters, the expressions of IL-1ß, iNOS, IL-6 and TNF-α were examined by Elisa, Immunohistochemistry, Quantitative real-time PCR (qRT-PCR), Western blot and Immunofluorescence analyses. We also examined the acetylpuerarin's effect on the activity of PKC-δ, IKKß and caspase-8/caspase-3 pathway. RESULTS: Acetylpuerarin exerted no significant cytotoxicity on BV2 microglia cells and was applied in all subsequent experiments. Acetylpuerarin treatment mitigated Aß25-35-induced morphological changes associated with microglia activation. Moreover, the expressions of caspase-8, cleaved caspase-3, PKC-δ, IKKß, iNOS, IL-1ß and TNF-α in Aß25-35-stimulated BV2 microglia cells were significantly suppressed by acetylpuerarin and in a dose-dependent manner. Additionally, the expression of IL-1ß in hippocampus and the level of IL-6 in serum of Aß1-42 treated rat were reduced by acetylpuerarin and in a concentration-dependent manner. CONCLUSION: Our results suggest that acetylpuerarin's anti-inflammation mechanism on AD may be mediated through the PKC-δ-dependent caspase signalling pathway.


Subject(s)
Caspases/drug effects , Encephalitis/drug therapy , Isoflavones/pharmacology , Protein Kinase C-delta/drug effects , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides , Animals , Cell Survival/drug effects , Cytokines/metabolism , Encephalitis/chemically induced , Female , Macrophage Activation/drug effects , Mice , Microglia/drug effects , Peptide Fragments , Rats , Rats, Wistar , Signal Transduction/drug effects
10.
Biochem Cell Biol ; 93(1): 16-27, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25453494

ABSTRACT

Matrix metalloproteinases (MMPs) have been regarded as major critical molecules assisting tumor cells during metastasis, for excessive ECM (ECM) degradation, and cancer cell invasion. In the present study, in vitro and in vivo assays were employed to examine the inhibitory effects of kaempferol, a natural polyphenol of flavonoid family, on tumor metastasis. Data showed that kaempferol could inhibit adhesion, migration, and invasion of MDA-MB-231 human breast carcinoma cells. Moreover, kaempferol led to the reduced activity and expression of MMP-2 and MMP-9, which were detected by gelatin zymography, real-time PCR, and western blot analysis, respectively. Further elucidation of the mechanism revealed that kaempferol treatment inhibited the activation of transcription factor activator protein-1 (AP-1) and MAPK signaling pathway. Moreover, kaempferol repressed phorbol-12-myristate-13-acetate (PMA)-induced MMP-9 expression and activity through suppressing the translocation of protein kinase Cδ (PKCδ) and MAPK signaling pathway. Our results also indicated that kaempferol could block the lung metastasis of B16F10 murine melanoma cells as well as the expression of MMP-9 in vivo. Taken together, these results demonstrated that kaempferol could inhibit cancer cell invasion through blocking the PKCδ/MAPK/AP-1 cascade and subsequent MMP-9 expression and its activity. Therefore, kaempferol might act as a therapeutic potential candidate for cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Kaempferols/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Metastasis/drug therapy , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation , Female , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Protein Kinase C-delta/drug effects , Transcription Factor AP-1/drug effects
11.
Res Vet Sci ; 95(3): 1125-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23993687

ABSTRACT

This study investigated the inhibitory effects of sanguinarine (SA) on PKC-CPI-17 pathway in rat intestinal smooth muscle cells (ISMC). Previous studies indicate that the inhibitory effects of SA on ISMC contraction are possibly mediated by the Ca(2+) influx. ISMC was treated with 1 µM SA for 24h remarkably inhibited the mRNA expression of m2 and m3 receptors. ISMC treated with 1 or 3 µM SA for 30 min significantly decreased the mRNA expression of PKC-δ, PKC-ε, PKC-η, and CPI-17. 1 µM SA could markedly inhibit carbachol (CCh)-mediated increase PKC-δ, PKC-η, and CPI-17 mRNA but had no effect in PKC-ε.Treatment of ISMC with SA (1 µM, 30 min) caused a decrease in protein expression of PKC-δ. However, the expression of CPI-17 was significantly inhibited in a time-dependent manner. These results demonstrate that the inhibitory effect of SA is coupled with alteration of PKC-mediated signal transduction and intracellular Ca(2+) concentration.


