Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Biosensors (Basel) ; 12(8)2022 Aug 19.
Article in English | MEDLINE | ID: mdl-36005053

ABSTRACT

The quantitative analysis of cell surface antigens has attracted increasing attention due to the antigenic variation recognition that can facilitate early diagnoses. This paper presents a novel methodology based on the optical "cell-tearing" and the especially proposed "dilution regulations" to detect variations in cell surface antigens. The cell attaches to the corresponding antibody-coated slide surface. Then, the cell-binding firmness between a single cell and the functionalized surface is assayed by optically tearing using gradually reduced laser powers incorporated with serial antibody dilutions. Groups B and B3 of red blood cells (RBCs) were selected as the experiment subject. The results indicate that a higher dilution called for lower power to tear off the cell binding. According to the proposed relative-quantitative analysis theory, antigenic variation can be intuitively estimated by comparing the maximum allowable dilution folds. The estimation result shows good consistency with the finding in the literature. This study suggests a novel methodology for examining the variation in cell surface antigens, expected to be widely capable with potential sensor applications not only in biochemistry and biophysics, but also in the micro-/nano- engineering field.


Subject(s)
Antigens, Surface , Optical Tweezers , Antibodies , Erythrocytes , Lasers
2.
Int J Mol Sci ; 21(16)2020 Aug 13.
Article in English | MEDLINE | ID: mdl-32823607

ABSTRACT

Pharmacologic intervention to affect the membrane lipid homeostasis of lipid rafts is a potent therapeutic strategy for cancer. Here we showed that gallic acid (GA) caused the complex formation of inactive Ras-related C3 botulinum toxin substrate 1 (Rac1)-phospho (p)-casein kinase 2 α (CK2α) (Tyr 255) in human tongue squamous carcinoma (TSC) cells, which disturbed the lipid raft membrane-targeting of phosphatidylinositol 3-kinase (PI3K)-Rac1-protein kinase B (Akt) signal molecules by inducing the association of p110α-free p85α with unphosphorylated phosphatase tensin homolog deleted on chromosome 10 (PTEN) in lipid rafts. The effects on induction of inactive Rac1-p-CK2α (Tyr 255) complex formation and attenuation of p-Akt (Ser 473), GTP-Rac1, glucose transporter-1 (GLUT-1) lipid raft membrane-targeting, and cell invasive activity by GA were counteracted either by CK2α short hairpin RNA or cellular-Src (c-Src) inhibitor PP1. PP1 treatment, GLUT-1 or constitutively active Rac1 ectopic-expression blocked GA-induced decreases in cellular glucose, sphingolipid and cholesterol of lipid raft membranes, p85α-p110α-GTP-Rac1 complexes, glucosylceramide synthase activity and increase in ceramide and p110α-free p85α-PTEN complex levels of lipid raft membranes, which reversed the inhibition on matrix metalloproteinase (MMP)-2/-9-mediated cell invasion induced by GA. Using transient ectopic expression of nuclear factor-kappa B (NF-κB) p65, MMP-2/-9 promoter-driven luciferase, and NF-κB-dependent luciferase reporter genes and NF-κB specific inhibitors or Rac1 specific inhibitor NSC23766, we confirmed that an attenuation of Rac1 activity by GA confers inhibition of NF-κB-mediated MMP-2/-9 expression and cell invasion. In conclusion, GA-induced c-Src activation is a key inductive event for the formation of inactive Rac1-p-CK2α (Tyr 255) complexes, which disturbed lipid raft compartment of PI3K and PTEN molecules by impairing Akt-regulated GLUT-1-mediated sphingolipid synthesis, and finally resulting in inhibition of TSC cell invasion.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Membrane Microdomains/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Sphingolipids/biosynthesis , Tongue Neoplasms/metabolism , src-Family Kinases/metabolism , Carcinoma, Squamous Cell/pathology , Cell Compartmentation , Cell Line, Tumor , Down-Regulation/drug effects , Enzyme Activation/drug effects , Epitopes/metabolism , Gallic Acid/pharmacology , Glucose Transporter Type 1/metabolism , Homeostasis/drug effects , Humans , Matrix Metalloproteinases/metabolism , Membrane Microdomains/drug effects , Models, Biological , NF-kappa B/metabolism , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/metabolism , Tongue Neoplasms/pathology , rac1 GTP-Binding Protein/metabolism
3.
Cells ; 9(2)2020 02 05.
Article in English | MEDLINE | ID: mdl-32033487

