Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Journal of Southern Medical University ; (12): 17-22, 2015.
Article in Chinese | WPRIM | ID: wpr-329181

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the expression of ASK1 and PRKCD in the process of monocyte differentiation, and explore their role in functional changes of hypersplenism spleen macrophages (Mφ) in portal hypertension (PH).</p><p><b>METHODS</b>U937 cells were stimulated to differentiate into monocyte/macrophage-like cells by cultivation in PMA and the mRNA expressions of ASK1 and PRKCD were detected by q-PCR and the changes of protein expression were identified by western blot analysis. The secretion of phagocytose related cytokines such as IL-10 and TNF-α were tested by ELISA, and the function of the macrophage-like cells were studied by chicken red blood cell phagocytose test.</p><p><b>RESULTS</b>The expressions of PRKCD and ASK1 mRNA were gradually decreased along with the cell differentiation, while the secretion of TNF-α was increased, IL-10 secretion reached a maximum at 24 h after PAM stimulation, and then gradually fell. The expression of ASK1 and p-ASK1 were rapidly increased compared with the non-stimulated U937 cells, while the expression of PRKCD and p-PRKCD were sightly declined. The phagocytose test show that U937 cells induced with PMA were able to swallow the chicken red blood cell.</p><p><b>CONCLUSION</b>Up-regulated protein expression of ASK1 and p-ASK1 and down-regulated protein expression of PRKCD and p-PRKCD in the process of PMA induced monocyte differentiation, are consist with the expression changes of splenic macrophage phagocytosis in hypersplenism, which leads to increased activity of Mφ.</p>


Subject(s)
Humans , Cell Differentiation , Down-Regulation , Hypersplenism , Hypertension, Portal , Interleukin-10 , Bodily Secretions , MAP Kinase Kinase Kinase 5 , Physiology , Macrophages , Cell Biology , Phagocytosis , Protein Kinase C-delta , Physiology , RNA, Messenger , Tumor Necrosis Factor-alpha , Bodily Secretions , U937 Cells
2.
Immune Network ; : 206-211, 2015.
Article in English | WPRIM | ID: wpr-186449

ABSTRACT

Pulmonary edema is a major cause of mortality due to acute lung injury (ALI). The involvement of protein kinase C-delta (PKC-delta) in ALI has been a controversial topic. Here we investigated PKC-delta function in ALI using PKC-delta knockout (KO) mice and PKC inhibitors. Our results indicated that although the ability to produce proinflammatory mediators in response to LPS injury in PKC-delta KO mice was similar to that of control mice, they showed enhanced recruitment of neutrophils to the lung and more severe pulmonary edema. PKC-delta inhibition promoted barrier dysfunction in an endothelial cell layer in vitro, and administration of a PKC-delta-specific inhibitor significantly increased steady state vascular permeability. A neutrophil transmigration assay indicated that the PKC-delta inhibition increased neutrophil transmigration through an endothelial monolayer. This suggests that PKC-delta inhibition induces structural changes in endothelial cells, allowing extravasation of proteins and neutrophils.


Subject(s)
Animals , Mice , Acute Lung Injury , Capillary Permeability , Endothelial Cells , Lung , Mortality , Neutrophils , Protein Kinase C-delta , Protein Kinases , Pulmonary Edema
3.
J. bras. nefrol ; 36(4): 542-544, Oct-Dec/2014. graf
Article in English | LILACS | ID: lil-731142

ABSTRACT

Introduction: When faced with violet, purple or purplish-blue urine, clinicians should consider urinary tract infection in their differential diagnosis. Case report: A 60-year-old woman with end-stage kidney disease and non-adherence to renal replacement therapy was admitted to our hospital for placement of hemodialysis catheter. During her hospitalization she had purple urine, and purple urine bag syndrome (PUBS) was diagnosed. She was effectively treated with antibiotics and her urine returned to a dark yellow color. Discussion: Although this condition is often easily treated, diagnosing PUBS in chronic renal patients probably means an increased serum concentration of indoxyl sulfate, metabolite that is involved in the progression of both CKD and cardiovascular disease. Conclusion: Hence, in the context of our renal patients, perhaps PUBS is not as benign as supposed. .


Subject(s)
Animals , Rats , Isoenzymes/metabolism , Membrane Glycoproteins/metabolism , Protein Kinase C/metabolism , Proteoglycans/metabolism , Amino Acid Sequence , Molecular Sequence Data , Phosphorylation , Protein Kinase C-alpha , Protein Kinase C-delta
4.
Chinese Journal of Preventive Medicine ; (12): 777-782, 2013.
Article in Chinese | WPRIM | ID: wpr-355788

