Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Neurosci Res ; 97(4): 444-455, 2019 04.
Article in English | MEDLINE | ID: mdl-30488977

ABSTRACT

Global cerebral ischemia that accompanies cardiac arrest is a major cause of morbidity and mortality. Protein Kinase C epsilon (PKCε) is a member of the novel PKC subfamily and plays a vital role in ischemic preconditioning. Pharmacological activation of PKCε before cerebral ischemia confers neuroprotection. The role of endogenous PKCε after cerebral ischemia remains elusive. Here we used male PKCε-null mice to assess the effects of PKCε deficiency on neurodegeneration after transient global cerebral ischemia (tGCI). We found that the cerebral vasculature, blood flow, and the expression of other PKC isozymes were not altered in the PKCε-null mice. Spatial learning and memory was impaired after tGCI, but the impairment was attenuated in male PKCε-null mice as compared to male wild-type controls. A significant reduction in Fluoro-Jade C labeling and mitochondrial release of cytochrome C in the hippocampus was found in male PKCε-null mice after tGCI. Male PKCε-null mice expressed increased levels of PKCδ in the mitochondria, which may prevent the translocation of PKCδ from the cytosol to the mitochondria after tGCI. Our results demonstrate the neuroprotective effects of PKCε deficiency on neurodegeneration after tGCI, and suggest that reduced mitochondrial translocation of PKCδ may contribute to the neuroprotective action in male PKCε-null mice.


Subject(s)
Hippocampus/metabolism , Ischemic Attack, Transient/metabolism , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/physiology , Animals , Brain/pathology , Cytosol/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Protein Kinase C-epsilon/metabolism , Spatial Learning , Spatial Memory
2.
J Alzheimers Dis ; 63(3): 1173-1189, 2018.
Article in English | MEDLINE | ID: mdl-29710707

ABSTRACT

Oxidative stress and amyloid-ß (Aß) oligomers have been implicated in Alzheimer's disease (AD). The growth and maintenance of neuronal networks are influenced by brain derived neurotrophic factor (BDNF) expression, which is promoted by protein kinase C epsilon (PKCɛ). We investigated the reciprocal interaction among oxidative stress, Aß, and PKCɛ levels and subsequent PKCɛ-dependent MnSOD and BDNF expression in hippocampal pyramidal neurons. Reduced levels of PKCɛ, MnSOD, and BDNF and an increased level of Aß were also found in hippocampal neurons from autopsy-confirmed AD patients. In cultured human primary hippocampal neurons, spherical aggregation of Aß (amylospheroids) decreased PKCɛ and MnSOD. Treatment with t-butyl hydroperoxide (TBHP) increased superoxide, the oxidative DNA/RNA damage marker, 8-OHG, and Aß levels, but reduced PKCɛ, MnSOD, BDNF, and cultured neuron density. These changes were reversed with the PKCɛ activators, bryostatin and DCPLA-ME. PKCɛ knockdown suppressed PKCɛ, MnSOD, and BDNF but increased Aß. In cultured neurons, the increase in reactive oxygen species (ROS) associated with reduced PKCɛ during neurodegeneration was inhibited by the SOD mimetic MnTMPyP and the ROS scavenger NAc, indicating that strong oxidative stress suppresses PKCɛ level. Reduction of PKCɛ and MnSOD was prevented with the PKCɛ activator bryostatin in 5-6-month-old Tg2576 AD transgenic mice. In conclusion, oxidative stress and Aß decrease PKCɛ expression. Reciprocally, a depression of PKCɛ reduces BDNF and MnSOD, resulting in oxidative stress. These changes can be prevented with the PKCɛ-specific activators.


Subject(s)
Alzheimer Disease/pathology , Brain-Derived Neurotrophic Factor/metabolism , Down-Regulation/physiology , Hippocampus/pathology , Neurons/metabolism , Protein Kinase C-epsilon/deficiency , Adjuvants, Immunologic/pharmacology , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Bryostatins/metabolism , Bryostatins/pharmacology , Cells, Cultured , Female , Fetus/anatomy & histology , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Metalloporphyrins/pharmacology , Mice , Middle Aged , Morpholinos/pharmacology , Protein Kinase C-epsilon/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Transfection , tert-Butylhydroperoxide/pharmacology
3.
PLoS One ; 12(8): e0182867, 2017.
Article in English | MEDLINE | ID: mdl-28783756

ABSTRACT

BACKGROUND: It has long been postulated that Protein Kinase C (PKC) is an important regulator of megakaryopoiesis. Recent contributions to the literature have outlined the functions of several individual PKC isoforms with regard to megakaryocyte differentiation and platelet production. However, the exact role of PKCε remains elusive. OBJECTIVE: To delineate the role of PKCε in megakaryopoiesis. APPROACH AND RESULTS: We used a PKCε knockout mouse model to examine the effect of PKCε deficiency on platelet mass, megakaryocyte mass, and bone marrow progenitor cell distribution. We also investigated platelet recovery in PKCε null mice and TPO-mediated signaling in PKCε null megakaryocytes. PKCε null mice have higher platelet counts due to increased platelet production compared to WT littermate controls (p<0.05, n = 8). Furthermore, PKCε null mice have more bone marrow megakaryocyte progenitor cells than WT littermate control mice. Additionally, thrombopoietin-mediated signaling is perturbed in PKCε null mice as Akt and ERK1/2 phosphorylation are enhanced in PKCε null megakaryocytes stimulated with thrombopoietin. Finally, in response to immune-induced thrombocytopenia, PKCε null mice recovered faster and had higher rebound thrombocytosis than WT littermate control mice. CONCLUSIONS: Enhanced platelet recovery could be due to an increase in megakaryocyte progenitor cells found in PKCε null mice as well as enhanced thrombopoietin-mediated signaling observed in PKCε deficient megakaryocytes. These data suggest that PKCε is a negative regulator of megakaryopoiesis.