Subject(s)
Benzophenanthridines/pharmacology , Isoquinolines/pharmacology , Muscle, Smooth/drug effects , Phosphoprotein Phosphatases/drug effects , Protein Kinase C/drug effects , Receptors, Muscarinic/drug effects , Signal Transduction/drug effects , Animals , Blotting, Western , Calcium/analysis , Intestines/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/chemistry , Protein Kinase C-delta/drug effects , Rats
12.
FASEB J ; 26(8): 3537-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22573912

ABSTRACT

Energy production in mitochondria is a multistep process that requires coordination of several subsystems. While reversible phosphorylation is emerging as the principal tool, it is still unclear how this signal network senses the workloads of processes as different as fuel procurement, catabolism in the Krebs cycle, and stepwise oxidation of reducing equivalents in the electron transfer chain. We previously proposed that mitochondria use oxidized cytochrome c in concert with retinol to activate protein kinase Cδ, thereby linking a prominent kinase network to the redox balance of the ETC. Here, we show that activation of PKCε in mitochondria also requires retinol as a cofactor, implying a redox-mechanism. Whereas activated PKCδ transmits a stimulatory signal to the pyruvate dehdyrogenase complex (PDHC), PKCε opposes this signal and inhibits the PDHC. Our results suggest that the balance between PKCδ and ε is of paramount importance not only for flux of fuel entering the Krebs cycle but for overall energy homeostasis. We observed that the synthetic retinoid fenretinide substituted for the retinol cofactor function but, on chronic use, distorted this signal balance, leading to predominance of PKCε over PKCδ. The suppression of the PDHC might explain the proapoptotic effect of fenretinide on tumor cells, as well as the diminished adiposity observed in experimental animals and humans. Furthermore, a disturbed balance between PKCδ and PKCε might underlie the injury inflicted on the ischemic myocardium during reperfusion. dehydrogenase complex.


Subject(s)
Energy Metabolism/drug effects , Homeostasis/drug effects , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Animals , Cell Line , Citric Acid Cycle , Enzyme Activation , Fenretinide/pharmacology , Mice , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Oxidative Phosphorylation/drug effects , Phosphorylation , Protein Kinase C-delta/drug effects , Protein Kinase C-epsilon/genetics , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Signal Transduction/drug effects , Vitamin A/metabolism , Zinc Fingers
13.
J Urol ; 186(6): 2434-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22019170

ABSTRACT

PURPOSE: Emerging evidence shows that the translocation of apoptosis related factors on cellular organelles, such as mitochondria, endoplasmic reticulum, Golgi apparatus and nucleus, has a crucial role in the apoptotic process. We characterized the effect of paclitaxel (Sigma®) on Golgi involved apoptosis in human hormone refractory prostate cancer. MATERIALS AND METHODS: FACScan™ flow cytometric analysis was used to determine cell cycle distribution and the subG1 (apoptosis) population. Protein expression and localization were detected by Western blot, confocal microscopic examination and the sucrose gradient separation technique. RESULTS: Paclitaxel induced Golgi apparatus disassembly and interaction between Golgi complexes and mitochondria. Discontinuous sucrose gradient fractionation was used to determine and collect Golgi containing fractions. Data revealed that paclitaxel induced an increase of Cdk1 activity and DR5 expression on the Golgi complex that was associated with increased cleavage of caspase-8, a DR5 downstream factor, and caspase-3 into catalytically active fragments. Data were validated by confocal immunofluorescence microscopy. Golgi associated effects were inhibited by the Cdk1 inhibitor roscovitine (Sigma), suggesting a critical role for Golgi-Cdk1. Also, paclitaxel caused an increase of nuclear but not of Golgi associated PKC-δ activity. The selective PKC-δ inhibitor rottlerin (Sigma) completely inhibited the increase of Golgi-Cdk1 activity, suggesting that nuclear PKC-δ served as an upstream regulator of Golgi-Cdk1. CONCLUSIONS: Data suggest that paclitaxel induces nuclear translocation and activation of PKC-δ, which in turn causes Golgi-Cdk1 activation, leading to Golgi associated DR5 up-regulation, and caspase-8 and 3 activation. Golgi mediated signaling cascades facilitate mitochondria involved apoptotic pathways and at least partly explain the anticancer activity of paclitaxel action.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , CDC2 Protein Kinase/physiology , Paclitaxel/pharmacology , Prostatic Neoplasms/enzymology , Protein Kinase C-delta/drug effects , Protein Kinase C-delta/physiology , Golgi Apparatus/enzymology , Humans , Male , Prostatic Neoplasms/drug therapy , Tumor Cells, Cultured
14.
Gastroenterology ; 141(5): 1720-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21704002