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effects of mRNA transcriptional and protein expressions of protein kinase Cδ (PKCδ) on the development of arsenic liver injury caused by coal-burning.</p><p><b>METHODS</b>Population study:133 arsenic exposures were selected as arsenic exposure groups including the ward non-patient group (25 cases) , no obvious hepatopathy group (38 cases) , mild (43 cases) and moderate to severe hepatopathy group (27 cases) from the area with endemic arsenism in Guizhou province. Another 34 healthy residents were selected as the control group in non-arsenic pollution village. The urine and peripheral blood were collected from the subjects. The arsenic contents in urine and mRNA expressions of PKCδ in peripheral blood were detected. Animal experiment study:thirty wistar rats were randomly by random number table divided into control group, drinking water arsenic poisoning group and coal-burning arsenic poisoning group (i.e., low, medium and high arsenic contaminated grain group) by random number table method, including 6 rats in each group. The control group was fed normally for 3 months, drinking water arsenic poisoning group and coal-burning arsenic poisoning groups were fed respectively with 10 mg/kg As2O3 solution and different concentrations (25, 50 and 100 mg/kg) of arsenic-containing feed which was persisted 3 months. The arsenic contents in urine, mRNA expression levels of PKCδ in peripheral blood and liver tissue and the protein expression levels of phosphorylated protein kinase Cδ(pPKCδ) in liver tissue were detected.</p><p><b>RESULTS</b>The median(quartile) of arsenic contents in urine were 25.58 (18.62-40.73), 56.66 (38.93-76.77), 64.90 (39.55- 98.37) and 75.47 (41.30-109.70) µg/g Cr respectively for the non-patient group, no obvious hepatopathy group, mild and moderate to severe hepatopathy group. The levels were higher than that in the control group (23.34 (17.84-37.45) µg/g Cr) (P < 0.05), except for the ward non-patient group. The arsenic contents in rat urine were 2223.61 (472.98-3976.73), 701.16 (194.01-1300.27), 1060.94 (246.33-2585.47) and 3101.11 (1919.97-5407.07) µg/g Cr, respectively for the drinking water arsenic poisoning group, the low, medium and high dosage arsenic grain contamination groups, all higher than that in the control group (94.32 (22.65-195.25) µg/g Cr) (P < 0.05) . The protein expressions of pPKCδ in liver tissue were 324.83 ± 25.06, 278.50 ± 30.57, 308.83 ± 34.67 and 326.33 ± 35.09, which were significantly higher than that in the control group (240.17 ± 28.07) (P < 0.05) . The protein expression levels of pPKCδ in liver cell membrane were 0.49 ± 0.06,0.33 ± 0.05,0.37 ± 0.06 and 0.50 ± 0.08, which were significantly higher than that in the control group (0.28 ± 0.04) (P < 0.05) . The protein expression levels of pPKCδ in liver cell cytoplasm were 0.38 ± 0.06,0.31 ± 0.05, 0.35 ± 0.05 and 0.36 ± 0.05, which were significantly higher than that in the control group (0.24 ± 0.05) (P < 0.05).</p><p><b>CONCLUSION</b>The arsenic may regulate protein expressions of pPKCδ and induce its membrane translocation, and cause the development of arsenic liver injury caused by coal-burning.</p>


Subject(s)
Animals , Female , Humans , Male , Rats , Arsenic , Urine , Arsenic Poisoning , Epidemiology , Metabolism , Case-Control Studies , China , Epidemiology , Coal , Environmental Exposure , Liver , Pathology , Liver Diseases , Protein Kinase C-delta , Metabolism , Rats, Wistar
5.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 303-310, 2012.
Article in English | WPRIM | ID: wpr-233162

ABSTRACT

Store-operated Ca(2+) channels (SOCs) are plasma membrane Ca(2+) permeable channels activated by depletion of intracellular Ca(2+) store. Ca(2+) entry through SOCs is known as store-operated Ca(2+) entry (SOCE), which plays an important role in the functional regulation of airway smooth muscle cells (ASMCs). Protein kinase C (PKC) has been shown to have an activating or inhibiting effect on SOCE, depending on cell types and PKC isoforms that are involved. In ASMCs, the effect of PKC on SOCE has not been elucidated so far. In this study, the role of PKC in the activation of SOCE in rat ASMCs was examined by using Ca(2+) fluorescence imaging technique. The results showed that acute application of PKC activators PMA and PDBu did not affect SOCE induced by the sarcoplasmic reticulum Ca(2+)-ATPase (SERCA) inhibitor thapsigargin. The non-selective PKC inhibitor chelerythrine significantly inhibited thapsigargin- and bradykinin-induced SOCE. RT-PCR assay identified PKCα, δ and ɛ isoforms in rat ASMCs. PKCα-selective inhibitor Gö6976 and PKCɛ-inhibiting peptide Epsilon-V1-2 had no effect on SOCE; by contrast, PKCδ-selective inhibitor rottlerin attenuated SOCE dramatically, suggesting that PKCδ was the major PKC isoform involved in the activation of SOCE in ASMCs. Moreover, PKC down-regulation by extended exposure to high doses of PMA or PDBu also reduced SOCE, confirming the essential role of PKC in the activation of SOCE in ASMCs. In addition, PKC down-regulation did not influence the expression of stromal interaction molecule 1 (STIM1) and Orai1, two elementary molecules in the regulation and activation of SOCs. These results identified PKCδ as an essential PKC isoform involved in the activation of SOCE, and confirmed that PKC regulates the function of ASMCs in a SOCE-dependent manner.