Subject(s)
Gene Knockout Techniques , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , Stem Cells/cytology , Stem Cells/metabolism , Thrombopoiesis , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Megakaryocytes/cytology , Megakaryocytes/drug effects , Mice , Platelet Count , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cells/drug effects , Thrombocytopenia/enzymology , Thrombocytopenia/immunology , Thrombopoiesis/drug effects , Thrombopoiesis/genetics , Thrombopoietin/pharmacology
4.
J Cell Mol Med ; 21(9): 2009-2021, 2017 09.
Article in English | MEDLINE | ID: mdl-28266127

ABSTRACT

Mitochondrial fission is critically involved in cardiomyocyte apoptosis, which has been considered as one of the leading causes of ischaemia/reperfusion (I/R)-induced myocardial injury. In our previous works, we demonstrate that aldehyde dehydrogenase-2 (ALDH2) deficiency aggravates cardiomyocyte apoptosis and cardiac dysfunction. The aim of this study was to elucidate whether ALDH2 deficiency promotes mitochondrial injury and cardiomyocyte death in response to I/R stress and the underlying mechanism. I/R injury was induced by aortic cross-clamping for 45 min. followed by unclamping for 24 hrs in ALDH2 knockout (ALDH2-/- ) and wild-type (WT) mice. Then myocardial infarct size, cell apoptosis and cardiac function were examined. The protein kinase C (PKC) isoform expressions and their mitochondrial translocation, the activity of dynamin-related protein 1 (Drp1), caspase9 and caspase3 were determined by Western blot. The effects of N-acetylcysteine (NAC) or PKC-δ shRNA treatment on glycogen synthase kinase-3ß (GSK-3ß) activity and mitochondrial permeability transition pore (mPTP) opening were also detected. The results showed that ALDH2-/- mice exhibited increased myocardial infarct size and cardiomyocyte apoptosis, enhanced levels of cleaved caspase9, caspase3 and phosphorylated Drp1. Mitochondrial PKC-ε translocation was lower in ALDH2-/- mice than in WT mice, and PKC-δ was the opposite. Further data showed that mitochondrial PKC isoform ratio was regulated by cellular reactive oxygen species (ROS) level, which could be reversed by NAC pre-treatment under I/R injury. In addition, PKC-ε inhibition caused activation of caspase9, caspase3 and Drp1Ser616 in response to I/R stress. Importantly, expression of phosphorylated GSK-3ß (inactive form) was lower in ALDH2-/- mice than in WT mice, and both were increased by NAC pre-treatment. I/R-induced mitochondrial translocation of GSK-3ß was inhibited by PKC-δ shRNA or NAC pre-treatment. In addition, mitochondrial membrane potential (∆Ψm ) was reduced in ALDH2-/- mice after I/R, which was partly reversed by the GSK-3ß inhibitor (SB216763) or PKC-δ shRNA. Collectively, our data provide the evidence that abnormal PKC-ε/PKC-δ ratio promotes the activation of Drp1 signalling, caspase cascades and GSK-3ß-dependent mPTP opening, which results in mitochondrial injury-triggered cardiomyocyte apoptosis and myocardial dysfuction in ALDH2-/- mice following I/R stress.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Myocardium/pathology , Protein Kinase C-epsilon/deficiency , Aldehyde Dehydrogenase, Mitochondrial/deficiency , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Animals , Apoptosis , Enzyme Activation , Isoenzymes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Permeability Transition Pore , Models, Biological , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
5.
Oncotarget ; 7(22): 33069-80, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27102301

ABSTRACT

PKCε is a transforming oncogene and a predictive biomarker of various human cancers. However, a precise in vivo link of PKCε to cancer induction, progression and metastasis remain undefined. To achieve these goals, we generated tissue specific conditional PKCε knockout mice (PKCε-CKO) using cre-lox technology. Homozygous PKCε(LoxP/LoxP) mice have normal body weight and phenotype. To determine what effect loss of PKCε would have on the prostate, the PKCε(LoxP/LoxP) mice were bred to probasin cre (PB-Cre4+) mice which express cre specifically in the prostate epithelium of postnatal mice. Western blot and immunohistochemical analyses showed reduced levels of PKCε specifically in the prostate of PKCε-CKO mice. Histopathological analyses of prostate from both PKCε(LoxP/LoxP) and prostate PKCε-CKO mice showed normal pathology. To determine the functional impact of prostate specific deletion of PKCε on prostate tumor growth, we performed an orthotopic xenograft study. Transgenic adenocarcinoma of the mouse prostate (TRAMP) cells (TRAMPC1, 2×106) were implanted in the prostate of PKCε-CKO mice. Mice were sacrificed at 6th week post-implantation. Results demonstrated a significant (P<0.05) decrease in the growth of TRAMPC1 cells-derived xenograft tumors in PKCε-CKO mice compared to wild type. To determine a link of PKCε to ultraviolet radiation (UVR) exposure-induced epidermal Stat3 phosphorylation, PKCε(LoxP/LoxP) mice were bred to tamoxifen-inducible K14 Cre mice. PKCε deletion in the epidermis resulted in inhibition of UVR-induced Stat3 phosphorylation. In summary, our novel PKCε(LoxP/LoxP) mice will be useful for defining the link of PKCε to various cancers in specific organ, tissue, or cells.