ABSTRACT

BACKGROUND & AIMS: Activation of protein kinase C (PKC) enzymes in liver and brain alters hepatic glucose metabolism, but little is known about their role in glucose regulation in the gastrointestinal tract. We investigated whether activation of PKC-δ in the duodenum is sufficient and necessary for duodenal nutrient sensing and regulates hepatic glucose production through a neuronal network in rats. METHODS: In rats, we inhibited duodenal PKC and evaluated whether nutrient-sensing mechanisms, activated by refeeding, have disruptions in glucose regulation. We then performed gain- and loss-of-function pharmacologic and molecular experiments to target duodenal PKC-δ; we evaluated the impact on glucose production regulation during the pancreatic clamping, while basal levels of insulin were maintained. RESULTS: PKC-δ was detected in the mucosal layer of the duodenum; intraduodenal infusion of PKC inhibitors disrupted glucose homeostasis during refeeding, indicating that duodenal activation of PKC-δ is necessary and sufficient to regulate glucose homeostasis. Intraduodenal infusion of the PKC activator 1-oleoyl-2-acetyl-sn-glycerol (OAG) specifically activated duodenal mucosal PKC-δ and a gut-brain-liver neuronal pathway to reduce glucose production. Molecular and pharmacologic inhibition of duodenal mucosal PKC-δ negated the ability of duodenal OAG and lipids to reduce glucose production. CONCLUSIONS: In the duodenal mucosa, PKC-δ regulates glucose homeostasis.


Subject(s)
Diglycerides/pharmacology , Duodenum/metabolism , Glucose/metabolism , Intestinal Mucosa/metabolism , Protein Kinase C-delta/metabolism , Animals , Duodenum/innervation , Homeostasis/physiology , Intestinal Mucosa/innervation , Male , Models, Animal , Neurons/physiology , Protein Kinase C-delta/drug effects , Rats , Rats, Sprague-Dawley
15.
J Leukoc Biol ; 90(2): 235-47, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21393419

