Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biotech Histochem ; 98(7): 501-507, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37501577

ABSTRACT

Glioma is the most common type of primary brain tumor; it exhibits great invasive capacity, morbidity and mortality. Protein kinase Cε (PKCε), a serine/threonine kinase, contributes to the development and progression of many cancers. We investigated whether knockdown of PKCε could affect the mitochondrial membrane potential of human glioma cell lines, U251 and U87, and the growth of U251 cell-derived tumors in nude mice. We found that the expression of PKCε was greater in human glioma tissues than in human normal brain tissues. Knockdown of PKCε reduced mitochondrial membrane potential in U251 and U87 cells. Knockdown of PKCε also suppressed the growth of tumors derived from U251 cells and induced apoptosis of U251 cells in vivo. Our findings indicate that PKCε is important for development and progression of glioma and may be a potential therapeutic target for glioma treatment.


Subject(s)
Glioma , Protein Kinase C-epsilon , Animals , Mice , Humans , Protein Kinase C-epsilon/metabolism , Protein Kinase C-epsilon/pharmacology , Mice, Nude , Membrane Potential, Mitochondrial , Cell Proliferation , Glioma/genetics , Glioma/drug therapy , Glioma/metabolism , Apoptosis , Cell Line, Tumor
2.
J Ethnopharmacol ; 303: 115953, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36442760

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ischemic stroke is a significant risk factor for human health, and Buyang Huanwu Decoction is a classical and famous Chinese formula for treating it, but without clear pharmacological mechanism. AIM OF THE STUDY: The aim of this study was to investigate that the molecular mechanism of BYHWD activation of the PKCε/Nrf2 signaling pathway to attenuate cerebral ischemia-reperfusion (I/R) oxidative damage. MATERIALS AND METHODS: The MCAO method was used to establish a brain I/R injury model in SD rats, and neurological deficits were evaluated by neurological function score. Neuronal damage was observed by Nissl staining and immunofluorescence detection of MAP2 expression. Oxidative damage was observed by ROS, SOD, GSH-PX, MDA, and 8-OHdG. Changes in mitochondrial membrane potential were detected by using the fluorescent probe JC-1. The Western blot analysis detected protein expression of PKCε, P-PKCε, total Nrf2, nuclear Nrf2, HO-1, and NQO1. RESULTS: BYHWD significantly enhanced neural function, reduced neuronal damage, inhibited the production of ROS, decreased MDA and 8-OHdG levels, increased SOD and GSH-PX activity to reduce oxidative damage, and restored mitochondrial membrane potential. BYHWD and Nrf2 activator TBHQ increased total Nrf2, nucleus Nrf2 protein expression, and its downstream HO-1 and NQO1 proteins, and the administration of the Nrf2 inhibitor brusatol reduced the enhancing effect of BYHWD. Meanwhile, BYHWD increased the expression of PKCε and P-PKCε and the administration of the PKCε inhibitor εV1-2 reduced the effect of BYHWD in increasing the expression of PKCε, P-PKCε, nuclear Nrf2, and HO-1, as well as promoting the effect of Nrf2 translocation to the nucleus. CONCLUSION: This study marks the first to demonstrate that BYHWD ameliorates oxidative damage and attenuates brain I/R injury by activating the PKCε/Nrf2/HO-1 pathway.


Subject(s)
Brain Ischemia , Reperfusion Injury , Animals , Rats , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cerebral Infarction , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Protein Kinase C-epsilon/metabolism , Protein Kinase C-epsilon/pharmacology , Rats, Sprague-Dawley , Reactive Oxygen Species , Reperfusion , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Signal Transduction , Superoxide Dismutase/metabolism
3.
Int Immunopharmacol ; 108: 108905, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35729836