Subject(s)
Prostatic Neoplasms/genetics , Protein Kinase C-epsilon/metabolism , Animals , Disease Progression , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Prostatic Neoplasms/pathology , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics
6.
J Mol Cell Cardiol ; 72: 281-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24713463

ABSTRACT

Up-regulation and activation of PYK2, a member of the FAK family of protein tyrosine kinases, is involved in the pathogenesis of left ventricular (LV) remodeling and heart failure (HF). PYK2 activation can be prevented by CRNK, the C-terminal domain of PYK2. We previously demonstrated that adenoviral-mediated CRNK gene transfer improved survival and LV function, and slowed LV remodeling in a rat model of coronary artery ligation-induced HF. We now interrogate whether cardiomyocyte-specific, transgenic CRNK expression prevents LV remodeling and HF in a mouse model of dilated cardiomyopathy (DCM) caused by constitutively active Protein Kinase Cε (caPKCε). Transgenic (TG; FVB/N background) mice were engineered to express rat CRNK under control of the α-myosin heavy chain promoter, and crossed with FVB/N mice with cardiomyocyte-specific expression of caPKCε to create double TG mice. LV structure, function, and gene expression were evaluated in all 4 groups (nonTG FVB/N; caPKCε(+/-); CRNK(+/-); and caPKCε×CRNK (PXC) double TG mice) at 1, 3, 6, 9 and 12mo of age. CRNK expression followed a Mendelian distribution, and CRNK mice developed and survived normally through 12mo. Cardiac structure, function and selected gene expression of CRNK mice were similar to nonTG littermates. CRNK had no effect on caPKCε expression and vice versa. PYK2 was up-regulated ~6-fold in caPKCε mice, who developed a non-hypertrophic, progressive DCM with reduced systolic (Contractility Index=151±5 vs. 90±4s(-1)) and diastolic (Tau=7.5±0.5 vs. 14.7±1.3ms) function, and LV dilatation (LV Remodeling Index (LVRI)=4.2±0.1 vs. 6.0±0.3 for FVB/N vs. caPKCε mice, respectively; P<0.05 for each at 12mo). In double TG PXC mice, CRNK expression significantly prolonged survival, improved contractile function (Contractile Index=115±8s(-1); Tau=9.5±1.0ms), and reduced LV remodeling (LVRI=4.9±0.1). Cardiomyocyte-specific expression of CRNK improves contractile function and slows LV remodeling in a mouse model of DCM.


Subject(s)
Cardiomyopathy, Dilated/genetics , Focal Adhesion Kinase 2/genetics , Myocytes, Cardiac/metabolism , Transgenes , Ventricular Function/physiology , Ventricular Remodeling , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Focal Adhesion Kinase 2/deficiency , Gene Expression Regulation , Heart Ventricles/metabolism , Heart Ventricles/pathology , Longevity , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Promoter Regions, Genetic , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , Protein Structure, Tertiary
7.
Exp Lung Res ; 38(8): 383-95, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22897707

ABSTRACT

Organic dust samples from swine confinement facilities elicit pro-inflammatory cytokine/chemokine release from bronchial epithelial cells and monocytes, dependent, in part, upon dust-induced activation of the protein kinase C (PKC) isoform, PKCε. PKCε is also rapidly activated in murine tracheal epithelial cells following in vivo organic dust challenges, yet the functional role of PKCε in modulating dust-induced airway inflammatory outcomes is not defined. Utilizing an established intranasal inhalation animal model, experiments investigated the biologic and physiologic responses following organic dust extract (ODE) treatments in wild-type (WT) and PKCε knock-out (KO) mice. We found that neutrophil influx increased more than twofold in PKCε KO mice following both a one-time challenge and 3 weeks of daily challenges with ODE as compared with WT mice. Lung pathology revealed increased bronchiolar and alveolar inflammation, lymphoid aggregates, and T cell influx in ODE-treated PKCε KO mice. Airway hyperresponsiveness to methacholine increased in PKCε KO + ODE to a greater magnitude than WT + ODE animals. There were no significant differences in cytokine/chemokine release elicited by ODE treatment between groups. However, ODE-induced nitric oxide (NO) production differed in that ODE exposure increased nitrate levels in WT mice but not in PKCε KO mice. Moreover, ODE failed to upregulate NO from ex vivo stimulated PKCε KO lung macrophages. Collectively, these studies demonstrate that PKCε-deficient mice were hypersensitive to organic dust exposure and suggest that PKCε is important in the normative lung inflammatory response to ODE. Dampening of ODE-induced NO may contribute to these enhanced inflammatory findings.