ABSTRACT

ATRA and a synthetic RAR agonist tamibarotene (Am80) induce granulocytic differentiation of human acute leukemia HL-60 cells and have been used in antineoplastic therapy. ATRA induces CD38 antigen during HL-60 cell differentiation, which interacts with CD31 antigen on the vascular EC surface and may induce disadvantages in the therapy. We here examined the mechanisms of the ATRA-mediated CD38 induction and compared the difference between ATRA- and tamibarotene-mediated induction. Tamibarotene-induced HL-60 cell adhesion to ECs was 38% lower than ATRA, and NB4 cell adhesion to ECs by tamibarotene was equivalent to ATRA, which induced CD38 gene transcription biphasically in HL-60 cells, the early-phase induction via DR-RARE containing intron 1, and the delayed-phase induction via RARE lacking the 5'-flanking region. In contrast to ATRA, tamibarotene induced only the early-phase induction, resulting in its lower CD38 induction than ATRA. A PKCδ inhibitor, rottlerin, and siRNA-mediated PKCδ knockdown suppressed the ATRA-induced CD38 promoter activity of the 5'-flanking region, whereas a RAR antagonist, LE540, or RAR knockdown did not affect it. Cycloheximide and rottlerin suppressed the delayed-phase induction of CD38 expression by ATRA but did not affect the early-phase induction. Moreover, ATRA, but not tamibarotene, induced PKCδ expression without affecting its mRNA stability. The diminished effect of tamibarotene on CD38-mediated HL-60 cell adhesion to ECs compared with ATRA is likely a result of the lack of its delayed-phase induction of CD38 expression, which may be advantageous in antineoplastic therapy.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Benzoates/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Leukemia/pathology , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology , Cell Adhesion/drug effects , Endothelial Cells/cytology , HL-60 Cells , Humans , Protein Kinase C-delta/drug effects , Protein Kinase C-delta/genetics , Retinoids/pharmacology
16.
Diabetologia ; 53(4): 717-29, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20020096

ABSTRACT

AIMS/HYPOTHESIS: Protein kinase C (PKC) regulates exocytosis in various secretory cells. Here we studied intracellular translocation of the PKC isoenzymes PKCalpha and PKCdelta, and investigated how activation of PKC influences glucagon secretion in mouse and human pancreatic alpha cells. METHODS: Glucagon release from intact islets was measured in static incubations, and the amounts released were determined by RIA. Exocytosis was monitored as increases in membrane capacitance using the patch-clamp technique. The expression of genes encoding PKC isoforms was analysed by real-time PCR. Intracellular PKC distribution was assessed by confocal microscopy. RESULTS: The PKC activator phorbol 12-myristate 13-acetate (PMA) stimulated glucagon secretion from mouse and human islets about fivefold (p < 0.01). This stimulation was abolished by the PKC inhibitor bisindolylmaleimide (BIM). Whereas PMA potentiated exocytosis more than threefold (p < 0.001), BIM inhibited alpha cell exocytosis by 60% (p < 0.05). In mouse islets, the PKC isoenzymes, PKCalpha and PKCbeta1, were highly abundant, while in human islets PKCeta, PKCepsilon and PKCzeta were the dominant variants. PMA stimulation of human alpha cells correlated with the translocation of PKCalpha and PKCdelta from the cytosol to the cell periphery. In the mouse alpha cells, PKCdelta was similarly affected by PMA, whereas PKCalpha was already present at the cell membrane in the absence of PMA. This association of PKCalpha in alpha cells was principally dependent on Ca(2+) influx through the L-type Ca(2+) channel. CONCLUSIONS/INTERPRETATION: PKC activation augments glucagon secretion in mouse and human alpha cells. This effect involves translocation of PKCalpha and PKCdelta to the plasma membrane, culminating in increased Ca(2+)-dependent exocytosis. In addition, we demonstrated that PKCalpha translocation and exocytosis exhibit differential Ca(2+) channel dependence.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Protein Kinase C-alpha/physiology , Protein Kinase C-delta/physiology , Protein Kinase C/metabolism , Adult , Aged , Animals , Exocytosis/drug effects , Exocytosis/physiology , Humans , Mice , Middle Aged , Protein Kinase C-alpha/drug effects , Protein Kinase C-delta/drug effects , Protein Transport , Tetradecanoylphorbol Acetate/pharmacology
17.
Biomed Pharmacother ; 63(10): 787-91, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19914793