ABSTRACT

Studies have confirmed that the heart is the main target organ of lipopolysaccharide (LPS) attacks, and 14-3-3γ and protein kinase C epsilon (PKCε) are the endogenous protective proteins. Puerarin (Pue) is the major bioactive ingredient isolated from the root of Pueraria lobata. It possesses many pharmacological properties, which has been widely used in the treatment and adjuvant therapy of cardio- and cerebrovascular diseases and cancer, etc. The study intended to explore the effects and mechanism of Pue pretreatment to protect myocardium against LPS injury. Adult mice and primary cultured neonatal rat cardiomyocytes were pretreated with Pue, and the injury model was made with LPS. Results showed that Pue pretreatment alleviated LPS-induced injury, as demonstrated by increased cell viability, decreased LDH activity and apoptosis, inhibited excess oxidative stress and the inflammatory cytokine release, and maintained mitochondrial function. Furthermore, Pue pretreatment upregulated 14-3-3γ expression, interacted with PKCε, which was phosphorylated and impelled migration to mitochondria, and then activated adaptive autophagy and protected the myocardium. However, pAD/14-3-3γ-shRNA or 3-MA (an autophagy inhibitor) could weaken the above effects of Pue pretreatment. Together, Pue pretreatment could activate adaptive autophagy by the 14-3-3γ/PKCε pathway and protect the myocardium against LPS injury.


Subject(s)
Heart Injuries , Isoflavones , Animals , Apoptosis , Autophagy , Isoflavones/pharmacology , Isoflavones/therapeutic use , Lipopolysaccharides/pharmacology , Mice , Myocytes, Cardiac/metabolism , Protein Kinase C-epsilon/metabolism , Protein Kinase C-epsilon/pharmacology , Rats , Rats, Sprague-Dawley
4.
Mediators Inflamm ; 2022: 2782080, 2022.
Article in English | MEDLINE | ID: mdl-35125963

ABSTRACT

METHODS: A rat hyperalgesia model was induced using an intraplantar injection of Freund's complete adjuvant (FCA) or an intrathecal injection of IL-6. Mechanical allodynia was evaluated using von Frey filament tests after intrathecal injections of T-5224 (c-Fos/AP-1 inhibitor), minocycline (Mino, a specific microglia inhibitor), L-2-aminoadipic acid (LAA, an astroglial toxin), PKCε inhibitor peptide, APTSTAT3-9R (STAT3 inhibitor), or anti-IL-6 antibody. The c-Fos, GFAP, Iba-1, PKCε, STAT3, pSTAT3Tyr705 and pSTAT3Ser727, and IL-6 expression at the spinal cord level was assessed by Western blot analysis. The interactive effects of PKCε and STAT3 were determined using immunofluorescence staining and immunoprecipitation in vivo and in vitro. Interleukin-6 promoter activity was examined using luciferase assays. RESULTS: T-5224, Mino, and LAA attenuated FCA- or IL-6-mediated inflammatory pain, with a decrease in c-Fos, GFAP, Iba-1, PKCε, and IL-6 expression. PKCε inhibitor peptide and APTSTAT3-9R reversed FCA-induced nociceptive behavior, while decreasing pSTAT3Ser727, IL-6, c-Fos, GFAP, and Iba-1 expression and PKCε and STAT3 coexpression. Interleukin-6 promoter activity increased in the presence of PKCε and STAT3. The interaction with PKCε increased on phosphorylating STAT3 at Ser727 but not at Tyr705. CONCLUSION: STAT3 phosphorylation at Ser 727 and the interaction with PKCε contribute to hyperalgesia via the IL-6-mediated signaling pathway, thus regulating neuron-glia crosstalk during inflammatory pain.


Subject(s)
Hyperalgesia , Interleukin-6 , Animals , Hyperalgesia/metabolism , Interleukin-6/metabolism , Neuroglia/metabolism , Neurons/metabolism , Phosphorylation , Protein Kinase C-epsilon/metabolism , Protein Kinase C-epsilon/pharmacology , Rats , Spinal Cord/metabolism
5.
Cardiovasc Res ; 106(1): 121-30, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25661082