Subject(s)
Bronchial Hyperreactivity/enzymology , Dust/immunology , Inflammation Mediators/metabolism , Lung/drug effects , Protein Kinase C-epsilon/biosynthesis , Animals , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/immunology , Bronchial Provocation Tests , Chemokines/metabolism , Disease Models, Animal , Environmental Exposure/adverse effects , Enzyme Activation/drug effects , Enzyme Activation/immunology , Female , Lung/metabolism , Lung/pathology , Male , Methacholine Chloride , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , T-Lymphocytes/pathology
8.
Diabetologia ; 55(10): 2789-2793, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22814763

ABSTRACT

AIMS/HYPOTHESIS: Protein kinase Cε (PKCε) is emerging as a key mediator of lipid-induced insulin resistance in liver and hepatic lipid metabolism itself. We investigated whether PKCε plays a role in other metabolic processes, to further examine its suitability as a therapeutic target. METHODS: We measured amino acid, organic acid and sugar levels by liquid and gas chromatography-mass spectrometry of liver extracts from chow and fat-fed wild-type (WT) and PKCε-deficient (Prkce(-/-)) mice. Fed and fasting glucose, ketone and fatty acid levels were measured in blood. Triacylglycerol levels and gluconeogenic and ketogenic enzyme expression were measured in liver. The effect of fasting on epididymal fat pad mass was also determined. RESULTS: Metabolomic analysis indicated that the short-term high-fat diet affected over 20 compounds, including a 50% reduction in the glucogenic amino acid alanine. Prkce deletion resulted only in a reduction of 4-hydroxyproline and aspartate and an increase in glutamate. However, upon fasting, Prkce(-/-) mice were better able to maintain blood glucose levels and also exhibited lower levels of the ketone ß-hydroxybutyrate compared with WT mice. Upon fasting, Prkce deletion also resulted in lower liver and plasma lipids and a smaller reduction in fat pad mass. CONCLUSIONS/INTERPRETATION: Metabolomic analysis provided new insights into the effects of a high-fat diet on liver metabolite levels. Glucose homeostasis under fasting conditions is improved in Prkce(-/-) mice, which, in turn, may reduce the mobilisation of lipid from adipose tissue, reducing the availability of ketogenic substrate in the liver. Together with the protection against fat-diet-induced glucose intolerance previously observed in the fed state, these findings indicate PKCε as a unique therapeutic target for the improvement of glucose homeostasis.


Subject(s)
Fasting/metabolism , Gluconeogenesis/physiology , Ketones/metabolism , Liver/metabolism , Protein Kinase C-epsilon/deficiency , Animals , Fatty Acids/metabolism , Gene Deletion , Hemostasis/physiology , Insulin Resistance/physiology , Mice , Mice, Knockout , Models, Animal , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/physiology
9.
Br J Pharmacol ; 164(2b): 755-71, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21486279

ABSTRACT

BACKGROUND AND PURPOSE: Cyclical phosphorylation and dephosphorylation of a key residue within the C-terminal domain of the activated type 5 metabotropic glutamate (mGlu5) receptor is believed to cause the synchronous, oscillatory changes in inositol 1,4,5-trisphosphate and Ca²âº levels observed in a variety of cell types. Here, we have attempted to better define the kinase and phosphatase enzymes involved in this modulation. EXPERIMENTAL APPROACH: Ca²âº and [³H]inositol phosphate ([³H]IP(x) ) measurements in astrocyte preparations have been used to evaluate the effects of pharmacological inhibition of protein kinase C (PKC) and protein phosphatase activities and small interfering RNA-mediated specific PKC isoenzymic knock-down on mGlu5 receptor signalling. KEY RESULTS: Ca²âº oscillation frequency or [³H]IP(x) accumulation in astrocytes stimulated by mGlu5 receptors, was concentration-dependently decreased by protein phosphatase-1/2A inhibition or by PKC activation. PKC inhibition also increased [³H]IP(x) accumulation two- to threefold and changed the Ca²âº response into a peak-plateau response. However, selective inhibition of conventional PKC isoenzymes or preventing changes in [Ca²âº](i) concentration by BAPTA-AM loading was without effect on mGlu5 receptor-stimulated [³H]IP(x) accumulation. Selective knock-down of PKCδ was without effect on glutamate-stimulated Ca²âº responses; however, selective PKCε knock-down in astrocytes changed Ca²âº responses from oscillatory into peak-plateau type. CONCLUSION AND IMPLICATIONS: These data confirm the acute regulation of mGlu5 receptor signalling by protein kinases and protein phosphatases and provide novel data pinpointing the isoenzymic dependence of this regulation in the native mGlu5 receptor-expressing rat cortical astrocyte. These data also highlight a potential alternative mechanism by which mGlu5 receptor signalling might be therapeutically manipulated.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Protein Kinase C-epsilon/metabolism , Protein Kinase C/metabolism , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism , Receptors, Metabotropic Glutamate/metabolism , Type C Phospholipases/metabolism , Animals , Calcium Signaling , Gene Knockdown Techniques/methods , Inositol Phosphates/metabolism , Isoenzymes , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 2/antagonists & inhibitors , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics , Signal Transduction
10.
Diabetologia ; 54(6): 1447-56, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21347625