Subject(s)
Animals , Male , Rats , Bronchi , Metabolism , Calcium , Metabolism , Calcium Channels , Calcium Signaling , Physiology , Cells, Cultured , Membrane Glycoproteins , Metabolism , Myocytes, Smooth Muscle , Metabolism , ORAI1 Protein , Protein Kinase C-delta , Metabolism , Rats, Sprague-Dawley , Stromal Interaction Molecule 1
6.
Journal of the Korean Society for Vascular Surgery ; : 61-65, 2011.
Article in Korean | WPRIM | ID: wpr-726665

ABSTRACT

PURPOSE: Protein kinase C (PKC) has been implicated in a wide variety of cellular processes. Although PKC-delta is implicated in cell growth inhibition, as well as in cell differentiation, apoptosis, and tumor suppression, its role in atherosclerosis remains unclear. This study aimed to identify the mechanism of PKC-delta in the development of atherosclerosis. METHODS: To induce atherosclerosis, we performed allograft transplantations on aortas in mice. At 2, 4, and 6 weeks after transplantation, grafted aortas were obtained to compare the degree of atherosclerosis between wild type and PKC-delta (-/-) aorta. Alloantibody levels in the recipient mice's blood were measured. Reverse transcriptase-polymerase chain reaction (RT-PCR) was performed to quantitatively measure chemokine and cytokine mRNA expression of the inflammation from the harvested aorta. RESULTS: Atherosclerosis was more severe in the PKC-delta (-/-) aorta than in the wild type aorta. Alloantibody levels were higher in the mice grafted with aorta from the PKC-delta (-/-) mice than in the mice grafted with aorta from the wild type mice. RT-PCR revealed higher expressions of MRP-2, MCP-1, MIP-1alpha, and IL-2 in the mice grafted with aorta from the PKC-delta (-/-) mice than the wild type mice. CONCLUSION: Aorta allograft transplantation is a useful modality for inducing atherosclerosis. PKC-delta may be a negative regulator of atherosclerosis.


Subject(s)
Animals , Mice , Aorta , Apoptosis , Atherosclerosis , Cell Differentiation , Chemokine CCL3 , Inflammation , Interleukin-2 , Protein Kinase C , Protein Kinase C-delta , Protein Kinases , RNA, Messenger , Transplantation, Homologous , Transplants
7.
Journal of the Korean Surgical Society ; : 235-241, 2011.
Article in English | WPRIM | ID: wpr-76451

ABSTRACT

PURPOSE: This study demonstrated that apoptosis induced by mycophenolic acid (MPA) is mediated by mitochondrial membrane potential transition (MPT) changes in Jurkat cells. METHODS: Cell viability and MPT changes were measured by flow cytometry. Western blotting was performed to evaluate the expression of Bcl-2 family proteins, Bid, truncated Bid (tBid), cytochrome c, voltage dependent anion channel (VDAC), poly ADP-ribose polymerase (PARP), and protein kinase C-delta (PKC-delta). The catalytic activity of caspase-9 and -3 was also measured. RESULTS: Cell viability was decreased in time- and dose-dependent manners. Bcl-2 protein expression was decreased, but Bax protein expression was identified. A decreased Bcl-XL /Bcl-XS ratio was also noted. The expression of tBid protein also increased in a time-dependent manner in Jurkat cells treated with MPA. While normal MPT appeared as orange fluorescence, abnormal MPT corresponded to green fluorescence. Green fluorescence increased as orange decreased in the MPA-treated cells. Significantly increased concentrations of MPA induced the release of cytosolic cytochrome c. MPA also augmented the catalytic activity of caspase-9 and caspase-3 in Jurkat cells. Our findings demonstrated that MPA-induced apoptosis is mediated by MPT changes accompanied by decreased Bcl-XL expression and the appearance of tBid protein. The release of cytosolic cytochrome c from mitochondria and increased catalytic activity of caspase-9 and caspase-3 were observed in MPA-treated Jurkat cells. CONCLUSION: These results suggest that mitochondrial dysfunction caused by MPA induces human T lymphocyte apoptosis.