ABSTRACT

BACKGROUND: Recently we have demonstrated that the cardioprotective potential of ischemic postconditioning (IPOC) against ischemia and reperfusion (I/R)-induced myocardial injury was markedly suppressed in hyperhomocysteinemic (Hhcy) rat hearts. The present study investigated the possible role of PKC-delta in Hhcy-induced suppression of myocardial infarct size-limiting effect of IPOC. METHODS: Isolated Langendorff's perfused normal and Hhcy rat hearts were subjected to 30-min global ischemia (I), followed by 120-min reperfusion (R). The myocardial damage was assessed by measuring the infarct size, and analyzing the release of LDH and CK-MB in coronary effluent. The oxidative stress in the heart was assessed by measuring lipid peroxidation and superoxide anion generation. RESULTS: The I/R produced myocardial injury in normal and Hhcy rat hearts by increasing myocardial infarct size, LDH and CK in coronary effluent and oxidative stress. Hhcy rat hearts exhibited enhanced I/R-induced myocardial injury and high oxidative stress as compared to normal rat hearts subjected to I/R. The IPOC (six brief episodes of I/R, 10s each) afforded cardioprotection against I/R-induced myocardial injury in normal rat hearts; but IPOC-mediated cardioprotection was abolished in Hhcy rat hearts. Treatment with rottlerin (10 microM), a selective inhibitor of PKC-delta, did not affect the cardioprotective effect of IPOC in normal rat hearts; but its treatment significantly restored the myocardial infarct size-limiting effect of IPOC in Hhcy rat hearts. CONCLUSION: The high oxidative stress produced in Hhcy rat hearts during reperfusion may activate PKC-delta, which may be responsible for impairing the infarct size-limiting potential of IPOC in Hhcy rat hearts.


Subject(s)
Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Protein Kinase C-delta/metabolism , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Creatine Kinase, MB Form/metabolism , Female , Hyperhomocysteinemia/complications , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation , Male , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Oxidative Stress , Protein Kinase C-delta/drug effects , Rats , Rats, Wistar , Superoxides/metabolism
18.
Cancer Res ; 69(10): 4260-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19417139

ABSTRACT

Acquired resistance to protein kinase C (PKC) modulators may explain the failure of clinical trials in patients with cancer. Herein, we established a human colon cancer cell line resistant to PEP005, a drug that inhibits PKCalpha and activates PKCdelta. Colo205-R cells, selected by stepwise exposure to PEP005, were >300-fold more resistant to PEP005 than parental Colo205-S cells and were cross-resistant to phorbol 12-myristate 13-acetate, bryostatin, bistratene A, and staurosporine. No PKCalpha or PKCdelta mutation was detected in Colo205-S and Colo205-R cells. Changes in Colo205-R cells were reminiscent of the epithelial-to-mesenchymal transition (EMT) phenotype. Accordingly, Colo205-R cells were more invasive than Colo205-S in Matrigel assays and in mouse xenografts. We also found an increased mRNA expression of several EMT genes, such as those encoding for transforming growth factor-beta and vimentin, along with a decreased mRNA expression of genes involved in epithelial differentiation, such as CDH1 (E-cadherin), CLDN4 (claudin 4), S100A4, and MUC1, in Colo205-R compared with Colo205-S cells in vitro and in vivo. Interestingly, high expression of ET-1 was shown in Colo205-R cells and correlated with low sensitivity to PEP005 and staurosporine in a panel of 10 human cancer cell lines. Inhibition of the ET-1 receptor ETR-A with bosentan restored the antiproliferative effects of PEP005 in Colo205-R cells and decreased the invasive properties of this cell line. Exogenous exposure to ET-1 and silencing ET-1 expression using small interfering RNA modulated cell signaling in Colo205-S and Colo205-R. In summary, acquired resistance to PEP005 was associated with expression of EMT markers and activates the ET-1/ETR-A cell signaling.


Subject(s)
Cell Survival/drug effects , Colonic Neoplasms/genetics , Diterpenes/pharmacology , Epithelial Cells/pathology , Mesoderm/pathology , Protein Kinase C-alpha/genetics , Protein Kinase C-delta/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Differentiation , Cell Division/drug effects , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Epithelial Cells/drug effects , Exons , Female , Humans , Matrix Metalloproteinases/drug effects , Matrix Metalloproteinases/metabolism , Mesoderm/drug effects , Mice , Mice, Nude , Neoplasm Invasiveness , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Protein Kinase C-alpha/drug effects , Protein Kinase C-delta/drug effects , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
19.
Inflamm Res ; 57(5): 216-29, 2008 May.
Article in English | MEDLINE | ID: mdl-18594782