ABSTRACT

AIMS/HYPOTHESIS: We examined the time-dependent effects of deletion of the gene encoding protein kinase C epsilon (Prkce) on glucose homeostasis, insulin secretion and hepatic lipid metabolism in fat-fed mice. METHODS: Prkce(-/-) and wild-type (WT) mice were fed a high-fat diet for 1 to 16 weeks and subjected to i.p. glucose tolerance tests (ipGTT) and indirect calorimetry. We also investigated gene expression and protein levels by RT-PCR, quantitative protein profiling (isobaric tag for relative and absolute quantification; iTRAQ) and immunoblotting. Lipid levels, mitochondrial oxidative capacity and lipid metabolism were assessed in liver and primary hepatocytes. RESULTS: While fat-fed WT mice became glucose intolerant after 1 week, Prkce(-/-) mice exhibited normal glucose and insulin levels. iTRAQ suggested differences in lipid metabolism and oxidative phosphorylation between fat-fed WT and Prkce(-/-) animals. Liver triacylglycerols were increased in fat-fed Prkce(-/-) mice, resulting from altered lipid partitioning which promoted esterification of fatty acids in hepatocytes. In WT mice, fat feeding elevated oxygen consumption in vivo and in isolated liver mitochondria, but these increases were not seen in Prkce(-/-) mice. Prkce(-/-) hepatocytes also exhibited reduced production of reactive oxygen species (ROS) in the presence of palmitate. After 16 weeks of fat feeding, however, the improved glucose tolerance in fat-fed Prkce(-/-) mice was instead associated with increased insulin secretion during ipGTT, as we have previously reported. CONCLUSIONS/INTERPRETATION: Prkce deletion ameliorates diet-induced glucose intolerance via two temporally distinct phenotypes. Protection against insulin resistance is associated with changes in hepatic lipid partitioning, which may reduce the acute inhibitory effects of fatty acid catabolism, such as ROS generation. In the longer term, enhancement of glucose-stimulated insulin secretion prevails.


Subject(s)
Dietary Fats/metabolism , Glucose/metabolism , Homeostasis/physiology , Lipid Metabolism/physiology , Liver/metabolism , Protein Kinase C-epsilon/deficiency , Animals , Gene Deletion , Insulin/metabolism , Mice , Mice, Knockout , Models, Animal , Protein Kinase C-epsilon/genetics , Reactive Oxygen Species/metabolism , Time Factors
11.
Circ Res ; 107(3): 365-73, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20538683

ABSTRACT

RATIONALE: Epidemiological studies demonstrate a clear association of adverse intrauterine environment with an increased risk of ischemic heart disease in adulthood. Hypoxia is a common stress to the fetus and results in decreased protein kinase C epsilon (PKCepsilon) expression in the heart and increased cardiac vulnerability to ischemia and reperfusion injury in adult offspring in rats. OBJECTIVES: The present study tested the hypothesis that fetal hypoxia-induced methylation of cytosine-phosphate-guanine dinucleotides at the PKCepsilon promoter is repressive and contributes to PKCepsilon gene repression in the heart of adult offspring. METHODS AND RESULTS: Hypoxic treatment of pregnant rats from days 15 to 21 of gestation resulted in significant decreases in PKCepsilon protein and mRNA in fetal hearts. Similar results were obtained in ex vivo hypoxic treatment of isolated fetal hearts and rat embryonic ventricular myocyte cell line H9c2. Increased methylation of PKCepsilon promoter at SP1 binding sites, -346 and -268, were demonstrated in both fetal hearts of maternal hypoxia and H9c2 cells treated with 1% O(2) for 24 hours. Whereas hypoxia had no significant effect on the binding affinity of SP1 to the unmethylated sites in H9c2 cells, hearts of fetuses and adult offspring, methylation of both SP1 sites reduced SP1 binding. The addition of 5-aza-2'-deoxycytidine blocked the hypoxia-induced increase in methylation of both SP1 binding sites and restored PKCepsilon mRNA and protein to the control levels. In hearts of both fetuses and adult offspring, hypoxia-induced methylation of SP1 sites was significantly greater in males than in females, and decreased PKCepsilon mRNA was seen only in males. In fetal hearts, there was significantly higher abundance of estrogen receptor alpha and beta isoforms in females than in males. Both estrogen receptor alpha and beta interacted with the SP1 binding sites in the fetal heart, which may explain the sex differences in SP1 methylation in the fetal heart. Additionally, selective activation of PKCepsilon restored the hypoxia-induced cardiac vulnerability to ischemic injury in offspring. CONCLUSIONS: The findings demonstrate a direct effect of hypoxia on epigenetic modification of DNA methylation and programming of cardiac PKCepsilon gene repression in a sex-dependent manner, linking fetal hypoxia and pathophysiological consequences in the hearts of adult offspring.


Subject(s)
Protein Kinase C-epsilon/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Western , DNA Methylation/genetics , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Epigenesis, Genetic/genetics , Female , Fetal Heart/physiopathology , Fetal Hypoxia/enzymology , Fetal Hypoxia/genetics , Gene Expression Regulation, Enzymologic , Male , Methylation , Pregnancy , Promoter Regions, Genetic/genetics , Protein Kinase C-epsilon/deficiency , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/metabolism
12.
Diabetologia ; 52(12): 2616-20, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19809797

ABSTRACT

AIMS/HYPOTHESIS: This study aimed to determine whether protein kinase C (PKC) delta plays a role in the glucose intolerance caused by a high-fat diet, and whether it could compensate for loss of PKCepsilon in the generation of insulin resistance in skeletal muscle. METHODS: Prkcd (-/-), Prkce (-/-) and wild-type mice were fed high-fat diets and subjected to glucose tolerance tests. Blood glucose levels and insulin responses were determined during the tests. Insulin signalling in liver and muscle was assessed after acute in vivo insulin stimulation by immunoblotting with phospho-specific antibodies. Activation of PKC isoforms in muscle from Prkce (-/-) mice was assessed by determining intracellular distribution. Tissues and plasma were assayed for triacylglycerol accumulation, and hepatic production of lipogenic enzymes was determined by immunoblotting. RESULTS: Both Prkcd (-/-) and Prkce (-/-) mice were protected against high-fat-diet-induced glucose intolerance. In Prkce (-/-) mice this was mediated through enhanced insulin availability, while in Prkcd (-/-) mice the reversal occurred in the absence of elevated insulin. Neither the high-fat diet nor Prkcd deletion affected maximal insulin signalling. The activation of PKCdelta in muscle from fat-fed mice was enhanced by Prkce deletion. PKCdelta-deficient mice exhibited reduced liver triacylglycerol accumulation and diminished production of lipogenic enzymes. CONCLUSIONS/INTERPRETATION: Deletion of genes encoding isoforms of PKC can improve glucose intolerance, either by enhancing insulin availability in the case of Prkce, or by reducing lipid accumulation in the case of Prkcd. The absence of PKCepsilon in muscle may be compensated by increased activation of PKCdelta in fat-fed mice, suggesting that an additional role for PKCepsilon in this tissue is masked.