ABSTRACT

The specialized cholesterol/sphingolipid-rich membrane domains termed lipid rafts are highly dynamic in the cancer cells, which rapidly assemble effector molecules to form a sorting platform essential for oncogenic signaling transduction in response to extra- or intracellular stimuli. Density-based membrane flotation, subcellular fractionation, cell surface biotinylation, and co-immunoprecipitation analyses of bichalcone analog ((E)-1-(4-Hydroxy-3-((4-(4-((E)-3-(pyridin-3-yl)acryloyl)phenyl)piperazin-1-yl)methyl)phenyl)-3-(pyridin-3-yl)prop-2-en-1-one (TSWU-BR4)-treated cancer cells showed dissociation between GRP78 and p85α conferring the recruitment of PTEN to lipid raft membranes associated with p85α. Ectopic expression of GRP78 could overcome induction of lipid raft membrane-associated p85α-unphosphorylated PTEN complex formation and suppression of GRP78PI3KAktGTP-Rac1-mediated and GRP78-regulated PERKNrf2 antioxidant pathway and cancer cell invasion by TSWU-BR4. Using specific inducer, inhibitor, or short hairpin RNA for ASM demonstrated that induction of the lipid raft membrane localization and activation of ASM by TSWU-BR4 is responsible for perturbing homeostasis of cholesterol and ceramide levels in the lipid raft and ER membranes, leading to alteration of GRP78 membrane trafficking and subsequently inducing p85α-unphosphorylated PTEN complex formation, causing disruption of GRP78PI3KAktGTP-Rac1-mediated signal and ER membrane-associated GRP78-regulated oxidative stress balance, thus inhibiting cancer cell invasion. The involvement of the enrichment of ceramide to lipid raft membranes in inhibition of NF-κB-mediated MMP-2 expression was confirmed through attenuation of NF-κB activation using C2-ceramide, NF-κB specific inhibitors, ectopic expression of NF-κB p65, MMP-2 promoter-driven luciferase, and NF-κB-dependent reporter genes. In conclusion, localization of ASM in the lipid raft membranes by TSWU-BR4 is a key event for initiating formation of ceramide-enriched lipid raft membrane platforms, which causes delocalization of GRP78 from the lipid raft and ER membranes to the cytosol and formation of p85α-unphosphorylated PTEN complexes to attenuate the GRP78-regulated oxidative stress balance and GRP78p85αAktGTP-Rac1NF-κBMMP-2-mediated cancer cell invasion.


Subject(s)
Chalcones/pharmacology , Heat-Shock Proteins/metabolism , Homeostasis , Membrane Lipids/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Ceramides/metabolism , Chalcones/chemistry , Cholesterol/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Endoplasmic Reticulum Chaperone BiP , Homeostasis/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Models, Biological , NF-kappa B/metabolism , Neoplasm Invasiveness , Oxidation-Reduction , Oxidative Stress/drug effects , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Sphingomyelin Phosphodiesterase
4.
Int J Mol Sci ; 20(9)2019 Apr 29.
Article in English | MEDLINE | ID: mdl-31035650