ABSTRACT

OBJECTIVE AND DESIGN: The process of neutrophil extravasation is involved in acute and chronic inflammatory diseases; however our understanding of the role of endothelial second messengers in the regulation of leukocyte emigration is still incomplete. MATERIALS AND METHODS: We investigated this using an in vitro model of neutrophil migration across human endothelial cells. RESULTS: Activation of endothelial protein kinase C (PKC) by either phorbol myristate acetate (PMA) or bryostatin-1 (a potent PKC delta and epsilon activator) completely abolished neutrophil migration mediated by either endothelial TNF-alpha stimulation or LTB4. Pretreatment with Gö-6983 (PKC alpha, beta, delta, inhibitor) prior to addition of bryostatin-1 restored LTB4 induced PMN migration, while pretreatment with Gö-6976 (PKC alpha and beta inhibitor) did not. PKC delta specific siRNA knockdown of PKC delta expression in endothelial cells also restored LTB4 induced PMN migration. In addition, PMA and bryostatin-1 both increased endothelial adhesion to the substratum that was also reversed using PKC delta siRNA knockdown of PKC delta expression. PMA and bryostatin-1 additionally altered the staining pattern of FAK[pY397], paxillin, and vinculin from a dot-like pattern to a dash-like pattern around the cell perimeter. While PMA reduced transendothelial resistance (TER), bryostatin-1 had no effect on TER. CONCLUSIONS: These observations show that endothelial PKC delta activation eliminates neutrophil transendothelial migration through a mechanism unrelated to endothelial barrier integrity. These data are consistent with PKC delta mediated increased cell substrate adhesion as a limiting factor for neutrophil transendothelial migration towards a chemoattractant.


Subject(s)
Cell Movement/physiology , Endothelium, Vascular/enzymology , Neutrophils/physiology , Protein Kinase C-delta/physiology , Antineoplastic Agents/pharmacology , Bryostatins/pharmacology , Carbazoles/pharmacology , Carcinogens/pharmacology , Cell Movement/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 1/metabolism , Humans , Leukotriene B4/pharmacology , Neutrophils/drug effects , Paxillin/metabolism , Protein Kinase C-delta/drug effects , RNA, Small Interfering/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vinculin/metabolism
20.
Eur J Pharmacol ; 591(1-3): 7-12, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18577379

ABSTRACT

Wogonin, a natural monoflavonoid, has been shown to have tumor therapeutic potential in vitro and in vivo. Recently many studies have focused on the induction of apoptosis of tumor cells by wogonin. In this study, we found that wogonin could induce differentiation and G1 phase arrest of human U-937 leukemia cells. The growth of U-937 cells incubated with wogonin was inhibited in a time- and concentration-dependent manner. After treatment with wogonin, U-937 cells exhibited the characteristics of mature granulocytes, such as increased cytoplasmic-to-nuclear ratio, enhanced prominence of cytoplasmic granules, membrane ruffling, a higher NBT-reducing ability, and an increased expression of CD11b. Moreover, wogonin could induce G1 phase arrest and influenced the expression of associated proteins. For example, the expression of phorsphorylated protein kinase C (PKC) delta, p21 increased, while that of cyclin D1/cyclin-dependent kinase (CDK) 4, p-Rb decreased. The upregulation of p21 could be reversed by rottlerin, an inhibitor of PKCdelta. Taken together, wogonin induced U-937 cells to undergo granulocytic differentiation and G1 phase arrest via PKCdelta phosphorylation-induced upregulation of p21 proteins.


Subject(s)
Flavanones/pharmacology , G1 Phase/drug effects , Lymphoma, Large B-Cell, Diffuse/metabolism , Protein Kinase C-delta/drug effects , Cell Differentiation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavanones/administration & dosage , Humans , Phosphorylation/drug effects , Protein Kinase C-delta/metabolism , Time Factors , U937 Cells , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...