Subject(s)
Dietary Fats/adverse effects , Glucose Intolerance/chemically induced , Protein Kinase C-delta/deficiency , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/metabolism , Animals , Blood Glucose/metabolism , Crosses, Genetic , Gene Deletion , Glucose Intolerance/blood , Glucose Intolerance/enzymology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Muscle, Skeletal/enzymology , Protein Kinase C-delta/genetics , Protein Kinase C-epsilon/genetics , Triglycerides/metabolism
13.
Cell Signal ; 21(11): 1680-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19632318

ABSTRACT

Phosphorylation and dephosphorylation of PKCs can regulate their activity, stability and function. We have previously shown that downregulation of PKC delta by tumor promoting phorbol esters was compromised when HeLa cells acquired resistance to cisplatin (HeLa/CP). In the present study, we have used these cells to understand the mechanism of PKC delta downregulation. A brief treatment of HeLa cells with phorbol 12,13-dibutyrate (PDBu) induced phosphorylation of PKC delta at the activation loop (Thr505), turn motif (Ser643), hydrophobic motif (Ser662) and Tyr-311 sites to a greater extent in HeLa/CP cells compared to HeLa cells. Prolonged treatment with PDBu led to downregulation of PKC delta in HeLa but not in HeLa/CP cells. The PKC inhibitor Gö 6983 inhibited PDBu-induced downregulation of PKC delta, decreased Thr505 phosphorylation and increased PKC delta tyrosine phosphorylation at Tyr-311 site. However, knockdown of c-Abl, c-Src, Fyn and Lyn had little effect on PKC delta downregulation and Tyr311 phosphorylation. Pretreatment with the phosphatidylinositol 3-kinase inhibitor Ly294002 and mTOR inhibitor rapamycin restored the ability of PDBu to downregulate PKC delta in HeLa/CP cells. Knockdown of mTOR and rictor but not raptor facilitated PKC delta downregulation. Depletion of PKC epsilon also enhanced PKC delta downregulation by PDBu. These results suggest that downregulation of PKC delta is regulated by PKC epsilon and mammalian target of rapamycin complex 2 (mTORC2).


Subject(s)
Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Transcription Factors/metabolism , Carbazoles/pharmacology , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm , Gene Knockdown Techniques , HeLa Cells , Humans , Indoles , Maleimides , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , RNA, Small Interfering/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
14.
Diabetes ; 58(8): 1826-34, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19401415

ABSTRACT

OBJECTIVE: Insufficient insulin secretion is a hallmark of type 2 diabetes, and exposure of beta-cells to elevated lipid levels (lipotoxicity) contributes to secretory dysfunction. Functional ablation of protein kinase C epsilon (PKCepsilon) has been shown to improve glucose homeostasis in models of type 2 diabetes and, in particular, to enhance glucose-stimulated insulin secretion (GSIS) after lipid exposure. Therefore, we investigated the lipid-dependent mechanisms responsible for the enhanced GSIS after inactivation of PKCepsilon. RESEARCH DESIGN AND METHODS: We cultured islets isolated from PKCepsilon knockout (PKCepsilonKO) mice in palmitate prior to measuring GSIS, Ca(2+) responses, palmitate esterification products, lipolysis, lipase activity, and gene expression. RESULTS: The enhanced GSIS could not be explained by increased expression of another PKC isoform or by alterations in glucose-stimulated Ca(2+) influx. Instead, an upregulation of the amplifying pathways of GSIS in lipid-cultured PKCepsilonKO beta-cells was revealed under conditions in which functional ATP-sensitive K(+) channels were bypassed. Furthermore, we showed increased esterification of palmitate into triglyceride pools and an enhanced rate of lipolysis and triglyceride lipase activity in PKCepsilonKO islets. Acute treatment with the lipase inhibitor orlistat blocked the enhancement of GSIS in lipid-cultured PKCepsilonKO islets, suggesting that a lipolytic product mediates the enhancement of glucose-amplified insulin secretion after PKCepsilon deletion. CONCLUSIONS: Our findings demonstrate a mechanistic link between lipolysis and the amplifying pathways of GSIS in murine beta-cells, and they suggest an interaction between PKCepsilon and lipolysis. These results further highlight the therapeutic potential of PKCepsilon inhibition to enhance GSIS from the beta-cell under conditions of lipid excess.