ABSTRACT

Citrate is a key intermediate of the tricarboxylic acid cycle and acts as an allosteric signal to regulate the production of cellular ATP. An elevated cytosolic citrate concentration inhibits growth in several types of human cancer cells; however, the underlying mechanism by which citrate induces the growth arrest of cancer cells remains unclear. The results of this study showed that treatment of human pharyngeal squamous carcinoma (PSC) cells with a growth-suppressive concentration of citrate caused cell cycle arrest at the G2/M phase. A coimmunoprecipitation study demonstrated that citrate-induced cell cycle arrest in the G2/M phase was associated with stabilizing the formation of cyclin B1-phospho (p)-cyclin-dependent kinase 1 (CDK1) (Thr 161) complexes. The citrate-induced increased levels of cyclin B1 and G2/M phase arrest were suppressed by the caspase-3 inhibitor Ac-DEVD-CMK and caspase-3 cleavage of mutant p21 (D112N). Ectopic expression of the constitutively active form of protein kinase B (Akt1) could overcome the induction of p21 cleavage, cyclin B1-p-CDK1 (Thr 161) complexes, and G2/M phase arrest by citrate. p85α-phosphatase and tensin homolog deleted from chromosome 10 (PTEN) complex-mediated inactivation of Akt was required for citrate-induced G2/M phase cell cycle arrest because PTEN short hairpin RNA or a PTEN inhibitor (SF1670) blocked the suppression of Akt Ser 473 phosphorylation and the induction of cyclin B1-p-CDK1 (Thr 161) complexes and G2/M phase arrest by citrate. In conclusion, citrate induces G2/M phase arrest in PSC cells by inducing the formation of p85α-PTEN complexes to attenuate Akt-mediated signaling, thereby causing the formation of cyclin B1-p-CDK1 (Thr 161) complexes.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Citric Acid/pharmacology , Class Ia Phosphatidylinositol 3-Kinase/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , PTEN Phosphohydrolase/metabolism , Pharyngeal Neoplasms/metabolism , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cyclin B1/metabolism , Humans , Models, Biological , Multiprotein Complexes/metabolism , Pharyngeal Neoplasms/genetics , Phosphorylation , Signal Transduction
5.
Eur J Pharmacol ; 855: 137-148, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31059711

ABSTRACT

Selective pharmacologic targeting of cell cycle regulators is a potent anti-cancer therapeutic strategy. Here, we show that caspase-3-mediated p21 cleavage involves p53 independent of triptolide (TPL)-induced S phase arrest in human type 1 nasopharyngeal carcinoma (NPC) cells. Coimmunoprecipitation studies demonstrated that TPL causes S phase cell cycle arrest by suppressing the formation of cyclin A-phosphor (p)-cyclin-dependent kinas 2 (CDK2) (Thr 39) complexes. Ectopic expression of constitutively active protein kinase B1 (Akt1) blocks the induction of S phase arrest and the suppression of cyclin A expression and CDK2 Thr 39 phosphorylation by TPL. Expression of the phosphomimetic mutant CDK2 (T39E) rescues the cells from TPL-induced S phase arrest, whereas phosphorylation-deficient CDK2 (T39A) expression regulates cell growth with significant S phase arrest and enhances TPL-triggered S phase arrest. Treatment with TPL induces an increase in the formation of complexes between unphosphorylated phosphatase and tensin homolog deleted from chromosome 10 (PTEN) and p85α in the plasma membrane. Decreased microRNA (miR)-144 expression and increased PTEN expression after TPL treatment were demonstrated, and TPL-enhanced p85α-PTEN complexes and inhibitory effects on Akt (Ser 473) phosphorylation and S phase arrest were suppressed by ectopic PTEN short hairpin RNA or miR-144 expression. Knockdown of endogenous miR-144 by miR-144 Trap upregulated PTEN expression and accordingly enhanced p85α-PTEN complex formation and S phase arrest. Collectively, the effect of TPL on S phase arrest in human NPC cells is likely to enhance the p85α-PTEN interaction in the plasma membrane by suppressing miR-144 expression, resulting in the attenuation of cyclin A-p-CDK2 (Thr 39) complex formation via Akt inactivation.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Diterpenes/pharmacology , Down-Regulation/drug effects , MicroRNAs/genetics , Nasopharyngeal Neoplasms/pathology , PTEN Phosphohydrolase/metabolism , Phenanthrenes/pharmacology , S Phase Cell Cycle Checkpoints/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Epoxy Compounds/pharmacology , Humans , Phosphorylation/drug effects , Proteolysis/drug effects , Threonine/metabolism , p21-Activated Kinases/metabolism
6.
Arch Pharm Res ; 42(8): 672-683, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31020545