Subject(s)
Humans , Adenosine Diphosphate Ribose , Apoptosis , bcl-2-Associated X Protein , BH3 Interacting Domain Death Agonist Protein , Blotting, Western , Caspase 3 , Caspase 9 , Cell Survival , Citrus sinensis , Cytochromes c , Cytosol , Flow Cytometry , Fluorescence , Jurkat Cells , Lymphocytes , Membrane Potential, Mitochondrial , Mitochondria , Mitochondrial Membranes , Mycophenolic Acid , Protein Kinase C-delta , Proteins
8.
Experimental & Molecular Medicine ; : 669-675, 2011.
Article in English | WPRIM | ID: wpr-73119

ABSTRACT

Cyclooxygenase-2 (COX-2) is an important enzyme in inflammation. In this study, we investigated the underlying molecular mechanism of the synergistic effect of rottlerin on interleukin1beta (IL-1beta)-induced COX-2 expression in MDA-MB-231 human breast cancer cell line. Treatment with rottlerin enhanced IL-1beta-induced COX-2 expression at both the protein and mRNA levels. Combined treatment with rottlerin and IL-1beta significantly induced COX-2 expression, at least in part, through the enhancement of COX-2 mRNA stability. In addition, rottlerin and IL-1beta treatment drove sustained activation of p38 Mitogen-activated protein kinase (MAPK), which is involved in induced COX-2 expression. Also, a pharmacological inhibitor of p38 MAPK (SB 203580) and transient transfection with inactive p38 MAPK inhibited rottlerin and IL-1beta-induced COX-2 upregulation. However, suppression of protein kinase C delta (PKC delta) expression by siRNA or overexpression of dominant-negative PKC delta (DN-PKC-delta) did not abrogate the rottlerin plus IL-1beta-induced COX-2 expression. Furthermore, rottlerin also enhanced tumor necrosis factor-alpha (TNF-alpha), phorbol myristate acetate (PMA), and lipopolysaccharide (LPS)-induced COX-2 expression. Taken together, our results suggest that rottlerin causes IL-1beta-induced COX-2 upregulation through sustained p38 MAPK activation in MDA-MB-231 human breast cancer cells.


Subject(s)
Female , Humans , Acetophenones/pharmacology , Benzopyrans/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cyclooxygenase 2/genetics , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Interleukin-1beta/immunology , MAP Kinase Signaling System/drug effects , Mallotus Plant/chemistry , NF-kappa B/immunology , Protein Kinase C-delta/antagonists & inhibitors , Reactive Oxygen Species/immunology , p38 Mitogen-Activated Protein Kinases/immunology
9.
West China Journal of Stomatology ; (6): 539-546, 2010.
Article in Chinese | WPRIM | ID: wpr-350289

ABSTRACT

<p><b>OBJECTIVE</b>To study the role of protein kinase C-delta (PKC-delta) in hyperthermia-induced apoptosis in human tongue squamous cell carcinoma Tca8113 cells.</p><p><b>METHODS</b>Tca8113 cells were treated at 43 degrees C in a heating water bath for 0, 40, 80, 120 min after pretreatment with Rottlerin, a specific inhibitor of PKC-delta, and equal volume dimethyl sulfoxide (DMSO) for 30 min, respectively. The cells were stained by propidium iodide (PI) and Rhodamine 123 to analysis apoptotic rate and the changes of mitochondrial transmembrane potential by flow cytometry (FCM). The total proteins were extracted for Western blotting analysis of activation and proteolysis of PKC-delta, and for colorimetric assay of relative activity of Caspase-3.</p><p><b>RESULTS</b>Hyperthermia could induce proteolysis and activation of PKC-delta, and this was attenuated by Rottlerin. Apoptotic rate, decreasing of mitochondrial transmembrane potential and activity of Caspase-3 which being induced by hyperthermia in Tca8113 cells were inhibited by PKC-delta specific inhibitor Rottlerin. There were significantly statistical differences in apoptosis rates, mitochondrial transmembrane potential and activity of Caspase-3 between Rottlerin- and non-Rottlerin-pretreated cells after hyperthermia for 40, 80, 120 min (P < 0.01).</p><p><b>CONCLUSION</b>Activated PKC-delta may facilitate hyperthermia-induced apoptosis in Tca8113 cells, and may be one of the mechanisms of apoptosis induced by hyperthermia.</p>


Subject(s)
Humans , Acetophenones , Apoptosis , Benzopyrans , Carcinoma, Squamous Cell , Protein Kinase C , Protein Kinase C-delta
10.
Journal of Southern Medical University ; (12): 1376-1378, 2010.
Article in Chinese | WPRIM | ID: wpr-336176

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the expressions of protein kinase C (PKC) isoforms in X-ray-exposed HepG2 cells and identify the PKC isoforms that induce radioresistance in HepG2 cells.</p><p><b>METHODS</b>Cultured HepG2 cells were divided into control group and 6 Gy radiation group for corresponding treatments. The fluorescence intensity (FI) and the percentage of positive cells were determined using flow cytometry.</p><p><b>RESULTS</b>The FI of PKCalpha and PKCdelta were 2.28 and 5.05 in the radiation group, respectively, significantly higher than those in the control group (P<0.05). The percentages of PKCalpha- and PKCdelta -positive cells were significantly higher in the radiation group than in the control group (P<0.05). The FI and the percentages of PKC zeta, gamma, epsilon, zeta positive cells were rather low and showed no significant differences between the two groups (P>0.05); PKCbeta expression was not detected in the two groups of cells. The apoptosis rates of the control and radiation groups were 1.73% and 20.90%, respectively.</p><p><b>CONCLUSION</b>PKCalpha and PKCdelta may be involved in protecting HepG2 cells from radiation-induced apoptosis.</p>