Subject(s)
Glucose/pharmacology , Insulin-Secreting Cells/physiology , Insulin/metabolism , Lipolysis/physiology , Protein Kinase C-epsilon/deficiency , Animals , Crosses, Genetic , Female , Gene Deletion , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Lipolysis/genetics , Male , Mice , Mice, Knockout , Protein Kinase C-epsilon/genetics
15.
Int Heart J ; 50(2): 191-206, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19367030

ABSTRACT

This experimental study investigated the impact of hyperglycemic control on left ventricular (LV) function using a model of diabetes mellitus (DM) (induced by streptozocin 60 mg/kg). Sixteen adult-Sprague Dawley rats were divided into group 1 (poor hyperglycemic control, n = 8) and group 2 (good hyperglycemic control, n = 8). Diabetic rats and 8 healthy rats serving as controls (group 3) were sacrificed on day 28 after DM induction. The results demonstrated that HbA(1C) on day 28 was higher in group 1 than in groups 2 and 3 (P < 0.0001). The mRNA expressions of MMP-9 and endothelin-1 were elevated in group 1 compared with that in groups 2 and 3 (P < 0.05), whereas PGC-1alpha and eNOS were lower in group 1 than in groups 2 and 3 (P < 0.05). The number of apoptotic nuclei was higher in group 1 than in groups 2 and 3 (P < 0.01). The integrated area (microm(2)) of connexin43 (Cx43), Cx43 protein expression, and LV function were lower in group 1 than in groups 2 and 3 (P < 0.05). Moreover, PKC-epsilon expression in the mitochondrial compartment was decreased in group 1 compared to that in groups 2 and 3 (P < 0.005).


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Heart Ventricles/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Myocardium/metabolism , Ventricular Function, Left/drug effects , Animals , Apoptosis/drug effects , Connexin 43/genetics , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Endothelin-1/biosynthesis , Endothelin-1/drug effects , Hemoglobin A/biosynthesis , Hemoglobin A/drug effects , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinase C-epsilon/deficiency , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA-Binding Proteins , Rats , Rats, Sprague-Dawley , Transcription Factors/deficiency
16.
Neuropsychopharmacology ; 34(7): 1733-42, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19158669

ABSTRACT

The cannabinoid CB1 receptor (CB1) is one of the most abundant G protein-coupled receptors in the brain, but little is known about the mechanisms that modulate CB1 receptor signaling. Here, we show that inhibition or null mutation of the epsilon isozyme of protein kinase C (PKCepsilon) selectively enhances behavioral responses to the CB1 agonist WIN55,212-2 in mice, but not to the structurally unrelated CB1 agonist CP55,940. Binding affinity for [(3)H] WIN55,212-2 was increased in brain membranes from PKCepsilon(-/-) mice compared with PKCepsilon(+/+) mice. There was no difference in binding of the inverse agonist [(3)H] SR141716A. In addition, repeated administration of WIN55,212-2 produced greater analgesic and thermal tolerance in PKCvarepsilon(-/-) mice compared with PKCepsilon(+/+)mice. These results indicate that PKCvarepsilon selectively regulates behavioral sensitivity, CB1 receptor binding and tolerance to WIN55,212-2.


Subject(s)
Behavior, Animal/drug effects , Benzoxazines/pharmacology , Drug Tolerance/genetics , Morpholines/pharmacology , Naphthalenes/pharmacology , Protein Kinase C-epsilon/metabolism , Receptor, Cannabinoid, CB1/agonists , Analgesia , Analgesics/pharmacology , Animals , Behavior, Animal/physiology , Competitive Bidding/methods , Cyclohexanols/pharmacology , Dose-Response Relationship, Drug , Drug Tolerance/physiology , Enzyme Inhibitors/pharmacology , Hypothermia/chemically induced , Hypothermia/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Peptides/pharmacology , Protein Binding/drug effects , Protein Kinase C-epsilon/deficiency , Serine/genetics , Serine/metabolism , Tritium/metabolism
17.
Arch Biochem Biophys ; 482(1-2): 104-11, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19022218

ABSTRACT

Steatosis is a critical stage in the pathology of alcoholic liver disease (ALD), and preventing steatosis could protect against later stages of ALD. PKCepsilon has been shown to contribute to hepatic steatosis in experimental non-alcoholic fatty liver disease (NAFLD); however, the role of PKCepsilon in ethanol-induced steatosis has not been determined. The purpose of this study was to therefore test the hypothesis that PKCepsilon contributes to ethanol-induced steatosis. Accordingly, the effect of acute ethanol on indices of hepatic steatosis and insulin signaling were determined in PKCepsilon knockout mice and in wild-type mice that received an anti-sense oligonucleotide (ASO) to knockdown PKCepsilon expression. Acute ethanol (6g/kg i.g.) caused a robust increase in hepatic non-esterified free fatty acids (NEFA), which peaked 1h after ethanol exposure. This increase in NEFA was followed by elevated diacylglycerols (DAG), as well as by the concomitant activation of PKCepsilon. Acute ethanol also changed the expression of insulin-responsive genes (i.e. increased G6Pase, downregulated GK), in a pattern indicative of impaired insulin signaling. Acute ethanol exposure subsequently caused a robust increase in hepatic triglycerides. The accumulation of triglycerides caused by ethanol was blunted in ASO-treated or in PKCepsilon(-/-) mice. Taken together, these data suggest that the increase in NEFA caused by hepatic ethanol metabolism leads to an increase in DAG production via the triacylglycerol pathway. DAG then subsequently activates PKCepsilon, which then exacerbates hepatic lipid accumulation by inducing insulin resistance. These data also suggest that PKCepsilon plays a causal role in at least the early phases of ethanol-induced liver injury.