ABSTRACT

Caloric restriction activates sirtuin 1 (SIRT1) and induces a variety of metabolic effects that are beneficial for preventing age-related disease. The present study screened a commercially available used drug library to develop small molecule activators of SIRT1 as therapeutics for treatment of metabolic disorders. Using an in vitro fluorescence assay, the cancer therapeutic camptothecin increased SIRT1 enzymatic activity by 5.5-fold, indicating it to be a potent SIRT1 activator. Camptothecin also elevated the nicotinamide adenine dinucleotide (NAD)+/NADH ratio and increased SIRT1 protein levels in differentiated C2C12 myogenic cells. Treatment of C2C12 myotubes with camptothecin increased phosphorylation of AMP-dependent kinase (AMPK) and acetyl-coenzyme A carboxylase, caused nuclear translocation and deacetylation of forkhead box O1 (FoxO1), increased transcription and protein expression of adipose triglyceride lipase (ATGL), decreased the amount of intracellular oil droplets, and significantly increased ß-oxidation of fatty acids. These in vitro data were confirmed in vivo as camptothecin treatment of C57BL/6J mice reduced fat and plasma triglyceride levels. All of the above camptothecin-induced alterations were attenuated by the SIRT1-specific inhibitor nicotinamide and/or 6-[4-(2-piperidin-1-ylethoxy) phenyl]-3-pyridin-4-ylpyrazolo [1,5-a]pyrimidin (compound C). Thus, camptothecin activation of SIRT1 promotes lipid catabolism through AMPK/FoxO1/ATGL signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Camptothecin/pharmacology , Forkhead Box Protein O1/metabolism , Lipase/metabolism , Sirtuin 1/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Structure-Activity Relationship
7.
Free Radic Res ; 52(11-12): 1348-1358, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29764240

ABSTRACT

Iron or oxygen regulates the stability of hypoxia inducible factor-1α (HIF-1α). We investigated whether ferrous glycinate would affect HIF-1α accumulation, aerobic glycolysis and mitochondrial energy metabolism in human A549 lung cancer cells. Incubation of A549 cells with ferrous glycinate decreased the protein levels of HIF-1α, which was abrogated by proteosome inhibitor, or prolyl hydroxylase inhibitor. The addition of ferrous glycinate decreased protein levels of glucose transporter-1, hexokinase-2, and lactate dehydrogenase A, and decreased pyruvate dehydrogenase kinase-1 (PDK-1) and pyruvate dehydrogenase (PDH) phosphorylation in A549 cells. Ferrous glycinate also increased the expression of the mitochondrial transcription factor A (TFAM), and the mitochondrial protein, cytochrome c oxidase (COX-IV). Silencing of HIF-1α expression mimicked the effects of ferrous glycinate on PDK-1, PDH, TFAM and COX-IV in A549 cells. Ferrous glycinate increased mitochondrial membrane potential and ATP production in A549 cells. These results suggest that ferrous glycinate may reverse Warburg effect through down regulating HIF-1α in A549 cells.


Subject(s)
Energy Metabolism/drug effects , Ferrous Compounds/pharmacology , Glycine/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , A549 Cells , Dose-Response Relationship, Drug , Ferrous Compounds/chemistry , Glycine/analogs & derivatives , Glycine/chemistry , Humans , Membrane Potential, Mitochondrial/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Kaohsiung J Med Sci ; 30(9): 477-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25224772

ABSTRACT

Healthcare professionals have a high risk of needlestick and sharps injuries (NSIs), which have a high potential for disease transmission. Ambulatory care follow up is essential, but is usually overlooked. This study aimed to investigate the annual and cumulative (age-, sex-, and subtype-specific) incidences of ambulatory care visits after NSIs. This study was also designed to evaluate the incidences of blood-borne diseases associated with NSIs among Taiwanese health professionals in Taiwan between 2004 and 2010. Data were obtained from the National Health Insurance Research Database, which contains anonymized records representing approximately 99% of the Taiwan population. A total of 4443 nurse healthcare workers (NHCWs) and 3138 non-nurse healthcare workers (NNHCWs), including physicians, medical technologists, and other health professionals were included in this longitudinal study. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated. The Mantel-Haenszel method was used to adjust for sex, age, and type of affiliation. Results showed that the annual incidence of ambulatory care visits of NHCWs increased from 0.7% in 2004 to 1.9% in 2010; this incidence was significantly higher than that of NNHCWs (from 0.3% in 2004 to 0.5% in 2010) in any yearly comparison (p < 0.05). The sex-adjusted 7-year cumulative incidence rate was 3.23 (95% CI = 1.23-8.45) in males and 3.92 (95% CI = 2.70-5.69) in females (p < 0.05). The age-adjusted 7-year cumulative incidence rate was 2.74 (95% CI = 1.99-3.77) and 2.14 (95% CI = 1.49-3.07) in subjects ≤ 30 and ≥31 years old, respectively (p < 0.0005). The affiliation-adjusted 7-year cumulative incidence rate was 1.89 (95% CI = 1.21-2.94) in medical centers and 3.33 (95% CI = 2.51-4.41) in nonmedical centers (p < 0.01). In conclusion, NSIs increased steadily from 2004 to 2010 in Taiwan with NHCWs having higher NSIs incidences than NNHCWs. A routine ambulatory care visit after NSIs can prevent blood-borne transmission, especially for NHCWs. Educational programs may be helpful for reducing the incidence of NSIs and increasing ambulatory care visit ratios after NSIs.