Subject(s)
Humans , Apoptosis , Physiology , Radiation Effects , Hep G2 Cells , Isoenzymes , Classification , Metabolism , Protein Kinase C-alpha , Metabolism , Protein Kinase C-delta , Metabolism , Radiation Tolerance , Signal Transduction , Physiology
11.
Experimental & Molecular Medicine ; : 699-708, 2008.
Article in English | WPRIM | ID: wpr-167143

ABSTRACT

Expression of protein kinase C-delta (PKC delta) is up-regulated by apoptosis-inducing stimuli. However, very little is known about the signaling pathways that control PKC delta gene transcription. In the present study, we demonstrate that JNK stimulates PKC delta gene expression via c-Jun and ATF2 in response to the anticancer agent doxorubicin (DXR) in mouse lymphocytic leukemia L1210 cells. Luciferase reporter assays showed that DXR-induced activation of the PKC delta promoter was enhanced by ectopic expression of JNK1, c-Jun, or ATF2, whereas it was strongly reduced by expression of dominant negative JNK1 or by treatment with the JNK inhibitor SP600125. Furthermore, point mutations in the core sequence of the c-Jun/ATF2 binding site suppressed DXR-induced activation of the PKC delta promoter. Our results suggest an additional role for a JNK signaling cascade in DXR-induced PKC delta gene expression.


Subject(s)
Animals , Mice , Activating Transcription Factor 2/physiology , Anthracenes/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Line, Tumor , Doxorubicin/pharmacology , Mitogen-Activated Protein Kinase 8/physiology , Mutation , Promoter Regions, Genetic , Protein Kinase C-delta/genetics , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Signal Transduction/physiology , Transcription, Genetic
12.
Acta Pharmaceutica Sinica ; (12): 338-341, 2006.
Article in Chinese | WPRIM | ID: wpr-271431

ABSTRACT

<p><b>AIM</b>To investigate the effects of sulfated polymannuroguluronate (SPMG), a novel candidate anti-AIDS drug in Phase II clinical trial, on Tat-induced release of proinflammatory cytokines (i.e., TNFalpha, IL-1beta and IL-6) and its related mechanism.</p><p><b>METHODS</b>The effects of SPMG on Tat induced TNFalpha (4 h), IL-1beta and IL-6 (6 h) secretion in THP-1 cells were measured by ELISA. Western blotting analysis was used to study the effects of SPMG on Tat induced PKCzeta, PKCtheta and PKCsigma phosphorylation.</p><p><b>RESULTS</b>SPMG (50 to 100 microg x mL(-1)) markedly suppressed TNFalpha, IL-1beta and IL-6 secretion in Tat activated THP-1 cells. In THP-1 cells the phosphorylation levels of PKCzeta, PKCtheta and PKCsigma significantly increased following Tat stimulation, and only PKCsigma phosphorylation levels was inhibited by SPMG (50 to 100 microg x mL(-1)).</p><p><b>CONCLUSION</b>SPMG suppresses the secretion of proinflammatory cytokines in THP-1 cells may be by inhibiting PKCsigma activation.</p>


Subject(s)
Humans , Cell Line, Tumor , Gene Products, tat , Pharmacology , Interleukin-1beta , Bodily Secretions , Interleukin-6 , Bodily Secretions , Isoenzymes , Metabolism , Phosphorylation , Polysaccharides , Pharmacology , Protein Kinase C , Metabolism , Protein Kinase C-delta , Metabolism , Protein Kinase C-theta , Tumor Necrosis Factor-alpha , Bodily Secretions
13.
The Journal of the Korean Rheumatism Association ; : 33-45, 2006.
Article in Korean | WPRIM | ID: wpr-203398