Subject(s)
Ethanol/toxicity , Fatty Liver/chemically induced , Liver Diseases, Alcoholic/enzymology , Protein Kinase C-epsilon/metabolism , Actins/genetics , Animals , DNA Primers , Fatty Liver/enzymology , Glucokinase/genetics , Liver Diseases, Alcoholic/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotides, Antisense , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/metabolism
18.
Genes Brain Behav ; 7(3): 323-33, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17908177

ABSTRACT

Corticotropin-releasing factor (CRF), its receptors, and signaling pathways that regulate CRF expression and responses are areas of intense investigation for new drugs to treat affective disorders. Here, we report that protein kinase C epsilon (PKCepsilon) null mutant mice, which show reduced anxiety-like behavior, have reduced levels of CRF messenger RNA and peptide in the amygdala. In primary amygdala neurons, a selective PKCepsilon activator, psiepsilonRACK, increased levels of pro-CRF, whereas reducing PKCepsilon levels through RNA interference blocked phorbol ester-stimulated increases in CRF. Local knockdown of amygdala PKCepsilon by RNA interference reduced anxiety-like behavior in wild-type mice. Furthermore, local infusion of CRF into the amygdala of PKCepsilon(-/-) mice increased their anxiety-like behavior. These results are consistent with a novel mechanism of PKCepsilon control over anxiety-like behavior through regulation of CRF in the amygdala.


Subject(s)
Amygdala/enzymology , Anxiety/psychology , Corticotropin-Releasing Hormone/physiology , Protein Kinase C-epsilon/metabolism , Amygdala/drug effects , Animals , Anxiety/genetics , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/pharmacology , Mice , Mice, Knockout , Neurons/enzymology , Neurons/physiology , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , RNA Interference , RNA, Messenger/genetics
19.
J Biol Chem ; 282(45): 33052-63, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17875639

ABSTRACT

Ethanol enhances gamma-aminobutyrate (GABA) signaling in the brain, but its actions are inconsistent at GABA(A) receptors, especially at low concentrations achieved during social drinking. We postulated that the epsilon isoform of protein kinase C (PKCepsilon) regulates the ethanol sensitivity of GABA(A) receptors, as mice lacking PKCepsilon show an increased behavioral response to ethanol. Here we developed an ATP analog-sensitive PKCepsilon mutant to selectively inhibit the catalytic activity of PKCepsilon. We used this mutant and PKCepsilon(-/-) mice to determine that PKCepsilon phosphorylates gamma2 subunits at serine 327 and that reduced phosphorylation of this site enhances the actions of ethanol and benzodiazepines at alpha1beta2gamma2 receptors, which is the most abundant GABA(A) receptor subtype in the brain. Our findings indicate that PKCepsilon phosphorylation of gamma2 regulates the response of GABA(A) receptors to specific allosteric modulators, and, in particular, PKCepsilon inhibition renders these receptors sensitive to low intoxicating concentrations of ethanol.


Subject(s)
Benzodiazepines/pharmacology , Ethanol/pharmacology , Protein Kinase C-epsilon/metabolism , Receptors, GABA-A/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Allosteric Regulation , Animals , Cells, Cultured , Enzyme Activation , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice , Mice, Knockout , Mutation/genetics , Phosphorylation/drug effects , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , Protein Kinase Inhibitors/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Pyridines/pharmacology , Receptors, GABA-A/genetics , Sensitivity and Specificity , Serine/genetics , Serine/metabolism , Zolpidem
20.
Neuropsychopharmacology ; 32(1): 127-36, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16541084

ABSTRACT

A low level of response to ethanol is associated with increased risk of alcoholism. A major determinant of the level of response is the capacity to develop acute functional tolerance (AFT) to ethanol during a single drinking session. Mice lacking protein kinase C epsilon (PKCepsilon) show increased signs of ethanol intoxication and reduced ethanol self-administration. Here, we report that AFT to the motor-impairing effects of ethanol is reduced in PKCepsilon (-/-) mice when compared with wild-type littermates. In wild-type mice, in vivo ethanol exposure produced AFT that was accompanied by increased phosphorylation of PKCepsilon and resistance of GABA(A) receptors to ethanol. In contrast, in PKCepsilon (-/-) mice, GABA(A) receptor sensitivity to ethanol was unaltered by acute in vivo ethanol exposure. Both PKCepsilon (-/-) and PKCepsilon (+/+) mice developed robust chronic tolerance to ethanol, but the presence of chronic tolerance did not change ethanol preference drinking. These findings suggest that ethanol activates a PKCepsilon signaling pathway that contributes to GABA(A) receptor resistance to ethanol and to AFT. AFT can be genetically dissociated from chronic tolerance, which is not regulated by PKCepsilon and does not alter PKCepsilon modulation of ethanol preference.


Subject(s)
Alcoholic Intoxication/metabolism , Central Nervous System Depressants/administration & dosage , Drug Tolerance/physiology , Ethanol/administration & dosage , Protein Kinase C-epsilon/physiology , Alcohol Drinking/genetics , Alcohol Drinking/metabolism , Alcoholic Intoxication/genetics , Alcoholic Intoxication/physiopathology , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal/drug effects , Blotting, Western/methods , Cerebellum/drug effects , Chlorides/metabolism , Conditioning, Operant/drug effects , Drug Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Protein Kinase C-epsilon/deficiency , Reaction Time/drug effects , Receptors, GABA/drug effects , Receptors, GABA/physiology , Reflex/drug effects , Rotarod Performance Test/methods , Self Administration/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...