Subject(s)
Ambulatory Care/statistics & numerical data , Health Personnel/statistics & numerical data , Health Surveys/statistics & numerical data , Needlestick Injuries/epidemiology , Adult , Age Factors , Communicable Diseases/etiology , Demography , Female , Humans , Incidence , Male , Needlestick Injuries/complications , Nurses/statistics & numerical data , Risk Factors , Taiwan/epidemiology
9.
Acta Pharmacol Sin ; 34(11): 1411-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24077634

ABSTRACT

AIM: To investigate the effects of punicalagin, a polyphenol isolated from Punica granatum, on human U87MG glioma cells in vitro. METHODS: The viability of human U87MG glioma cells was evaluated using MTT assay. Cell cycle was detected with flow cytometry analysis. The levels of Bcl-2, cleaved caspase-9, cleaved poly(ADP-ribose) polymerase (PARP), phosphor-AMPK and phosphor-p27 at Thr198 were measured using immunoblot analyses. Caspase-3 activity was determined with spectrophotometer. To determine autophagy, LC3 cleavage and punctate patterns were examined. RESULTS: Punicalagin (1-30 µg/mL) dose-dependently inhibited the cell viability in association with increased cyclin E level and decreased cyclin B and cyclin A levels. The treatment also induced apoptosis as shown by the cleavage of PARP, activation of caspase-9, and increase of caspase-3 activity in the cells. However, pretreatment of the cells with the pan-caspase inhibitor z-DEVD-fmk (50 µmol/L) did not completely prevent the cell death. On the other hand, punicalagin treatment increased LC3-II cleavage and caused GFP-LC3-II-stained punctate pattern in the cells. Suppressing autophagy of cells with chloroquine (1-10 µmol/L) dose-dependently alleviated the cell death caused by punicalagin. Punicalagin (1-30 µg/mL) also increased the levels phosphor-AMPK and phosphor-p27 at Thr198 in the cells, which were correlated with the induction of autophagic cell death. CONCLUSION: Punicalagin induces human U87MG glioma cell death through both apoptotic and autophagic pathways.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Glioma/drug therapy , Hydrolyzable Tannins/pharmacology , AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Glioma/pathology , Humans , Hydrolyzable Tannins/administration & dosage , Hydrolyzable Tannins/isolation & purification , Lythraceae/chemistry , Phosphorylation
10.
Neuro Oncol ; 15(10): 1330-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23911596

ABSTRACT

BACKGROUND: A shift in glucose metabolism from oxidative phosphorylation to anaerobic glycolysis is the biochemical hallmark of malignant cancer cells. METHODS: In the present study, we demonstrated that Nodal stimulated the expression of glycolytic enzymes and decreased reliance on mitochondrial oxidative phosphorylation in human glioma cancer cells. The shift in glucose metabolism was mediated by induction of the hypoxia-inducible factor (HIF). RESULTS: Nodal protein expression was shown to be correlated with expression levels of glucose transporter (Glut)-1, hexokinase (HK)-II, pyruvate dehydrogenase kinase (PDK)-1, the phosphorylation level of pyruvate dehydrogenase (PDH), glucose uptake, and lactate accumulation in human glioma cells. These effects were inversely correlated with mitochondrial oxygen consumption and ATP production. Knockdown of Nodal expression with specific small hairpin RNA reduced Glut-1, HK-II, and PDK-1 expressions and PDH phosphorylation. Nodal knockdown also reduced glucose uptake and lactate generation, which in turn increased mitochondrial membrane potential (Ψ), O2 utilization, and ATP synthesis. The ectopic expression of Nodal in low-expressing Nodal glioma cells resulted in the opposite results compared with those of Nodal knockdown glioma cells. Treatment of cells with recombinant Nodal increased HIF-1 expression, and this effect was regulated at the transcriptional level. Blockage of the Nodal receptor by a pharmacological inhibitor or Nodal knockdown in U87MG cells decreased HIF-1α expression. Furthermore, HIF-1α knockdown in U87MG cells decreased Glut-1, HK-II, and PDK-1 expressions and PDH phosphorylation, which were similar to results in Nodal knockdown cells. CONCLUSION: Taken together, these results suggest that Nodal affects energy metabolism through HIF-1α.