ABSTRACT

OBJECTIVE: CD27 is a member of the tumor necrosis factor receptor (TNFR) superfamily and is expressed on T, B, and NK cells. The signaling via CD27 plays pivotal roles in T-T and T-B interaction. CD27 is a useful marker in assessing the number of circulation B cells and B cell subsets because it permits one step identification of the major B cell compartments, CD27- naive and CD27+ memory B cells as well as CD27high plasma cells. We have analyzed the mechanisms underlying the regulation of CD27 expression. METHODS: Isolation B cells and Raji cells were cultured with PMA. The levels of cell surface CD27 and CD 27 mRNA were analyzed by FACs staining and RT-PCR. Raji cells were cultured with phorbol 12-myristate 13-acetate (PMA), with or without pretreated shedding inhibitor BB3103 and TAPI-1. sCD27 was measured in culture supernatant by ELISA. Cell lysates were analyzed for PKC isotype activation by Western blot. We used PKC inhibitor Ly379196 and rottlen. RESULTS: Membrane expression of CD27 was down-regulated after PMA stimulation without cytotoxic effect in B cells. Furthermore, PMA treatment could directly reduce CD27 mRNA without intermediate protein synthesis in B cells. In contrast, PMA treatment induced soluble form of CD27 (sCD27), which was shed from the cell surface and was found in PMA treatment B cell culture supernatant. PMA-induced sCD27 proteins were decreased with shedding inhibitor BB3103 and TAPI-1. PMA-induced down regulation of CD27 expressions were quenched with protein kinase C (PKC) inhibitor Staurosporin, PKC-beta inhibitor rottlerin and PKC-delta inhibitor Ly379196. CONCLUSION: These data suggest that PMA-induced activation of PKC plays a crucial role in down-regulation of the expression of the CD27 and up-regulation of the shedding of the CD27 in human B cells.


Subject(s)
Humans , B-Lymphocyte Subsets , B-Lymphocytes , Blotting, Western , Cell Culture Techniques , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Killer Cells, Natural , Membranes , Memory , Plasma Cells , Protein Kinase C , Protein Kinase C-delta , Protein Kinases , Receptors, Tumor Necrosis Factor , RNA, Messenger , Up-Regulation
14.
Acta Physiologica Sinica ; (6): 269-274, 2006.
Article in Chinese | WPRIM | ID: wpr-265455

ABSTRACT

Cardiac hypertrophy is an adaptive process to an increased hemodynamic overload. However, the adaption may lead to the fragility of myocardium facing pathological stimuli. In the present study, experiments were designed to explore the susceptibility of hypertrophic myocardiocytes to apoptotic stimuli and the role of protein kinase Cdelta (PKCdelta) during the transition from hypertrophy to apoptosis. Endothelin-1 (ET-1)-treated cardiomyocytes were used as model of cardiac hypertrophy. Angiotensin II (Ang II) was used as an apoptotic stimulus. Cell surface area was measured to determine the extent of hypertrophy. The apoptotic rate in cardiomyocytes was detected by Hoechst 33258. (1) Cell surface area was increased by 42.5% and 67.3% following 1 nmol/L and 10 nmol/L ET-1 treatment, respectively, as compared with serum-free cultured myocytes. So the mildly and moderately hypertrophic myocyte models were set up. (2) Apoptotic rates in serum-free cultured, mildly and moderately hypertrophic myocytes after Ang II treatment were (15.54+/-1.32) %, (20.65+/-1.40) % and (29.33+/-3.52) %, respectively. It is suggested that hypertrophic myocytes are more susceptive to apoptotic stimulus. (3) Rottlerin, a specific inhibitor of PKCdelta depressed apoptotic rates induced by Ang II to (15.88+/-2.25) % in mildly hypertrophic myocytes and to (15.01+/-1.37) % in moderately hypertrophic myocytes; but rottlerin did not affect apoptotic rate induced by Ang II in serum-free cultured myocytes. These results suggest that inhibition of PKCdelta can reduce Ang II-induced apoptosis of hypertrophic cardiomyocytes and that PKCdelta is possibly involved in the apoptotic process of hypertrophic cardiomyocytes.


Subject(s)
Animals , Rats , Angiotensin II , Pharmacology , Animals, Newborn , Apoptosis , Physiology , Cardiomegaly , Pathology , Cell Enlargement , Endothelin-1 , Pharmacology , Heart Failure , Myocytes, Cardiac , Cell Biology , Pathology , Primary Cell Culture , Protein Kinase C-delta , Physiology , Rats, Sprague-Dawley
15.
Chinese Journal of Oncology ; (12): 564-567, 2006.
Article in Chinese | WPRIM | ID: wpr-236909

ABSTRACT

<p><b>OBJECTIVE</b>To evaluate the effect of PKC-delta inhibitor Rottlerin on human colon cancer cells and its mechanism.</p><p><b>METHODS</b>Human colon cancer cell line SW1116 cells were treated with Rottlerin. The transcriptional level of DNA methyltransferase (Dnmt)1, Dnmt3a and Dnmt3b was detected by real-time RT-PCR. Cell cycle distribution was evaluated by flow cytometry (FCM). In addition, cellular morphological changes were examined by light microscopy.</p><p><b>RESULTS</b>PKC-delta inhibitor decreased the expression of Dnmt1, Dnmt3a mRNA, up-regulated APC, p21(WAF1) and p16(INK4A) mRNA. Demonstarted by flow cytometry, Rottlerin increased the percentage of cell cycle G0/G1 phase cell numbers (P = 0.02) and decreased the percentage of cell cycle G2/M phase cell numbers (P = 0.01). Remarkable changes of cellular morphology were observed under light microscope: The volume and cytoplasm of cells treated with Rottlerin were increased. The cell contour was not very clear, and mitotic figures were less frequently seen.</p><p><b>CONCLUSION</b>PKC-delta inhibitor Rottlerin inhibites cell division and proliferation of the colon cancer SW1116 cells through regulating DNA methylation and blocking the signaling pathway of mitogen-activated protein kinase (MAPK).</p>