ABSTRACT

AIMS: Sphingosylphosphorylcholine (SPC) elicits vasoconstriction at micromolar concentrations. At lower concentrations (≤1 µmol/L), however, it does not constrict intrapulmonary arteries (IPAs), but strongly potentiates vasoreactivity. Our aim was to determine whether this also occurs in a systemic artery and to delineate the signalling pathway. METHODS AND RESULTS: Rat mesenteric arteries and IPAs mounted on a myograph were challenged with ∼25 mmol/L [K+] to induce a small vasoconstriction. SPC (1 µmol/L) dramatically potentiated this constriction in all arteries by ∼400%. The potentiation was greatly suppressed or abolished by inhibition of phospholipase C (PLC; U73122), PKCε (inhibitory peptide), Src (PP2), and NADPH oxidase (VAS2870), and also by Tempol (superoxide scavenger), but not by inhibition of Rho kinase (Y27632). Potentiation was lost in mesenteric arteries from p47(phox-/-), but not NOX2(-/-), mice. The intracellular superoxide generator LY83583 mimicked the effect of SPC. SPC elevated reactive oxygen species (ROS) in vascular smooth muscle cells, and this was blocked by PP2, VAS2870, and siRNA knockdown of PKCε. SPC (1 µmol/L) significantly reduced the EC50 for U46619-induced vasoconstriction, an action ablated by Tempol. In patch-clamped mesenteric artery cells, SPC (200 nmol/L) enhanced Ba2+ current through L-type Ca2+ channels, an action abolished by Tempol but mimicked by LY83583. CONCLUSION: Our results suggest that low concentrations of SPC activate a PLC-coupled and NOX1-mediated increase in ROS, with consequent enhancement of voltage-gated Ca2+ entry and thus vasoreactivity. We speculate that this pathway is not specific for SPC, but may also contribute to vasoconstriction elicited by other G-protein coupled receptor and PLC-coupled agonists.


Subject(s)
Calcium Channels/drug effects , Mesenteric Arteries/physiology , NADH, NADPH Oxidoreductases/physiology , Phosphorylcholine/analogs & derivatives , Pulmonary Artery/physiology , Reactive Oxygen Species/metabolism , Sphingosine/analogs & derivatives , Vasoconstriction/drug effects , Animals , Calcium Channels/physiology , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Drug , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mesenteric Arteries/drug effects , Mice , Mice, Knockout , Models, Animal , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , NADPH Oxidases/pharmacology , NADPH Oxidases/physiology , Phosphorylcholine/pharmacology , Protein Kinase C-epsilon/pharmacology , Pulmonary Artery/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/pharmacology , Spin Labels , Type C Phospholipases/pharmacology , Vasoconstriction/physiology
6.
Exp Cell Res ; 330(2): 277-286, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25433270

ABSTRACT

RATIONALE: Vessel formation is a crucial event in tissue repair after injury. Thus, one assumption of innovative therapeutic approaches is the understanding of its molecular mechanisms. Notwithstanding our knowledge of the role of Protein Kinase C epsilon (PKCε) in cardio-protection and vascular restenosis, its role in vessel progenitor differentiation remains elusive. OBJECTIVE: Given the availability of PKCε pharmacological modulators already tested in clinical trials, the specific aim of this study is to unravel the role of PKCε in vessel progenitor differentiation, with implications in vascular pathology and vasculogenesis. METHODS AND RESULTS: Mouse Peri-Vascular Adipose Tissue (PVAT) was used as source of mesenchymal vessel progenitors. VEGF-induced differentiation of PVAT cells down-regulates both PKCε and p-PAK1 protein expression levels. PKCε overexpression and activation: i) reduced the expression levels of SMA and PECAM in endothelial differentiation of PVAT cells; ii) completely abrogated tubules formation in collagen gel assays; iii) increased the expression of p-PAK1. CONCLUSION: PKCε negatively interferes with vessel progenitor differentiation via interaction with PAK-1.


Subject(s)
Adipose Tissue/cytology , Endothelial Cells/cytology , Neovascularization, Physiologic/physiology , Protein Kinase C-epsilon/metabolism , p21-Activated Kinases/biosynthesis , Actins/biosynthesis , Adventitia/cytology , Animals , Calcium-Binding Proteins/biosynthesis , Cell Differentiation , Cells, Cultured , Coronary Restenosis/enzymology , Down-Regulation , Enzyme Activation , Mice , Microfilament Proteins/biosynthesis , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Protein Kinase C-epsilon/biosynthesis , Protein Kinase C-epsilon/pharmacology , Smad Proteins/biosynthesis , Vascular Endothelial Growth Factor A/metabolism , Calponins
7.
Neuroscience ; 159(2): 618-28, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19167461