Subject(s)
Energy Metabolism , Glioma/metabolism , Glioma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , Nodal Protein/metabolism , Adenosine Triphosphate/metabolism , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glucose/metabolism , Glycolysis , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lactic Acid/metabolism , Luciferases/metabolism , Membrane Potential, Mitochondrial , Nodal Protein/antagonists & inhibitors , Nodal Protein/genetics , Oxygen Consumption , RNA, Small Interfering/genetics , Tumor Cells, Cultured
11.
Arch Pharm Res ; 36(12): 1541-51, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23657807

ABSTRACT

Monacolin K is the secondary metabolite isolated from Monascus spp. It is the natural form of lovastatin, which is clinically used to reduce the synthesis of cholesterol by inhibiting 3-hydroxy-3-methylglutaryl coenzyme A reductase. In the present study, monacolin K increased protein expression of SIRT1 and phosphorylation level of AMP-activated protein kinase (AMPK) in HepG2 cells. Through activation of SIRT1/AMPK pathway, monacolin K increased phosphorylation of acetyl CoA carboxylase and caused nuclear translocation of forkhead box O1. The western blotting results showed that monacolin K increased expression of adipose triglyceride lipase but decreased abundances of fatty acid synthase (FAS) and sterol regulatory element-binding protein 1 (SREBP1). Monacolin K also decreased the intracellular accumulation of lipids as demonstrated by Oil Red O staining. In addition, the immunostaining showed that monacolin K prevented the nuclear translocation of SREBP1, indicating the association with down-regulation of FAS. All the demonstrated effects of monacolin K were counteracted by nicotinamide or compound C, the inhibitors of SIRT1 or AMPK. In summary, monacolin K reduces the lipid content through SIRT1/AMPK pathway in HepG2 cells, which promotes catabolism and inhibits anabolism of lipid.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anticholesteremic Agents/pharmacology , Lipid Metabolism/physiology , Lovastatin/pharmacology , Signal Transduction/physiology , Sirtuin 1/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Signal Transduction/drug effects
12.
BMC Musculoskelet Disord ; 14: 144, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23617330

ABSTRACT

BACKGROUND: Musculoskeletal disorders (MSDs) represent the leading causes of occupational injuries among nursing staff. This population-based study was designed to assess the incidence and age-specific incidence of MSDs among a Taiwanese nurse cohort compared with non-nurses. DESCRIPTION: Data from the Taiwan National Health Insurance Research Database were used to identify MSDs in the study population. A total of 3914 nurses with a diagnosis of MSD were included, together with 11,744 non-nurses as a comparison group. The comparison subjects were randomly selected at a ratio of 3:1 relative to the nurse population and were matched by gender and age. The incidence of MSDs was calculated for the study group, with nurse-to-reference risk ratios presented as odds ratios with 95% confidence intervals (CIs). During the period 2004-2010, 3004 MSDs occurred among the nurses (76.24%) and 7779 (65.79%) in the non-nurses. The annual incidence of MSDs for the nurses increased from 28.35% in 2006 to 33.65% in 2010. The nurse-to-reference risk ratio was 1.27 (95% CI 1.19-1.35) in 2004 and 1.46 (1.37-1.55) in 2010. Herniation of intervertebral disc, lumbago, rotator cuff syndrome, medial epicondylitis, trigger finger and carpal tunnel syndrome were the most common problems. CONCLUSIONS: Nurses are at higher risk of MSDs and the trend is increasing. Incorrect work-related posture/movement, psychological issues and the rolling shift system may be the major causes of MSDs among nurses in Taiwan.