Subject(s)
Humans , Acetophenones , Pharmacology , Adenomatous Polyposis Coli Protein , Genetics , Benzopyrans , Pharmacology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms , Genetics , Metabolism , Pathology , Cyclin-Dependent Kinase Inhibitor p16 , Genetics , Cyclin-Dependent Kinase Inhibitor p21 , Genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases , Genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic , Protein Kinase C-delta , RNA, Messenger , Genetics , Metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
16.
Experimental & Molecular Medicine ; : 310-319, 2006.
Article in English | WPRIM | ID: wpr-51258

ABSTRACT

Myristoylated alanine-rich C kinase substrate (MARCKS) is a widely distributed protein kinase C (PKC) substrate and has been implicated in actin cytoskeletal rearrangement in response to extracellular stimuli. Although MARCKS was extensively examined in various cell culture systems, the physiological function of MARCKS in the central nervous system has not been clearly understood. We investigated alterations of cellular distribution and phosphorylation of MARCKS in the hippocampus following kainic acid (KA)-induced seizures. KA (25 mg/kg, i.p.) was administered to eight to nine week-old C57BL/6 mice. Behavioral seizure activity was observed for 2 h after the onset of seizures and was terminated with diazepam (8 mg/kg, i.p.). The animals were sacrificed and analyzed at various points in time after the initiation of seizure activity. Using double-labeling immunofluorescence analysis, we demonstrated that the expression and phosphorylation of MARCKS was dramatically upregulated specifically in microglial cells after KA-induced seizures, but not in other types of glial cells. PKC alpha, beta I, beta II and delta, from various PKC isoforms examined, also were markedly upregulated, specifically in microglial cells. Moreover, immunoreactivities of phosphorylated MARCKS were co-localized in the activated microglia with those of the above isoforms of PKC. Taken together, our in vivo data suggest that MARCKS is closely linked to microglial activation processes, which are important in pathological conditions, such as neuroinflammation and neurodegeneration.


Subject(s)
Mice , Animals , Up-Regulation/drug effects , Time Factors , Seizures/chemically induced , Protein Kinase C-delta/analysis , Protein Kinase C-alpha/analysis , Protein Kinase C/analysis , Protein Biosynthesis/drug effects , Phosphorylation/drug effects , Microscopy, Confocal , Microglia/cytology , Mice, Inbred C57BL , Membrane Proteins/analysis , Kainic Acid/toxicity , Isoenzymes/analysis , Intracellular Signaling Peptides and Proteins/analysis , Immunohistochemistry
17.
Korean Journal of Anatomy ; : 21-30, 2005.
Article in Korean | WPRIM | ID: wpr-655573

ABSTRACT

Our previous study demonstrated that a bacterial siderophore, deferoxamine (DFO), could trigger inflammatory signals in human intestinal epithelial cells as a single stimulus, leading to IL-8 production via ERK1/2 and p38 phosphorylation and NF-kappa B-independent mechanism. In the present study, we proved that a novel protein kinase C (PKC)isoform, PKCdelta, is necessary for DFO-induced IL-8 production. Pretreatment of HT-29 cells with rottlerin showed remarkable inhibition of DFO-induced IL-8 production. In contrast, a conventional PKC inhibitor Go6976 did not show significant inhibition of DFO-induced IL-8 production. DFO induced strong phosphorylation of PKCdelta in the epithelial cells. Overexpression of PKCdelta resulted in enhanced PKCdelta phosphorylation, while transfection with dominant-negative PKCdelta vector failed DFO-induced phosphorylation. In addition, transfection of HT-29 cells with siRNA targeting endogenous PKCdelta, which suppressed PKCdelta expression, attenuated DFO-induced IL-8 production. These results demonstrate that PKCdelta plays an important role in regulating iron chelator-induced IL-8 production in human intestinal epithelial cells.