ABSTRACT

Ca(v)2.2 high voltage-gated calcium channels are regulated by phorbol-12-myristae, 13-acetate (PMA) via Ser/Thr protein kinase C (PKC) phosphorylation sites in the I-II linker and C-terminus of the alpha(1) 2.2 subunit. Here we show that PMA enhancement of Ca(v)2.2 currents expressed in Xenopus oocytes can be blocked by inhibitors of PKC betaII or PKC epsilon isozymes, as shown previously for Ca(v)2.3 currents, and that microinjection of PKC betaII or PKC epsilon isozymes in the oocytes expressing the WT Ca(v)2.2 channels increases the basal barium current (I(Ba)). The I-V plot shows a large increase in current amplitude with PKC betaII and PKC epsilon isozymes with only a small shift in the peak I(Ba) in the hyperpolarizing direction. The potentiation of Ca(v)2.2 currents by microinjection of PKC betaII and PKC epsilon isozymes was not altered by the inhibition of G proteins with GDPbetaS. The combination of isozyme specific inhibitors with previously generated Ser/Thr to Ala mutants of alpha(1) 2.2 subunit revealed that PKC betaII or PKC epsilon isozymes (but not PKC alpha or delta) can provide full enhancement through the stimulatory site (Thr-422) in the I-II linker but that PKC epsilon is better at decreasing channel activity through the inhibitory site Ser-425. The enhancing effect of PKC betaII or epsilon at Thr-422 is dominant over the inhibitory effect at Ser-425. Injected PKC betaII also enhances Ca(v)2.2 current when any of the potential stimulatory sites (Ser-1757, Ser-2108 and Ser-2132) are available in the C-terminus. PKC epsilon provides lesser enhancement with C-terminal sites and only with Ser-2108 and Ser-2132. Sites Ser-1757 and Ser-2132, but not Ser-2108, are dominant over the inhibitory site Ser-425. Collectively, these results reveal a hierarchy of regulatory sites in Ca(v)2.2 channels. Site-specific regulation by different PKC isozymes may allow graded levels of channel activation and susceptibility or resistance to subsequent stimulatory events.


Subject(s)
Calcium Channels, N-Type/metabolism , Gene Expression Regulation/physiology , Protein Kinase C-epsilon/metabolism , Protein Kinase C/metabolism , Xenopus Proteins/metabolism , Animals , Aspartic Acid/genetics , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/genetics , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Microinjections/methods , Mutation/genetics , Oocytes , Patch-Clamp Techniques , Phorbol Esters/pharmacology , Phosphorylation/genetics , Protein Kinase C/genetics , Protein Kinase C/pharmacology , Protein Kinase C beta , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/pharmacology , Protein Structure, Tertiary/physiology , RNA, Small Interfering/pharmacology , Serine/genetics , Serine/metabolism , Thionucleotides/pharmacology , Threonine/genetics , Xenopus Proteins/drug effects , Xenopus Proteins/genetics , Xenopus laevis
8.
Blood ; 107(2): 508-13, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16166586

ABSTRACT

Apoptosis plays a central role in the regulation of the size of the hematopoietic stem cell pool as well as in the processes of cell differentiation along the various hematopoietic lineages. TRAIL is a member of the TNF family of cytokines with a known apoptogenic role against a variety of malignant cells and an emerging role in the modulation of normal hematopoiesis. Here we worked on the hypothesis that PKCepsilon could act as a switch of the cellular response to TRAIL during erythropoiesis. We demonstrate that EPO-induced erythroid CD34 cells are insensitive to the apoptogenic effect of TRAIL at day 0 due to the lack of specific receptor expression. From day 3 onward, erythroid cells express surface death receptors and become sensitive to TRAIL up to day 7/8 when, notwithstanding death-receptor expression, the EPO-driven up-regulation of PKCepsilon intracellular levels renders differentiating erythroid cells resistant to TRAIL likely via Bcl-2 up-regulation. Our conclusion is that in human CD34 cells, EPO promotes a series of events that, being finely regulated in their kinetics, restricts the sensitivity of these cells to TRAIL to a specific period of time, which therefore represents the "TRAIL window" for the negative regulation of erythroid-cell numbers.


Subject(s)
Antineoplastic Agents/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , Membrane Glycoproteins/metabolism , Protein Kinase C-epsilon/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Antigens, CD34/metabolism , Blotting, Western , Erythroid Precursor Cells/cytology , Erythropoietin/pharmacology , Flow Cytometry , Humans , Kinetics , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...