Subject(s)
Musculoskeletal Diseases/epidemiology , Nurses , Occupational Injuries/epidemiology , Population Surveillance , Adult , Cohort Studies , Female , Humans , Male , Musculoskeletal Diseases/diagnosis , Occupational Injuries/diagnosis , Population Surveillance/methods , Risk Factors , Taiwan/epidemiology , Young Adult
13.
Biochem Pharmacol ; 84(4): 522-31, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22687626

ABSTRACT

Fenofibrate, a fibric acid derivative, is known to possess lipid-lowering effects. Although fenofibrate may activate peroxisome proliferator-activated receptor (PPAR)α and regulate the transcription of several genes, the underlying mechanisms are poorly understood. In this study, we demonstrated that incubation of C2C12 myotubes with fenofibrate increased adipose triglyceride lipase (ATGL) expression and suppressed fatty acid synthase (FAS) level, thereby decreasing intracellular triglyceride accumulation when cells were incubated at high-glucose condition. Fenofibrate increased the phosphorylation of AMP-activated protein kinase (AMPK), which subsequently increased fatty acid ß-oxidation. AMPK phosphorylation was reduced by pretreatment with GW9662 (a PPARα inhibitor), suggesting that AMPK may be a downstream effector of PPARα. Pretreatment with compound C (an AMPK inhibitor) or GW9662 blocked fenofibrate-induced ATGL expression and the lipid-lowering effect. Our results suggest that AMPK is as an upstream regulator of ATGL. With further exploration, we demonstrated that fenofibrate stimulated FoxO1 translocation from the cytosol to nuclei by immunefluorescence assay, chromatin immuneprecipitation assay, and reporter assay. Furthermore, oral administration of fenofibrate ameliorated the body weight, visceral fat and serum biochemical indexes in db/db mice. Taken together, our results suggest that the lipid-lowering effect of fenofibrate was achieved by activating PPARα and AMPK signaling pathway that resulted in increasing ATGL expression, lipolysis, and fatty acid ß-oxidation.


Subject(s)
AMP-Activated Protein Kinases/physiology , Fenofibrate/pharmacology , Forkhead Transcription Factors/physiology , Hypolipidemic Agents/pharmacology , Lipase/physiology , Lipid Metabolism , Muscle Fibers, Skeletal/metabolism , PPAR alpha/physiology , Acetyl-CoA Carboxylase/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Lipase/genetics , Male , Mice , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Transport , Signal Transduction
14.
Mol Carcinog ; 49(9): 783-97, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20607722

ABSTRACT

Aloe-emodin (AE), extracted from the rhizome of Rheum palmatum, has an anti-proliferative effect on different human cancer cell lines. Nonetheless, the underlying mechanism by which AE inhibits nasopharyngeal carcinoma (NPC) cell invasion is still unclear. The results of this study show that treatment of NPC cells with growth suppressive concentrations of AE caused cell cycle arrest at the S-G(2)/M phase. Coimmunoprecipitation and small interfering RNA (siRNA) studies demonstrated that AE-induced cell cycle arrest in NPC cells was associated with increasing levels of cyclin B1 bound to cyclin-dependent kinase 1. The inhibition of NPC cell invasion by AE was evidenced through the suppression of matrix metalloproteinases-2 (MMP-2) expression. MMP-2 promoter activity and cell invasion were inhibited by p38 mitogen-activated protein kinase (MAPK) siRNA, inhibitor 4-(4-Fluorophenyl)-2-[4-(methylsulfinyl)phenyl]-5-(4-pyridyl)-1H-imidazole (SB203580), and AE, but not by JNK siRNA and inhibitor 1,9-pyrazoloanthrone. Treatment with AE, SB203580, NF-kappaB inhibitors N-p-tosyl-(L)-phenylalanine chloromethyl ketone (TPCK) and pyrrolidine dithiocarbamate (PDTC) or transfection with p38 MAPK siRNA significantly inhibited NF-kappaB transcriptional activity. In addition, TPCK and PDTC treatment inhibited the expression and promoter activity of MMP-2 and thereby significantly inhibited cell invasion activity. The involvement of p38 MAPK activity in NF-kappaB-mediated MMP-2 function was further confirmed through the attenuation of p38 MAPK by SB203580 and NF-kappaB ectopic expression. Collectively, our results indicate that AE inhibits invasion of NPC cells by suppressing the expression of MMP-2 via the p38 MAPK-NF-kappaB signaling pathway.