Subject(s)
Humans , Deferoxamine , Epithelial Cells , HT29 Cells , Interleukin-8 , Iron , Phosphorylation , Protein Kinase C , Protein Kinase C-delta , Protein Kinases , RNA, Small Interfering , Transfection
18.
Chinese Journal of Biotechnology ; (12): 300-304, 2005.
Article in Chinese | WPRIM | ID: wpr-305281

ABSTRACT

Protein kinase Cdelta (PKCdelta) is a member of protein kinase C family, which possess phospholipid-dependent serine and threonine kinase activity. PKCdelta is a potential drug target of diabetes and some cancers. The abnormal activation of PKCdelta can arouse diabetes and some cancers. Therefore the specific inhibitors of PKCdelta can be applied in the research and development of the drug candidate of these diseases. The present aim is to obtain active recombinant PKCdelta from COS1 cells. For cloning of mouse PKCdelta a pair of specific primers were designed based on the published sequence of this gene. The cDNA of full coding region was obtained by RT-PCR. The amplified cDNA was subsequently cloned into FLAG-tagged pcDNA3.0 and its sequence was confirmed by DNA sequencing analysis. FLAG-tagged pcDNA3.0-PKCdelta was transfected into COS1 cells. A cell strain which can stably express PKCdelta was obtained by G418 screening. FLAG-tagged PKCdelta in the supernant of COS1 cells extracts was absorbed by anti-FLAG resin and eluted by FLAG peptide. The purified protein appeared as a single band on both SDS-PAGE and western blotting, indicating that it was chemical and antigenic pure. By kinase assay, the recombinant PKCdelta was active. Positive inhibitor, staurosporine, was used to prove the enzyme could be greatly inhibited. Several compounds have been found to inhibit the enzyme, which indicates the preliminary application in drug lead compounds screening.


Subject(s)
Animals , Humans , Mice , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Drug Delivery Systems , Drug Evaluation, Preclinical , Protein Kinase C-delta , Genetics , Recombinant Proteins , Genetics , Staurosporine , Pharmacology
19.
Journal of the Korean Surgical Society ; : 194-200, 2003.
Article in Korean | WPRIM | ID: wpr-125361

ABSTRACT

PURPOSE: The purpose of this study was to investigate the effect of exogenous nitric oxide (NO) on the proliferation of gastric carcinoma cells and the signaling pathways that regulate these responses. METHODS: MKN-28 cells were obtained from the Korean Cell Line Bank (KCLB) and maintained in DMEM culture media. The effect of sodium nitroprusside (SNP), a NO donor, on the proliferation of a serum-starved gastric carcinoma cell line, MKN-28, was examined by [3H]thymidine incorporation. Western blot was performed to analyze the translocation of protein kinase C (PKC)-deltafrom the cytosol to the plasma membrane of the MKN-28 cells. RESULTS: The proliferation of MKN-28 cells was significantly increased by SNP. It was also found that the proliferation was significantly inhibited by the protein kinase A (PKA) inhibitor, KT5720, and the protein kinase G inhibitor (PKG), KT5823, in SNP-treated cells. The SNP-induced proliferation was also inhibited by the PKC-deltaspecific inhibitor, rottlerin (1mu), but was increased by the PKC-beta inhibitor, Go6976 (1muM). The amount of translocated PKC-deltaprotein in the plasma membrane from the cytosol increased time-dependently after treating the cells with SNP, suggesting that NO activates PKC-delta Anti-inflammatory drugs, including dexamethasone, aspirin, indomethacin, mephenamic acid, and acetaminophen inhibited the SNP-induced proliferation of the cells and blocked of PKC-deltaactivation. CONCLUSION: NO stimulates the proliferation of serum- starved gastric cancer cells. The NO-induced proliferation may be mediated by PKC-delta The inhibitory effect of anti-inflammatory drugs on cell proliferation may be related to the inhibition of PKC-deltaactivity.


Subject(s)
Humans , Acetaminophen , Aspirin , Blotting, Western , Cell Line , Cell Membrane , Cell Proliferation , Culture Media , Cyclic AMP-Dependent Protein Kinases , Cyclic GMP-Dependent Protein Kinases , Cytosol , Dexamethasone , Indomethacin , Nitric Oxide , Nitroprusside , Protein Kinase C , Protein Kinase C-delta , Protein Kinases , Stomach Neoplasms , Tissue Donors
20.
Acta Academiae Medicinae Sinicae ; (6): 471-473, 2002.
Article in Chinese | WPRIM | ID: wpr-350082

ABSTRACT

<p><b>OBJECTIVE</b>To study the function of 5 single nucleotide polymorphisms (SNPs) of the PRKCZ gene, a susceptibility gene for type 2 diabetes in Han population of North China, in the pathogenesis of the disease.</p><p><b>METHODS</b>Bioinformatic methods and reporter gene activity determination were used to analyze the function of the 5 SNPs.</p><p><b>RESULTS</b>The reporter gene activities of different alleles of 2 SNPs, rs427811 and rs809912, were obviously different, which implies that these 2 SNPs might be susceptibility loci of the disease.</p><p><b>CONCLUSION</b>The PRKCZ gene is further confirmed to be a susceptibility gene for type 2 diabetes in Han population of North China. Two SNPs in the gene play a role in the pathogenesis of the disease by affecting the expression level of PRKCZ gene.</p>


Subject(s)
Humans , Alleles , Asian People , Diabetes Mellitus, Type 2 , Genetics , Ethnicity , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Protein Kinase C , Genetics , Protein Kinase C-delta
SELECTION OF CITATIONS
SEARCH DETAIL