Subject(s)
NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Anthraquinones , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/pharmacology , Cell Cycle/drug effects , Cell Cycle/genetics , Cyclin B1/metabolism , Down-Regulation/drug effects , Humans , Imidazoles/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/pharmacology , NF-kappa B/genetics , NF-kappa B/pharmacology , Nasopharyngeal Neoplasms/genetics , Pyridines , Signal Transduction/drug effects , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
15.
Cancer Lett ; 291(1): 46-58, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19942342

ABSTRACT

Aloe-emodin (AE), a natural, biologically active compound from the rhizome of Rheum palmatum, has been shown to induce apoptosis in several cancer cell lines in vitro. However, its molecular mechanism of action in the apoptosis induction of human nasopharyngeal carcinoma (NPC) cells has not been explored. This study shows that AE induced G(2)/M phase arrest by increasing levels of cyclin B1 bound to Cdc2, and also caused an increase in apoptosis of NPC cells, which was characterized by morphological changes, nuclear condensation, DNA fragmentation, caspase-3 activation, cleavage of poly (ADP-ribose) polymerase (PARP) and increased sub-G(1) population. Treatment of NPC cells with AE also resulted in a decrease in Bcl-X(L) and an increase in Bax expression. Ectopic expression of Bcl-X(L) but not Bcl-2 or small interfering RNA (siRNA)-mediated attenuation of Bax suppressed AE-induced apoptotic cell death. AE-induced loss of mitochondrial membrane potential (MMP) and increase in cellular Ca(++) content, reactive oxygen species (ROS) and apoptotic cell death were suppressed by the treatment of cyclosporin A (CsA) or caspase-8 inhibitor Z-IETD-FMK. Co-treatment with caspase-9 inhibitor Z-LEHD-FMK could inhibit AE-induced cell death and the activation of caspase-3 and -9. In addition, suppression of caspase-8 with the specific inhibitor Z-IETD-FMK inhibited AE-induced the activation of Bax, the cleavage of Bid, the translocation of tBid to the mitochondria and the release of cytochrome c, apoptosis-inducing factor (AIF) and Endo G from the mitochondria and subsequent apoptosis. Taken together, these results indicate that the caspase-8-mediated activation of the mitochondrial death pathway plays a critical role in AE-induced apoptosis of NPC cells.


Subject(s)
Anthraquinones/pharmacology , Apoptosis/drug effects , Caspase 8/physiology , Mitochondria/drug effects , Nasopharyngeal Neoplasms/drug therapy , CDC2 Protein Kinase , Caspase 3/physiology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin B/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinases , Humans , Mitochondria/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/physiology , bcl-X Protein/physiology
16.
Cell Biol Int ; 32(1): 151-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17900936

ABSTRACT

The human TAO kinase 1 (hTAOK1) is a member of the Ste20 group of kinases with the kinase domain located at the N-terminus. The rat homologue, originally named TAO1, has been demonstrated to be highly expressed in brain. In this study, the human TAO kinase 1 was transfected into human neuroblastoma SH-SY5Y cells and its biological effects on the cell morphology were observed by co-expressing the enhanced green fluorescent protein (EGFP). It was found that after 16 h of transfection the cells had shrunk, and finally became rounded when transfected with wild-type or mutant K57A genes encoding either the kinase domain (residues 1-376) or the full-length molecule (residues 1-1001). Thirty-four hours after transfection, cells floated and apoptotic bodies were observed after nuclear staining with DAPI. On the other hand, the cells that were transfected with the gene encoding the C-terminal regulatory region (residues 377-1001) of hTAOK1, appeared to remain unchanged. In order to know the signaling events involved in the above biological phenomena, caspase-3-like activities of the transfected cells were measured in the absence or presence of JNK inhibitor SP600125, in which caspase-3 and JNK (C-jun-N-terminal kinase) are both known to be critical components of the neuronal apoptosis. The results showed that the apoptotic cells exhibited elevated caspase-3-like activity, which could be reduced by SP600125 to some extent. It is concluded that human TAO kinase 1 induces apoptosis in SH-SY5Y cells and the kinase domain is essential, but its catalytic activity seems to be dispensable in this case.


Subject(s)
Apoptosis/physiology , MAP Kinase Kinase Kinases/metabolism , Neuroblastoma/metabolism , Anthracenes/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Neuroblastoma/pathology , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL